Research Models
Find A Model By Name
Search Research Models
Search Results
318 Models
263 Visualizations
AD-related Research Models
Phenotypes Examined
- Plaques
- Tangles
- Neuronal Loss
- Gliosis
- Synaptic Loss
- Changes in LTP/LTD
- Cognitive Impairment
When visualized, these phenotypes will distributed over a 18 month timeline demarcated at the following intervals: 3mo, 6mo, 9mo, 1yr, 15mo, 18mo+.
3xTg
Observed
-
Plaques at 26
Extracellular Aβ deposits by 6 months in the frontal cortex, predominantly layers 4 and 5 and progress with age (Oddo et al., 2003).
-
Tangles at 52
By 12 months extensive tau immunoreactivity in CA1 neurons of the hippocampus, particularly pyramidal neurons, later in the cortex. No tau pathology at 6 months (Oddo et al., 2003).
-
Gliosis at 30
Increased density of GFAP immunoreactive astrocytes and IBA-1 immunoreactive microglia compared with wild-type mice at 7 months (Caruso et al., 2013). Development of gliosis may occur earlier.
-
Changes in LTP/LTD at 26
By 6 months decreased LTP compared with wild type controls. Impairment in basal synaptic transmission. No change at 1 month of age (Oddo et al., 2003).
-
Cognitive Impairment at 17
Cognitive impairment manifests at 4 months as a deficit in long-term retention and correlates with the accumulation of intraneuronal Aβ in the hippocampus and amygdala, but plaques and tangles are not yet apparent (Billings et al., 2005).
Absent
No Data
-
Neuronal Loss at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Psen1, APP, MAPT | APP K670_M671delinsNL (Swedish), MAPT P301L, PSEN1 M146V | Psen1: Knock-In; APP: Transgenic; MAPT: Transgenic | Alzheimer's Disease | Age-related, progressive neuropathology including plaques and tangles. Extracellular Aβ deposits by 6 months in frontal cortex, more extensive by 12 months. No tau pathology at 6 months, but evident at 12 months. Synaptic dysfunction, including LTP deficits, prior to plaques and tangles. |
Cognitive impairment by 4 months. Impairments first manifest as a retention/retrieval deficit and not as a learning deficit, and occur prior to plaques and tangles. Deficits in both spatial and contextual based paradigms. Clearance of intraneuronal Aβ by immunotherapy rescues the early cognitive deficits in a hippocampal-dependent task. |
5xFAD (B6SJL)
Observed
-
Plaques at 8
Extracellular amyloid deposition begins around 2 months, first in the subiculum and layer V of the cortex. Aβ42 also accumulates intraneuronally in an aggregated form within the soma and neurites starting at 1.5 months.
-
Neuronal Loss at 24
Neuron loss in cortical layer V and subiculum.
-
Gliosis at 8
Gliosis begins at 2 months.
-
Synaptic Loss at 16
Levels of the presynaptic marker synaptophysin begin to decline by 4 months; levels of syntaxin, another presynaptic marker, and PSD-95, a postsynaptic marker, decline by 9 months
-
Changes in LTP/LTD at 24
Basal synaptic transmission and LTP in hippocampal area CA1 begin to deteriorate between 4 and 6 months
-
Cognitive Impairment at 18
Impaired spatial working memory in the Y-maze test and impaired remote memory stabilization in a contextual-fear-conditioning test by 4 to 5 months of age.
Absent
-
Tangles at
Absent.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid pathology starting at 2 months, including amyloid plaques. Accumulation of intraneuronal Aβ before amyloid deposition. Gliosis and synapse degeneration. Neuron loss in cortical layer 5 and subiculum. No neurofibrillary tangles. |
Age-dependent memory deficits including spatial memory, stress-related memory, and memory stablization. Motor phenotype. |
5xFAD (C57BL6)
Observed
-
Plaques at 8
Amyloid plaques observed in hippocampus, cortex, thalamus, and spinal cord.
-
Neuronal Loss at 52
Approximate 40 percent loss of layer V pyramidal neurons at one year.
-
Gliosis at 8
Microgliosis and astrogliosis are associated with amyloid plaques; microgliosis is associated with vascular damage.
-
Synaptic Loss at 24
Spine density was reduced in pyramidal neurons in somatosensory and prefrontal cortices, but not in the hippocampi, of 5xFAD mice crossed with mice expressing yellow fluorescent protein (YFP mice), compared with mice expressing YFP alone.
-
Changes in LTP/LTD at 8
While spike-timing-dependent long-term potentiation was induced in layer V neurons from wild-type mice, the same stimulation protocol induced long-term depression in neurons from 5xFAD mice.
-
Cognitive Impairment at 24
Impairments of spatial working memory and reduced anxiety emerge between 3 and 6 months and worsen with age.
Absent
No Data
-
Tangles at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid pathology starting at 2 months, including amyloid plaques. Accumulation of intraneuronal Aβ before amyloid deposition. Gliosis and synapse degeneration. Neuron loss in cortical layer V. |
Age-dependent memory deficits, motor phenotype, and reduced anxiety. |
A7 APP transgenic
Observed
-
Plaques at 39
These mice develop progressive amyloid deposition in the cerebral cortex by 9-12 months. By 21 months of age amyloid pathology is extensive.
Absent
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP T714I (Austrian) | APP: Transgenic | Alzheimer's Disease | Progressive amyloid deposition in the cerebral cortex by approximately 9-12 months. |
Unknown. |
AAV-AD
Observed
-
Plaques at 128
Amyloid plaques and cerebral amyloid angiopathy observed 30 months post-injection.
-
Changes in LTP/LTD at 40
Deficits in LTP as Schaffer collateral-CA1 synapse at 10 months (8 months post-injection). LTD similar to controls.
-
Cognitive Impairment at 40
AAV-AD spent less time in the target quadrant of the Morris water maze in probe tests administered 3 and 5 days after training.
Absent
-
Gliosis at
No astrogliosis observed up to 30 months post-injection.
No Data
-
Tangles at
Immunostaining with monoclonal antibodies AT8 and AT100 suggests the presence of (pre)tangle-like structures 30 months post-injection.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP V717I (London), PSEN1 M146L (A>C) | APP: Virus; PSEN1: Virus | Alzheimer's Disease | Amyloid plaques and cerebral amyloid angiopathy observed 30 months post-injection. Anti-phospho-tau immunostaining suggests the presence of (pre)tangle-like structures. No astrogliosis seen up to 30 months post-injection. |
Compared with control rats at 8 months post-injection, AAV-AD spent less time in the target quadrant of the Morris water maze in probe tests administered 3 and 5 days after training and less time in the center of the open field. |
AAV-sTREM2 5xFAD
Observed
-
Plaques at 28
When examined at 7 months of age, amyloid plaque burdens in the hippocampus and cortex of AAV-sTREM2 5xFAD mice were about half those of 5xFAD mice injected with control vector.
-
Gliosis at 28
The number of plaque-associated microglia was increased in AAV-sTREM2 5xFAD mice examined at 7 months of age.
-
Changes in LTP/LTD at 24
Long-term potentiation at Shaeffer collateral-CA1 synapses is impaired in 5xFAD mice. AAV-mediated over expression of sTREM2 rescued LTP in AAV-sTREM2 5xFAD mice.
-
Cognitive Impairment at 24
5xFAD mice show impaired learning and memory in the Morris water maze, but AAV-sTREM2 5xFAD mice performed as well as non-transgenic controls.
Absent
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
TREM2, APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | TREM2: Virus; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | AAV-mediated over expression of sTREM2 decreased amyloid plaque burdens and increased numbers of plaque-associated microglia in the brains of 5xFAD mice. |
5xFAD mice show impaired learning and memory in the Morris water maze, but AAV-sTREM2 5xFAD mice performed as well as non-transgenic controls. |
AAV-sTREM2 PS19
Observed
-
Synaptic Loss at 28
AAV-mediated expression of sTREM2 protected against hippocampal synapse loss in PS19 mice.
-
Changes in LTP/LTD at 29
LTP at Shaeffer collateral-CA1 synapses was slightly enhanced in hippocampal slices from 7-month-old AAV-sTREM2 PS19 mice, compared with PS19 mice who had received control vector.
-
Cognitive Impairment at 30
AAV-mediated expression of sTREM2 improved performance of PS19 mice in the Morris water maze and Y-maze.
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
TREM2, MAPT | MAPT P301S | TREM2: Virus; MAPT: Transgenic | Frontotemporal Dementia, Alzheimer's Disease | AAV-mediated expression of sTREM2 protected against hippocampal synapse loss in PS19 mice. |
AAV-mediated expression of sTREM2 improved performance of PS19 mice in the Morris water maze and Y-maze. |
Abca7*A1527G/APOE4/Trem2*R47H
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Abca7, APOE, Trem2 | TREM2 R47H | Abca7: Knock-In; APOE: Knock-In; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
Abca7 KO/APOE4/Trem2*R47H
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Abca7, APOE, Trem2 | TREM2 R47H | Abca7: Knock-Out; APOE: Knock-In; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
ADanPP
Observed
-
Plaques at 9
Vascular amyloid deposits and punctate parenchymal aggregates first occur in the hippocampus and increase with age, spreading throughout the brain, including the cortex, amygdala, thalamus, and brainstem in hemizygous mice.
-
Gliosis at 17
Astrogliosis and microgliosis increase with age and increasing ADan-amyloid deposition.
-
Cognitive Impairment at 78
The only ages tested were 6 months and 18-20 months. Mice 18-20 months of age exhibited both motor and spatial learning defects in the Morris water maze, and increased anxiety in the open field test. No impairments were observed in 6 month-old mice.
Absent
-
Tangles at
Absent.
-
Neuronal Loss at
Absent.
No Data
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
ITM2B (BRI2) | BRI2: Familial Danish Dementia (FDD) duplication | ITM2B (BRI2): Transgenic | Familial Danish Dementia, Alzheimer's Disease, Cerebral Amyloid Angiopathy | ADan deposition starts in the hippocampus and meningeal vessels at 2 months and increases with age. By 18 months, deposition is widespread. The majority of amyloid deposits are associated with the vasculature, where they destroy the integrity of the vessel wall and lead to microhemorrhages. Parenchymal amyloid plaques surrounded by microglia and dystrophic neurites are also present. |
Impaired performance in Morris water maze, due to a combination of both motor deficits (i.e. reduced swim speed) and spatial learning deficits reported at 18-20 months. Open field test at 18-20 months also showed an anxiety-related phenotype. |
AD-BXD
Observed
-
Plaques at 24
Transgenic AD-BXD mice develop amyloid plaques by 6 months of age, the earliest age examined. The extent of plaque deposition is strain-dependent.
-
Gliosis at 25
Strain-dependent gliosis by 6 months.
-
Cognitive Impairment at 60
In the AD-BXD population as a whole, transgenic mice performed similarly to non-transgenic littermates in a contextual fear-conditioning test at 6 months, but were impaired at 14 months. The age of onset and severity of impairment are strain-dependent.
Absent
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Transgenic AD-BXD mice develop amyloid plaques by 6 months of age, although the extent of plaque deposition is strain-dependent. |
Transgenic AD-BXD mice exhibit cognitive deficits, assessed using contextual fear conditioning. The age of onset and severity of impairment are strain-dependent. |
APOE2 Knock-In, floxed (CureAlz)
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE | APOE: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
APOE2 Knock-In (JAX)
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE | APOE: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
APOE3 Knock-In, floxed (CureAlz)
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE | APOE: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
APOE3 Knock-In (JAX)
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE | APOE: Knock-In | Alzheimer's Disease | No data. |
No data. |
APOE3 Knock-In (Lamb)
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE | APOE: Knock-In | Alzheimer's Disease, Traumatic Brain Injury | Unknown. |
Unknown. |
APOE4 Knock-In, floxed (CureAlz)
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE | APOE: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
APOE4 Knock-In (JAX)
Observed
Absent
-
Cognitive Impairment at
At 2 and 12 months of age, APOE4 KI mice perform similarly to wild-type mice in tests of locomotor activity, motor coordination, and working memory.
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE | APOE: Knock-In | Alzheimer's Disease | No data. |
No data. |
APP21
Observed
-
Neuronal Loss at 76
Necrotic neurons in hippocampus and cortex of female rats.
-
Gliosis at 64
Activated (MHCII-positive) microglia present in white matter tracts at 15 months.
-
Cognitive Impairment at 12
Male rats show deficits in Morris water maze as early as 3 months of age. Females show deficits in Barnes maze at 14 months of age.
Absent
-
Plaques at
Do not spontaneously develop amyloid pathology, but can serve as hosts for exogenously seeded amyloid deposits.
No Data
-
Tangles at
“Flame-shaped” profiles in hippocampal neurons of 18- to 19-month-old female rats revealed by hematoxylin-and-eosin-staining.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP V717F (Indiana) | APP: Transgenic | Alzheimer's Disease | No plaques to 30 months of age. Necrotic neurons in hippocampus and cortex by 19 months, in females; neuron loss not observed in cortices of 19-month males. |
Male rats show deficits in Morris water maze as early as 3 months of age. Females show deficits in Barnes maze at 14 months of age. |
APP23
Observed
-
Plaques at 26
Congophillic, dense-core amyloid plaques first appear at 6 months, and increase in size and number with age. Amyloid plaques can occupy more than 25% of the neocortex and hippocampus in 24 month-old mice (Sturchler-Pierrat et al., 1997; Calhoun et al., 1998).
-
Neuronal Loss at 61
Neuronal loss (14-28%) has been reported in the CA1 region of the hippocampus in 14-18 month old mice (Calhoun et al., 1998).
-
Gliosis at 26
Activated microglia in close proximity to dense amyloid plaques (Stalder et al., 1999). Upregulation of neuroinflammatory markers and activation of astrocytes and macrophages. Age-associated increase in components of the complement system, namely C1q and C3, at later ages (9 and 18 months, respectively) (Reichwald et al., 2009).
-
Cognitive Impairment at 13
Spatial memory defects in Morris Water maze at 3 months and progresses with age (Van dam et al., 2003; Kelly et al., 2003).
Absent
-
Tangles at
Dystrophic neurites containing hyperphopshorylated tau surounds Aβ plaques, but no neurofibrillary tangles are observed (Sturchler-Pierrat et al., 1997).
-
Synaptic Loss at
Neocortical synapses were examined in mice as old as 24 months of age; no evidence of alterations in the number of synapses or levels of synaptophysin were observed (Boncristiano et al., 2005).
-
Changes in LTP/LTD at
LTP in the hippocampus and prefrontal cortex is normal at all ages studied: 3, 6, 9, 12, 18 and 24 months (Roder at al., 2003).
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish) | APP: Transgenic | Alzheimer's Disease, Cerebral Amyloid Angiopathy | Aβ deposits first observed at 6 months. Congophilic plaques increase in size and number with age and are surrounded by activated microglia, astrocytes, and dystrophic neurites containing hyperphosphorylated tau (although no neurofibrillary tangles). Neuronal loss in the CA1 region of the hippocampus. Mice also develop CAA, and microhemorrages occur at later ages. |
Spatial memory defects in Morris Water maze at 3 months and progresses with age. Memory deficits in passive avoidance were observed in 25 month-old mice, but not at younger ages. |
APP23 x PS1-R278I
Observed
-
Plaques at 26
By 6 months of age amyloid plaques accumulate in the cortex and hippocampus. A high percentage of plaques are thioflavin-S –positive cored plaques.
-
Gliosis at 39
Astrocytosis in the vicinity of plaques in the hippocampus and cortex by 9 months.
-
Cognitive Impairment at 13
Short-term memory deficits are apparent by 3 to 4 months as measured by the Y maze.
Absent
-
Tangles at
Not observed.
No Data
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | PSEN1 R278I | APP: Transgenic; PSEN1: Knock-In | Alzheimer's Disease | Amyloid deposition by 6 months of age in the cortex and hippocampus. Abundant reactive astrocytes in the vicinity of plaques. Elevated Aβ43 in the brain by 3 months. High density of cored plaques. Pyroglutamate Aβ (N3pE-Aβ) associated with amyloid plaques. |
Short-term memory deficits apparent by 3-4 months as measured by the Y maze. |
APP751SL/PS1 KI
Observed
-
Plaques at 11
Aβ deposition at 2.5 months compared to 6 months in APPSL mice. At 6 months, numerous compact Aβ deposits in the cortex, hippocampus, and thalamus, whereas in age-matched APPSL mice only very few deposits restricted mainly to the subiculum and deeper cortical layers. At 10 months, deposits increased in distribution, density, and size in both models (Casas et al., 2004).
-
Neuronal Loss at 23
Some cell loss detectable as early as 6 months in female mice. At 10 months extensive neuronal loss (>50%) is present in the CA1/2 hippocampal pyramidal cell layer. SNeuronal loss also occurs in the frontal cortex and cholinergic system (Casas et al., 2004; Christensen et al., 2008; Christensen et al., 2010).
-
Gliosis at 11
Astrogliosis occurs in parallel with Aβ deposition, starting around 2.5 months, and in proximity to Aβ-positive neurons (Wirths et al., 2010).
-
Synaptic Loss at 24
At 6 months, levels of pre- and post-synaptic markers are reduced (Breyhan et al., 2009).
-
Changes in LTP/LTD at 28
At 6 months there is a large reduction of long-term potentiation and disrupted paired pulse facilitation. No deficit at 4 months (Breyhan et al., 2009).
-
Cognitive Impairment at 27
Age-dependent impairments in working memory as measured by the Y maze and T-maze continuous alternation task. No deficit at 2 months, but deficits at 6 and 12 months compared to PS1KI littermates (Wirths et al., 2008).
Absent
-
Tangles at
Absent.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP V717I (London), PSEN1 M233T, PSEN1 L235P | APP: Transgenic; PSEN1: Knock-In | Alzheimer's Disease | Acceleration of extracellular Aβ deposition compared to the single transgenics. Age-dependent neuronal loss in the hippocampus with extensive neuronal loss in the CA1/2 at 10 months with detection as early as 6 months in female mice. Intraneuronal Aβ and thioflavin-S-positive deposits before neuronal loss. Astrogliosis in proximity of Aβ-positive neurons. |
Age-dependent impairments in working memory as measured by the Y maze and T-maze continuous alternation task. No deficit at 2 months, but deficits at 6 and 12 months compared to PS1KI littermates. |
APPDutch
Observed
-
Gliosis at 126
Microgliosis develops after the onset of CAA pathology and is prominent in areas adjacent to amyloid-laden vessels. There is also widespread activation of astrocytes in neocortical regions affected by CAA. These changes have been reported at 29 months of age, although the actual onset of gliosis may occur earlier than has been examined.
Absent
-
Plaques at
No plaques are observed, but CAA develops at 22-24 months.
-
Tangles at
Absent.
No Data
-
Neuronal Loss at
Unknown.
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
-
Cognitive Impairment at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP E693Q (Dutch) | APP: Transgenic | Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch type, Cerebral Amyloid Angiopathy, Alzheimer's Disease | Increased Aβ40/42 ratio. Extensive vascular Aβ deposition starting at 22-24 months appearing first in leptomeningeal vessels followed by cortical vessels, leading to smooth muscle cell degeneration, hemorrhages, and neuroinflammation. Parenchymal amyloid plaques are not observed. |
Unknown. |
APP E693Δ-Tg (Osaka)
Observed
-
Neuronal Loss at 104
Neuronal loss, as measured by NeuN staining, was observed in the CA3 region of the hippocampus at 24 months of age. Neuronal loss was not detected in the cerebral cortex at this time.
-
Gliosis at 52
At 12 months of age, microgliosis is seen in transgenic mice, as measured by the presence of Iba-1 staining in the hippocampus and cortex. Astrocytosis, as measured by GFAP-reactivity, increased starting around 18 months of age in these regions.
-
Synaptic Loss at 34
Starting around eight months of age, transgenic mice exhibit a decrease in synaptic density in the CA3 region of the hippocampus as measured by synaptophysin staining.
-
Changes in LTP/LTD at 35
By eight months of age, transgenic mice exhibit reduced short term plasticity as measured by paired-pulse facilitation in addition to reduced LTP as elicited by high frequency stimulation to the perforant pathway.
-
Cognitive Impairment at 36
By 8 months of age, transgenic mice exhibit memory impairment in the Morris water maze compared to mice expressing equivalent levels of wild-type human APP.
Absent
-
Plaques at
Extracelluar amyloid plaques are not observed out to 24 months; however, Aβ accumulates within neurons of the hippocampus and cerebral cortex starting around eight months of age.
-
Tangles at
Overt tangle pathology is not observed out to 24 months of age, but abnormal tau phosphorylation is observed starting around eight months of age.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP E693del (Osaka) | APP: Transgenic | Alzheimer's Disease | Age-dependent accumulation of Aβ oligomers within hippocampal and cortical neurons, but negligible deposits of extracellular amyloid. Abnormal tau phosphorylation, but no overt tangle pathology. Synaptic loss and gliosis in hippocampus and cerebral cortex. Late neuronal loss in the CA3 region of the hippocampus. |
Memory impairment by eight months as measured by the Morris water maze. Specifically, reduced spatial reference memory in the Morris water maze compared to mice expressing comparable levels of wild-type human APP. |
App knock-in (humanized Aβ)
Observed
Absent
-
Plaques at
None observed at 3 months.
-
Tangles at
None observed at 3 months.
-
Neuronal Loss at
None observed at 3 months.
No Data
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
App | App: Knock-In | Alzheimer's Disease | No plaques, neurofibrillary tangles, or neuron loss observed at three months, the oldest age reported. |
Unknown. |
App knock-in (humanized Aβ)
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
App | App: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
App knock-in (humanized Aβ) (Leuven)
Observed
Absent
-
Plaques at
No plaques observed up to 2 years of age.
-
Tangles at
No tangles observed up to 2 years of age.
No Data
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
App | App: Knock-In | Alzheimer's Disease | No plaques or tangles were observed up to two years of age. |
Unknown. |
App knock-in (humanized Aβ) (Leuven); Psen1 knock-in (M139T)
Observed
Absent
-
Plaques at
No plaques observed up to 2 years of age.
-
Tangles at
No tangles observed up to 2 years of age.
No Data
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
App, Psen1 | PSEN1 M139T | App: Knock-In; Psen1: Knock-In | Alzheimer's Disease | No plaques or tangles were observed up to 2 years of age. |
Unknown. |
App knock‐in (Icelandic mutation and humanized Aβ)
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
App | APP A673T (Icelandic) | App: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
App knock‐in (Swedish mutation and humanized Aβ)
Observed
Absent
-
Plaques at
None observed at 3 months.
-
Tangles at
None observed at 3 months.
-
Neuronal Loss at
None observed at 3 months.
No Data
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
App | APP K670_M671delinsNL (Swedish) | App: Knock-In | Alzheimer's Disease | No plaques, neurofibrillary tangles, or neuron loss observed at 3 months, the oldest age reported. |
Unknown. |
App KO/APOE4/Trem2*R47H
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE, App, Trem2 | TREM2 R47H | APOE: Knock-In; App: Knock-Out; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
APP NL-F Knock-in
Observed
-
Plaques at 26
Homozygotes develop amyloid plaques starting at 6 months in the cortex and hippocampus. Heterozygotes develop amyloidosis after 24 months. Plaques contained Aβ1-42 and pyroglutamate Aβ (Aβ3(pE)-42); Aβx-40 was a minor species.
-
Gliosis at 26
Microglia and activated astrocytes accumulate with age, starting around 6 months of age, concurrent with plaque formation.
-
Synaptic Loss at 39
Reduced synaptophysin and PSD95 immunoreactivities associated with Aβ plaques at 9-12 months.
-
Cognitive Impairment at 78
Memory impairment in homozygous mice at 18 months as measured by the Y maze test. APPNL/NL mice (with Swedish mutation only) were unimpaired at this age. No significant deficit was seen in the Morris water maze at 18 months.
Absent
-
Tangles at
Absent; although elevated levels of phosphorylated tau are observed in dystrophic neurites around plaques.
-
Neuronal Loss at
Absent.
No Data
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP I716F (Iberian) | APP: Knock-In | Alzheimer's Disease | Elevated Aβ peptides accumulating into plaques starting at 6 months. Microgliosis and astrocytosis, especially around plaques. Reduced synaptophysin and PSD-95 indicative of synaptic loss. No tangle pathology or neurodegeneration. |
Memory impairment by 18 months as measured by the Y maze. No significant impairment in the Morris water maze. |
APP NL-G-F Knock-in
Observed
-
Plaques at 9
Aggressive amyloidosis; plaques develop in homozygous mice starting at 2 months with near saturation by 7 months. Aβ deposition at 4 months in heterozygous mice. Cortical and subcortical amyloidosis present.
-
Gliosis at 9
Microglia and activated astrocytes accumulate with age starting around 2 months, especially around plaques in a manner concurrent with plaque formation.
-
Synaptic Loss at 17
Reduction of synaptophysin and PSD95 immunoreactivities associated with Aβ plaques in both cortical and hippocampal areas.
-
Cognitive Impairment at 26
Memory impairment in homozygous mice by 6 months of age as measured by the Y maze.
Absent
-
Tangles at
Absent; although phosphorylated tau is elevated in dystrophic neurites around plaques.
-
Neuronal Loss at
Absent.
No Data
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP I716F (Iberian), APP E693G (Arctic) | APP: Knock-In | Alzheimer's Disease | Aggressive amyloidosis with deposition in the cortex beginning at 2 months and approaching saturation by 7 months. Aβ deposition in heterozygous mice at 4 months. Subcortical amyloidosis. Exacerbated microgliosis and astrocytosis compared to APPNL-F mice. Reduced synaptophysin and PSD-95 indicative of synaptic loss. No tangle pathology or neurodegeneration. |
Memory impairment by 6 months as measured by the Y maze. |
App NL-G-F Knock-in Rat
Observed
-
Plaques at 4
Amyloid plaques apparent as early as 1 month in homozygous knock-ins, 4 months in heterozygotes. Amyloid pathology progresses more rapidly in females than males.
-
Neuronal Loss at 52
Reduced brain weight, fewer neurons in the hippocampus and cortex, and enlarged lateral ventricles seen at 12 months.
-
Gliosis at 24
Astrogliosis and microgliosis, particularly pronounced around amyloid plaques, were observed in homozygous knock-in rats at 6 months of age. Gliosis was also seen in year-old heterozygotes.
-
Synaptic Loss at 24
Decreased levels of the presynaptic marker synaptophysin and the postsynaptic marker PSD-95 in knock-in rats. Quantitative electron microscopy showed reductions in synaptic density, area, and perimeters in the hippocampus, entorhinal cortex and prefrontal cortex of knock-in brains.
-
Cognitive Impairment at 20
Deficits in the Morris Water Maze task and a paired associate learning task as early as 5 and 7 months of age, respectively.
Absent
-
Tangles at
No neurofibrillary tangles through 22 months of age, but increases in tau phosphorylation, aggregation, and conformational changes.
No Data
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
App | APP K670_M671delinsNL (Swedish), APP E693G (Arctic), APP I716F (Iberian) | App: Knock-In | Alzheimer's Disease | Amyloid plaques apparent as early as 1 month in homozygous knock-ins. No neurofibrillary tangles through 22 months of age, but increases in tau phosphorylation, aggregation, and conformational changes. Astrogliosis, microgliosis, synapse and neuron loss. |
Deficits in the Morris Water Maze task and a paired associate learning task as early as 5 and 7 months of age, respectively. |
AppNL-G-F/MAPT double knock-in
Observed
-
Plaques at 8
Plaques observed at 2 months.
-
Gliosis at 16
Astrogliosis and microgliosis observed by 4 months.
-
Cognitive Impairment at 52
Deficits in the Y-maze test of working memory at 12 months of age.
Absent
-
Tangles at
No neurofibrillary tangles observed up to 24 months of age.
-
Neuronal Loss at
No neurodegeneration observed up to 24 months of age.
No Data
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
App, MAPT | APP K670_M671delinsNL (Swedish), APP I716F (Iberian), APP E693G (Arctic) | App: Knock-In; MAPT: Knock-In | Alzheimer's Disease | Amyloid plaques, plaque-associated neuritic dystrophy, and neuroinflammation, similar to AppNL-G-F. |
Deficits in the Y-maze test of working memory, similar to AppNL-G-F. |
APPPS1
Observed
-
Plaques at 6
Aβ deposition begins at 6 weeks of age in the cortex and 3-4 months of age in the hippocampus (Radde et al., 2006).
-
Neuronal Loss at 74
Global neuron loss is not observed, but modest neuron loss was found in the granule cell layer of the dentate gyrus and other subregions with high neuronal density in 17-month old animals (Rupp et al., 2011).
-
Gliosis at 6
Activated microglia around Aβ deposits at 6 weeks as well as increased astrogliosis (Radde et al., 2006). Levels of CCL2 and TNFα increase at later ages (Lee et al., 2010).
-
Synaptic Loss at 10
Dendritic spine loss around plaques reported to begin approximately 4 weeks after plaque formation and continue for several months (Bittner et al., 2012).
-
Changes in LTP/LTD at 35
Hippocampal CA1 LTP normal at 4.5 months of age, but impaired at 8 and 15 months of age (Gengler et al., 2010).
-
Cognitive Impairment at 30
Cognitive deficits in spatial learning and memory in the Morris water maze reported at 7 months (Serneels et al., 2009). Impaired reversal learning of a food-rewarded four-arm spatial maze task observed at 8 months (Radde et al., 2006).
Absent
-
Tangles at
Phosphorylated tau-positive neuritic processes around plaques have been observed, but no mature tangles (Radde et al., 2006).
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1 L166P | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaque deposition starts at approximately 6 weeks in the neocortex. Amyloid deposits in the hippocampus appear at 3-4 months, and in the striatum, thalamus and brainstem at 4-5 months. Phosphorylated tau-positive neuritic processes have been observed in the vicinity of all congophilic amyloid deposits, but no fibrillar tau inclusions are seen.
|
Cognitive deficits in spatial learning and memory in the Morris water maze reported at 7 months. Impaired reversal learning of a food-rewarded four-arm spatial maze task at 8 months. |
APP+PS1
Observed
-
Plaques at 76
Abundant plaques in hippocampus and subiculum, scattered plaques in cortex.
-
Neuronal Loss at 76
Necrotic neurons in hippocampus and cortex.
-
Cognitive Impairment at 40
Deficits in Barnes maze at 10 months.
Absent
No Data
-
Tangles at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP V717F (Indiana), PSEN1 L166P | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques, cerebral amyloid angiopathy, and necrotic neurons in hippocampus and cortex by 19 months of age. |
Deficits in Barnes maze by 10 months of age. |
APP/PS1/rTg21221
Observed
-
Plaques at 35
Cortical plaques observed between 8-10 months. Plaques larger than in control mice not expressing human tau.
-
Neuronal Loss at 36
Neuronal loss observed adjacent to plaques relative to more distal areas.
-
Gliosis at 37
Increased astrocytosis adjacent to plaques relative to more distal areas.
-
Synaptic Loss at 40
Decreased synapse density adjacent to plaques relative to more distal areas.
Absent
-
Tangles at
No tangles. Aggregates of misfolded and phosphorylated tau observed between 8-10 months.
No Data
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1, MAPT | APP K670_M671delinsNL (Swedish), PSEN1: deltaE9 | APP: Transgenic; PSEN1: Transgenic; MAPT: Transgenic | Alzheimer's Disease | Tau accumulations, dystrophic neurites, astrocytosis, neuronal loss, and synapse loss were more pronounced adjacent to cortical plaques. Tangles were not observed. |
No data. |
AppSAA Knock-in
Observed
-
Plaques at 16
Amyloid plaques seen in AppSAA homozygous mice from 4 months of age and heterozygous mice at 16 months of age.
-
Gliosis at 16
Plaque-associated microgliosis observed by 4 months of age.
Absent
-
Tangles at
AT8-positive dystrophic neurites, but no neurofibrillary tangles, detected in AppSAA homozygous mice at 8 months of age.
No Data
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
App | APP K670_M671delinsNL (Swedish), APP E693G (Arctic), APP T714I (Austrian) | App: Knock-In | Alzheimer's Disease | Homozygotes: Amyloid plaques and plaque-associated microgliosis from 4 months of age; cerebral amyloid angiopathy and dystrophic neurites from 8 months of age. Heterozygotes: Amyloid plaques at 16 months of age. |
Unknown. |
APPsw/0; Pdgfrβ+/-
Observed
-
Plaques at 39
By 9 months of age APPsw/0;Pdgfrβ+/- mice have an elevated plaque load in the cortex and hippocampus compared with age matched APPsw/0;Pdgfrβ+/+. littermates. They also have extensive cerebral amyloid angiopathy.
-
Neuronal Loss at 39
Progressive neuronal degeneration including reduced neurite density and reduced neuronal number in the cortex and hippocampus of APPsw/0; Pdgfrβ+/- mice at at nine months compared to age-matched APPsw/0; Pdgfrβ+/+ littermates.
-
Cognitive Impairment at 41
At nine months, APPsw/0;Pdgfrβ+/- mice perform poorly on several hippocampal-dependent behavioral tests including burrowing, nest construction, and novel object recognition, compared with age-matched APPsw/0;Pdgfrβ+/+ littermates.
Absent
No Data
-
Tangles at
Although mature neurofibrillary tangles were not observed by 9 months (the oldest age assessed), the mice develop significant tau pathology, including tau hyperphosphorylation in cortical and hippocampal neurons. Pre-tangle pathology is observed, including neuronal caspase-cleaved tau, and conformational changes as indicated by the conformation-specific antibody MC1.
-
Gliosis at
Unknown.
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PDGFRB | APP K670_M671delinsNL (Swedish) | APP: Transgenic; PDGFRB: Knock-Out | Alzheimer's Disease | Amyloid plaques; elevated brain interstitial human and murine Aβ due to reduced clearance of soluble Aβ, cerebral amyloid angiopathy, tau hyperphosphorylation and related pathology. Neurite loss and neuronal loss in the cortex and hippocampus. |
Age-associated cognitive impairment as measured by hippocampal-dependent tasks, including nest building, burrowing, and novel object recognition. |
APPSwDI x NOS2 Knock-out
Observed
-
Plaques at 49
Aβ deposits by 52 weeks. Particularly dense Aβ immunoreactivity in the subiculum and thalamus, including in the cerebral microvessels (Wilcock et al., 2008).
-
Tangles at 49
Extensive tau pathology by 52 weeks, including intraneuronal aggregates of hyperphosphorylated tau. Increased phosphorylated tau in bigenic mice compared to APPSwDI mice (Wilcock et al., 2008).
-
Neuronal Loss at 52
Significant neuron loss by 52 weeks in the hippocampus and subiculum, especially of neuropeptide Y neurons. Numerous Fluoro-Jade C+ neurons: 30% loss in the hippocampus, 35% loss in the subiculum (Wilcock et al., 2008).
-
Cognitive Impairment at 53
Impairments in spatial memory by 52-56 weeks as measured by the radial arm maze and the Barnes maze. Bigenic mice more impaired than APPSwDI (Wilcock et al., 2008).
Absent
No Data
-
Gliosis at
Unknown.
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, NOS2 | APP K670_M671delinsNL (Swedish), APP E693Q (Dutch), APP D694N (Iowa) | APP: Transgenic; NOS2: Knock-Out | Alzheimer's Disease | Plaques especially in the thalamus and subiculum. Aggregated, hyperphosphorylated tau tangles. Neuronal loss especially of NPY neurons in the hippocampus and subiculum. More severe pathology than Tg-SwDI alone. |
Severe learning and memory deficits. Impaired spatial memory compared to Tg-SwDI as measured by the radial arm maze and the Barnes maze at 52-56 weeks. |
APP(Swedish) (R1.40)
Observed
-
Plaques at 59
By 13.5 months homozygous mice develop both parenchymal and vascular amyloid deposits which first appear in the frontal cortex. No Aβ deposition at 5 months (Lehman et al., 2003).
-
Gliosis at 61
Reactive astrocytes and microglia in 14-16 month old animals (Kulnane et al., 2001).
Absent
-
Tangles at
No mature tangles, but some changes in phosphorylated tau.
-
Changes in LTP/LTD at
Absent.
No Data
-
Neuronal Loss at
Unknown.
-
Synaptic Loss at
Unknown.
-
Cognitive Impairment at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish) | APP: Transgenic | Alzheimer's Disease | By 14-16 months, homozygotes have diffuse and compact Aβ deposits in the frontal cortex, by 18-20 months plaques throughout the cortex and olfactory bulb with occasional deposits in the corpus callosum and hippocampus. No tangles, but some changes in phosphorylated tau. Reactive astrocytes and microglia by 14-16 months. |
Unknown. |
APPSwe (line C3-3)
Observed
-
Plaques at 104
Some plaque formation at advanced age (24-26 months) (Savonenko et al., 2003).
Absent
-
Cognitive Impairment at
Normal reference and working memory up to 12-14 months on congenic background (Savonenko et al., 2003).
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish) | APP: Transgenic | Alzheimer's Disease | Age-associated increase in Aβ40 and Aβ42 and some amyloid deposition at advanced age. |
Congenic animals showed normal reference and working memory up to 12-14 months. |
APPSwe/PSEN1(A246E)
Observed
-
Plaques at 39
By 9 months of age, amyloid plaques develop in the hippocampus and subiculum, later extending to the cortex (Borchelt et al., 1997). The striatum and thalamus are relatively spared out to 18 months of age. Amyloid pathology is more severe in female mice, with a greater amyloid burden measured at 12 and 17 months of age (Wang et al., 2003).
-
Gliosis at 52
By one year of age, reactive gliosis is observed in the cortex and hippocampus and is associated with dystrophic neurites (Borchelt et al., 1997).
-
Cognitive Impairment at 48
Age-associated cognitive impairment, as measured by the Morris water maze, was observed in 11 to 12-month-old males. Both acquisition and retention were impaired. No impairment at 3-4 months of age. At both time points mice performed normally on a position discrimination task in the T-maze (Puoliväli et al., 2002).
Absent
-
Tangles at
Not observed.
-
Neuronal Loss at
There was no difference in neuronal numbers in the cingulate cortex compared with wild-type mice (Xiang et al., 2002).
No Data
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1 A246E | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques by 9 months, starting in the hippocampus and subiculum. Plaques later develop in the cortex; the striatum and thalamus are relatively spared. Amyloid pathology is more severe in females. Dystrophic neurites and gliosis in the cortex and hippocampus. |
Poor nest building. Reduced retention in a learned passive avoidance task. Increased immobility time in forced swim task. Age-associated impairment in acquisition and retention in the Morris water maze. No impairment in a position discrimination T-maze task. |
APPSwe/PSEN1dE9 (C3-3 x S-9)
Observed
-
Plaques at 26
Plaques are present in the hippocampus and cortex by 6 months of age.
-
Cognitive Impairment at 78
Age-related cognitive deficits. Episodic memory appears to be more sensitive than reference memory. No differences at 6 months of age, but detectable at 18 months (Savonenko et al., 2005).
Absent
-
Tangles at
Not observed.
No Data
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1: deltaE9 | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Elevated Aβ42 and plaques in the hippocampus and cortex. No tangles. Reduced cholinergic markers. |
Age-related cognitive deficits; episodic memory more sensitive than reference memory. No differences at 6 months, but detectable at 18 months. |
APPswe/PSEN1dE9 (C57BL6)
Observed
-
Plaques at 16
Amyloid plaques begin to emerge in the cortex at about 4 months of age and in the hippocampus at about 6 months.
-
Gliosis at 17
Plaque-associated astrogliosis and microgliosis are evident by 4 and 8 months, respectively.
-
Synaptic Loss at 18
Synapse loss in the hippocampus occurs by 4 months.
-
Cognitive Impairment at 40
Deficits in the Morris water maze emerge between 6 and 10 months and worsen with age.
Absent
-
Tangles at
Not observed.
-
Neuronal Loss at
Neuron loss has not been observed in mice up to 12 months of age.
No Data
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1: deltaE9 | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques begin to emerge in the cortex at about 4 months of age and in the hippocampus at about 6 months. Gliosis and dystrophic neurites are associated with plaques. Amyloid angiopathy has been observed in the retina. |
Hyperactivity is apparent by 6 months. Deficits in the Morris water maze emerge between 6 and 10 months and worsen with age. |
APPswe/PSEN1dE9 (line 85)
Observed
-
Plaques at 26
Occasional Aβ deposits can be found by 6 months, with abundant plaques in the hippocampus and cortex by 9 months (Jankowsky et al., 2004) and a progressive increase in plaques up to 12 months (Garcia-Alloza et al., 2006).
-
Neuronal Loss at 35
Neuronal loss observed adjacent to plaques relative to more distal areas.
-
Gliosis at 26
Minimal astrocytosis at 3 months; significant astrocytosis by 6 months, especially in areas around plaques. Extensive GFAP+ staining at 15 months and later throughout the cortex (Kamphuis et al., 2012).
-
Synaptic Loss at 17
In the B6 congenic mice, age-dependent loss of synaptophysin, synaptotagmin, PSD-95, and Homer immunoreactivity in the hippocampus by 4 months (Hong et al., 2016).
-
Changes in LTP/LTD at 13
Transient long-term potentiation (t-LTP) is reduced by 3 months. The degree of impairment is not related to age from 3 to 12 months (Volianskis et al., 2008).
-
Cognitive Impairment at 52
Impairment in the Morris water maze at 12 months, specifically during acquisition of the hidden platform sub-task and the probe trial, but not in the visible platform test (Lalonde et al., 2005). At 13 months the mice commit more errors in the Morris water maze, but not at 7 months (Volianskis et al., 2008).
Absent
-
Tangles at
Not observed.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1: deltaE9 | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Occasional Aβ deposits by 6 months with abundant plaques in the hippocampus and cortex by 9 months and a progressive increase in plaques up to 12 months. No tangles. Decrease in synaptic markers and increase in complement immunoreactivity. |
Cognitive impairment (e.g., deficits in spatial memory and contextual memory). Changes in spontaneous behavior (e.g., nest-building, burrowing). |
APP(V642I)KI
Observed
-
Cognitive Impairment at 117
Impairments at the water finding task at age 27-29 months, a test of long-term memory. No differences in the open field test of the elevated plus maze indicating no difference in general behavioral patterns, activity level, or emotional state.
Absent
-
Plaques at
Absent.
-
Tangles at
Absent.
-
Neuronal Loss at
Absent.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP V717I (London) | APP: Transgenic | Alzheimer's Disease | Increased Aβ42(43) relative to Aβ40 at 29 months, but without neuritic plaques, neurofibrillary tangles, massive neuronal loss, or brain atrophy. |
At 27-29 months mice displayed long-term memory deterioration. Acquisition of spatial memory is slightly affected, but no deterioration in short-term working memory. No difference in open field test or elevated plus maze suggesting no difference in overall behavioral patterns or activity levels. |
APP(V717I)
Observed
-
Plaques at 43
Plaques start in the cortex and subiculum at ~10 months. Diffuse amyloid deposits and compact neuritic plaques at 13-18 months especially in the hippocampus and cortex, with occasional deposits in the thalamus and fimbria, external capsule, pontine nuclei, and white matter (Moechars et al., 1999). Prominent amyloid deposits in brain vessels after 15 months (Van Dorpe et al, 2000).
-
Gliosis at 43
GFAP, microglial activation, and other markers of brain inflammation are elevated by 10 months.
-
Changes in LTP/LTD at 26
Significant deficit in LTP in CA1 region of the hippocampus at 6 months.
-
Cognitive Impairment at 26
From the age of 6 months, spatial and non-spatial orientation and memory deficits by Morris water maze and other tests. Also deficits in associative learning.
Absent
-
Tangles at
Dystrophic neurites containing hyperphosphorylated tau, but no tangle pathology.
-
Neuronal Loss at
Absent.
No Data
-
Synaptic Loss at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP V717I (London) | APP: Transgenic | Alzheimer's Disease, Cerebral Amyloid Angiopathy | Plaques start in the subiculum, spreading to the frontal cortex as dense and diffuse aggregates. Prominent amyloid deposits in brain vessels after 15 months. Microbleeds. Amyloid-associated inflammation. CSF Aβ42/Aβ40 ratio decreases from 15 months. Dystrophic neurites containing hyperphosphorylated tau, but no tangle pathology. |
From the age of 6 months, spatial and non-spatial orientation and memory deficits by Morris water maze. Impaired associative learning. Increased agitation/anxiety from 8 weeks. Reduced ambulation, especially with age. Hyperactivity and aggression. |
APP(V717I) x PS1(A246E)
Observed
-
Plaques at 17
Plaques start in cortex, hippocampus and subiculum at 4-6 months.
-
Gliosis at 20
Elevated GFAP, microglial activation, and other markers of brain inflammation increase as of 4.5 months.
-
Changes in LTP/LTD at 26
Significant deficit in LTP in CA1 region of the hippocampus at 6 months.
-
Cognitive Impairment at 22
From the age of 5 months, spatial and non-spatial orientation and memory deficits by Morris water maze and other tests. Also deficits in associative learning.
Absent
-
Tangles at
Dystrophic neurites containing hyperphosphorylated murine tau, but no tangle pathology.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP V717I (London), PSEN1 A246E | APP: Multi-transgene; PSEN1: Transgenic | Alzheimer's Disease, Cerebral Amyloid Angiopathy | Soluble, oligomeric Aβ at 2 months and increases with age. Amyloid plaques at 6-9 months, earlier than APP(V717I) single transgenics. Plaques start in the subiculum and spread to the frontal cortex. Amyloid-associated inflammation. CAA pathology at 8 months; microbleeds at 12-15 months. Dystropic neurites containing hyperphosphorylated tau, but no tangle pathology. |
From the age of 5 months, spatial and non-spatial orientation and memory deficits by Morris Water Maze. Impaired associative learning, hyperactivity, anxiety, and aggression. |
Arc48 (APPSw/Ind/Arc)
Observed
-
Plaques at 9
Parenchymal neuritic plaques by 2 months with prominent plaque deposition in the hippocampus by 3-4 months. Abundant mature thioflavin-S positive plaques with dystrophic neurites by 10-12 months (Cheng et al., 2007).
-
Gliosis at 13
Reactive astrocytosis at 3-4 months in the dentate gyrus as demonstrated by GFAP immunoreactivity (Cheng et al., 2007).
-
Cognitive Impairment at 13
At 3-4 months the Arc48 mouse was able to learn a task involving escape to a cued platform in the Morris water maze, but were impaired in the ability to use extramaze cues to navigate to the hidden platform (Cheng et al., 2007).
Absent
-
Tangles at
Absent.
No Data
-
Neuronal Loss at
Unknown.
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP V717F (Indiana), APP E693G (Arctic) | APP: Transgenic | Alzheimer's Disease, Cerebral Amyloid Angiopathy | Parenchymal neuritic plaques by 2 months accompanied by dystrophic neurites. Prominent hippocampal Aβ deposition by 3-4 months. Relatively low Aβ42/Aβ40 ratio. Comparable cerebrovascular amyloid deposition to J20. |
At 3-4 months the Arc48 mouse was able to learn a task involving escape to a cued platform in the Morris water maze, but had an impaired ability to use extramaze cues to navigate to the hidden platform. |
ArcAβ
Observed
-
Plaques at 39
Between 9 and 15 months of age β-amyloid plaques became prominent. Plaques had a characteristic dense core morphology which differed from the cotton wool-like structure of plaques seen with the Swedish mutation alone (Knobloch et al., 2007).
-
Changes in LTP/LTD at 15
LTP is severely impaired in slices from 3.5 and 7.5 month old mice. LTP and basal synaptic transmission were normal in slices from one month old mice (Knobloch et al., 2007).
-
Cognitive Impairment at 26
Cognitive impairment measured from the age of 6 months in the Morris water maze and Y-maze, as well as in active avoidance behavior (Knobloch et al., 2007).
Absent
-
Tangles at
Absent.
No Data
-
Synaptic Loss at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP E693G (Arctic) | APP: Transgenic | Alzheimer's Disease | At 6 months intracellular punctate deposits of Aβ abundant in cortex and hippocampus, but overt β-amyloid plaques not apparent until 9-15 months. Severe CAA also present at this age with dense Aβ aggregates in blood vessels walls and spreading into the parenchyma. | Cognitive impairments from the age of 6 months measured in the Morris water maze and Y-maze. |
ARTE10
Observed
-
Plaques at 13
Robust and reliable plaque pathology as early as 3 months in homozygotes, 5 months in hemizygotes. Plaques start in the anterior neocortex and subiculum, spreading to other brain regions (e.g. hippocampus, thalamus, amygdala). Congophilic dense-core plaques are abundant, with lower levels of diffuse plaques and some cerebral amyloid angiopathy.
-
Gliosis at 22
Glial activation, including reactive astrocytes and activated microglia, is present in areas around plaques by 5 months of age in homozygous animals, later in hemizygotes.
-
Synaptic Loss at 13
Decreased expression of synaptophysin mRNA in the brain by 3-4 months of age in both hemizygous and homozygous animals.
-
Cognitive Impairment at 52
Select, paradigm-dependent, deficits in learning and memory, especially episodic memory, by 12 months in homozygous and hemizygous mice.
Absent
-
Tangles at
No tangles or neuropil threads, but some hyperphosphorylated tau by eight months in dystrophic neurites.
-
Neuronal Loss at
Outright neuronal loss has not been documented, but substantial degeneration of dendritic arbors occurs by 10-14 months of age in hippocampal neurons.
No Data
-
Changes in LTP/LTD at
Unknown; however, hippocampal neurons exhibit substantial changes in electrophysiological properties by 10-14 months of age, including hyperexcitability in the form of increased firing of action potentials and a more efficient transition from solitary firing to bursting.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1 M146V | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Robust early plaque development (by 3 months in homozygotes, 5 months in hemizygotes), predominantly congophilic dense-core amyloid plaques surrounded by dystrophic neurites and gliosis. Some diffuse plaques and cerebral amyloidosis. No tau tangles. Neurons have reduced dendritic length, surface area, and branches. |
Age-related learning and memory deficits, especially episodic memory, in select paradigm-specific tasks by 12 months. |
Atg16LΔWD
Observed
-
Neuronal Loss at 104
Apparent neuron loss in hippocampi of 2-year-old mice (fewer neurons, increased levels of cleaved caspase-3, and increased numbers of TUNEL-positive neurons).
-
Gliosis at 104
Microgliosis in the hippocampi of 2-year-old mice.
-
Changes in LTP/LTD at 104
Impaired long-term potentiation at CA3-CA1 synapses.
-
Cognitive Impairment at 104
Deficits in the sucrose preference test, spontaneous alternation in the Y-maze, and novel object recognition test.
Absent
-
Plaques at
Intracellular and extracellular Aβ deposits, but no dense-core plaques, in 2-year-old mice.
No Data
-
Tangles at
No data.
-
Synaptic Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Atg16l1 | Atg16l1: Knock-Out | Alzheimer's Disease | Intracellular and extracellular Aβ deposits, but no dense-core plaques. Microgliosis and neuron loss in 2-year-old mice. |
Deficits in the sucrose-preference test, spontaneous alternation in the Y-maze, and novel object recognition test. |
BACE1 cKO (Hu, Yan) X 5xFAD
Observed
-
Plaques at 11
Accumulate up to day 120, but to a lesser degree than in control 5xFAD, then recede thereafter.
-
Gliosis at 11
Reactive astrocytes and microglia accumulate up to day 120, but to a lesser degree than in control 5xFAD, then recede thereafter.
-
Changes in LTP/LTD at 40
Deficit in LTP at Schaffer collateral–CA1 synapses, but less severe than in control 5xFAD mice.
Absent
-
Cognitive Impairment at
Cued and contextual fear conditioning normal, tested at eight to 10 months of age.
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Bace1, APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | Bace1: Conditional Knock-out; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques, reactive astrocytes and microglia, and dystrophic neurites accumulate up to day 120, but to a lesser degree than in control 5xFAD (5xFAD mice homozygous for a floxed Bace1 gene), then recede thereafter. |
Normal contextual and cued fear conditioning, tested at 8 to 10 months of age. |
Bace1 conditional knock-out (adult, whole body) (Vassar)
Observed
Absent
-
Changes in LTP/LTD at
LTP at Schaffer collateral–CA1 synapses was similar in slices obtained from 12-month BACE1-deficient and control mice.
-
Cognitive Impairment at
Normal learning and memory in the Morris water maze, normal alternation in the Y-maze test of working memory, and normal cued and contextual fear conditioning when tested at 9 months of age.
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Bace1 | Bace1: Conditional Knock-out | Alzheimer's Disease | Defects in axonal organization. |
Normal learning and memory in the Morris water maze, normal alternation in the Y-maze test of working memory, and normal cued and contextual fear conditioning; possible hyperactivity in novel situations. |
BACE1 conditional knock-out (Hu, Yan)
Observed
-
Changes in LTP/LTD at 40
Long-term potentiation at Schaffer collateral–CA1 synapses impaired in slices obtained from 10- to 12-month-old mice.
Absent
-
Plaques at
Not observed.
-
Tangles at
Not observed.
-
Neuronal Loss at
Not observed.
-
Gliosis at
No astrogliosis at 1-2 months.
-
Cognitive Impairment at
Contextual and cued fear conditioning normal at 8-10 months.
No Data
-
Synaptic Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Bace1 | Bace1: Conditional Knock-out | Alzheimer's Disease | No gross morphological changes observed. |
BACE1-deficient mice and Bace1fl/fl mice performed similarly in tests of contextual and cued fear conditioning at 8 to 10 months of age. |
Bace1 conditional knock-out (neonatal, forebrain) (Vassar)
Observed
-
Cognitive Impairment at 24
Delayed learning, but normal memory, in the Morris water maze; normal alternation in the Y-maze test of working memory, normal cued and contextual fear conditioning.
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Bace1 | Bace1: Conditional Knock-out | Alzheimer's Disease | Hypomyelination and defects in axon organization. |
Delayed learning, but normal memory, in the Morris water maze; normal alternation in the Y-maze test of working memory, normal cued and contextual fear conditioning. Hyperactivity in when placed in novel environments. |
Bace1 conditional knockout (Tesco)
Observed
-
Changes in LTP/LTD at 60
Deficit in long-term potentiation at Schaffer collateral–CA1 synapses in slices from 14-month-old animals that had received tamoxifen between 8 and 12 weeks of age.
Absent
-
Cognitive Impairment at
Animals that had received tamoxifen between 8 and 12 weeks of age were tested at 4–5 or 12–14 months. Tamoxifen-treated mice performed similarly to vehicle-treated controls in the Y-maze, contextual fear conditioning, pre-pulse inhibition, open field, and light-dark transition tests.
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Bace1 | Bace1: Conditional Knock-out | Alzheimer's Disease | None observed: hippocampal mossy fiber organization and sciatic-nerve myelination were normal. |
Performed similarly to controls in a battery of tests (Y-maze, contextual fear conditioning, pre-pulse inhibition, open field, and light-dark transition task). |
BRI-Aβ42 (BRI2-Aβ42)
Observed
-
Plaques at 13
Detergent-insoluble amyloid-β and cored plaques as early as three months in the cerebellum. Variable forebrain pathology later with extracellular Aβ plaques in the hippocampus and entorhinal/piriform cortices by 12 months. Extensive congophillic amyloid angiopathy.
-
Gliosis at 13
Plaque-associated reactive gliosis as measured by GFAP immunostaining.
Absent
-
Tangles at
Absent.
-
Neuronal Loss at
Absent.
-
Cognitive Impairment at
On a mixed (C57/B6//C3H) background hemizygous mice have intact cognition as measured by fear conditioning at 12 months and 14-17 months despite accumulating amyloid.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Transgenic | Alzheimer's Disease, Cerebral Amyloid Angiopathy | Detergent-insoluble amyloid-β appearing with age and cored plaques as early as 3 months in the cerebellum. Variable forebrain pathology later with extracellular Aβ plaques in the hippocampus and entorhinal/piriform cortices at 12 months. Age-associated congophillic amyloid angiopathy. No tangles or neuronal loss. |
On a mixed (C57/B6//C3H) background hemizygous mice have intact cognition as measured by fear conditioning at 12 months and 14-17 months despite accumulating amyloid. |
CAST.APP/PS1
Observed
-
Plaques at 32
Thioflavin S-positive amyloid plaques are present in the cortex and hippocampus by 8 months of age, with more severe plaque pathology in females than in males.
-
Neuronal Loss at 34
Compared with their non-transgenic littermates, CAST.APP/PS1 mice have fewer neurons in area CA1 of the hippocampus. Cortical neuron numbers do not differ between the genotypes.
-
Gliosis at 33
Plaque-associated microgliosis observed by 8 months.
-
Cognitive Impairment at 31
Deficits in short-term memory by 8 months in males (data from females unavailable).
Absent
-
Tangles at
Not observed.
No Data
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1: deltaE9 | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques, plaque-associated gliosis, cerebral amyloid angiopathy; possible neuron loss in hippocampal area CA1. |
Transgenic mice are hyperactive. Working memory (spontaneous alternation in the Y-maze) is normal at 7 to 8 months, but short-term memory (tested in the Y-maze) is impaired in males (data from females is not available, as wild-type females are unable to perform this test). |
Ceacam1 KO/APOE4/Trem2*R47H
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE, Ceacam1, Trem2 | TREM2 R47H | APOE: Knock-In; Ceacam1: Knock-Out; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
Clasp2*L163P/APOE4/Trem2*R47H
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Clasp2, APOE, Trem2 | TREM2 R47H | Clasp2: Knock-In; APOE: Knock-In; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
Dp(16)1Yey/+
Observed
-
Neuronal Loss at 56
Loss of vulnerable neurons (neurons in layer II of entorhinal cortex, catecholaminergic neurons in the locus coeruleus and the basal forebrain at advanced ages.
-
Synaptic Loss at 37
Critical synaptic proteins were altered, including syntaxin 1A and SNAP25.
-
Changes in LTP/LTD at 8
Magnitude of hippocampal LTP following theta burst stimulation is lower than the WT controls around 2-4 months of age.
-
Cognitive Impairment at 9
Cognitive impairments were observed at 2-4 months of age, as well as at more advanced ages.
Absent
-
Plaques at
None.
-
Tangles at
None.
No Data
-
Gliosis at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Multi-transgene | Down's Syndrome, Alzheimer's Disease | Neuronal loss in the entorhinal cortex, locus coeruleus, and the basal forebrain magnocellular complex; increased tau pathology and increased astrocyte and microglia levels. |
Impairments in contextual memory, spatial learning, novel object recognition memory, and vocalizations. |
Dp1Tyb
Observed
-
Gliosis at 14
Increased density of microglia in the hippocampus at 14 weeks of age.
-
Cognitive Impairment at 8
Exploratory behavior is impaired at 8 weeks of age. Fear memory is impaired at 10 months. Spatial working memory is impaired at 3-months of age.
Absent
-
Plaques at
No amyloid-β plaque deposition is observed in hippocampus at 12-months of age.
-
Tangles at
Not observed.
No Data
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Knock-In | Down's Syndrome, Alzheimer's Disease | No amyloid plaques. At 14 weeks, smaller and rounder brain, reduced medial prefrontal cortex and dorsal hippocampus volumes, reduced density of neurons, and increased density of microglia in the hippocampus. Altered hippocampal metabolites including glutamine and the glutamine/glutamate ratio. Reduced IB4 neurons in the DRG. |
Exploratory behavior is impaired at 8 weeks of age. Fear memory is impaired at 10 months. Motor function is impaired at 12-15 weeks of age. Sleep architecture is disrupted at 16-weeks of age. Spatial working memory is impaired at 3-months of age. |
Dp9Tyb
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Down's Syndrome, Alzheimer's Disease | Unknown. |
No locomotor defects as assessed by Rotarod at 12 weeks of age. |
E2FAD
Observed
-
Plaques at 17
Plaques develop in the subiculum and deep cortical layers by 4 months.
-
Gliosis at 26
Microgliosis and astrocytosis in the subiculum and cortex at 6 months.
-
Synaptic Loss at 17
Protein levels of NMDA receptor subunits decreased from 2 to 6 months.
Absent
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
E2FAD mice had performance in learning and memory tasks comparable to E3FAD animals and better than E4FAD mice.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE, APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | APOE: Knock-In; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques starting at 4 months and increasing with age. Gliosis and loss of synaptic proteins. |
In the Y maze and Morris water maze, E2FAD mice performed better than E4FAD mice, and were comparabile to E3FAD mice. |
E3FAD
Observed
-
Plaques at 17
Plaques develop in the subiculum and deep cortical layers by 4 months.
-
Gliosis at 26
Microgliosis and astrocytosis in the subiculum and cortex at 6 months.
-
Synaptic Loss at 17
Protein levels of NMDA receptor subunits decreased from 2 to 6 months.
Absent
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
E3FAD mice had performance in learning and memory tasks comparable to E4FAD and E2FAD animals.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE, APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | APOE: Knock-In; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques starting at 4 months and increasing with age. Gliosis and loss of synaptic proteins. |
In the Y maze and Morris water maze E3FAD mice performed better than E4FAD mice, and were comparabile to E2FAD mice. |
E4FAD
Observed
-
Plaques at 17
Plaques develop in the subiculum and deep cortical layers by 4 months.
-
Gliosis at 26
Microgliosis and astrocytosis in the subiculum and cortex at 6 months.
-
Synaptic Loss at 17
Decreased protein levels of PSD95 and NMDA receptor subunits by 4 months.
-
Cognitive Impairment at 8
Modest learning deficits in the Morris water maze by 2 months. Progressive decrease in performance on learning and memory tasks.
Absent
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE, APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | APOE: Knock-In; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques starting at 4 months and increasing with age. Gliosis and loss of synaptic proteins. |
Age-dependent learning and memory deficits in the Y maze and Morris water maze. |
hAbeta/APOE4/Trem2*R47H (LOAD2)
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE, APP, Trem2 | TREM2 R47H | APOE: Knock-In; APP: Knock-In; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
hAbeta-loxP-KI
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
hAβ-KI
Observed
-
Synaptic Loss at 78
Fewer synaptophysin-immunoreactive puncta, but similar numbers of PSD95-immunoreactive puncta, in knock-in mice compared with wild-type mice.
-
Changes in LTP/LTD at 78
Impaired theta-burst-induced LTP at Schaffer collateral-CA1 synapses, by 18 months of age.
-
Cognitive Impairment at 40
Differed from wild-type mice in the contextual fear conditioning test by 10 months of age and in the novel object recognition task by 14 months.
Absent
-
Plaques at
No plaques observed through 22 months of age, using immunohistochemical, thioflavin-S or Congo red stains.
-
Neuronal Loss at
Neuron numbers in hippocampal CA1 were similar in 22-month hAβ-KI and wild-type mice, although hippocampal volume was decreased in the knock-in mice.
-
Gliosis at
Neither microgliosis nor astrogliosis was observed through 22 months of age.
No Data
-
Tangles at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
App | App: Knock-In | Alzheimer's Disease | No amyloid plaques observed through 22 months of age. Accelerated formation of OC-immunoreactive and Periodic Acid Schiff-positive granules. |
Differed from wild-type mice in the contextual fear conditioning test by 10 months of age and in the novel object recognition task by 14 months. |
hCR1 KI on APOE4/Trem2
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Cr2, CR1, CR2, APOE, Trem2 | TREM2 R47H | Cr2: Knock-Out; CR1: Knock-In; CR2: Knock-In; APOE: Knock-In; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
htau
Observed
-
Tangles at 39
Aggregated tau and paired helical filaments detectable at nine months by immunoelectron microscopy, although paired helical filaments of aggregated insoluble tau can be isolated from brain tissue as early as two months. Tau first redistributes from axons to cell bodies. Hyperphosphorylated tau begins to accumulate by six months, and increases further by 13 and 15 months (Andorfer et al., 2003).
-
Neuronal Loss at 43
Decrease in cortical thickness and reduced cell number between 10 and 14 months of age. Increased ventricle size increased from age eight months to 18 months. Decrease in the thickness of the corpus callosum (Andorfer et al., 2005).
-
Changes in LTP/LTD at 52
In hippocampal slices, LTP induced by high frequency stimulation (HFS) was normal at four months but abolished by 12 months. LTP induced by theta burst stimulation (TBS) was normal at both ages. Paired-pulse ratio (PPR) was unaffected at four months, but increased at 12 months compared with controls, suggesting a decrease in probability of transmitter release (Polydoro et al., 2009).
-
Cognitive Impairment at 26
Abnormal spatial learning in six-month-old mice compared with control mice (Phillips et al., 2011). Normal object recognition and spatial learning and memory by MWM at four months, but deficits by 12 months (Polydoro et al., 2009). Impaired burrowing relative to control mice occurs by four months (Geiszler et al., 2016).
Absent
No Data
-
Gliosis at
Unknown.
-
Synaptic Loss at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT: Knock-Out; MAPT: Transgenic | Alzheimer's Disease, Frontotemporal Dementia | Age-associated tau pathology, including redistribution of tau to cell bodies and dendrites, phosphorylated tau, accumulation of aggregated paired helical filaments, and ultimately thioflavin-S positive neurofibrillary tangles. Pathology most severe in neocortex and hippocampus, and minimal in the brain stem and spinal cord. Some neuronal loss. |
Normal object-recognition memory and spatial learning/memory (as assessed by the Morris Water Maze) at four months, but impaired at 12 months (Polydoro et al., 2009). |
hTau-A152T
Observed
-
Neuronal Loss at 87
Neuron loss in the hippocampus was observed by 20 months.
-
Gliosis at 17
Astrocytosis, but no differences in microglia.
-
Cognitive Impairment at 74
In the Morris water maze, performance was impaired after 17 months of age. Nest building was impaired at 10-14 months. Social interaction, anxiety, exploratory behavior, and motor functions were unaltered.
Absent
-
Tangles at
Abnormal accumulations of soluble tau were observed, but not tangles or tangle-like structures.
-
Changes in LTP/LTD at
Unchanged at 20 months.
No Data
-
Plaques at
No data.
-
Synaptic Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT A152T | MAPT: Transgenic | Alzheimer's Disease, Frontotemporal Dementia, Other Tauopathy | Tangles or dense tau inclusions not observed. Abnormal accumulations of soluble tau. Age-dependent neuronal loss was observed in the hippocampus. |
Age-dependent learning and memory deficits in the Morris water maze. Nest building impaired. Social interaction, anxiety, exploratory behavior, and motor functions were normal. |
hTau-AT (hTau40-AT)
Observed
-
Tangles at 13
Tangles in hippocampus, cortex, and spinal cord starting at 3 months with age-dependent increases. Hyperphosphorylation, conformation changes, and mislocalization.
-
Neuronal Loss at 52
Neuron loss in the hippocampus and cortex at 12 months.
-
Gliosis at 43
Astrocytosis and microgliosis at 10 months.
-
Synaptic Loss at 87
Synaptophysin, but not PSD95, decreased in hippocampus and cortex at 12 months. By Golgi staining, spines unchanged in CA1 at 10 months, increased in CA3 at 12 months, and decreased in CA1 and CA3 at 16 months.
-
Cognitive Impairment at 70
No change at 10 months but at 16 months deficits in learning and memory (Morris water maze).
Absent
-
Changes in LTP/LTD at
Unchanged at 12 months.
No Data
-
Plaques at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT A152T | MAPT: Knock-In | Alzheimer's Disease, Frontotemporal Dementia, Other Tauopathy | Tangles in hippocampus, cortex, and spinal cord at 3 months with age-dependent increases. Tau hyperphosphorylation, conformation changes, and mislocalization observed. Age-dependent loss of synapses. |
Age-dependent learning and memory deficits in the Morris water maze. Motor functions normal. |
hTau.P301S
Observed
-
Tangles at 17
Neurofibrillary tangles detected as early as 4 months of age.
-
Neuronal Loss at 13
Neuronal loss starting at 3 months. Loss is especially prominent in the spinal cord with notable loss of superficial cortical neurons as well (Hampton et al., 2010).
-
Gliosis at 22
Astrocytosis, as measured by GFAP reactivity, in 6 month-old animals. Microglial activation in the brain stem and spinal cord of 5 month-old animals by OX42 staining (Bellucci et al., 2004).
-
Cognitive Impairment at 11
Memory deficit starting at 2.5 months as assessed by the Morris water maze (Xu et al., 2014), but no deficit at 2 months (Scattoni et al., 2010).
Absent
-
Plaques at
Absent.
No Data
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT P301S | MAPT: Transgenic | Alzheimer's Disease, Frontotemporal Dementia, Other Tauopathy | Age-dependent hyperphosphorylation of tau and conformational changes leading to neurofibrillary tanglelike pathology in the cerebral cortex, hippocampus, brain stem, and spinal cord. Neurodegeneration, especially in the spinal cord, accompanied by astrocytosis. |
Early motor impairment, including abnormal clasping and rotarod deficit at 4 months, with nearly complete deficit at 5 months. Deficits progress to severe paraparesis. Disinhibition and hyperactivity at 2 to 3 months. |
hTREM2-KI
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
TREM2 | TREM2: Knock-In | Alzheimer's Disease |
hTREM2-R47H_KI
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
TREM2 | TREM2 R47H | TREM2: Knock-In | Alzheimer's Disease |
Il1rap KO/APOE4/Trem2*R47H
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE, Il1rap, Trem2 | TREM2 R47H | APOE: Knock-In; Il1rap: Knock-Out; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
J20 (PDGF-APPSw,Ind)
Observed
-
Plaques at 22
At 5-7 months of age diffuse amyloid-β plaques deposit in the dentate gyrus and neocortex. Amyloid deposition is progressive with widespread plaques by 8-10 months. Aβ puncta are deposited in the hippocampus as early as 1 month (Hong et al., 2016).
-
Neuronal Loss at 12
Cell loss varies by brain region. No significant neuronal loss was observed in the CA3 region of the hippocampus at 6, 12, 24 and 36 weeks of age nor in the CA1 region at 6 weeks; however, at 12, 24, and 36 weeks significant neuronal loss was observed in the CA1 region compared to age-matched wild-type animals (Wright et al., 2013).
-
Gliosis at 24
At 24 and 36 weeks a significant increase in the number of reactive GFAP+ astrocytes and CD68+ microglia was observed in the hippocampi of J20 mice compared to age-matched wild-type controls. No significant difference was observed at 6 and 12 weeks (Wright et al., 2013).
-
Synaptic Loss at 15
Age-dependent loss of synaptophysin, synaptotagmin, PSD-95, and homer immunoreactivity in the hippocampus by 3 months; synapse loss was confirmed by electron microscopy. No significant difference was seen at 1 month (Hong et al., 2016).
-
Changes in LTP/LTD at 13
Basal synaptic transmission is impaired between 3-6 months; extracellularly recorded field EPSPs at the Schaffer collateral to CA1 synapse in acute hippocampal slices were on average smaller in amplitude than those seen in wild-type mice. Significant deficits in LTP at the Schaffer collateral–CA1 synapse compared with control mice at 3-6 months (Saganich et al., 2006).
-
Cognitive Impairment at 16
Deficits in spatial memory and learning appear as the mice age. By 4 months, J20 mice demonstrate spatial reference memory deficits as measured by the radial arm maze (Wright et al., 2013) and Morris water maze (Cheng et al., 2007).
Absent
-
Tangles at
Absent.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP V717F (Indiana) | APP: Transgenic | Alzheimer's Disease | Age-dependent formation of Aβ plaques. Dystrophic neurites associated with plaques. No tangles. Variable cell loss. Decrease in synaptic markers and increase in complement immunoreactvity. |
Learning and memory deficits are age-dependent and may appear as early as 16 weeks. Hyperactivity and increased time in the open arm of the elevated plus maze than wild-type mice indicating lower levels of anxiety, but has not been universally replicated. |
JNPL3(P301L)
Observed
-
Tangles at 20
Neurofibrillary tangles develop in an age and gene-dose dependent manner; as early as 4.5 months in homozygotes and 6.5 months in heterozygotes. Tangles and Pick-body-like neuronal inclusions in the amygdala, septal nuclei, preoptic nuclei, hypothalamus, midbrain, pons, medulla, deep cerebellar nuclei and spinal cord (Lewis et al., 2000).
-
Neuronal Loss at 43
Neuronal loss, especially in the spinal cord, most prominent in the anterior horn (Lewis et al., 2000).
-
Gliosis at 43
Astrogliosis (as measured by GFAP reactivity) in brainstem, diencephalon, and basal telencephalon by 10 months (Lewis et al., 2000).
Absent
-
Plaques at
Absent.
No Data
-
Cognitive Impairment at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT P301L | MAPT: Transgenic | Frontotemporal Dementia, Progressive Supranuclear Palsy, Alzheimer's Disease | Age and gene-dose dependent development of neurofibrillary tangles as early as 4.5 months in homozygotes and 6.5 months in heterozyotes. Tangles and Pick-body-like inclusions in the amygdala, hypothalamus, pons, medulla, and spinal cord among other areas. Neuronal loss, especially in the spinal cord. |
By 10 months, 90% developed motor and behavioral disturbances including limb weakness, hunched posture, decrease in grooming and vocalization. |
Kif21b*T82T/APOE4/Trem2*R47H
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Kif21b, APOE, Trem2 | TREM2 R47H | Kif21b: Knock-In; APOE: Knock-In; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
MAPT 10IVS+16 C>T
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT, MAPT-AS1, Mapt | MAPT IVS10+16 C>T | MAPT: Knock-In; MAPT-AS1: Knock-In; Mapt: Knock-Out | Frontotemporal Dementia | Unknown. |
Unknown. |
MAPT(H1.0)-GR
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT, MAPT-AS1, Mapt | MAPT: Knock-In; MAPT-AS1: Knock-In; Mapt: Knock-Out | Alzheimer's Disease, Frontotemporal Dementia, Other Tauopathy | Unknown. |
Unknown. |
MAPT(H1.0*N279K)-GR
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT, MAPT-AS1, Mapt | MAPT N279K | MAPT: Knock-In; MAPT-AS1: Knock-In; Mapt: Knock-Out | Frontotemporal Dementia | Unknown. |
Unknown. |
MAPT(H1.0*)P301L-GR
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT, MAPT-AS1, Mapt | MAPT P301L | MAPT: Knock-In; MAPT-AS1: Knock-In; Mapt: Knock-Out | Frontotemporal Dementia | Unknown. |
No overt behavioral phenotypes |
MAPT(H2.1)-GR
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT, MAPT-AS1, Mapt | MAPT: Knock-In; MAPT-AS1: Knock-In; Mapt: Knock-Out | Alzheimer's Disease, Frontotemporal Dementia, Other Tauopathy | Unknown. |
Unknown. |
MAPT knock-in
Observed
Absent
-
Plaques at
No amyloid plaques at 24 months of age.
-
Tangles at
No neurofibrillary tangles at 24 months of age.
-
Neuronal Loss at
Neurodegeneration not apparent up to 2 years of age.
-
Gliosis at
No astrogliosis or microgliosis observed at 24 months.
-
Cognitive Impairment at
At 12 months of age, MAPT knock-in mice perform similarly to wild-type mice in the Y-maze test of working memory (only males tested).
No Data
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT: Knock-In | Alzheimer's Disease, Other Tauopathy | No evidence of increased neuroinflammation, neuronal death, or brain atrophy in MAPT knock-in mice, compared with wild-type mice. |
MAPT knock-in mice perform similarly to wild-type mice in the Y-maze test of working memory. |
McGill-R-Thy1-APP
Observed
-
Plaques at 24
Amyloid plaques present in homozygotes, appearing in hippocampus at 6 months and cortex at 13 months. Plaques are generally absent in hemizygotes.
-
Neuronal Loss at 72
A 22 percent reduction in the number of neurons was seen in the subiculum of homozygous transgenic rats at 18 months.
-
Gliosis at 24
Microgliosis and astrogliosis observed in homozyogotes.
-
Synaptic Loss at 80
Reduction in cholinergic synaptic boutons seen at 20 months in homozygous transgenic rats.
-
Changes in LTP/LTD at 14
Impairments in long-term potentiation in CA1 by 3.5 months of age.
-
Cognitive Impairment at 12
Deficits in Morris water maze and fear conditioning test are apparent by 3 months of age in both hemizygous and homozygous transgenic rats.
Absent
No Data
-
Tangles at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP V717F (Indiana) | APP: Transgenic | Alzheimer's Disease | Hemizygotes: intraneuronal Aβ in hippocampus and cortex by one week, but no plaques even at advanced ages. Homozygotes: intraneuronal Aβ in hippocampus and cortex by one week; amyloid plaques in hippocampus beginning at 6 months, in cortex beginning at 13 months. |
Cognitive deficits are apparent by three months of age in both hemizygous and homozygous transgenic rats. |
Mthfr*C677T/APOE4/Trem2*R47H
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Mthfr, APOE, Trem2 | TREM2 R47H | Mthfr: Knock-In; APOE: Knock-In; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
mThy-1 3R Tau (line 13)
Observed
-
Tangles at 34
Pick-body like inclusions of aggregated tau appeared in the hippocampus and cortex by 8-10 months. Inclusions were positive for Bielchowsky silver stain but negative for Gallyas-silver stain and Thioflavin-S.
-
Neuronal Loss at 34
Neuronal loss occurred by 8-10 months as evidenced by decreased NeuN staining in the dentate gyrus and CA3 regions of the hippocampus. Neocortical volume also decreased.
-
Gliosis at 35
Astrogliosis was seen by 8-10 months in the neocortex and hippocampus. Some GFAP+ astrocytes also contained 3R tau.
-
Cognitive Impairment at 26
By 6-8 months memory impairment was evident as a failure to habituate to a novel environment. This deficit was not present at 3-4 months. At 8-10 months, transgenics also took longer than wild-type mice to find the hidden platform in the Morris water maze.
Absent
-
Plaques at
Absent.
No Data
-
Synaptic Loss at
Synapto-dendritic damage manifested as reduced dendritic density, reduced MAP2 immunoreactivity, and accumulation of 3R tau in dendrites.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT L266V, MAPT G272V | MAPT: Transgenic | Frontotemporal Dementia, Pick's disease, Alzheimer's Disease | Accumulation of 3R tau in neurons of the cortex and hippocampus. Pick body-like tau aggregates and neuronal loss in the hippocampus and cortex. Astrogliosis, with some 3R tau in GFAP-positive astrocytes. Synapto-dendritic changes and mitochondrial pathology. |
Age-related memory and motor deficits as assessed by habituation to a novel environment, the Morris water maze, and the round beam test. |
mThy1-hAPP751 (TASD41)
Observed
-
Plaques at 13
Plaques start at 3-6 months in the frontal cortex and become widespread with age, affecting the piriform and olfactory cortices, hippocampus, and thalamus (Rockenstein et al., 2001; Havas et al., 2011).
-
Gliosis at 27
Inflammation related to activated microglia (increased CD11) and reactive astrocytes (increased GFAP) is significant by 6 months and increases with age.
-
Synaptic Loss at 52
Dystrophic neurites and synaptic loss starting at 12 months.
-
Cognitive Impairment at 26
Cognitive impairment observed by 6 months by Morris Water Maze (Rockenstein et al., 2005).
Absent
-
Tangles at
Absent.
-
Neuronal Loss at
Absent.
No Data
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP V717I (London) | APP: Transgenic | Alzheimer's Disease | Age-dependent increases in Aβ40 and Aβ42, with Aβ42 > Aβ40. Plaques at an early age, starting at 3-6 months in the frontal cortex. At 5-7 months, size and number of plaques increased in the frontal cortex, and dense amyloid deposits appear in hippocampous, thalamus, and olfactory region. |
Age-associated impairment in spatial memory and learning in the water maze task and habituation in the hole-board task, with significant deficits at 6 months of age. Some gender-specific differences in open field exploration. |
NSE-APP751
Observed
-
Plaques at 8
Aβ deposits were observed as early as two months of age. These deposits were diffuse and extracellular and had a “cotton-like” appearance. Classic mature plaques were not observed.
-
Gliosis at 95
Gliosis was noted in a single 22-month-old animal with extensive Aβ deposits (Higgins et al., 1994).
-
Cognitive Impairment at 52
Deficits in spatial memory and learning appear as the mice age. At 12 months the mice demonstrate learning and memory deficits as measured by a water-maze task and in spontaneous alternation in a Y maze (Moran et al., 1995). At six months cognition is largely normal.
Absent
-
Tangles at
Classic tangles were not observed, but aberrant tau immunoreactivity was observed as early as two months.
-
Neuronal Loss at
Cell death was not formally assessed, however, overt neuronal death was not seen.
No Data
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP: Transgenic | Alzheimer's Disease | Age-dependent increase in Aβ deposits and tau immunoreactivity. |
Learning and memory deficits are age-dependent as assessed on spontaneous alternation in a Y maze and in the water-maze task. |
PA-Rab5
Observed
-
Neuronal Loss at 28
Loss of basal forebrain cholinergic neurons, beginning at 7 months.
-
Synaptic Loss at 34
Loss of spines in CA3 and dentate gyrus regions of the hippocampus, observed in 8.5-month-old mice.
-
Changes in LTP/LTD at 24
Pronounced defect in LTD and slight impairment in LTD at Schaffer collateral-CA1 synapses in hippocampal slices from 6-month-old mice.
-
Cognitive Impairment at 24
When tested at 6 months of age, the performance of PA-Rab5 mice differed from wild-type controls in a novel object recognition test.
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Gliosis at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
RAB5A | RAB5A: Transgenic | Alzheimer's Disease | Enlarged Rab5-positive endosomes, tau hyperphosphorylation, synapse loss in hippocampus, and loss of basal forebrain cholinergic neurons. |
When tested at 6 months of age, the performance of PA-Rab5 mice differed from wild-type controls in a novel object recognition test. |
PDAPP(line109)
Observed
-
Plaques at 26
In heterozygous mice no plaque pathology at 4-6 months. At 6-9 months mice begin to exhibit deposits of human Aβ in the hippocampus, corpus callosum, and cerebral cortex. Plaques become more extensive with age and vary in size and structure including diffuse irregular plaques and compact cored plaques (Games et al., 1995).
-
Gliosis at 26
GFAP-positive astrocytes and activated microglia associated with plaques (Games et al., 1995).
-
Synaptic Loss at 35
Decreased synaptic density in the dentate gyrus as measured by synaptophysin immunoreactivity. Also decreased dendritic density as measured by MAP2 immunoreactivity (Games et al., 1995).
-
Changes in LTP/LTD at 17
Alterations in LTP induced by theta burst stimulation at 4-5 months which is prior to plaque formation; although the potentiation immediately after TBS was comparable to control mice, the potentiation decayed more rapidly in PDAPP mice. Also paired pulse facilitation was enhanced. Responses to high frequency stimulation bursts were distorted (Larson et al., 1999).
-
Cognitive Impairment at 13
Deficits in a variety of memory paradigms from a young age. Robust deficits in the radial arm maze at 3 months (deficits appear before amyloid plaque deposits). Object recognition, 6, 9-10 months. Operant learning, 3, 6 months (Dodart et al., 1999).
Absent
-
Tangles at
No paired helical filaments or aggregates, but phosphorylated tau immunoreactivity is observed in dystrophic neurites after 14 months (Masliah et al., 2001).
-
Neuronal Loss at
Absent.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP V717F (Indiana) | APP: Transgenic | Alzheimer's Disease | Amyloid plaques in the hippocampus, cerebral cortex. Gliosis. Dystrophic neurites. Decreased synaptic and dendritic density in the hippocampus. |
Deficits in a variety of memory paradigms from a young age. Deficits in the radial arm maze at 3 months (before plaques), object recognition, operant learning, spatial reference memory (starting at 3-4 months), cued fear conditioning at 11 months. |
PDGF-APP(WT) (line I5)
Observed
-
Synaptic Loss at 9
By 2-4 months of age, there is a decrease in synaptophysin-immunoreactive presynaptic terminals compared to nonTg controls. Synaptophysin immunoreactivity decreases further with age.
Absent
-
Tangles at
Not observed.
-
Neuronal Loss at
Not observed.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP: Transgenic | Alzheimer's Disease | Expression of human APP in the brain especially in the neocortex and hippocampus. No plaques up to 24 months. |
Unknown. |
PLB1-triple (hAPP/hTau/hPS1)
Observed
-
Gliosis at 52
Increased inflammation (GFAP labelling) detected at 12 months in cortex and hippocampus (Platt, unpublished observation).
-
Changes in LTP/LTD at 26
Impairments in long-term and short-term hippocampal plasticity. LTP following theta-burst stimulation decayed faster and paired-pulse facilitation was reduced relative to wild-type mice at both six and 12 months of age. Synaptic transmission impacted at 12 months.
-
Cognitive Impairment at 22
Social recognition memory was impaired by five months and further impaired by 12 months. Similarly, object recognition memory was impaired by eight months. Spatial learning impairments were seen later; at 12 months deficits in spatial acquisition learning were seen in the open field water maze that were not apparent at 5 months.
Absent
-
Plaques at
Sparse plaques out to 21 months of age. Only marginally increased compared with wild-types and overall very low compared to over-expression models. However, Aβ accumulated intracellularly and also formed oligomers.
-
Tangles at
No overt tangle pathology; however, hyyperphosphorylated tau accumulated in the hippocampus and cortex from six months of age.
-
Neuronal Loss at
Absent.
No Data
-
Synaptic Loss at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, MAPT, PSEN1 | APP V717I (London), APP K670_M671delinsNL (Swedish), PSEN1 A246E, MAPT P301L, MAPT R406W | APP: Multi-transgene; MAPT: Multi-transgene; PSEN1: Multi-transgene | Alzheimer's Disease | Age-related neuropathology including intraneuronal and oligomeric Aβ accumulation and hyperphosphorylated tau in the hippocampus and cortex from six months. Minimal amyloid plaques up to 21 months. Subtle tau pathology, but no overt tangles. Cortical hypometabolism with increased metabolic activity in basal forebrain and ventral midbrain by FDG-PET/CT. |
Cognitive deficits in recognition memory and spatial learning emerging between five and 12 months. Impairments in hippocampal plasticity. |
PLB4 (hBACE1)
Observed
-
Gliosis at 52
Increased GFAP-positive astrocytes at 12 months of age in the dentate gyrus, CA1 region of the hippocampus, and the piriform cortex. Gliosis is suspected to begin earlier than 12 months.
-
Cognitive Impairment at 13
Impaired spatial representation in a habituation task by 3 months of age. By 6 months, impaired learning and memory by a variety of tasks including the Y-maze, Morris water maze, and a test of the social transmission of food preference. These effects appear to be distinct from reduced motor activity and reduced anxiety.
Absent
-
Plaques at
Plaques virtually absent, minimal small sparse plaques. However, prominent extracellular Aβ staining surrounding neuronal cell bodies, including Aβ multimers (e.g. Aβ*56 and Aβ hexamers).
-
Tangles at
Preliminary analysis did not find abnormal phosphorylation or conformational changes in tau.
No Data
-
Neuronal Loss at
Unknown.
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
BACE1 | BACE1: Transgenic | Alzheimer's Disease | Elevated extracellular multimeric Aβ, including Aβ*56 and Aβ hexamers, in the absence of plaques. At 12 months of age, astrogliosis was observed in a region- and genotype-dependent manner, especially in the dentate gyrus, hippocampal CA1, and piriform cortex. No overt tau pathology. |
Largely intact motor coordination and gait (Rotarod, CatWalk). Age-associated changes in multiple measures of learning and memory. Early deficits in habituation to a novel environment and semantic-like memory (three-four months). Impaired spatial learning and long-term reference (Morris water maze) and working memory (Y-maze) at six months, distinct from reduced locomotor activity and anxiety. |
Plcγ2-P522R knock-in
Observed
-
Gliosis at 24
Astrogliosis revealed by GFAP immunohistochemistry in 6-month-old males. Microglial activation revealed by TSPO PET imaging in year-old females.
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Plcg2 | Plcg2: Knock-In | Alzheimer's Disease, Dementia with Lewy Bodies, Frontotemporal Dementia | Hypertrophic astrocytes in the hippocampi, revealed by GFAP immunohistochemistry. Microglial activation revealed by TSPO PET imaging. |
Unknown. |
Plcg2 KO
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Plcg2 | Plcg2: Knock-Out | Alzheimer's Disease | Unknown. |
Unknown. |
Plcg2*M28L/APOE4/Trem2*R47H
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE, Plcg2, Trem2 | TREM2 R47H | APOE: Knock-In; Plcg2: Knock-In; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
Plcg2*M28L x 5xFAD
Observed
-
Plaques at 30
Diffuse and compact amyloid plaques observed in mice studied at 7.5 months of age. Higher plaque burdens than 5xFAD.
-
Gliosis at 31
Microgliosis observed in mice studied at 7.5 months of age.
-
Synaptic Loss at 32
Decreased basal synaptic transmission, lower frequencies and amplitudes of spontaneous excitatory postsynaptic currents and spontaneous inhibitory postsynaptic currents recorded in hippocampal CA1 region, compared with wild-type mice.
-
Changes in LTP/LTD at 33
Impaired LTP at Schaffer collateral-CA1 synapses, compared with wild-type.
-
Cognitive Impairment at 24
Deficits in working memory (decreased spontaneous alternation in the Y-maze), compared with wild-type.
Absent
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Plcg2, APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | Plcg2: Knock-In; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Plaque burdens in the cortex and subiculum were elevated in 5xFADM28L mice but microglia showed less interaction with plaques, compared with 5xFAD. |
Six-month-old 5xFADM28L and 5xFAD mice showed similar deficits in working memory, assessed in the Y-maze. |
Plcg2*P522R
Observed
-
Gliosis at 24
Plcg2*P522R knock-in mice had a slightly higher density of Iba1-positive microglia than wild-type mice. Microglia in the knock-in animals were simpler in shape—with less ramified processes—and contained a greater density of puncta immunoreactive for the lysosomal marker CD68, compared with wild-type microglia.
Absent
-
Synaptic Loss at
Synapse number in hippocampal CA1—assessed as the density of puncta immunoreactive for the presynaptic marker bassoon or the postsynaptic marker PSD95—did not differ between Plcg2*P522R and wild-type mice. However, a slight decrease in the number of thin spines was observed in mutation carriers, while numbers of stubby and mushroom spines did not differ between the genotypes.
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Plcg2 | Plcg2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
Plcg2*P522R x 5xFAD
Observed
-
Plaques at 30
Diffuse and compact amyloid plaques observed in mice studied at 7.5 months of age. Lower plaque burdens than 5xFAD.
-
Gliosis at 31
Microgliosis observed in mice studied at 7.5 months of age.
Absent
-
Synaptic Loss at
No deficits in synaptic transmission—including basal synaptic transmission, frequencies and amplitudes of spontaneous excitatory postsynaptic currents and spontaneous inhibitory postsynaptic currents, and AMPA/NMDA current ratios—recorded in hippocampal CA1 region of 7.5-month-old mice.
-
Changes in LTP/LTD at
Normal LTP at Schaffer collateral-CA1 synapses at 7.5 months of age.
-
Cognitive Impairment at
Normal working memory (spontaneous alternation in the Y-maze) at 6 months of age.
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Plcg2, APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | Plcg2: Knock-In; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Plaque burdens in the cortex and subiculum were lower in 5xFADP522R mice and microglia showed increased interaction with plaques, compared with 5xFAD. |
The PLCγ2 P522R variant protected against deficits in the Y-maze test of working memory in 5xFAD mice. |
Plcg2*P522R x APP NL-G-F
Observed
-
Plaques at 24
ThioflavinS-positive amyloid plaques observed in mice studied at 6 months of age. Higher plaque burdens than APPNL-G-F.
-
Gliosis at 24
Microgliosis observed in mice studied at 6 months of age. Attenuated microglia-plaque interactions in the hippocampus, compared with APPNL-G-F.
Absent
-
Synaptic Loss at
The P522R variant attenuated the synapse loss observed in APPNL-G-F mice with wild-type PLCγ2.
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Plcg2, App | APP K670_M671delinsNL (Swedish), APP I716F (Iberian), APP E693G (Arctic) | Plcg2: Knock-In; App: Knock-In | Alzheimer's Disease | Sex- and region-dependent increases in plaque burden, and decreases in microglia-plaque interactions, in Plcg2*P552R x APPNL-G-F mice, compared with APPNL-G-F. |
Unknown. |
PS19 with humanized TREM2 (common variant)
Observed
-
Tangles at 37
Tangles revealed using antibody PG5 at 9 months.
-
Neuronal Loss at 38
At 9 months, atrophy of hippocampus and entorhinal/piriform cortex and pronounced ventricular expansion. Thinning of the granule cell layer of the dentate gyrus and pyramidal cell layer of the piriform cortex, compared with PS19 mice carrying TREM2-R47H.
-
Gliosis at 39
Elevated expression of markers of astroglial and microglial reactivity, compared with PS19 mice carrying the R47H variant of TREM2.
-
Synaptic Loss at 40
Fewer synapses and more dystrophic synapses, compared with PS19 mice carrying the R47H variant of TREM2.
Absent
No Data
-
Plaques at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT, TREM2, Trem2 | MAPT P301S | MAPT: Transgenic; TREM2: Transgenic; Trem2: Knock-Out | Alzheimer's Disease, Frontotemporal Dementia | Brain atrophy by 9 months of age. Increased microgliosis, astrogliosis and synapse loss, compared with PS19 mice carrying TREM2 with the R47H mutation. |
Not known. |
PS19 with humanized TREM2 (R47H)
Observed
-
Tangles at 36
Tangles revealed using antibody PG5 at 9 months.
Absent
No Data
-
Plaques at
No data.
-
Neuronal Loss at
No data relative to wild-type mice, but at 9 months of age, the volumes of the hippocampus and entorhinal/piriform cortex are larger, and the granule cell layer of the dentate gyrus and pyramidal cell layer of the piriform cortex are thicker, in PS19-TREM2R47H mice, compared with PS19 mice carrying the common variant of human TREM2.
-
Gliosis at
At 9 months of age, decreased expression of markers of astroglial and microglial reactivity, compared with PS19 mice carrying the common variant of TREM2, but no data relative to wild-type mice.
-
Synaptic Loss at
At 9 months of age, more synapses and fewer dystrophic synapses, compared with PS19 mice carrying the common variant of TREM2, but no data relative to wild-type mice.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT, TREM2, Trem2 | MAPT P301S, TREM2 R47H | MAPT: Transgenic; TREM2: Transgenic; Trem2: Knock-Out | Alzheimer's Disease, Frontotemporal Dementia | Decreased brain atrophy, microgliosis, astrogliosis, and synapse loss, compared with PS19 mice carrying the common variant of TREM2. |
Not known. |
PS1 conditional Knock-out
Observed
-
Cognitive Impairment at 22
Mild impairment of spatial learning and memory in the Morris water maze observed in 5 month-old mice (Yu et al., 2001).
Absent
-
Plaques at
Absent.
-
Tangles at
Absent.
-
Neuronal Loss at
Absent.
-
Changes in LTP/LTD at
Mice at 3-6 months of age exhibit normal paired-pulse facilitation, LTP, and LTD in the Schaffer collateral pathway of the hippocampus (Yu et al., 2001).
No Data
-
Gliosis at
Unknown.
-
Synaptic Loss at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
PSEN1 | PSEN1: Conditional Knock-out | Alzheimer's Disease | Reduction in Aβ40 and Aβ42 peptides; accumulation of APP C-terminal fragments. |
Subtle but significant deficits in long-term spatial memory in the Morris water maze. |
PS2APP
Observed
-
Plaques at 26
Age-associated development of plaques: none at 3 months, overt Aβ deposition at approximately 6 months, with heavy plaque load in the hippocampus, frontal cortex, and subiculum at 10 months (Ozmen et al., 2009; Weidensteiner et al. 2009).
-
Gliosis at 26
Gliosis at 6 months (personal communication, Laurence Ozmen).
-
Changes in LTP/LTD at 43
A strong increase in LTP and post-tetanic potentiation induced by tetanic stimulation in hippocampal slices of 10 month-old animals compared to wild-type mice (Poirier et al., 2010).
-
Cognitive Impairment at 35
Cognitive impairment is detected by the Morris water maze (probe trial 2) at 8 and 12 months of age, not at 3 months (personal communication Laurence Ozmen).
Absent
-
Tangles at
Absent.
No Data
-
Neuronal Loss at
Unknown.
-
Synaptic Loss at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN2 | APP K670_M671delinsNL (Swedish), PSEN2 N141I | APP: Transgenic; PSEN2: Transgenic | Alzheimer's Disease | Age-associated development of plaques: none at 3 months, overt Aβ deposition in the brain at approximately 6 months, with heavy plaque load in the hippocampus, frontal cortex, and subiculum at 10 months. Aβ deposits in blood vessels were sporadic, mainly in large vessels. Cerebral amyloid deposits correlate with levels of the human APP transcript at 12 months. |
Cognitive impariment detected by the Morris water maze at 8 and 12 months of age, but not at 3 months. |
PS2APP (PS2(N141I) x APPswe)
Observed
-
Plaques at 39
Rare amyloid deposits at 5 months, with consistent deposits in the subiculum and frontolateral cortices by 9 months. Plaques increase in number and distribution over time, spreading throughout the neocortex and hippocampus as well as the amygdala and thalamic and pontine nuclei (Richards et al., 2003).
-
Gliosis at 39
An inflammatory response indicated by the presence of activated microglia and astrocytes begins around 9 months. The onset, distribution, and abundance of activated microglia and astrocytes correlate with Aβ deposition.
-
Cognitive Impairment at 35
Age-associated cognitive impairment from 8 months with impaired acquisition of spatial learning in the water maze (Richards et al., 2003).
Absent
-
Tangles at
Absent.
-
Changes in LTP/LTD at
No difference in LTP in the dentate gyrus at 3 and 10 months compared to wild-type mice (Richards et al., 2003).
No Data
-
Neuronal Loss at
Unknown.
-
Synaptic Loss at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN2 | APP K670_M671delinsNL (Swedish), PSEN2 N141I | APP: Transgenic; PSEN2: Transgenic | Alzheimer's Disease | Rare amyloid deposits at 5 months, with consistent deposits in the subiculum and frontolateral cortices by 9 months. Plaques increase in number and distribution with time, spreading throughout the neocortex and hippocampus as well as the amygdala and thalamic and pontine nuclei. The distribution and abundance of activated microglia and astrocytes correlate with Aβ deposition. |
Mice develop age-associated cognitive impairment from 8 months with impaired acquisition of spatial learning in the water maze. |
PS/APP
Observed
-
Plaques at 26
Large amounts of Aβ accumulate in the cerebral cortex and hippocampus, starting around 6 months and increasing with age. Other brain regions are affected later. Both diffuse and fibrillar plaques form (Gordon et al., 2002).
-
Neuronal Loss at 79
Neuronal loss in the CA1 region of the hippocampus has been reported at 22 months accompanied by reduced glucose utilization (Sadowski et al., 2004).
-
Gliosis at 26
GFAP-positive astrocytes appear first in the cortex in the vicinity of the developing Aβ deposits. Numbers increase with age, becoming confluent. Numbers of resting microglia (positive for complement receptor-3) increase in the vicinity of deposits at 6 months, but activated microglia (positive for MHC-II) are negligible before 12 months and more variable (Gordon et al., 2002).
-
Cognitive Impairment at 12
Double and single transgenic mice had reduced spontaneous alternation performance in a “Y” maze, a test of spatial memory, at 12-14 weeks, before substantial Aβ deposition (Holcomb et al., 1998). Progressive age-related cognitive impairment is seen later in select tasks (e.g. water maze acquisition and radial arm water maze working memory)(Arendash et al., 2001).
Absent
-
Tangles at
Neurofibrillary tangles are not associated with this model, but hyperphosphorylated tau is detected, starting at 24 weeks, appearing as punctate deposits near amyloid deposits in the cortex and hippocampus (Kurt et al., 2003).
No Data
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1 M146L (A>C) | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Aβ accumulates in the cerebral cortex and hippocampus starting ~6 months and increasing with age. Other regions affected later. Deposition occurs in white matter, cerebrovasculature, and grey matter in the form of diffuse and fibrillar plaques. Fibrillar deposits are associated with dystrophic neurites and GFAP-positive astrocytes at ~ 6 months with later microglial activation. |
Progressive impairment between 5–7 and 15–17 months in some tests of cognitive performance, but not others. No change in anxiety levels. |
PS cDKO
Observed
-
Neuronal Loss at 9
Significant increase (about 8-fold) in apoptotic neurons at 2 months of age, although the total number of cortical neurons is not significantly altered due to the low basal level of apoptosis in the cerebral cortex. By 4 months of age, the cumulative loss of cortical neurons reaches about 9 percent of all cortical neurons.
-
Gliosis at 17
Astrogliosis and microgliosis; up-regulation of GFAP and other inflammatory markers are observed in the neocortex and hippocampus at 6 months, and this increases with age (Wines-Samuelson et al., 2010, Beglopoulos et al., 2004).
-
Synaptic Loss at 26
Reduction in synaptophysin immunoreactivity in hippocampal CA1 pyramidal neurons by 6 months. Reduction in dendritic spines by 9 months (Saura et al., 2004).
-
Cognitive Impairment at 9
Deficits in the Morris water maze and contextual fear conditioning are mild at 2 months, but become more severe with age (Saura et al., 2004).
Absent
-
Plaques at
Absent.
-
Tangles at
Tangles are absent, but hyperphosphorylation of tau has been reported in 9 month-old mice.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
PSEN1, PSEN2 | PSEN1: Conditional Knock-out; PSEN2: Knock-Out | Alzheimer's Disease | At 2 months the number of apoptotic neurons is elevated about 8-fold. By 6 months, about 18 percent of of cortical neurons are lost. Up-regulation of inflammatory markers and progressive astrogliosis and microgliosis in the neocortex and hippocampus. |
Impairments in hippocampal learning and memory as indicated by Morris water maze and contextual fear conditioning evident by 2 months and worsens with age. |
Psen1 L435F knock-in
Observed
Absent
-
Plaques at
No plaques at 15 days of age.
-
Neuronal Loss at
No neuron loss at 15 days of age.
-
Gliosis at
No astrogliosis or microgliosis at 15 days of age.
No Data
-
Tangles at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Psen1, App | PSEN1 L435F | Psen1: Knock-In; App: Knock-In | Alzheimer's Disease | None observed in 15-day-old rats. |
Unknown. |
PWK.APP/PS1
Observed
-
Plaques at 32
Thioflavin S-positive amyloid plaques are present in the cortex and CA1 region of the hippocampus by 8 months of age, with females having more plaques in the cortex than males.
-
Gliosis at 33
Plaque-associated microgliosis observed by 8 months.
Absent
-
Tangles at
Not observed.
-
Neuronal Loss at
Not observed.
-
Cognitive Impairment at
Working memory and short-term memory were intact at 7 to 8 months, as assessed by tests in the Y-maze.
No Data
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1: deltaE9 | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques and plaque-associated gliosis by 8 months. |
Transgenic mice are hyperactive and aggressive. Working memory and short-term memory are intact at 7 to 8 months, as assessed by tests in the Y-maze. |
rTg9191
Observed
-
Plaques at 35
Plaques emerge first in the cerebral cortex, starting around 8 months of age. This is followed by plaques in the hippocampus at 10.5 to 12.5 months of age. Some dense core plaques develop.
-
Neuronal Loss at 9
Expression of the tetracycline transactivator (tTA) resulted in reduced forebrain weight and smaller dentate gyri in rTg9191 mice compared to non-Tg littermates. This effect was also observed in mice expressing tTA alone, and is thought to be a developmental effect, as it was observed even in young mice (e.g., 2-6 months of age).
-
Gliosis at 104
rTg9191 mice develop reactive gliosis (astrocytosis and microgliosis) in the vicinity of dense-core plaques by 24 months of age.
Absent
-
Tangles at
Tangles are not observed, but hyperphosphorylated tau develops with age.
-
Cognitive Impairment at
No transgene-related deficits seen in Morris water maze (4, 12, 21, 24 months of age) or fixed consecutive number test (23 months of age).
No Data
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP V717I (London) | APP: Transgenic | Alzheimer's Disease | Age-associated pathology in the cerebral cortex and hippocampus starting at 8 and 10½-12½ months of age, respectively. Gliosis and hyperphosphorylated tau in the vicinity of dense-core plaques. Fibrillar oligomeric species, e.g., Aβ dimers. |
No transgene-related deficits seen in Morris water maze (4, 12, 21, 24, months of age) or fixed consecutive-number (23 months of age) tests. |
rTgTauEC
Observed
-
Tangles at 78
By 18 months of age, Gallyas silver-positive staining is observed, indicative of paired helical filaments. This is followed by thioflavin-S staining at 24 months. Tau pathology develops first in neurons of the medial EC expressing human tau, followed by neurons in the dentate gyrus, CA1 and CA2/3(de Calignon et al., 2012).
-
Neuronal Loss at 83
Neuronal loss is detectable by 24 months of age in areas with transgene expression (e.g. layer II of the EC and parasubiculum), compared with age-matched mice expressing only tTA. Significant neuronal loss was not observed at 21 months (de Calignon et al., 2012).
-
Gliosis at 104
Microglial activation and astrogliosis by 24 months of age, in conjunction with axonal degeneration and neuronal loss (de Calignon et al., 2012).
-
Synaptic Loss at 104
By 24 months of age pre- and post-synaptic densities were reduced in the middle third of the molecular layer of the dentate gyrus as measured by synapsin-1 and PSD-95 staining (de Calignon et al., 2012).
-
Changes in LTP/LTD at 70
At 16 months of age, subtle differences in electrophysiological properties have been observed in the perforant pathway, including a decrease in LTP and an increase in the probability of neurotransmitter release (Polydoro et al., 2014).
-
Cognitive Impairment at 70
Very mild and specific deficits in contextual fear conditioning at 16 months of age, but no deficits in the radial arm maze (Polydoro et al., 2014).
Absent
-
Plaques at
Absent.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT P301L | MAPT: Transgenic | Frontotemporal Dementia, Alzheimer's Disease | Propagating tau pathology starting in the entorhinal cortex and spreading to regions functionally connected to the EC (e.g., dentate gyrus). Neurodegeneration and axonal degeneration, first in EC and parasubiculum. Gliosis and synaptic loss. |
Subtle cognitive deficit in contextual fear conditioning, but not in the radial arm maze, at 16 months. Mild specific deficit in locomotor activity in the open field test. |
rTg(tauP301L)4510
Observed
-
Tangles at 17
Pretangles as early as 2.5 months. Argyrophilic tangle-like inclusions in cortex by 4 months and in hippocampus by 5.5 months.
-
Neuronal Loss at 24
Decreased (~60%) CA1 hippocampal neurons by 5.5 months with significant loss in brain weight. Progressive loss of neurons and brain weight in 7 and 8.5 month mice with ~23% of CA1 pyramidal cells remaining at 8.5 months. Gross atrophy of the forebrain by 10 months.
-
Synaptic Loss at 35
Significant loss of dendritic spines at 8-9 months (~30% decrease in spine density in somatosensory cortex).
-
Cognitive Impairment at 11
Retention of spatial memory (Morris Water Maze) became impaired from 2.5 to 4 months. No significant motor impairments up to 6 months. Spatial memory improved when transgene suppressed by dox.
Absent
-
Plaques at
Absent.
No Data
-
Changes in LTP/LTD at
LTP at the Schaffer collateral-CA1 synapse is normal at 1.3 months, but impaired at 4.5 months.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT P301L | MAPT: Transgenic | Alzheimer's Disease, Frontotemporal Dementia | Argyrophilic tangle-like inclusions in cortex by 4 months and in hippocampus by 5.5 months. Decreased CA1 neurons (~60 percent) by 5.5 months. Gross forebrain atrophy by 10 months. The number of CA1 neurons stabilized after a brief (six to eight week) suppression of transgenic tau. |
Spatial memory impairments by 2.5 to 4 months. No significant motor impairment up to 6 months of age. When the transgene was suppressed with dox at 2.5 months, spatial memory improved. |
SHR24
Observed
-
Tangles at 38
Argyrophilic neurofibrillary tangles accumulate in cortex, hippocampus, thalamus, and brainstem.
-
Synaptic Loss at 60
Decreased levels of synaptophysin and a decreased number of synaptic vesicles per synapse in animals at the end of the lifespan of this line.
Absent
-
Neuronal Loss at
No neuron loss was observed in the hippocampi or cortices of male rats examined at 15 month of age.
No Data
-
Plaques at
No data.
-
Gliosis at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
Sensorimotor deficits and abnormal reflexes observed as early as 3.5 months, but no data available from cognitive tests.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT: Transgenic | Alzheimer's Disease | Neurofibrillary tangles accumulate in cortex, hippocampus, thalamus, and brainstem, beginning at 9 to 10 months. No neuron loss was observed in the hippocampus or cortex. |
SHR24 rats exhibit age-dependent impairments in several neurobehavioral tests; hind-limb clasping during the tail-hang test is one of the earliest abnormalities to appear, evident by 3.5 months of age. |
SHR318
Observed
-
Tangles at 38
Argyrophilic neurofibrillary tangles are particularly prominent in the brainstem and spinal cord.
-
Cognitive Impairment at 18
At 4.5 months, rats show normal learning, but deficits in spatial memory, in the Morris water maze.
Absent
-
Neuronal Loss at
Neuron numbers in the hippocampi and brainstem gigantocellular reticular nucleus do not differ between 10.5-month SHR318 rats and non-transgenic rats.
No Data
-
Plaques at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT: Transgenic | Alzheimer's Disease | Neurofibrillary tangles first appear at 9 months and are particularly prominent in the brainstem and spinal cord. Axonal degeneration is observed in the brainstem and spinal cord of 10- to 12-month animals. |
At 4.5 months, rats show normal learning, but deficits in spatial memory, in the Morris water maze. Reflexes and sensorimotor coordination are impaired at 7 months. |
SHR72
Observed
-
Tangles at 30
Neurofibrillary tangles, demonstrated by Gallyas silver stain, are present in the brainstem and spinal cord.
-
Gliosis at 29
Astrogliosis and microgliosis are present in brainstem regions bearing neurofibrillary tangles.
Absent
-
Neuronal Loss at
Although neuron loss has not been documented, chromatolytic neurons and damaged axons were seen in the brains of 7-month animals, particularly in the brainstem reticular formation.
No Data
-
Plaques at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
Sensorimotor deficits and abnormal reflexes observed as early as 3 months, but no data available from cognitive tests.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT: Transgenic | Alzheimer's Disease | Neurofibrillary tangles, demonstrated by Gallyas silver stain, were found the brainstems and spinal cords of terminal stage (7- to 8-month old) animals. Chromatolytic neurons and damaged axons were also observed at this stage. |
Sensorimotor deficits and loss of muscle strength are apparent at 3 months. This stage lasted about three months, and then rats experienced a rapid, dramatic decline in neurological function, succumbing within several days. |
SHRSP/FAD
Observed
-
Plaques at 69
Diffuse amyloid plaques observed at 16-18 months, the only age examined to date.
-
Tangles at 70
Occasional neurons appear to contain globose neurofibrillary tangles, as revealed by immunostaining using an antibody directed against tau phosphorylated at serine 422, an epitope found in paired helical filaments.
-
Gliosis at 71
Hypertrophied microglia and elevated levels of GFAP observed at 16-18 months, the only age examined to date.
-
Cognitive Impairment at 72
Working memory deficits as assessed by novel object recognition, but not as assessed by spontaneous alternation in the Y-maze, at 16-18 months, the only age examined to date.
Absent
-
Synaptic Loss at
Levels of SNAP25, synaptophysin, and drebrin do not differ from non-hypertensive, non-transgenic rats at 16-18 months, the only age examined to date.
No Data
-
Neuronal Loss at
A reduction in calbindin staining might reflect a loss of inhibitory neurons. Levels of caspase-cleaved actin, a marker of apoptosis, are elevated, compared with non-hypertensive, non-transgenic rats.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1: deltaE9 | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease, Vascular Dementia | Amyloid plaques, microgliosis, and possible astrogliosis. Occasional neurons appear to contain paired helical filament tau. Demyelination. Reduced calbindin immunoreactivity and increased levels of caspase-cleaved actin may indicate neuron loss. |
Hyperactive. Working memory deficits as assessed by novel object recognition, but not as assessed by spontaneous alternation in the Y-maze. |
Snx1*D465N/APOE4/Trem2*R47H
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Snx1, APOE, Trem2 | TREM2 R47H | Snx1: Knock-In; APOE: Knock-In; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
Sorl1*A528T
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Sorl1 | SORL1 A528T (SNP 13) | Sorl1: Knock-In | Alzheimer's Disease |
Sorl1*A528T/APOE4/Trem2*R47H
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Sorl1, APOE, Trem2 | TREM2 R47H, SORL1 A528T (SNP 13), APOE C130R (ApoE4) | Sorl1: Knock-In; APOE: Knock-In; Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
SORL1 transgenic (Cre-inducible)
Observed
Absent
-
Changes in LTP/LTD at
LTP at Schaffer collateral-CA synapses was similar in hippocampal slices from 3-month-old Rosa26Tg/+ and wild-type mice. The application of Aβ oligomers impaired LTP in slices from wild-type mice but did not affect LTP in SORL1 transgenic mice.
-
Cognitive Impairment at
Three-month-old Rosa26Tg/+ SORL1 transgenic mice performed similarly to wild-type mice in the acquisition and retention phases of the Morris Water Maze test. Hippocampal injection of Aβ oligomers prevented wild-type mice from learning the location of the escape platform but did not affect the performance of the transgenic mice.
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
SORL1 | SORL1: Transgenic | Alzheimer's Disease | Unknown. |
Normal performance in the Morris Water Maze. |
SORLA-deficient
Observed
Absent
-
Plaques at
No amyloid plaques observed up to 10 months of age. When SORLA-deficient mice are crossed with APP transgenic models of amyloidosis, amyloid deposition is accelerated, compared with the parental APP transgenic line.
No Data
-
Tangles at
No data.
-
Neuronal Loss at
Neuron loss was not seen in the substantia nigra and ventral tegmental areas, assessed at 5 weeks and 45 weeks. Data on neuron numbers are not available from other brain regions. Nigrostriatal connectivity appears to be disrupted in SORLA-deficient mice.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No differences in LTP were observed in hippocampal slices from 10- to 12-month-old Sorl1-/- mice and slices from littermates heterozygous for the Sorl1 deletion (Rohe et al., 2008). It is not known whether LTP in these genotypes differs from that of wild-type mice.
-
Cognitive Impairment at
Compared with wild-type mice, SORLA-deficient mice exhibited more arm entries and more time spent in the open arms of the elevated plus maze—behaviors interpreted as evidence of hyperactivity and reduced anxiety. Hyperactivity was also noticed in the open field test.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Sorl1 | Sorl1: Knock-Out | Alzheimer's Disease | Mice do not generate amyloid plaques. Disrupted nigrostriatal connectivity and thinner inner nuclear layer of the retina. |
Hyperactivity and reduced anxiety, compared with wild-type mice. |
TAS10 (thy1-APPswe)
Observed
-
Plaques at 52
Fibrillar amyloid plaques develop by 12 months in the cortex and hippocampus.
-
Gliosis at 26
Astrogliosis and microgliosis underway by 6 months of age in the dentate gyrus.
-
Synaptic Loss at 104
TAS10 mice initially have more synapses than non-Tg mice; specifically, greater numbers of synapses per neuron were documented at 12 and 18 months of age. However, by 24 months of age, TAS10 mice have fewer synapses than non-Tg mice.
-
Cognitive Impairment at 26
Deficits in spatial learning present by 6 months of age as measured by the Morris water maze. No difference from non-Tg at 2 months of age. Deficits in Y maze at 12 months. No deficit in fear conditioning up to 24 months of age.
Absent
-
Tangles at
Absent.
-
Neuronal Loss at
Qualitative difference in neuronal numbers at 24 months in specific regions of the hippocampus, but no significant neuronal loss.
-
Changes in LTP/LTD at
At 12 to 14 months of age, deficits in basal synaptic transmission have been observed in the CA1 region, but short- and long-term synaptic plasticity are relatively normal (Brown et al., 2005).
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish) | APP: Transgenic | Alzheimer's Disease | Age-related accumulation of Aβ in the hippocampus and cortex leading to plaque deposition by 12 months of age. Early gliosis and dystrophic neurites, not limited to the vicinity around plaques. Changes in synaptic morphology and number, along with increased number of lysosomes. |
Deficits in spatial memory prior to Aβ deposition, including deficits in the Morris water maze by 6 months Deficits in spontaneous alternation behavior in the Y maze by 12 months. No deficit in fear conditioning. |
TASTPM (TAS10 x TPM)
Observed
-
Plaques at 26
Aβ begins to deposit at 3 months of age, with fibrillar plaques evident by 6 months in the cerebral cortex and hippocampus. Some vascular amyloid is also observed. Plaque pathology is more severe in female mice.
-
Gliosis at 28
Greater numbers of reactive astrocytes and microglia by 6 months of age in the hippocampus and cortex, predominantly near amyloid plaques.
-
Cognitive Impairment at 26
Age-dependent impairment in object recognition memory starting around 6 months of age for both sexes. No impairment at 3 to 4 months of age.
Absent
-
Tangles at
Absent.
-
Neuronal Loss at
Minimal neuronal loss up to 10 months of age. Some signs of loss in the immediate vicinity of plaques in the hippocampus (Howlett et al., 2008).
No Data
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1 M146V | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Aβ deposits beginning at 3 months of age, with fibrillar plaques by 6 months in the cerebral cortex and hippocampus. Some vascular amyloid. Plaques surrounded by dystrophic neurites and reactive glia. No tangles or neuronal loss. Female mice have more rapid and severe amyloid pathology. |
Age-dependent impairment in object recognition memory starting around 6 months of age. |
Tau35
Observed
-
Tangles at 35
Abnormally phosphorylated tau detected at two months and by eight months tau was mislocalized and misfolded and dystrophic neurites were observed. Tangle-like structures observed in the hippocampus by 14 months.
-
Synaptic Loss at 61
At 14 months synapsin1 protein levels were decreased but synaptophysin levels remained at wild-type levels.
-
Cognitive Impairment at 36
In the Morris water maze, Tau35 had the same performance as wild-type animals at six months but developed progressive deficits by eight months.
Absent
-
Gliosis at
Gliosis was not observed at 14 months.
No Data
-
Plaques at
Unknown.
-
Neuronal Loss at
Cell death was not formally assessed, however, overt neuronal death was not seen in the hippocampus.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT: Transgenic | Progressive Supranuclear Palsy, Alzheimer's Disease, Frontotemporal Dementia, Other Tauopathy | Progressive tau pathology in the hippocampus, including abnormally phosphorylated and misfolded tau, mislocalized tau, and tangle-like structures. Dystrophic neurites. |
Impaired spatial learning and memory in the Morris water maze. Early motor impairments, including abnormal limb clasping, Rotarod deficits and decreased grip strength. |
Tau4RTg2652
Observed
-
Cognitive Impairment at 13
Deficits in spatial learning and memory as indicated by performance in the Barnes maze at multiple time points (3, 6, 11 months of age).
Absent
-
Plaques at
Absent.
-
Tangles at
Absence of mature neurofibrillary tangles, but extensive pretangle pathology throughout the brain (e.g. phospho-tau).
-
Neuronal Loss at
Absent.
No Data
-
Gliosis at
Unknown.
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT: Transgenic | Frontotemporal Dementia, Other Tauopathy, Alzheimer's Disease | Extensive pretangle pathology throughout the brain (e.g. phospho- tau) but no mature neurofibrillary tangles and only mild oligomeric tau, restricted to the CA1 region of the hippocampus. Dystrophic neurites and axonal pathology (spheroids). No overt neuronal loss. |
Motor deficits develop with age, including decreased grip strength and impaired Rotarod performance. Cognitive deficits, indicative of impaired spatial learning and memory, as assessed by the Barnes maze. |
Tau609 (Tau 10 + 16)
Observed
-
Tangles at 65
Gallyas silver-positive intracellular inclusions of hyperphosphorylated tau aggregates in the entorhinal cortex at 15 months, and in the hippocampus and cerebral cortex at 24 months, but not at 18 months.
-
Neuronal Loss at 65
Significant loss of NeuN-positive neurons in layer II of the entorhinal cortex at 15 months, and in the hippocampal CA1 region at 24 months, compared with non-Tg controls. No difference in the hippocampus at 18 months.
-
Gliosis at 52
At 12 months of age, Iba1-positive cells are observed. GFAP is observed at 24 months of age.
-
Synaptic Loss at 28
Reduced synaptic density at 6 months of age in select hippocampal areas compared to non-Tg mice and those expressing wild-type human tau. Densities in other areas were comparable until later ages (i.e., 24 months).
-
Changes in LTP/LTD at 26
Some changes in basal synaptic transmission and significant impairment of LTP evident by 6 months of age in some regions of the hippocampus.
-
Cognitive Impairment at 26
Deficits in spatial reference memory by 6 months of age as measured by the Morris water maze. No difference from non-Tg littermates at 4 months of age.
Absent
-
Plaques at
Absent.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT IVS10+16 C>T | MAPT: Transgenic | Frontotemporal Dementia, Alzheimer's Disease, Other Tauopathy | Aggregated tau in neurons of the entorhinal cortex, hippocampus, and cerebral cortex at advanced ages. Intraneuronal accumulation of tau oligomers in the hippocampus. Neuronal loss in the entorhinal cortex and hippocampus. Gliosis. Some hippocampal areas affected by age-related synaptic dysfunction and reduced synaptic density. |
Impaired spatial reference memory as measured by the Morris water maze by 6 months of age. |
TauA152T-AAV
Observed
-
Neuronal Loss at 10
Neuron loss in cortex, seen at 3 months.
-
Gliosis at 11
Astrogliosis, but not microgliosis, seen at 3 months.
-
Cognitive Impairment at 12
Deficits in contextual and cued fear conditioning, seen at 3 months.
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT A152T | MAPT: Virus | Alzheimer's Disease, Frontotemporal Dementia, Other Tauopathy | Neuron loss and astrogliosis were observed in the cortices of 3-month-old mice. |
Compared with GFP-AAV controls, TauA152T-AAV mice showed deficits in contextual and cued fear conditioning, increased hyperactivity, and decreased rearing in the open-field test, and spent more time in the open arms of the elevated plus maze. TauA152T-AAV mice also exhibited motor impairment on the Rotarod. |
TauC3 (Transgenic caspase-cleaved tau)
Observed
-
Synaptic Loss at 6
Reduced levels of synaptic proteins as early as 1.3 months, including synaptophysin. Further reductions in 3 and 6-month-old animals.
-
Cognitive Impairment at 6
Learning and memory impairments as early as 1.3 months in several behavioral tests including the Y-maze, passive avoidance, and novel object recognition.
Absent
-
Plaques at
Amyloid plaques were absent.
-
Tangles at
Neurofibrillary tangles were not observed; however, hyperphosphorylated tau occurred early in the form of oligomers and aggregates.
-
Neuronal Loss at
No significant neurodegeneration by 12 months of age.
-
Gliosis at
No significant astrogliosis in the hippocampus or cortex by 12 months of age.
No Data
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT: Transgenic | Alzheimer's Disease, Frontotemporal Dementia | No significant cell loss or astrogliosis in the brain. Age-dependent reduction in synaptic proteins (e.g. synaptophysin, PSD95) by 1.3 to 3 months of age. Hyperphosphorylated tau oligomers and aggregates. |
Learning and memory deficits by 1.3 to 3 months of age, as assessed by the Y-maze and passive avoidance tests. No significant motor impairment. |
TauΔK280 ("Proaggregation mutant")
Observed
-
Tangles at 104
Mature tangles are observed only at advanced age (>24 months), but extensive pre-tangle pathology develops with as little as three months of transgene expression. This includes mislocalization of tau to the somatodendritic compartment, conformational changes indicative of aggregation, and hyperphosphorylation (e.g. Ser 262, Ser 356).
-
Synaptic Loss at 57
Electron microscopy showed a moderate decrease in spine synapses in the CA1 region of the hippocampus following 13 months of gene expression.
-
Changes in LTP/LTD at 52
Impaired hippocampal LTP in the CA1 and CA3 areas.
-
Cognitive Impairment at 70
Cognitive deficits in the Morris water maze and in passive-avoidance paradigms.
Absent
-
Plaques at
Absent.
-
Neuronal Loss at
Absent.
No Data
-
Gliosis at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT K280del | MAPT: Transgenic | Alzheimer's Disease, Frontotemporal Dementia | Abundant pre-tangle pathology, but only rare mature tangles, and only at advanced ages. Tau pathology included mislocalization of tau to the somatodendritic compartment, aggregation, and hyperphosphorylation. |
Unknown. |
Tau P301L
Observed
-
Tangles at 35
Hyperphosphorylation, conformational changes, and aggregation of tau resulting in tangle-like pathology by 8 months.
-
Gliosis at 30
Astrogliosis by 7 months.
-
Changes in LTP/LTD at 26
Deficit in LTP in CA1 region of the hippocampus at 6 months, but enhanced LTP in the dentate gyrus at a young age (8-10 weeks).
-
Cognitive Impairment at 22
Age-associated deficit in two cognitive tests that do not depend heavily on motor ability, the passive avoidance task (significant deficit starting at 5 months, but not 2 or 3 months of age) and a novel object recognition task (significant deficit at 9 months, but not at 2, 3, 5, or 7 months of age) (Maurin et al., 2014).
Absent
-
Plaques at
Absent.
No Data
-
Neuronal Loss at
Unknown.
-
Synaptic Loss at
Unknown at advanced age. Young mice (1-2 months) have a significantly higher spine maturation index than controls. At 4-6 months, the spine maturation index remains high in the hippocampus, but is reduced to control levels in the cortex. Note, these results were generated using the progeny of Tau P301L x transgenic Thy1-YFP (Kremer et al., 2011).
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT P301L | MAPT: Transgenic | Alzheimer's Disease, Frontotemporal Dementia | Pathologic hyperphosphorylation and conformational change of parenchymal tau in brain tissues starting at 7 months. Tangle-like pathology is mainly observed in the brain stem and spinal cord, and to a lesser extent in the midbrain and cerebral cortex. Age-dependent increase in total tau in CSF. |
Age-associated deficits in a passive avoidance task (starting at 5 months) and a novel object recognition task (starting at 9 months). At a young age (~2 months) outperforms wild-type littermates in object recognition memory. Progressive motor impairment and reduced activity, accompanied by increased clasping of hind and then forelimbs around seven months. |
TauP301L-AAV
Observed
-
Tangles at 24
Argyrophilic, Thioflavin S-positive neurofibrillary tangles in cortex and hippocampus.
-
Gliosis at 12
Astrogliosis and microgliosis observed at 3 months.
-
Cognitive Impairment at 24
Deficits in cued and contextual fear conditioning observed at 6 months.
Absent
-
Neuronal Loss at
No cortical neuron loss at 6 months.
No Data
-
Plaques at
No data.
-
Synaptic Loss at
The accumulation of a PSD95 fragment suggests the possibility of synaptic abnormalities, although synaptic structure and function have not been assessed directly.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT P301L | MAPT: Virus | Alzheimer's Disease, Frontotemporal Dementia, Other Tauopathy | Neurofibrillary tangles and gliosis, but no cortical neuron loss, at 6 months of age. |
Hyperactivity in the open field, decreased time spent in the center of open field, more time spent in the open arms of the elevated plus maze, and deficits in cued and contextual fear conditioning at 6 months of age. |
Tau P301S (Line PS19)
Observed
-
Tangles at 23
Neurofibrillary tangles in the neocortex, amygdala, hippocampus, brain stem and spinal cord at six months with progressive accumulation (Yoshiyama et al., 2007).
-
Neuronal Loss at 39
Neuron loss in the hippocampus and entorhinal cortex by nine to12 months, as well as in the amygdala and neocortex becoming more severe by 12 months (Yoshiyama et al., 2007).
-
Gliosis at 11
Microgliosis at three months, especially in the white matter of the brain and spinal cord. Increased microgliosis by six months in white and gray matter of the hippocampus, amygdala, entorhinal cortex, and spinal cord. Microglial activation precedes astrogliosis (Yoshiyama et al., 2007).
-
Synaptic Loss at 13
Synaptophysin immunoreactivity decreased progressively from three to six months in the CA3 region of the hippocamus. Impaired synaptic function (Yoshiyama et al., 2007).
-
Changes in LTP/LTD at 26
Reduced LTP in the CA1 region of the hippocampus at six months. Altered basal synaptic transmission (smaller fiber volley amplitude, fEPSP slopes, and amplitudes) (Yoshiyama et al., 2007). Impaired hippocampal LTP as measured in freely moving mice (Lasagna-Reeves, 2016).
-
Cognitive Impairment at 27
Impairments in spatial learning and memory ability in the Morris water maze in six-month-old animals (Takeuchi et al., 2011). Impaired memory in assays of contextual fear conditioning (Lasagna-Reeves 2016).
Absent
-
Plaques at
Absent.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT P301S | MAPT: Transgenic | Alzheimer's Disease, Frontotemporal Dementia | Neuron loss and brain atrophy by eight to 12 months, especially in the hippocampus and spreading to the neocortex and entorhinal cortex. Neurofibrillary tangles in the neocortex, amygdala, hippocampus, brain stem, and spinal cord. Neuroinflammation with microgliosis and astrocytosis. |
Impairments in spatial memory and learning ability in Morris water maze. Paralysis at seven to 10 months associated with a hunched-back posture followed by feeding difficulties. About 80 percent mortality by 12 months with median survival of about nine months. |
TauPS2APP
Observed
-
Plaques at 17
Rare amyloid plaques at 4 months, plaques become more abundant with age. By 8 months the number of amyloid plaques increases considerably in the subiculum and the CA1 region of the hippocampus (Grueninger et al., 2010).
-
Tangles at 70
Abnormally phosphorylated tau is detectable at 4 months in both TauPS2APP and tau single transgenic mice especially in the subiculum, amygdala, and the CA1 region of the hippocampus. Tau pathology increases with age with numerous tangle-like deposits in the hippocampus confirmed by Gallyas silver staining at 16 months (Grueninger et al., 2010).
-
Cognitive Impairment at 17
Impairment is not age-associated and does not progress from age 4 months to 12 months (Grueninger et al., 2010).
Absent
-
Neuronal Loss at
No overt neuronal loss in the hippocampus at 16 months (Grueninger et al., 2010).
No Data
-
Gliosis at
Unknown.
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, MAPT, PSEN2 | APP K670_M671delinsNL (Swedish), MAPT P301L, PSEN2 N141I | APP: Transgenic; MAPT: Transgenic; PSEN2: Transgenic | Alzheimer's Disease | Phosphorylated tau accumulation in the subiculum and the CA1 region of the hippocampus at 4 months. Neurofibrillary tangles in these regions as well as the amygdala. Amyloid plaques. Dystrophic neurites and neuropil threads containing abnormally phosphorylated tau. No overt neuronal loss. |
Impaired spatial learning in the Morris water maze at 4 months but impairment is not progressive between 4 and 12 months and appears to be independent of pathology. |
Tau R406W transgenic
Observed
-
Tangles at 78
Congophilic tau inclusions in a subset of forebrain neurons around 18 months of age. Detected by Congo red, thioflavin S, and Gallyas silver stain.
-
Cognitive Impairment at 70
Impairments in the contextual and cued fear conditioning test at 16–23 months compared with wild-type littermates. No detectable sensorimotor deficits.
Absent
-
Plaques at
Absent.
No Data
-
Neuronal Loss at
Unknown.
-
Gliosis at
Unknown.
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT R406W | MAPT: Transgenic | Frontotemporal Dementia, Alzheimer's Disease | Argyrophilic and congophilic tau inclusions in neurons of the forebrain with age. Detectable with Congo red, thioflavin-S and Gallyas silver stain. Congophilic tau inclusions also in the hippocampus and amygdala. Mainly straight tau filaments. |
Impairments in contextual and cued fear conditioning at 16–23 months compared with wild-type littermates. No detectable sensorimotor deficits. |
TauRDΔK280 (“Proaggregation mutant”)
Observed
-
Tangles at 9
Tau tangles and aggregates with as little as 2-3 months of transgene expression. Tangles start in the entorhinal cortex and amygdala and spread to the neocortex by 15 months. Heterogeneous tangle morphology, including flame-shaped.
-
Neuronal Loss at 22
Neuronal loss in the dentate gyrus (granule neurons) following 5 months of transgene expression. Shrinkage of the molecular layer of the hippocampus.
-
Gliosis at 91
Astrogliosis in the hilus region of the hippocampus after 21 months of transgene expression. Additional increases in GFAP-positive astrocytes in the entorhinal and piriform cortices.
-
Synaptic Loss at 41
Hippocampal synaptic loss as indicated by multiple measures following 9.5 months of transgene expression. Reduced synaptophysin immunoreactivity and reduced number of spine synapses as measured by electron microscopy.
-
Changes in LTP/LTD at 43
Multiple deficits in synaptic plasticity, including deficits in LTP and LTD, after 10 months of transgene expression. Functional changes are associated with structural synaptic changes, local calcium dysregulation, and a decrease in the synaptic vesicle pool.
-
Cognitive Impairment at 43
Learning and memory impairments are apparent after 10 months of transgene expression as assessed by the Morris water maze and passive avoidance tasks.
Absent
-
Plaques at
Absent.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT K280del | MAPT: Transgenic | Alzheimer's Disease, Frontotemporal Dementia | Tau aggregates and tangles as early as 2-3 months after gene expression. Gallyas silver-positive neurons abundant in the entorhinal cortex and amygdala, spreading to the neocortex by 15 months. “Ballooned” neurons. Astrogliosis. Synaptic structural changes and reduced synaptic number. Hippocampal neuronal loss. |
Reversible learning and memory deficits in the Morris water maze and passive avoidance test. No significant motor deficit, although slight reduction in Rotarod performance. |
Tau V337M
Observed
-
Tangles at 48
Fibrillar staining in the hippocampus of 11 month old animals by Congo red birefringence. Absent in 4 month old mice, indicating the formation of these neurofilament-like structures occurs between 4 and 11 months (Tanemura et al., 2001).
-
Neuronal Loss at 43
Evidence of hippocampal neuronal degeneration in 10 month old animals: irregularly shaped neurons with tau pathology that stained with propidium iodide. As characteristics of apoptosis were not observed, the neurons were thought to be undergoing non-apoptotic atrophic degeneration (Tanemura et al., 2002).
-
Changes in LTP/LTD at 65
In hippocampal slices there was an attenuation of the amplitude of Schaffer collateral evoked hippocampal depolarization (Tanemura et al., 2002).
-
Cognitive Impairment at 48
Behavioral abnormalities measured in 11 month-old mice. They spent more time in the open arms of the elevated plus maze and had greater overall locomoter activity. No differences in the Morris water maze compared with non-transgenic mice, suggesting the transgenic animals retain spatial recognition abilities (Tanemura et al., 2002).
Absent
-
Plaques at
Absent.
No Data
-
Gliosis at
Unknown.
-
Synaptic Loss at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT V337M | MAPT: Transgenic | Alzheimer's Disease, Frontotemporal Dementia | SDS-insoluble tau aggregates in hippocampus. Degenerating neurons in the hippocampus containing phosphorylated and ubiquitinated tau aggregates with β-sheet structure. |
Higher overall spontaneous locomotion than non-transgenic littermates in elevated plus maze. No differences in the Morris water maze. |
TBA42
Observed
-
Plaques at 52
Very rare extracellular Aβ deposits.
-
Neuronal Loss at 52
Age-dependent neuronal loss in the CA1 region of the hippocampus. No difference from wild-type mice at 3 and 6 months of age, but approximately 35% loss at 12 months of age.
-
Gliosis at 52
Marked gliosis in the hippocampus as measured by GFAP staining at 12 months.
-
Cognitive Impairment at 54
Age-dependent deficits in working and spatial reference memory at 12 months, but not at 3 and 6 months.
Absent
-
Tangles at
Absent.
No Data
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP: Transgenic | Alzheimer's Disease | Intraneuronal accumulation of Aβ peptides in the hippocampus by 3 months and in cerebellar nuclei by 6 months. Marked gliosis in the hippocampus by 12 months. Very rare extracellular Aβ deposits. |
Age-dependent behavioral deficits, including working memory as assessed by the cross maze at 12 months, but not at 3 or 6 months. Early and persistent decrease in anxiety in the elevated plus maze. Comparable to wild-type in general motor coordination at 3 and 6 months as indicated by the balance-beam test, but impairment at 12 months. |
TcMAC21
Observed
-
Changes in LTP/LTD at 14
Reduction in LTP compared to controls.
-
Cognitive Impairment at 13
Behavioral (Morris water maze and open field) and physiological results showed significant deficits in learning, memory, and synaptic plasticity. No alteration in nesting abilities.
Absent
-
Plaques at
At 15–24 months of age, no amyloid plaques detected.
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Multi-transgene | Down's Syndrome, Alzheimer's Disease | Elevated APP, Aβ40, and Aβ42 in brain at 15–24 months, but no amyloid plaque pathology. Enlarged lateral ventricles and superior colliculus, and smaller cerebellum. |
Learning and memory deficits. |
Tg12099 rat
Observed
-
Tangles at 26
Neurofibrillary tangles visualized by Bielschowsky silver staining were present in the piriform and entorhinal cortices of terminal Tg12099 rats homozygous for the transgene. Tangles also stained with ThioflavinS. Tangles were absent in hemizygous rats.
-
Neuronal Loss at 39
Pronounced neurodegeneration in the forebrains of Tg12099 homozygotes at terminal stages. Neuronal loss and concordant atrophy of piriform/entorhinal cortices were first observed between 9 and 12 months and continued to worsen with age.
-
Gliosis at 27
Increased GFAP immunoreactivity in the brains of terminal Tg12099 homozygotes compared with aged hemizygotes. Astrogliosis matches the spatiotemporal pattern of tau deposition, becoming apparent in the amygdala and entorhinal cortex as early as 6 to 7 months of age.
Absent
No Data
-
Plaques at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT P301S | MAPT: Transgenic | Frontotemporal Dementia, Alzheimer's Disease | Tau pathology, including neurofibrillary tangles, forebrain neurodegeneration, and astrogliosis in homozygotes. Hemizygotes exhibit little pathology, save for a few AT8-immunoreactive neurons in the entorhinal cortices of aged animals. |
Ataxia, bradykinesia, and seizures reported in aging homozygotes. |
Tg2576
Observed
-
Plaques at 48
Numerous parenchymal Aβ plaques by 11-13 months.
-
Gliosis at 43
Increase in microglial density and size in plaque-forming areas of the brain including the hippocampus, frontal cortex, entorhinal cortex, and occipital cortex in 10-16 month old hemizygotes (Frautschy et al., 1998).
-
Synaptic Loss at 20
Dendritic spine loss by 4.5 months In the CA1 region of the hippocampus (Lanz et al., 2003).
-
Changes in LTP/LTD at 22
By 5 months, there was a decline in LTP in the dentate gyrus after perforant path stimulation compared to wild-type; impairment was not observed at 2 months (Jacobsen et al., 2006). Both the CA1 and dentate gyrus of aged mice (>15 months) are impaired (Chapman et al., 1999). Differences have been observed between the Schaffer collateral and mossy fiber pathways (Jung et al., 2011).
-
Cognitive Impairment at 26
Impaired spatial learning, working memory, and contextual fear conditioning at <6 months although other studies have reported normal cognition at this age with progressive impairment by >12 months.
Absent
-
Tangles at
Absent.
-
Neuronal Loss at
Absent or very limited.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish) | APP: Transgenic | Alzheimer's Disease | Numerous parenchymal Aβ plaques by 11-13 months with some vascular amyloid. Oxidative lipid damage, astrogliosis and microgliosis. No tangles or neuronal loss. |
Impaired spatial learning, working memory, and contextual fear conditioning reported at <6 months although other studies have reported normal cognition at this age with progressive impairment by >12 months. |
Tg2576/Tau(P301L) (APPSwe-Tau)
Observed
-
Plaques at 39
Plaques develop gradually with age. No plaques at 5 months. Very few small plaques at 6 and 7 months. By 9 months plaques scattered throughout the cortex, hippocampus and amygdala, continue to increase at 12 months. Similar distribution as Tg2576.
-
Tangles at 13
Neurofibrillary tangles in the spinal cord and pons as early as 3 months, but more consistent and numerous by 6 months. Tangles morphologically similar to those in JNPL3 mice but older bigenic female mice had a marked increase in neurofibrillary tangles in limbic areas by 6 months, especially the olfactory cortex, entorhinal cortex and amygdala (Lewis et al., 2001).
-
Gliosis at 13
Reactive astrocytes and microglia as early as 3 months in the hippocampus as measured by GFAP and CD45. Increased astrocytosis with age especially in limbic areas with the most neurofibrillary tangles. Microglia especially concentrated around plaques at 9 and 12 months (Lewis et al., 2001).
Absent
No Data
-
Synaptic Loss at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, MAPT | APP K670_M671delinsNL (Swedish), MAPT P301L | APP; MAPT: Transgenic | Alzheimer's Disease | Gradual appearance of plaques; by 9 months plaques are scattered throughout the cortex, hippocampus, and amygdala similar to Tg2576. Tau pathology more extensive than JNPL3. Astrocytosis and microgliosis. |
Motor disturbances similar to JNPL3, with identical range in age of onset. Reduced vocalization and decreased grooming. |
Tg4-42
Observed
-
Neuronal Loss at 35
Age- and dose-dependent neuronal loss in the hippocampus CA1 region of hemizygous and homozygous mice. Compared with wild-type, hemizygous mice had 38% neuronal loss at 8 months, and 49% loss at 12 months. No difference at 3 months.
-
Gliosis at 9
Reactive microglia and astrocytes in the hippocampus starting at 2 months.
-
Synaptic Loss at 37
Altered synaptophysin staining in the CA3 region of the hippocampus. More pronounced in homozygous mice than hemizygous mice at 8 months.
-
Cognitive Impairment at 35
Spatial reference memory is impaired as assessed by Morris water maze at 8 months in homozygous mice and 12 months in hemizygous mice. Deficit is age-dependent and is not detected at 3 months. Impaired contextual fear conditioning at 12 months.
Absent
-
Plaques at
Absent.
-
Tangles at
Absent.
No Data
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP: Transgenic | Alzheimer's Disease | Aβ4-42 is dectable starting at two months, predominantly in the CA1 region of the hippocampus, but also in the occipital cortex, piriform cortex, striatum, and superior colliculus. Age- and dose-dependent hippocampal neuronal loss is seen in the CA1 region as well as microgliosis and astrogliosis. |
Age-dependent spatial learning deficit as demonstrated in the Morris water maze, specifically, the absence of a preference for the target quadrant starting at eight months in homozygous mice and at 12 months in hemizygous mice. Impaired contextual fear conditioning. |
tg-APPSwe
Observed
-
Plaques at 52
Plaques are detectable at approximately 12 months and are heterogeneous in morphological structure and size, as well as in terms of fluorescence emitted when stained with luminescent polymers (conformational amyloid ligands)(Philipsson et al., 2009).
-
Gliosis at 52
Microgliosis and astrogliosis are most prominent in the hippocampus, but also found locally around deposits in the cerebral cortex and in thalamus at approximately 12 months (Philipsson et al., 2009).
Absent
-
Tangles at
Absent.
-
Neuronal Loss at
Absent.
No Data
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
-
Cognitive Impairment at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish) | APP: Transgenic | Alzheimer's Disease | Extracellular amyloid deposition begins at ~12 months. Intraneuronal Aβ aggregates at ~6 months. Extracellular pathology, both cerebrovascular amyloid angiopathy (CAA) and congophilic parenchymal plaques, mainly found in the cerebral cortex, hippocampus and thalamus. Aβ-burden in cerebral cortex is approximately 1.0% (at 12 months) and 2.8% (at 18 months). |
Unknown. |
Tg-ArcSwe
Observed
-
Plaques at 22
Extracellular amyloid plaque deposition starts at around 5-6 months of age (Lord et al., 2006) and is most consistently present in the cerebral cortex, hippocampus, and thalamus (Lillehaug et al., 2013).
-
Gliosis at 26
Microgliosis and astrogliosis most prominent in the hippocampus, but also locally around deposits in the cerebral cortex and thalamus.
-
Cognitive Impairment at 17
Transgene-dependent spatial learning impairment in the Morris water maze (4-8 months) (Lord et al., 2009) and in an Intellicage-based Passive Avoidance test (16 months)(Codita et al., 2010).
Absent
-
Tangles at
Absent.
-
Neuronal Loss at
Absent.
No Data
-
Synaptic Loss at
Unknown.
-
Changes in LTP/LTD at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP E693G (Arctic) | APP: Transgenic | Alzheimer's Disease, Cerebral Amyloid Angiopathy | Strong intraneuronal Aβ aggregation starting at 1 month and increasing with age. Extracellular amyloid plaque at 5-6 months, most consistent in the cerebral cortex, hippocampus, and thalamus. Congophilic parenchymal plaques are predominant, but some mice show marked CAA, particularly in the thalamus. |
Mild spatial learning deficits at 4-8 months in Morris water maze and impaired functioning in a passive avoidance test at 16 months. |
TgCRND8
Observed
-
Plaques at 13
Amyloid deposition progresses with age. Thioflavin S-positive amyloid deposits at 3 months; dense cored plaques and neuritic pathology by 5 months. Plaques appear first in the subiculum, amygdala and frontal cortex, spread to the dentate gyrus, the olfactory bulb, and later thalamus, cerebral vasculature, and striatum, followed by the cerebellum and brain stem (Chishti et al., 2001).
-
Neuronal Loss at 26
Variable cell loss by region. No difference in overall cell count, but fewer hippocampal neurons at 6 months (Brautigam et al., 2012).
-
Gliosis at 13
Microglia activation appears simultaneously with Aβ deposition, with only rare activated microglia at 9-10 weeks, but by 13-14 weeks microglia cluster around Aβ deposits in the cerebral cortex and hippocampus; numerous by 20 weeks. Robust astrogliosis slightly later with clusters of GFAP+ astrocytes emerging around plaques at 13-14 weeks (Dudal et al., 2004).
-
Synaptic Loss at 26
Reduced synaptophysin immunoreactivity in the vicinity of plaques at 6 months (Adalbert et al., 2009).
-
Changes in LTP/LTD at 26
In hippocampal slices from 6- to 12-month-old mice basal excitatory synaptic transmission (as assessed by I/O relationships) and LTP at CA1 are reduced in TgCRND8 mice compared with wild-type mice (Kimura et al., 2012).
-
Cognitive Impairment at 13
Early impairment in acquisition and learning reversal in the reference memory version of the Morris water maze, present by 3 months (Chishti et al., 2001).
Absent
-
Tangles at
Neurofibrillary tangles are absent (Chishti et al., 2001). Tau is hyperphosphorylated, nitrosylated and aggregated at 7-12 months especially in the neocortex, dentate gyrus, and the CA1 and CA3 areas of the hippocampus (Bellucci et al., 2007).
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP V717F (Indiana) | APP: Transgenic | Alzheimer's Disease | Rapid, early plaque development, with thioflavin S-positive amyloid deposits at 3 months; dense cored plaques and neuritic pathology by 5 months. Plaques become more extensive with age. More Aβ42 than Aβ40. Activated microglia appear concurrently with plaques, whereas GFAP+ astrocytes follow later, about 13-14 weeks. Dystrophic neurites at 5 months . |
Early impairment in acquisition and learning reversal in the reference memory version of the Morris water maze by 3 months. Cognitive deficits in the step-down inhibitory avoidance test at 7 months but not at 2 months. Similar to wild-type in motility, exploratory activity, or neuromuscular function at 7 months as evaluated by the rotarod, hole board and grip strength tests. |
TgDimer
Observed
-
Changes in LTP/LTD at 28
LTP decays more rapidly, compared with wild-type mice.
-
Cognitive Impairment at 28
Learning deficits in the Morris Water Maze.
Absent
-
Plaques at
None observed through 24 months.
-
Tangles at
None observed through 24 months.
-
Neuronal Loss at
None observed through 24 months.
-
Gliosis at
No transgene-related gliosis, but slight age-associated gliosis—also seen in wild-type mice—at 24 months.
No Data
-
Synaptic Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP S679C | APP: Transgenic | Alzheimer's Disease, MCI due to AD | Intracellular Aβ immunoreactivity in the hippocampus and cortex, beginning by 12 months. No amyloid plaques, hyperphosphorylated tau, microgliosis, astrogliosis, or neuron loss through 24 months. |
Learning deficits, as well as indicators of increased anxiety and depression, by 7 months. |
TgF344-AD
Observed
-
Plaques at 24
Age-dependent accumulation of amyloid plaques in hippocampus and cortex between 6 and 26 months of age.
-
Tangles at 64
Structures similar in appearance to neurofibrillary tangles revealed by Gallyas staining and immunostaining using an antibody directed against phospo-tau.
-
Neuronal Loss at 64
Approximate 40 percent loss of neurons in hippocampus and cortex by 16 months.
-
Gliosis at 23
Microgliosis and astrogliosis are apparent by 6 months.
-
Cognitive Impairment at 25
Deficits in reversal learning in the Morris water maze apparent by 6 months.
Absent
No Data
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1: deltaE9 | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques, microgliosis, and astrogliosis by 6 months. Neurofibrillary tangle-like structures at 16 months. Approximate 40 percent loss of neurons in hippocampus and cortex by 16 months. |
Earliest reported deficits are in reversal learning in the Morris water maze, apparent by 6 months. |
Tg-SwDI (APP-Swedish,Dutch,Iowa)
Observed
-
Plaques at 13
Hemizygotes progressively accumulate insoluble Aβ40 and Aβ42, especially within brain microvessels starting at 3 months. Amyloid-β deposits in the subiculum, hippocampus, and cortex at ~3 months. By ~6 months deposits become more numerous and appear in the olfactory bulb and thalamic region as well, with deposits throughout most of the forebrain by 12 months (Davis et al., 2004).
-
Gliosis at 26
Pronounced increase in the number of GFAP-positive astrocytes and activated microglia with age (6-24 months) especially in the thalamus and subiculum and to a lesser extent in the cortex (Miao et al., 2005).
-
Cognitive Impairment at 13
Impaired learning and memory in the Barnes maze task at 3, 9, and 12 months; beginning at 3 months took longer to find the escape hole. No difference in mobility, strength or coordination (Xu et al., 2007).
Absent
-
Tangles at
Absent.
No Data
-
Neuronal Loss at
Unknown.
-
Synaptic Loss at
Unknown.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP | APP K670_M671delinsNL (Swedish), APP E693Q (Dutch), APP D694N (Iowa) | APP: Transgenic | Alzheimer's Disease, Cerebral Amyloid Angiopathy, Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch type | Hemizygotes progressively accumulate insoluble Aβ40 and Aβ42, especially within brain microvessels starting at 3 months. Fibrillar Aβ in micovessels around 6 months. Diffuse plaque-like deposits around 3 months in the subiculum, hippocampus and cortex. Aβ deposits throughout the forebrain by 12 months. |
Impaired learning and memory in the Barnes maze task at 3, 9, and 12 months. Beginning at 3 months transgenic mice took longer to find the escape hole. No difference in mobility, strength or coordination. |
THY-Tau22
Observed
-
Tangles at 13
Heterozygous animals develop tau pathology starting at 3-6 months. Pathology becomes more severe and widespread with age. Neurofibrillary tangle-like inclusions occur (Gallyas and MC1+) along with rare ghost tangles and paired helical filament-like structures (Schindowski et al., 2006).
-
Neuronal Loss at 52
Loss of cells in the CA1 region of the hippocampus from 12 months as measured by DAPI staining and Nissl/cresyl-violet (Schindowski et al., 2006). Also, a significant reduction in the number of choline acetyltransferase (ChAT)-immunopositive cholinergic neurons in the medial septum has been reported (Belarbi et al., 2011).
-
Gliosis at 13
Age-dependent increase in the number of GFAP+ astrocytes in the hippocampus (hilus, CA1, CA3), cerebral cortex, corpus callosum (Schindowski et al., 2006).
-
Changes in LTP/LTD at 39
Altered paired pulse facilitation (PPF), a form of presynaptic short-term plasticity in 9-10 month old heterozygous animals: PPF increased at 10 ms. Also at this age, impaired maintenance of long term depression as compared with wild-type littermates (Van der Jeugd et al., 2011). Deficit in basal synaptic transmission in the hippocampus, but normal LTP (Schindowski et al., 2006).
-
Cognitive Impairment at 26
Non-spatial memory affected as early as 6 months; spatial memory impaired only after 9 months (Van der Jeugd et al., 2013). Impaired appetitive responding (Lo et al., 2013).
Absent
-
Plaques at
Absent.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT G272V, MAPT P301S | MAPT: Transgenic | Frontotemporal Dementia, Alzheimer's Disease | A variety of tau pathologies starting at 3 months, including neurofibrillary tangle-like inclusions, rare ghost tangles, and paired helical filament-like structures. Hyperphosphorylation of tau on many epitopes (e.g. AT8, AT100, AT180, AT270, 12E8, tau-pSer396, and AP422) and mild astrogliosis. |
Increased anxiety and delayed learning from 3 months, and reduced spatial memory at 10 months. No changes in overall motor activity and no gross motor deficits. Increased depression-like and aggressive behavior, co-occurring with disturbances in nocturnal activity. |
TMHT (Thy-1 mutated human tau)
Observed
-
Tangles at 17
Tangles at 4 months and progress with age.
-
Cognitive Impairment at 22
Cognitive impairment by 5 months as measured by the Morris Water Maze.
Absent
-
Plaques at
Absent.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
MAPT | MAPT V337M, MAPT R406W | MAPT: Transgenic | Alzheimer's Disease | Increased total tau, and phosphorylated tau (Thr181, Ser199, Thr231) in amygdala and hippocampus starting at 3 months. |
Spatial memory deficits starting at 5 months (Morris water maze). Olfactory deficits at 5 months (Buried food test). No motor deficits (rota rod, beam walk) or depressive behavior (forced swim test). |
TREM2-BAC
Observed
Absent
-
Plaques at
Not observed at 7 months.
-
Gliosis at
No microgliosis was observed at 7 months.
-
Changes in LTP/LTD at
Normal LTP at 10 months.
-
Cognitive Impairment at
Normal contextual fear conditioning at 10 months.
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
TREM2 | TREM2: Transgenic | Alzheimer's Disease | No obvious neuropathology is observed at 4, 7 and 11 months of age. |
Normal contextual fear conditioning at 10 months of age. |
TREM2-BAC X 5xFAD
Observed
-
Plaques at 28
Observed at 7 months, the youngest age examined.
-
Gliosis at 28
Microgliosis observed; however, fewer plaque-associated microglia and altered microglial morphology (more ramified processes) compared with 5xFAD at 7 months, the only age examined.
Absent
-
Cognitive Impairment at
5xFAD/TREM2 mice perform comparably to wild-type mice in a contextual fear conditioning test, while 5xFAD mice are impaired.
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
TREM2, APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | TREM2: Transgenic; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques with plaque-associated microgliosis. Reduced plaque burden, altered microglial and plaque morphology, and less severe plaque-associated neuritic dystrophy, compared with 5xFAD. |
5xFAD/TREM2 mice perform comparably to wild-type mice in a contextual fear conditioning test, while 5xFAD mice are impaired. |
Trem2-H157Y knock-in
Observed
-
Changes in LTP/LTD at 24
Enhanced paired-pulse facilitation and long-term potentiation at Schaffer collateral-CA1 synapses in 6-month-old mice homozygous for the H157Y Trem2 allele.
Absent
-
Gliosis at
Microglial density and morphology did not differ between carriers of the H157Y variant and wild-type mice at 6 months of age.
-
Synaptic Loss at
Levels of the presynaptic marker synaptophysin and the postsynaptic markers PSD95 and GLUR2 in the cortices of 6-month-old mice did not differ between carriers of the H157Y variant and wild-type mice.
-
Cognitive Impairment at
The H157Y mutation did not affect the performances of 6-month-old mice on a battery of behavioral tests: the open-field test to assess anxiety, cued and contextual fear conditioning to measure associative memory, and the Y-maze test to measure spatial working memory.
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | TREM2 H157Y | Trem2: Knock-In | Alzheimer's Disease | Microglial density and morphology did not differ between carriers of the H157Y variant and wild-type mice. |
Trem2-H157Y knock-in did not differ from wild-type mice at 6 months of age in tests of anxiety, associative memory, and spatial working memory. |
Trem2-H157Y x 5xFAD
Observed
-
Plaques at 16
Decreased plaque burdens and densities in 5xFAD;Trem2H157Y/H157Y compared with 5xFAD;Trem2+/+ at 8.5 months, but genotypes similar at 4 months.
-
Gliosis at 16
Decreased microgliosis and astrogliosis in 5xFAD;Trem2H157Y/H157Y compared with 5xFAD;Trem2+/+ at 8.5 months, but genotypes similar at 4 months.
Absent
-
Synaptic Loss at
Cortical levels of synaptophysin and PSD95 did not differ between genotypes at 8.5 months.
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2, APP, PSEN1 | TREM2 H157Y, APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | Trem2: Knock-In; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Age-dependent effects on amyloid-β pathology and gliosis. At 4 months, plaque burdens, microgliosis, and astrogliosis were similar among genotypes. By 8.5 months, amyloid burdens, microgliosis, and astrogliosis were reduced in homozygous carriers of the H157Y variant, compared with 5xFAD mice homozygous for wild-type Trem2. |
Unknown. |
TREM2, humanized (common variant)
Observed
Absent
-
Gliosis at
Expression of DAM (disease-associated microglia) genes is low at 8.5 months, suggesting that microglia are in a resting or homeostatic state.
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
TREM2, Trem2 | TREM2: Transgenic; Trem2: Knock-Out | Alzheimer's Disease | Unknown. |
Unknown. |
TREM2, humanized (common variant) X 5XFAD
Observed
-
Plaques at 34
Plaques observed in 8.5-month-old mice, only age reported thus far.
-
Gliosis at 34
Microgliosis observed in 8.5-month-old mice, only age reported thus far.
Absent
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2, TREM2, APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | Trem2: Knock-Out; TREM2: Transgenic; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques surrounded by activated microglia. |
No data. |
TREM2, humanized (R47H)
Observed
Absent
-
Gliosis at
Expression of DAM (disease-associated microglia) genes is low at 8.5 months, suggesting that microglia are in a resting or homeostatic state.
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
TREM2, Trem2 | TREM2 R47H | TREM2: Transgenic; Trem2: Knock-Out | Alzheimer's Disease | Unknown. |
Unknown. |
TREM2, humanized (R47H) X 5XFAD
Observed
-
Plaques at 34
Plaques observed in 8.5-month-old mice, the only age reported thus far.
-
Gliosis at 34
Microgliosis observed in 8.5-month-old mice, the only age reported thus far. Fewer plaque-associated microglia in mice expressing the R47H variant, compared with the common variant of human TREM2.
Absent
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2, TREM2, APP, PSEN1 | TREM2 R47H, APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | Trem2: Knock-Out; TREM2: Transgenic; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Lower density of activated microglia surrounding amyloid plaques in 5XFAD mice expressing the R47H variant of human TREM2 compared with those expressing the common variant. |
No data. |
TREM2-IPD
Observed
-
Gliosis at 12
At 3 months of age, TREM2-IPD mice had more Tmem119-positive microglia and a greater percentage of proliferating microglia than mice expressing wild-type Trem2.
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | Trem2: Knock-In | Alzheimer's Disease | At 3 months of age, TREM2-IPD mice had more Tmem119-positive microglia and a greater percentage of proliferating microglia than mice expressing wild-type Trem2. |
Unknown. |
Trem2-IPDxAPP23xPS45
Observed
-
Plaques at 12
Greater numbers of plaques, particularly small plaques, and larger areas occupied by plaques in 3-month-old Trem2-IPDxAPP23xPS45 mice, compared with APP23xPS45 animals. These genotype-dependent differences disappeared by 7 months.
-
Gliosis at 13
Increased microgliosis in the vicinity of plaques in 3-month-old Trem2-IPDxAPP23xPS45 mice, compared with APP23xPS45 mice.
-
Synaptic Loss at 14
Compared with APP23xPS45 mice, 3-month-old Trem2-IPDxAPP23xPS45 mice had fewer and smaller puncta stained for the presynaptic marker Sv2a (synaptic vesicle glycoprotein 2A) in the vicinity of plaques. These genotype-dependent differences disappeared by 7 months.
Absent
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2, APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1 G384A | Trem2: Knock-In; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques, plaque-associated neuritic dystrophies, microgliosis. Pathology exacerbated in Trem2-IPDxAPP23xPS45 mice, compared with APP23xPS45 mice expressing wild-type Trem2, at an early—but not late—stage of plaque deposition. |
Unknown. |
Trem2 KO (Colonna)
Observed
Absent
-
Gliosis at
No spontaneous gliosis, but impaired microglial response to injury.
-
Cognitive Impairment at
Not observed.
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | Trem2: Knock-Out | Nasu-Hakola Disease, Frontotemporal Dementia, Alzheimer's Disease | Microglial number remains constant and microglial size decreases with age in the corpus callosum of Trem2 KO mice, while microglial number increases and microglial size remains stable in wild-type mice. |
No cognitive/behaviorial deficits observed. |
Trem2 KO (Colonna) x 5XFAD
Observed
-
Plaques at 16
Plaques present by 4 months, the earliest age studied.
-
Neuronal Loss at 32
Loss of cortical layer V neurons by 8 months, the earliest age studied.
-
Gliosis at 16
MIcrogliosis by 4 months, the earliest age studied.
Absent
No Data
-
Tangles at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2, APP, PSEN1 | APP K670_M671delinsNL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V | Trem2: Knock-Out; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Compared with 5XFAD, mice deficient in TREM2 show an age- dependent increase in amyloid accumulation in the hippocampus, more severe plaque-associated neuritic dystrophy, and exaggerated neuron loss in the cortex. Microglial containment of plaques is compromised in TREM2-deficient animals. Microglia accumulate autophagosomes. |
No data. |
Trem2 KO (Colonna) x PS19
Observed
-
Gliosis at 36
Microgliosis and astrogliosis by 9 months (the earliest age studied).
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2, MAPT | MAPT P301S | Trem2: Knock-Out; MAPT: Transgenic | Frontotemporal Dementia, Alzheimer's Disease | Microgliosis, astrogliosis, and brain atrophy in Trem2-/-PS19 mice are greatly attenuated compared with Trem2+/+PS19 animals. |
No data. |
Trem2 KO (JAX)
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | Trem2: Knock-Out | Alzheimer's Disease, Frontotemporal Dementia, Nasu-Hakola Disease | No data.. |
No data. |
Trem2 KO (KOMP)
Observed
Absent
-
Cognitive Impairment at
At six months, mice perform normally in the open-field test, elevated plus maze, three-chamber social-interaction test, and contextual and cued fear-conditioning test.
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | Trem2: Knock-Out | Nasu-Hakola Disease, Frontotemporal Dementia, Alzheimer's Disease | No data. |
At six months, mice perform normally in the open-field test, elevated plus maze, three-chamber social-interaction test, and contextual and cued fear-conditioning test. |
Trem2 KO (KOMP) x APPPS1
Observed
-
Plaques at 9
Plaques are observed by 2 months.
-
Gliosis at 9
Gliosis is observed by 2 months.
Absent
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2, APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1 L166P | Trem2: Knock-Out; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Reduced plaque burden at early stages of plaque deposition but increased plaque burden at later stages, fewer plaque-associated myeloid cells and astrocytes, less phospho-tau in plaque-associated dystrophic neurites, compared with APPPS1. |
No data. |
Trem2 KO (KOMP) x htau
Observed
-
Gliosis at 24
Microgliosis observed by 6 months, younger ages were not studied.
Absent
-
Neuronal Loss at
Neuron loss not observed in cortex or hippocampal field CA3 at 6 months of age; later ages were not studied.
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Mapt, MAPT, Trem2 | Mapt: Knock-Out; MAPT: Transgenic; Trem2: Knock-Out | Nasu-Hakola Disease, Alzheimer's Disease, Frontotemporal Dementia | Tau phosphorylation and aggregation in the cortex are enhanced in htau mice lacking TREM2, but reactive microglia are smaller and their processes have fewer branches. |
No data. |
Trem2*R47H(HSS)
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | TREM2 R47H | Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
Trem2 R47H KI (Haass)
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | TREM2 R47H | Trem2: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
Trem2 R47H KI (JAX)
Observed
Absent
-
Plaques at
Not observed in cortex or hippocampus up to 24 months of age.
-
Tangles at
Not observed in cortex or hippocampus up to 24 months of age.
-
Neuronal Loss at
Not observed in cortex or hippocampus up to 24 months of age.
-
Cognitive Impairment at
Locomotor activity, motor coordination, and working memory similar to wild-type at 2 and 12 months of age.
No Data
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | TREM2 R47H | Trem2: Knock-In | Alzheimer's Disease | No neuron loss, amyloid plaques, or neurofibrillary tangles were observed in mice up to 24 months of age. |
Locomotor activity, motor coordination, and working memory similar to wild-type at 2 and 12 months of age. |
Trem2 R47H KI (Lamb/Landreth)
Observed
Absent
-
Plaques at
No 6E10- or Thioflavin S-positive amyloid plaques were observed at 4 months of age.
No Data
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | TREM2 R47H | Trem2: Knock-In | Alzheimer's Disease | No 6E10- or Thioflavin S-positive amyloid plaques were observed at 4 months of age. |
Unknown. |
Trem2 R47H KI (Lamb/Landreth) X APPPS1-21
Observed
-
Plaques at 16
Reduction in the number and burden of fibrillar amyloid plaques in the hippocampus, compared with APPPS1-21 mice homozygous for wild-type Trem2.
-
Gliosis at 16
Fewer plaque-associated myeloid cells in APPPS1-21;Trem2+/R47H, compared with APPPS1-21 mice homozygous for wild-type Trem2.
Absent
-
Tangles at
Tangles were not observed at 4 months of age, but hyperphosphorylated tau was detected in dystrophic neurites surrounding plaques.
-
Neuronal Loss at
No differences in neuron number in cotical layer V in APPPS1-21;Trem2+/R47H mice relative to APPPS1-21 mice homozygous for wild-type Trem2, at 4 months of age.
No Data
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2, APP, PSEN1 | TREM2 R47H, APP K670_M671delinsNL (Swedish), PSEN1 L166P | Trem2: Knock-In; APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Reduction in the number and burden of fibrillar amyloid plaques in the hippocampus, fewer plaque-associated myeloid cells, and worse plaque-associated neuritic dystrophy, compared with APPPS1-21 mice homozygous for wild-type Trem2. |
Unknown. |
Trem2 R47H KI x APOE4 (LOAD1)
Observed
Absent
-
Plaques at
Not observed in cortex or hippocampus up to 24 months of age.
-
Tangles at
Not observed in cortex or hippocampus up to 24 months of age.
-
Neuronal Loss at
Not observed in cortex or hippocampus up to 24 months of age.
-
Gliosis at
Microgliosis not observed in cortex or hippocampus up to 24 months of age.
-
Cognitive Impairment at
Age-related changes in locomotor activity, motor coordination, and working memory, but no genotype-dependent differences through 24 months of age, compared with wild-type mice.
No Data
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APOE, Trem2 | TREM2 R47H, APOE C130R (ApoE4) | APOE: Knock-In; Trem2: Knock-In | Alzheimer's Disease | No neuron loss, amyloid plaques, neurofibrillary tangles, vascular leakage, myelin loss, or reactive microglia in mice up to 24 months of age. |
Age-related changes in locomotor activity, motor coordination, and working memory, but no genotype-dependent differences through 24 months of age, compared with wild-type mice. |
Trem2 R47H knock-in
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2, App | TREM2 R47H | Trem2: Knock-In; App: Knock-In | Alzheimer's Disease | Unknown. |
Unknown. |
Trem2*R47H(NSS)
Observed
-
Synaptic Loss at 50
Synapse loss, assessed by co-localization of the pre-synaptic marker bassoon and postsynaptic marker PSD95, by 12 months.
-
Changes in LTP/LTD at 52
Impaired basal synaptic transmission and LTP, by 12 months.
Absent
-
Plaques at
Not observed.
-
Tangles at
Not observed.
-
Gliosis at
Similar numbers of Iba1-immunoreactive microglia in Trem2*R47HNSS and wild-type hippocampi and cortices, but differences in microglial morphology at 4 months that are gone by 12 months.
No Data
-
Neuronal Loss at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | TREM2 R47H | Trem2: Knock-In | Alzheimer's Disease | Changes in microglial morphology at 4 months but not 12 months, compared with wild-type. |
Unknown. |
TREM2-sol
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | Trem2: Knock-In | None | Unknown. |
Unknown. |
Trem2 T66M KI
Observed
-
Gliosis at 30
Fewer clusters of Iba1-immunoreactive microglia in Trem2 T66M mice compared with wild-type mice, becoming statistically significant at 7 months of age.
Absent
-
Plaques at
Not observed.
-
Tangles at
Not observed.
No Data
-
Neuronal Loss at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | TREM2 T66M | Trem2: Knock-In | Frontotemporal Dementia | Age-related microglial activation seen in wild-type mice is absent in homozygotes. |
No data. |
Trem2 Y38C KI
Observed
Absent
No Data
-
Plaques at
No data.
-
Tangles at
No data.
-
Neuronal Loss at
No data.
-
Gliosis at
No data.
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
-
Cognitive Impairment at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Trem2 | TREM2 Y38C | Trem2: Knock-In | Frontotemporal Dementia | No data. |
No data. |
Ts65Dn
Observed
-
Neuronal Loss at 26
By 6 months, there is a loss of basal forebrain cholinergic neurons in the medial septal nucleus. From 10-11 months, a decrease in ChAT+ motor neurons are observed. Additional deficits include reduced brain volume, impaired neurogenesis, decreased neuronal density, and abnormal dendritic spine morphology, which are present in earlier stages of development.
-
Gliosis at 42
Ts65Dn mice show a developmental shift from neuronal to astrocytic lineage, leading to an increased percentage of astroglial cells in the cortex and hippocampus. By 10 to 18 months, an elevated density of CD45+ microglia cells are found in the hippocampus and basal forebrain, with IBA1 upregulation at 12 months and reduced expression of the homeostatic microglial marker P2RY12 at 15 months.
-
Synaptic Loss at 65
Ts65Dn mice have more inhibitory synapses, and fewer excitatory synapses. Synaptic density is decreased in both the hippocampus and neocortex, accompanied by enlarged pre-synaptic boutons and spines. Changes in the physical distribution of afferent inputs also occur.
-
Changes in LTP/LTD at 26
Ts65Dn mice demonstrate impaired hippocampal long-term potentiation (LTP) due to excessive GABA-mediated inhibition.
-
Cognitive Impairment at 13
Ts65Dn mice exhibit reduced attention, and deficits in hippocampal-dependent functions, including contextual fear conditioning, working memory, and long-term spatial memory.
Absent
-
Plaques at
Although Ts65Dn mice exhibit age-related increase in APP and Aβ levels in the cortex and hippocampus, they do not show plaque pathology. However, an increase in soluble Aβ oligomers and small amyloidal extracellular inclusions in the deep granular cell layer of the cerebellum has been reported.
-
Tangles at
Although Ts65Dn mice exhibit increased tau phosphorylation, they do not develop neurofibrillary tangles.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Multi-transgene | Down's Syndrome, Alzheimer's Disease | Elevation of APP and soluble Aβ in the brain without Aβ plaque pathology. Changes to tau 3R/4R splicing, augmenting tau mRNA stability and tau phosphorylation. Basal forebrain neurodegeneration; loss of NE neurons in locus coeruleus; hippocampal abnormalities; cholinergic neuron loss; reduced number of proliferating cells. Neuroinflammation and oxidative stress markers. |
Spatial learning and memory deficits, developmental delay in sensorimotor milestones, locomotor hyperactivity, lack of behavioral inhibition, and stereotypic behavior. Delayed motor acquisition, impaired coordination, reduced attention. |
WSB.APP/PS1
Observed
-
Plaques at 32
Thioflavin S-positive amyloid plaques are present in the cortex and CA1 region of the hippocampus by 8 months of age, with females having more plaques in the cortex than males.
-
Neuronal Loss at 34
Compared with their non-transgenic littermates, female WSB.APP/PS1 mice have fewer neurons in the cortex and in CA1. Neuron numbers in male mice do not differ between the genotypes.
-
Gliosis at 33
Plaque-associated microgliosis observed by 8 months.
-
Cognitive Impairment at 31
Deficits in short-term memory by 8 months in females (data from males unavailable).
Absent
-
Tangles at
Not observed.
No Data
-
Synaptic Loss at
No data.
-
Changes in LTP/LTD at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
APP, PSEN1 | APP K670_M671delinsNL (Swedish), PSEN1: deltaE9 | APP: Transgenic; PSEN1: Transgenic | Alzheimer's Disease | Amyloid plaques, plaque-associated gliosis, cerebral amyloid angiopathy; possible neuron loss in cortex and hippocampal area CA1 in females. |
Transgenic mice are hyperactive. Working memory (spontaneous alternation in the Y-maze) is normal at 7 to 8 months, but short-term memory (tested in the Y-maze) is impaired in females (data from males is not available, as wild-type males are unable to perform this test). |
PD-related Research Models
Phenotypes Examined
- Neuronal Loss
- Dopamine Deficiency
- α-synuclein Inclusions
- Neuroinflammation
- Mitochondrial Abnormalities
- Motor Impairment
- Non-Motor Impairment
When visualized, these phenotypes will distributed over a 18 month timeline demarcated at the following intervals: 3mo, 6mo, 9mo, 1yr, 15mo, 18mo+.
Atp13a2-flox Mouse
Observed
-
Neuroinflammation at 4
Germline Atp13a2 KO mice exhibit progressively worsening astrogliosis starting as early as 1 month of age, but in mice with adult-onset Atp13a2 KO in the ventral midbrain, astrogliosis and microglial reactivity were present only transiently after AAV-Cre delivery.
-
Motor Impairment at 39
Germline Atp13a2 null mice exhibit some motor defects on the open-field test (starting at 9 months of age) and in the tail suspension test (at 18 months of age), but other motor measures (including balance beam and Rotarod performance) did not differ from wild-type mice.
-
Neuronal Loss at 52
No cell loss in the substantia nigra pars compacta (SNpc) in germline Atp13a2 null mice by 18 months of age, but in mice with adult-onset Atp13a2 KO in the ventral midbrain (via AAV-Cre delivery), dopaminergic neuronal degeneration in the SN was observed 10 months after injection (i.e., in 12- to 16-month-old mice).
Absent
-
Dopamine Deficiency at
No differences observed between germline Atp13a2 null mice and wild-type mice in tyrosine hydroxylase levels (western blot) in the striatum or midbrain at 18 months of age.
-
α-synuclein Inclusions at
α-synuclein pathology absent in germline KO mice at 18 months of age, and also absent in mice with ventral midbrain, adult-onset Atp13a2 deletion.
No Data
-
Non-Motor Impairment at
No data.
-
Mitochondrial Abnormalities at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Atp13a2 | Atp13a2: Knock-Out | Parkinson's Disease | Adult-onset Atp13a2 KO in the ventral midbrain (via AAV-Cre delivery) resulted in neuropathology, including dopaminergic neuronal degeneration in the SN, which was not observed in germline Atp13a2 KO mice. |
Motor function partially impaired in germline Atp13a2 KO mice, including defects on the open-field and tail suspension tests, but not on the balance beam or Rotarod. |
DJ-1 KO Rat
Observed
-
Non-Motor Impairment at 17
Olfactory detection enhanced (16 mos). Short-term memory abnormal (4.5, 15 mos). Appetitive instrumental learning normal (4, 6, 8 mos). Coping behavior (forced-swim test) impaired (6 mos). No anxiety-like behavior (elevated plus maze; 4, 8, 17 mos), less anxiety on light-dark box (6, 8 mos). No sucrose preference at 9 mos. Sensorimotor function (adhesive removal) unaffected (4, 7, 13 mos).
-
Motor Impairment at 17
Abnormalities in gait and strength, vocalizations, and tongue movements were observed. By 4 months, the rats exhibited abnormal paw positioning and a shorter stride. Males showed impaired licking, longer and more frequent ultrasonic vocalizations, and an accelerated decrease in average call intensity with age. Fine motor skills were also impaired in KO versus wild-type rats by 7 months of age.
-
Mitochondrial Abnormalities at 13
At 3 months of age, the mitochondrial proteome in DJ-1 KO rats was differentially expressed compared to wild-type rats. Mitochondrial respiration was also increased in KO versus wild-type rats
-
Neuronal Loss at 26
Age-related decreases in TH-positive dopaminergic neurons were reported in the substantia nigra and locus coeruleus reaching approximately 50 percent by 8 months of age. No change was found in TH-immunoreactivity in the ventral tegmental area or striatum.
Absent
-
Dopamine Deficiency at
Striatal dopamine level was increased 2-3 fold in KO rats compared to wild-type levels at 8 months of age. Dopaminergic innervation of the dorsal striatum was intact in DJ-1 KO rats at 4 and 6 months of age compared to wild-type rats. Basal levels of dopamine metabolites and evoked levels of dopamine in the striatum were not different between KO and wild-type rats.
-
α-synuclein Inclusions at
Staining for α-synuclein revealed no increase in the striatum or in any other brain region assessed.
No Data
-
Neuroinflammation at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Park7 (DJ1) | Park7 (DJ1): Knock-Out | Parkinson's Disease | Age-related decrease in dopaminergic neurons in the substantia nigra and locus coeruleus; approximately 50 percent reduction by 8 months. Striatal dopamine and serotonin levels elevated 2-3fold over wild-type levels. No change was found in TH-immunoreactivity in the ventral tegmental area or striatum. |
Enhanced olfactory detection, abnormal short-term memory, impaired coping behavior, reduced anxiety on light-dark box. Abnormalities in gait, paw positioning, strength, vocalizations, and tongue movements. In males, impaired licking, longer and more frequent ultrasonic vocalizations, and accelerated decrease in average call intensity with age. |
Gba1 D409V KI Mouse (Grabowski)
Observed
-
Non-Motor Impairment at 17
Memory is impaired starting at 4 months in homozygous KI mice based on the novel object recognition and contextual fear-conditioning tests. In D409V/null mice, memory was impaired at 9 months; heterozygous KI mice did not exhibit memory deficits at 6 months. Anxiety- and compulsive-like behaviors were perturbed in D409V/null mice at 6 months, based on the marble burying test.
-
α-synuclein Inclusions at 18
Progressive α-synuclein accumulation starting at 6 months of age in homozygous and heterozygous KI mice as well as in D409V/null mice, and may be present even as early as 4 months of age, in the forebrain and hippocampus.
-
Motor Impairment at 14
Homozygous KI mice do not exhibit gait abnormalities or locomotion based on the open-field test, at 8 and 12 months, respectively. However, D409V/null mice exhibit perturbances in gait as early as 3 months of age.
Absent
-
Neuroinflammation at
No neuroinflammation observed at 12 months of age in the hippocampus based on GFAP and Iba-1 immunostaining.
-
Neuronal Loss at
No neurodegeneration in the hippocampus, striatum, or substantia nigra at 12 months of age.
No Data
-
Dopamine Deficiency at
No data.
-
Mitochondrial Abnormalities at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Gba1 | Gba1: Knock-In | Parkinson's Disease | No neurodegeneration in the hippocampus, striatum, or substantia nigra at 12 months of age. No neuroinflammation observed at 12 months of age in the hippocampus based on GFAP and Iba-1 immunostaining. Progressive α-synuclein accumulation starting as early as 4 months of age in the forebrain and hippocampus. |
Memory impaired starting at 4 months in homozygous KI mice and at 9 months in D409V/null mice. Anxiety- and compulsive-like behaviors perturbed in D409V/null mice at 6 months. Gait and locomotion normal in homozygous KI mice at 8 and 12 months, but impaired gait in D409V/null mice as early as 3 months. |
Gba1 D409V KI Mouse (MJFF)
Observed
-
Dopamine Deficiency at 55
Dopamine levels did not differ at 4, 8, and 12 months of age, but dopamine turnover (ratio of DOPAC and HVA to dopamine) tended to increase, though the increase was only significant at 12 months of age.
-
Non-Motor Impairment at 54
Cognitive performance was impaired in 12-month-old heterozygous KI mice (but not at 3, 6, or 9 months), based on the Morris water maze and Y-maze. Anxiety-like behavior (based on the open-field test) did not differ at 12 months.
-
α-synuclein Inclusions at 54
Homozygous KI mice have higher levels of soluble monomeric α-synuclein in the hippocampus at 12 months than heterozygous KI mice and wild-type controls. Levels of pathologic phosphorylated form pS129 do not differ between homozygous KI mice and controls in the substantia nigra, cortex, or hippocampus.
-
Neuroinflammation at 54
Data are mixed on levels of GFAP and Iba-1 immunostaining in KI mice brain. One study in homozygous KI mice found no differences in the striatum and substantia nigra at 4, 8, or 12 months of age; another found decreased GFAP staining in the substantia nigra at 12 months; and a third study (het mice) found increased GFAP and Iba-1 in the hippocampus at 12 months.
-
Motor Impairment at 36
Homozygous D409V KI mice generally exhibit motor function similar to wild-type controls (open-field, Rotarod, grip strength, swim velocity). However, a couple of exceptions found in one study were greater grip strength force at 12 months of age and transiently increased locomotor activity on the open-field test at 8 months of age.
Absent
-
Neuronal Loss at
No differences in the number of dopaminergic neurons in the substantia nigra pars compacta were found between homozygous KI mice and wild-type mice at 4, 8, and 12 months of age.
No Data
-
Mitochondrial Abnormalities at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Gba1 | Gba1: Knock-In | Parkinson's Disease | No deficits in dopaminergic neuron numbers in the substantia nigra pars compacta between homozygous KI mice and wild-type mice. Dopamine levels did not differ, but dopamine turnover (ratio of DOPAC and HVA to dopamine) tended to increase at older ages. Mixed findings on GFAP and Iba-1 staining across brain regions in KI mice. |
Motor function is largely intact in KI mice, apart from transient increases in locomotor activity and increased grip strength at 8 and 12 months, respectively. Cognitive performance was impaired at 12 months of age, but not at younger ages. Anxiety-like behavior was not affected. |
Gba1 L444P KI Mouse (JAX)
Observed
-
α-synuclein Inclusions at 52
α-synuclein deposition reported in the striatum of 1-year-old mice in one study, but no differences in α-synuclein levels found in brain extracts of 6- and 14-month-old KI mice in another study.
-
Neuroinflammation at 53
Increased GFAP immunoreactivity observed in the striatum of 1-year-old mice in one study, but no differences in striatal GFAP or Iba-1 immunostaining observed in another study of 14-month-old KI mice.
Absent
-
Motor Impairment at
No deficits in motor balance, as detected by Rotarod and balance beam tests, observed in 16-month-old L444P KI mice.
-
Neuronal Loss at
No loss in dopaminergic cell numbers in the substantia nigra at 14 months of age.
No Data
-
Dopamine Deficiency at
No data.
-
Non-Motor Impairment at
No data.
-
Mitochondrial Abnormalities at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Gba1 | Gba1: Knock-In | Parkinson's Disease | No loss of dopaminergic cells in the substantia nigra at 14 months. Inconsistent results regarding α-synuclein deposition and striatal GFAP immunostaining. |
No deficits in motor balance, as detected by Rotarod and balance beam tests, observed in 16-month-old L444P KI mice. |
Gba1 L444P KI Mouse (MMRRC)
Observed
-
Non-Motor Impairment at 13
Impaired contextual, but not cued, fear conditioning at 3 months of age in heterozygous KI mice. No deficits in olfaction (buried pellet test) or on the novel object recognition test at 24 months of age.
-
Neuroinflammation at 104
GFAP staining was comparable in heterozygous KI and wild-type mice at 8 and 24 months of age. Iba1 staining, however, was increased in 24-month-old heterozygous KI mice, but only in the granule cell layer of the olfactory bulb.
-
Mitochondrial Abnormalities at 35
By 8 months of age, heterozygous KI mice have impaired mitochondrial structure (smaller) and function (lower levels of mitochondrial DNA) in the midbrain. Mitochondrial function from cultured cortical neurons also impaired (increased reactive oxygen species generation, decreased mitochondrial complex I enzyme activity, decreased oxygen consumption rate).
Absent
-
Dopamine Deficiency at
Levels of dopamine, DOPAC (3,4-dihydroxyphenylacetic acid), HVA (homovanillic acid), or their ratio (to assess dopamine turnover) were similar in saline-treated heterozygous KI and wild-type mice at 8 months of age.
-
α-synuclein Inclusions at
α-synuclein levels are increased in the ventral midbrain in heterozygous L444P KI mice at 8 months, as well as in other brain regions assessed at 24 months. Another study, however, reported no differences in total synuclein levels at 3 months, but a decrease in soluble phosphorylated α-synuclein. There is no evidence of α-synuclein aggregates in this model.
-
Motor Impairment at
No differences between heterozygous KI and wild-type mice in open-field test performance at 3 months. At 8 months, heterozygous KI mice also performed at similar levels to wild-type controls on pole and grip strength tests. Heterozygous KI mice may perform better on the pole test at younger (3 months) ages. Pole test performance was also similar between genotypes at 24 months of age.
-
Neuronal Loss at
No deficits in the number of TH-positive neurons in the substantia nigra pars compacta or in the density of TH-immunopositive fibers in the striatum in 8-month-old heterozygous L444P KI mice.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Gba1 | Gba1: Knock-In | Parkinson's Disease | Dopaminergic neuron numbers are intact in the substantia nigra. Some alterations in soluble α-synuclein levels, but no findings of aggregation. Levels of dopamine and its metabolites are not perturbed in heterozygous KI mice. Neuroinflammation as measured by GFAP is not observed in heterozygous KI mice, but Iba1 staining may be increased in some regions. |
Largely no differences in motor function between heterozygous KI and wild-type mice from 3 to 24 months of age. Impaired contextual, but not cued, fear conditioning at 3 months of age in heterozygous KI mice, but no deficits in olfaction or on the novel object recognition test at 24 months of age. |
HGBA L444P Tg on Gba1 KO Mouse
Observed
Absent
-
α-synuclein Inclusions at
Older HGBA L444P /Gba−/− mice do not show evidence of α-synuclein inclusions.
-
Motor Impairment at
No obvious phenotypic features noted.
-
Neuronal Loss at
Older HGBA N370S/Gba−/− mice do not show evidence of neuropathology.
No Data
-
Dopamine Deficiency at
-
Non-Motor Impairment at
-
Neuroinflammation at
-
Mitochondrial Abnormalities at
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
GBA1, Gba1 | GBA1: Transgenic; Gba1: Knock-Out | Parkinson's Disease | Older mice do not show evidence of neuropathology or α-synuclein inclusions. Significant progressive elevations in central nervous system glucosylsphingosine, but not glucosylceramide, were noted. |
No obvious phenotypic features noted. |
HGBA N370S Tg on Gba1 KO Mouse
Observed
Absent
-
α-synuclein Inclusions at
Older HGBA N370S/Gba−/− mice do not show evidence of α-synuclein inclusions.
-
Motor Impairment at
No obvious features noted.
-
Neuronal Loss at
Neuronal Loss Older HGBA N370S/Gba−/− mice do not show evidence of neuropathology.
No Data
-
Dopamine Deficiency at
-
Non-Motor Impairment at
-
Neuroinflammation at
-
Mitochondrial Abnormalities at
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
GBA1, Gba1 | GBA1: Transgenic; Gba1: Knock-Out | Parkinson's Disease | Older transgenic mice do not show evidence of neuropathology or α-synuclein inclusions. Significant progressive elevations in central nervous system glucosylsphingosine, but not in glucosylceramide, were noted. |
No obvious features noted. |
LRRK2 R1441C KI Mouse
Observed
-
Non-Motor Impairment at 30
Acoustic startle reflex equal to wild-type mice at 12 months of age. Intracellular protein transport impaired in primary cultured cells. PKA activity is elevated in the striatum. Ciliation in striatal cholinergic neurons is decreased at 7 months of age and primary cilia formation is perturbed in the somatosensory cortex.
Absent
-
Dopamine Deficiency at
Basal levels of striatal dopamine, DOPAC, and HVA were comparable between KI and wild-type mice at 3, 12, and 23 months of age. However, evoked dopamine release in the striatum was reduced in adult heterozygous KI mice.
-
α-synuclein Inclusions at
No abnormal accumulation of α-synuclein observed at 3, 12, and 22 months of age in the substantia nigra pars compacta or locus coeruleus.
-
Neuroinflammation at
GFAP immunoreactivity was normal at 12 and 22 months of age. However, upon α-synuclein fibril injection, KI mice exhibited increased infiltration of pro-inflammatory monocytes into the brain.
-
Motor Impairment at
Spontaneous locomotor activity (open-field test) equal to wild-type mice at 3, 12, and 24 months of age. Involuntary motor movement (Rotarod) equal to wild-type mice 3 and 12 months of age.
-
Neuronal Loss at
No loss of dopaminergic (TH-immunoreactive) neurons in the substantia nigra pars compacta at 12 and 22 months of age. No loss of TH-immunoreactive neurons in the locus coeruleus.
No Data
-
Mitochondrial Abnormalities at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
LRRK2 | LRRK2: Knock-In | Parkinson's Disease | No loss of dopaminergic (TH+) neurons in the substantia nigra pars compacta at 12 and 22 months. No loss of TH-immunoreactive neurons in the locus coeruleus. Basal levels of striatal dopamine, DOPAC, and HVA were comparable between KI and wild-type mice at 3, 12, and 23 months. However, evoked dopamine release in the striatum was reduced in adult heterozygous KI mice. |
Acoustic startle reflex equal to wild-type mice at 12 months of age. Motor learning impaired upon antagonism of dopamine receptors (D1 and D2). |
LRRK2 G2019S KI Mouse
Observed
-
Non-Motor Impairment at 16
Altered responses to social-defeat stress (males, 3-4 mos) which correlated with changes in striatal plasticity and intrinsic membrane excitability. Attention deficits, slower information processing, impaired goal-directed learning in 2-6-month-old male KI mice, but cognitive flexibility and novel objective recognition are intact (2-6 mos). Perturbed sleep behavior at 8-10 mos.
-
Motor Impairment at 81
A battery of motor tests revealed no baseline deficits at 3-4, 12-13, and 18-19 months of age. However, an increased locomotor response after amphetamine challenge is observed at 18 months. Motor defects are exacerbated following a manganese stressor.
Absent
-
Dopamine Deficiency at
Striatal dopamine levels do not differ at 2 months of age, and neither do tyrosine hydroxylase levels in the substantia nigra.
-
Neuronal Loss at
The cytoarchitecture of the neocortex, striatum, hippocampus, and elsewhere is normal in Nissl-stained brain sections of 3-4 month-old mice, and striatal levels of tyrosine hydroxylase are similar to those of controls at P21. In another study, tyrosine hydroxylase levels are reduced in the striatum and midbrain at 2 months of age.
No Data
-
α-synuclein Inclusions at
No data.
-
Neuroinflammation at
No data.
-
Mitochondrial Abnormalities at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
LRRK2 | LRRK2: Knock-In | Parkinson's Disease | Levels of phospho-substrates of LRRK2 (e.g., Rab10) are increased in the brain. Endocytosis and axonal transport defects in neurons. Cholinergic innervation density is lower in the prelimbic/infralimbic cortical areas and dorsomedial striatum, but not in the dorsal lateral geniculate nucleus in 2-6-month-old males. Microglial immunostaining is similar in the striatum and midbrain at 8 weeks |
Attention deficits, slower information processing speeds, and impaired goal-directed learning are evident in 2-6-month-old mice—deficits rescued by systemic administration of the acetylcholinesterase inhibitor donepezil. Cognitive flexibility and novel objective recognition similar to controls. Sleep behavior is perturbed at 8-10 months of age. |
LRRK2 G2019S Mouse (BAC Tg)
Observed
-
Dopamine Deficiency at 52
Age-related decline in striatal dopamine content. Levels were decreased at 12 months of age, but not significantly different from controls at 6 months of age. Also, decreased dopamine metabolite homovanillic acid (HVA).
-
Non-Motor Impairment at 26
Tg mice spend less time in the REM sleep phase at 12 and 18 months of age. Age-dependent increase in plasma corticosterone (present starting at 6-8 months of age). Nuclear envelope integrity is perturbed in dopaminergic neurons at 12 months.
-
Neuroinflammation at 8
Application of α-synuclein fibrils leads to exacerbated responses (more inclusions and greater infiltration of pro-inflammatory monocytes).
-
Motor Impairment at 78
Behavior in hemizygous mice was comparable to littermate controls in terms of activity levels (open-field test) and coordination (beam-walk test) at 6 and 12 months, , but not at 18 months of age, when Tg mice develop motor deficits (Rotarod).
-
Mitochondrial Abnormalities at 0
Primary cultured cells from Tg mice exhibit mitochondrial fragmentation and membrane depolarization.
Absent
-
α-synuclein Inclusions at
No evidence of α-synuclein inclusions up to 18 months of age. However, there is mixed evidence on whether cultured mutant hippocampal neurons have increased levels of α-synuclein protein. After exposure to exogenous α-synuclein fibrils, mutant neurons developed more α-synuclein inclusions than non-Tg neurons.
-
Neuronal Loss at
No evidence of neuronal or other cell death in any brain region, including the cortex, striatum, and hippocampus. There was no difference in the number of dopaminergic neurons in the substantia nigra compared to littermate controls at 6 or 12 months.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
LRRK2 | LRRK2 G2019S | LRRK2: Transgenic | Parkinson's Disease | Brain appears normal. No neuronal or cell death at 12 months. Impaired neurite motility and synaptic vesicle endocytosis in cultured neurons. No increase in α-synuclein or ubiquitin levels or aggregation; however, cultured neurons developed more inclusions when exposed to exogenous α-synuclein fibrils. Decreased striatal dopamine content, decreased evoked release. |
Apparently normal behavior. No change in activity level or motor coordination at 12 months. Motor deficits appear at 18 months. |
LRRK2 G2019S Mouse (Tg)
Observed
-
Non-Motor Impairment at 45
Anxiety/depression-like symptoms were observed at 10-12 months of age.
-
α-synuclein Inclusions at 52
Around 2 years of age, mice did not exhibit abnormalities in α-synuclein in the ventral midbrain, striatum, or cerebral cortex. However, one study found α-synuclein accumulation in whole brain lysates of 12- to 19-month-old transgenic mice.
-
Neuroinflammation at 63
Around 2 years of age, mice did not have GFAP abnormalities in the ventral midbrain, striatum, or cerebral cortex. However, activated microglia were reported in the striatum at 14 months, and CD68 and TNF-α levels were increased in whole brains at 4-6 months. Others have not observed differences in Iba-1 staining (microglial marker) at 6, 12, or 18 months in the striatum or substantia nigra.
-
Motor Impairment at 35
Rotarod performance deteriorated in 14- to 18-month-old mice, but minor deficits are already observed as early as 8 months of age. Muscle weakness observed on the hanging wire test by 8 months of age. No change in pre-pulse inhibition of the acoustic startle reflex.
-
Mitochondrial Abnormalities at 63
Increased numbers and condensation of mitochondria in striatal microglia were reported at 14 months. Abnormally high levels of condensed mitochondria were also observed in cortical and striatal neurons at 17-18 months.
-
Neuronal Loss at 83
By 19-21 months, mice lose 18 percent of TH-positive dopaminergic neurons in the substantia nigra pars compacta and 14 percent of dopaminergic dendrites in the substantia nigra pars reticulata. At 1-2 months neuronal numbers were normal. Some authors do not see differences in TH staining up to 2 years of age. No abnormal neuronal loss is observed in the ventral tegmental area or cerebellum.
Absent
-
Dopamine Deficiency at
At 14-15 months of age, hemizygous mice had normal levels of striatal dopamine, DOPAC, and HVA. However, in the olfactory bulb, levels of HVA and DOPAC were lower, but dopamine was unchanged.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
LRRK2 | LRRK2 G2019S | LRRK2: Transgenic | Parkinson's Disease | Age-dependent dopaminergic neuron degeneration in the substantia nigra, though reports are mixed. No reduction in striatal dopaminergic terminals or dopamine levels. Some reports of α-synuclein accumulation. Abnormal mitochondria in striatal neurons and microglia; accumulation of autophagic vacuoles. Evidence for activated striatal microglia and increased levels of CD68 and TNF-α in whole brain. |
Deterioration of Rotarod performance in 14- to 18-month-old mice. Muscle weakness observed on the hanging wire test by 8 months of age. No change in pre-pulse inhibition of the acoustic startle reflex. Anxiety/depression-like symptoms at 10-12 months. |
LRRK2 G2019S Rat (BAC Tg)
Observed
-
Non-Motor Impairment at 26
Bone marrow myeloid progenitor numbers were decreased, but suppressive myeloid cells were increased at 6 to 11 months of age
-
Motor Impairment at 35
Mild abnormalities in motor behavior. Slightly more postural instability at 8 months of age (but not at 4 and 12 months). Slightly more rearing events at 12 months, but not at younger ages.
Absent
-
Dopamine Deficiency at
No change in striatal dopamine levels. No change in 3,4-dihydroxyphenylacetic acid (DOPAC) levels. No change in the rate of dopamine turnover. At 12 months of age Tg rats exhibited higher levels of striatal homovanillic acid (HVA).
-
α-synuclein Inclusions at
Under basal conditions no α-synuclein inclusions were observed.
-
Neuroinflammation at
No increase in Iba-1 positive microglia or GFAP-positive astrocytes in the substantia nigra at 12 months of age. However, iNOS expression was elevated in nigral dopaminergic neurons.
-
Neuronal Loss at
No overt loss of dopaminergic neurons in the substantia nigra out to 12 months of age.
No Data
-
Mitochondrial Abnormalities at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
LRRK2 | LRRK2 G2019S | LRRK2: Transgenic | Parkinson's Disease | No overt neurodegeneration out to 12 months of age. Elongated dopaminergic neurons. Elevated oxidative and nitrosative stress. No evidence of gliosis. No α-synuclein inclusions until challenged with exogenous α-synuclein. No change in dopamine levels. |
Mild abnormalities in motor behavior. Slightly more postural instability at 8 months of age (but not at 4 and 12 months). Slightly more rearing events at 12 months, but not at younger ages. |
Lrrk2 KO Mouse
Observed
-
Neuroinflammation at 87
Striatal staining of GFAP, a marker of reactive astrocytosis, did not differ between control and KO mice, but cells positive for Iba1 staining, a marker of activated microglia, were moderately enlarged in the striatum of 20 -month-old KO mice. Cx3cr1 mRNA levels higher in KO mouse brains.
-
Motor Impairment at 52
Motor behavior is generally intact up to 18 months based on Rotarod and open field tests. However, some age-dependent effects are observed on the open field test: 12 -month-old mice traveled longer distances and had higher walking speeds versus controls, which was not apparent in 3- or 24-month-old mice. Older (24 months) mice had deficits in motor skill learning as measured by Rotarod.
-
Mitochondrial Abnormalities at 9
Adult (9 - to 23-week-old) Lrrk2 KO mice exhibit enhanced mitophagy in dopaminergic neurons of the substantia nigra pars compacta, as detected by an increase in the number of mitolysosomes.
Absent
-
Dopamine Deficiency at
Levels of TH in the striatum are equal between genotypes in 18- to 24-month-old mice.
-
Non-Motor Impairment at
No differences were observed between KO and wild-type mice across 6 to 24 months of age on several behavioral tests, including the elevated plus maze for anxiety-like behavior, the buried treat test to measure hyposmia, the grip strength test for forelimb strength, or working memory as measured by spontaneous alternation.
-
α-synuclein Inclusions at
No abnormal accumulation of α-synuclein in the cell bodies of striatal neurons observed in 20-month-old KO mice.
-
Neuronal Loss at
Neuronal Loss No differences between KO and wild-type mice up to 24 months of age in the number of tyrosine hydroxylase (TH)–positive cells in the substantia nigra pars compacta. No neurodegeneration markers observed in the striatum and cortex at 20 months. Cerebral cortex and dorsal (but not ventral) striatum volumes reduced at 12 months.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Lrrk2 | Lrrk2: Knock-Out | Parkinson's Disease | No loss in the number of TH–positive cells in the substantia nigra pars compacta up to 24 months. Cerebral cortex and dorsal striatum volumes reduced at 12 months. Striatal spiny projection neurons were enlarged and the frequency of nuclear invaginations was increased at 12 months, suggesting premature aging. Dendritic morphology perturbed at 12 months. |
No differences between KO and wild-type mice up to 24 months on several behavioral tests, including the elevated plus maze for anxiety-like behavior, the buried treat test to measure hyposmia, the grip strength test for forelimb strength, or working memory as measured by spontaneous alternation. Older (24-month-old) mice showed deficits in motor skill learning as measured by Rotarod. |
Lrrk2 KO Rat
Observed
-
Non-Motor Impairment at 5
Abnormalities occur in peripheral organs, most notably the kidney, but also the liver, lung, and spleen. Changes are progressive, although they do not appear to shorten lifespan. The earliest reported alterations occur in the kidneys at 1 month of age.
Absent
-
Dopamine Deficiency at
Basal levels of dopamine metabolites (3,4-dihydroxyphenylacetic and homovanillic acid) do not differ between Lrrk2 KO and wild-type rats at 4, 8, and 12 months of age. Evoked release of dopamine also does not differ between KO and wild-type rats.
-
Neuroinflammation at
When challenged with LPS or α-synuclein overexpression, Lrrk2 KO rats show lower levels of pro-inflammatory CD68-positive myeloid cells in the substantia nigra than wild-type rats.
-
Motor Impairment at
Assessment of Rotarod performance revealed no impairment at 12 months of age compared with wild-type rats.
-
Neuronal Loss at
Under basal conditions, the number of TH-positive cells in the substantia nigra is comparable between Lrrk2 KO and wild-type rats. When challenged with LPS or α-synuclein overexpression, Lrrk2 KO rats develop significantly less neurodegeneration in the substantia nigra than wild-type rats.
No Data
-
α-synuclein Inclusions at
No data.
-
Mitochondrial Abnormalities at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Lrrk2 | Lrrk2: Knock-Out | Parkinson's Disease | Not observed. Protection against dopaminergic cell loss under conditions involving LPS or α-synuclein overexpression in the substantia nigra. No changes in basal or evoked release of dopamine. |
One assessment of Rotarod performance revealed no impairment at 12 months of age compared with wild-type rats. |
LRRK2 R1441C Mouse (Tg - Conditional)
Observed
Absent
-
Dopamine Deficiency at
HPLC analysis of striata from 10-month-old mice revealed no significant differences in the levels of dopamine or its metabolites DOPAC and HVA.
-
Non-Motor Impairment at
Olfactory function, as assessed by the ability to locate buried food, was normal out to 20 months of age.
-
α-synuclein Inclusions at
Immunohistochemical analysis of the brain at 22 months did not reveal abnormalities in α-synuclein, and no proteinaceous inclusions were seen.
-
Neuroinflammation at
Immunohistochemical analysis of the brain at 22 months found GFAP and Iba1 immunoreactivity comparable to control levels.
-
Motor Impairment at
Around 20 months of age, R26-LRRK2 mice behaved normally, exhibiting no deficits in locomotor activity (open-field test), motor coordination (Rotarod), or gait (digital CatWalk system).
-
Neuronal Loss at
In the substantia nigra pars compacta, there was no difference in the number of TH-positive neurons or the total number of Nissl-positive neurons at 12 and 22 months.
No Data
-
Mitochondrial Abnormalities at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
LRRK2 | LRRK2 R1441C | LRRK2: Transgenic | Parkinson's Disease | No neurodegeneration in the brain. No proteinaceous inclusions of α-synuclein, ubiquitin, or tau. No reactive gliosis. No change in dopamine levels. Subtle morphological abnormalities in dopaminergic and non-dopaminergic neuronal nuclei, including altered nuclear envelope. |
No overt behavioral differences. Activity levels and Rotarod performance are normal into advanced age. |
LRRK2 WT Mouse (BAC Tg)
Observed
-
Dopamine Deficiency at 52
Striatal dopamine levels, as measured by PET imaging with [18F]FDOPA uptake, are higher in WT-OX versus non-Tg mice.
-
Motor Impairment at 52
WT-OX mice (12 months) are hyperactive on several parameters of the open-field test. Gait analysis (Cat-Walk system) was also perturbed relative to non-Tg controls. However, the number of rears did not differ.
Absent
-
Mitochondrial Abnormalities at
Mitochondrial morphology and levels of proteins involved in mitochondrial fission (Drp1 and Fis1) are normal at 12 months of age.
No Data
-
Non-Motor Impairment at
No data.
-
α-synuclein Inclusions at
No in vivo data, but α-syn colocalization with LAMP-2 is increased in cultured neurons from WT-OX mice.
-
Neuroinflammation at
No data.
-
Neuronal Loss at
No data on neuron numbers are available, but neurite length is reduced in primary hippocampal neurons and primary nigral tyrosine hydroxylase-positive neurons of WT-OX mice versus non-Tg mice.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
LRRK2 | LRRK2: Transgenic | Parkinson's Disease | Overtly normal brain structure. Intact, but shorter, neurites. |
Motor hyperactivity at 12 months of age. |
MCI-Park Mouse
Observed
-
Dopamine Deficiency at 5
Profound loss of evoked dopamine release in the dorsolateral striatum as early as 30 days. In contrast, somatodendritic dopamine release in the SN did not differ between genotypes at 30 days, but was dramatically reduced by 60 days.
-
Non-Motor Impairment at 2
Impaired associative learning (Y-maze test) at 30 days . Impaired sleep functions starting at 6 weeks of age, with significantly altered sleep-wake patterns (total, NREM, and REM sleep), increased sleep fragmentation, and altered EEG activity.
-
Motor Impairment at 6
Striatal motor learning (adhesive removal test) was impaired starting at 30 days. Rearing in the cylinder testing was impaired at 40 days. Total distance travelled was decreased by 60 days on the open-field test. By 100 days, splayed hindlimbs, abnormal paw placement, and alterations in stride observed.
-
Mitochondrial Abnormalities at 3
By 20 days mitochondria were in an oxidative phosphorylation deficit. Altered mitochondria structure, but not mitochondrial density, was observed at 35 days in dopaminergic neurons of the SN. Metabolic reprogramming to a glycolytic-predominant state of mitochondria was indicated by alterations in expression of genes and functional pharmacologic experiments.
-
Neuronal Loss at 4
TH expression decreased at 30 days in the dorsal striatum. By age 60 days, TH expression decreased in substantia nigra dopaminergic neurons. No neurodegeneration was observed in axons, cell bodies, or dendritic arbors of SN dopaminergic neurons at 60 days. By 120 to 150 days, neurodegeneration is present and about 40% of SN dopaminergic neurons are lost.
Absent
No Data
-
α-synuclein Inclusions at
No data.
-
Neuroinflammation at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Conditional Knock-out | Parkinson's Disease | Progressive reductions in TH expression in the striatum and SN. Overt neuron loss in the SN only at older ages (>120 days). Electrophysiological measurement of SN dopaminergic neurons showed altered pacemaking activity and burst spiking. Altered expression of many neuronal genes. Loss of evoked dopamine release in the dorsolateral striatum at 30 days of age. |
Impaired associative learning and striatal motor learning at 30 days. Impaired rearing at 40 days. Decreased total distance travelled by 60 days. By 100 days, splayed hindlimbs, abnormal paw placement, and alterations in stride. Impaired sleep functions starting at 6 weeks. |
Parkin KO Mouse
Observed
-
Dopamine Deficiency at 11
Levels of striatal dopamine and metabolites DOPAC and HVA were normal at 6, 12, 18, and 24 months. In another study, striatal extracellular dopamine was increased, as measured by no-net-flux microdialysis, in 8-9-month-old mice. In a third study, evoked striatal dopamine release was reduced in striatal slices of 2-4-month-old mice.
-
Non-Motor Impairment at 9
Novel object recognition was decreased at 4-5 months of age and reduced sociability, increased repetitive behaviors, and deficits in communication were present at 2-3 months of age. Outcomes from the forced swim test, time spent investigating novel odors, latency to find buried food, the Barnes maze test, hot plate test, Morris water maze appear unaffected.
-
Motor Impairment at 19
On the beam traversal task, KO mice displayed deficits starting at 2-4 months of age. General behavior (beam breaks) on the open-field test did not differ at 6, 12, and 18 months of age. Findings on the Rotarod suggest no differences or that KO mice may have enhanced performance.
-
Mitochondrial Abnormalities at 7
Mitochondrial defects begin at 7 weeks. Proteomic analyses reveal differences in ventral midbrain lysates of proteins involved in mitochondrial function. Respiratory and antioxidant capacity, mitophagy, and mitochondrial DNA are affected. Mitochondrial structure appears intact in the brain, but is affected in heart tissue.
Absent
-
α-synuclein Inclusions at
Inclusions of α-synuclein were not observed in any brain region.
-
Neuroinflammation at
Spinal cord staining of GFAP did not differ between non-transgenic and parkin KO mice at 130 days of age.
-
Neuronal Loss at
In the substantia nigra, the number of dopaminergic neurons (as detected by TH staining) did not differ between parkin KO and wild-type mice up to 24 months. Dopaminergic projections in the striatum were also normal.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Park2 | Park2: Knock-Out | Parkinson's Disease | No loss of dopaminergic neurons in the substantia nigra. Striatal dopamine levels may be increased basally, but reduced when evoked. Striatal medium spiny neurons had reduced synaptic excitability at 6–9 months and impaired LTP and LTD at 8 weeks, but other passive and active membrane properties were unaffected. No α-synuclein inclusions or astrogliosis. |
Outcomes from the forced swim test, time spent investigating novel odors, latency to find buried food, the Barnes maze test, hot plate test, Morris water maze appeared unaffected. In contrast, novel object recognition was decreased at 4-5 months and reduced sociability, increased repetitive behaviors, and deficits in communication were present at 2-3 months. |
Parkin KO Rat
Observed
-
Non-Motor Impairment at 9
Orientation to an olfactory stimulus was normal. At 2 months of age, male KO rats had a greater preference for methamphetamine than wild-type rats based on self-administration and place preference tests.
-
Mitochondrial Abnormalities at 14
Alterations in mitochondrial protein expression in synaptic and nonsynaptic striatal samples of 3-month-old KO rats.
-
Neuronal Loss at 35
A small, non-significant reduction in dopaminergic neurons was observed in the substantia nigra at 8 months of age.
Absent
-
Dopamine Deficiency at
No differences in striatal dopamine levels at 4, 6, or 8 months. Altered dopaminergic transmission factors in the striata, including MAO, β-phenylethylamine, trace amine-associated receptor 1, and postsynaptic dopamine D2 receptors in 2-month-old KO rats. Striatal dopamine metabolite levels decreased with age in KO rats, showing lower levels at 12 months than at 8 months.
-
α-synuclein Inclusions at
There was no increase in α-synuclein protein in the striatum or any other brain region assessed.
-
Motor Impairment at
No behavioral deficits were detected at 4, 6, and 8 months of age. Motor functioning, including performance on the Rotarod, was intact. However, at 2 months, male KO rats made fewer small stereotypic movements, such as scratching and grooming, than wild-type controls.
No Data
-
Neuroinflammation at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Park2 | Park2: Knock-Out | Parkinson's Disease | No significant changes in dopaminergic neurons in the substantia nigra nor striatal dopamine levels, but alterations in dopaminergic signaling were detected at an early age, as were disruptions in mitochondrial protein expression in striatum. No increase in α-synuclein. Evoked release of striatal glycine greater at 12 months versus wild-type rats. |
No behavioral deficits detected at 4, 6, and 8 months of age. However, at 2 months, male KO rats made fewer small stereotypic movements, such as scratching and grooming, than wild-type controls. At 2 months, male KO rats had a greater preference for methamphetamine than wild-type rats. |
Parkin Q311X Mouse (BAC Tg)
Observed
-
Dopamine Deficiency at 69
Surviving nigral neurons at 16 months of age had reduced tyrosine hydroxylase expression. By 19-21 months, striatal concentrations of dopamine and the dopamine metabolite DOPAC were decreased compared with non-Tg littermates.
-
Non-Motor Impairment at 68
Autophagy and lysosomal dysfunction in mutant mice at 16-17 months of age.
-
Non-Motor Impairment at 71
Autophagy and lysosomal dysfunction in mutant mice at 16-17 months of age.
-
α-synuclein Inclusions at 72
Lewy body-like inclusions were not observed at any age, however, mutant mice exhibit age-dependent accumulation of proteinase-K resistant endogenous α-synuclein in the substantia nigra at 16 months of age.
-
Motor Impairment at 70
Behavior was fairly normal at 3 months, but motor abnormalities were detected by 16 months of age, including hypoactivity and deficits in coordination and in motor response to sensory stimuli.
-
Mitochondrial Abnormalities at 4
Mitochondrial dysfunction observed as early as 1 month of age, based on electron microscopy (e.g., lacking an outer membrane, swollen) and expression of the short isoform of OPA1.
-
Neuronal Loss at 26
Progressive loss of dopaminergic neurons in the substantia nigra, starting as early as 6 months of age. About 40 percent loss by 16 months of age with a corresponding decrease in dopaminergic projections to the striatum. Neurons in the ventral tegmental area were relatively spared.
Absent
No Data
-
Neuroinflammation at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Park2 | Parkin Q311X | Park2: Transgenic | Parkinson's Disease | Degeneration of dopaminergic neurons in the SN and nerve terminals in the striatum. Reduced dopamine in the striatum. Accumulation of proteinase-K resistant α-synuclein and oxidative protein damage. Dysfunction in the burst-firing pattern activity of dopaminergic SN neurons and increased expression of markers for excitotoxic damage. |
Late-onset hypoactivity (about 16 months of age), other modest changes in motor behavior and coordination in tests that included traversing a beam or removing adhesive. |
Parkin S65A KI Mouse
Observed
-
Motor Impairment at 53
Impaired performance on the raised balance beam at 12 and 18 months of age in homozygous KI mice. No deficits in Rotarod performance or gait analysis.
-
Mitochondrial Abnormalities at 52
Mitochondrial respiration (respiratory control ratio) was impaired in an age-dependent manner—at 12 months, but not at 3 months—in homozygous KI mice. No deficits in basal mitophagy.
Absent
-
Dopamine Deficiency at
No differences in levels of striatal dopamine and 3,4-DOPAC, nor in their ratio, between 18-month-old homozygous KI mice and wild-type mice.
-
Neuroinflammation at
Immunolabeling of astrocytes (GFAP) and microglia (Iba1) did not differ between homozygous Parkin KI mice and wild-type mice.
-
Neuronal Loss at
No deficits in striatal anatomy or volume or in nigrostriatal innervation in 18-month-old homozygous KI mice.
No Data
-
Non-Motor Impairment at
No data.
-
α-synuclein Inclusions at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Park2 | Park2 S65A | Park2: Knock-In | Parkinson's Disease | No evidence of nigrostriatal neuropathology in 18-month-old homozygous mice. |
Motor dysfunction on the raised balance beam by 12 months of age. No deficits in Rotarod performance or gait. |
PINK1 G309D (PINK1-/-) Mouse (KI)
Observed
-
Dopamine Deficiency at 39
Decreased dopamine concentration in the striatum by 9 months of age.
-
Neuroinflammation at 81
Expression of factors involved in Toll-like receptor signaling were increased in the cerebellum, as were astrocytic and microglial markers in the corticospinal tract and striatum at 18 months.
-
Motor Impairment at 70
At 16 months of age Pink1-/- mice exhibited decreased spontaneous locomotor activity. Strength and coordination were intact.
-
Mitochondrial Abnormalities at 13
By 3 months of age the mice exhibited a mitochondrial import defect. This phenotype was more severe at 6 months and import was reduced nearly 50% by 12 months of age. By 6 months, ATP production, respiration, and mitochondrial membrane potential were also reduced.
Absent
-
Non-Motor Impairment at
Mutant mice performed similarly to wild-type mice in tests assessing the startle reflex, sweating, and anxiety.
-
α-synuclein Inclusions at
No Lewy body-like inclusions or α-synuclein aggregates in the brainstem or substantia nigra, but expression levels of α-synuclein are altered in brainstem/midbrain.
-
Neuronal Loss at
Neuronal loss was not observed at 18 months of age (total neuronal population and TH-positive subset).
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
PINK1 | Pink1 G309D | PINK1: Knock-In | Parkinson's Disease | No neuronal loss. No Lewy bodies or α-synuclein aggregates, but α-synuclein expression change in brainstem/midbrain. Low dopamine levels. Mitochondrial dysfunction (e.g., reduced ATP, reduced respiratory activity). Increase in factors involved in Toll-like receptor signaling in the cerebellum, and increased astrocytic and microglial markers in the corticospinal tract and striatum. |
Reduced spontaneous locomotor activity in open-field test. No difference in strength or coordination. |
PINK1 KO Mouse
Observed
-
Non-Motor Impairment at 0
Modest vocalization deficits observed at 4-6 months. Reduced BDNF levels in the midbrain and cortex at 10 months. Cardiac hypertrophy observed at 2 and 6 months of age.
-
Motor Impairment at 23
Reduced spontaneous locomotor activity and skill reported at 3-6 months.
-
Mitochondrial Abnormalities at 9
Altered shape, density, and movement of dendritic mitochondria observed in cultured primary neurons from embryonic mice. Also, an abnormal rise in serum cytokines in response to acute mitochondrial stress was reported in vivo. By 2 months of age, mitochondrial dysfunction observed in cardiomyocytes.
Absent
-
Dopamine Deficiency at
Overall striatal levels of dopamine did not significantly differ from levels in wild-type mice at 2-3 months or 8-9 months of age.
-
Neuronal Loss at
No decrease in the number of dopaminergic neurons in the substantia nigra at 2-3 months or 8-9 months of age. Neuronal morphology also grossly intact.
No Data
-
α-synuclein Inclusions at
No data.
-
Neuroinflammation at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Pink1 | Pink1: Knock-Out | Parkinson's Disease | Normal numbers of dopaminergic neurons and tyrosine hydroxylase levels in substantia nigra at 8-9 months of age. Alterations in the dendrites of midbrain dopaminergic neurons and cultured cortical neurons. Altered shape, density, and movement of dendritic mitochondria in cultured primary neurons from embryonic mice. Reduced BDNF levels in the midbrain and cortex at 10 months. |
Reduced spontaneous locomotor activity and skill at 3-6 months. Modest vocalization deficits at 4-6 months. |
Pink1 KO Rat
Observed
-
Non-Motor Impairment at 9
Nociception alterations in male KO rats observed at 6 to 10 months of age, indicating thermal hyperalgesia. This effect was present in female KO rats at 2 months of age, but not at older ages. Abnormalities in ventilation frequency were also observed in male KO rats. Defects in ultrasonic vocalizations starting at 2 months of age in male and female KO rats.
-
α-synuclein Inclusions at 18
Alpha-synuclein aggregates were found as early as 4 months of age and increased in number up to 12 months. Areas affected include the periaqueductal gray, substantia nigra pars compacta, locus coeruleus, nucleus ambiguous, cortex, thalamus, and striatum.
-
Motor Impairment at 5
Abnormalities in gait, coordination, and strength. By 5 weeks, KOs had increased foot slips on the tapered balance beam, at 7 weeks they showed hind limb fatigue, which progressed to hind limb dragging, and by 2 months they exhibited alterations in oromotor behaviors. Deficits in gait may be transient. Partial reversal of motor impairment by Levodopa.
-
Mitochondrial Abnormalities at 18
Alterations in mitochondrial metabolites and mitochondrial protein expression were reported as early as 4 months of age in cortex and striatum. Oxygen consumption rates were elevated in striatal mitochondria isolated from 9-month-old rats, but not in non-synaptic samples from 3-month-old rats.
-
Neuronal Loss at 11
Age-related decrease in tyrosine hydroxylase (TH)-positive dopaminergic neurons in the substantia nigra. A reduction of 25 and 50 percent at 6 months and 8 months, respectively. Deficits in TH staining in the substantia nigra have been observed as early as at 2.5 months of age. While some studies did not see any changes in TH-positive cells in the striatum, others have observed a 15% loss.
Absent
-
Dopamine Deficiency at
One study found striatal dopamine levels were increased two- to threefold in Pink1 KO rats compared with wild-type levels at 8 months of age, whereas another reported a slight decrease at this age. In the dorsal striatum, KO rats have age-dependent differences in basal and evoked dopamine levels, but no differences were observed compared to wild-type rats.
No Data
-
Neuroinflammation at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Pink1 | Pink1: Knock-Out | Parkinson's Disease | Age-related decrease in dopaminergic neurons in the substantia nigra; greater than 50 percent reduction at eight months. Alterations in striatal dopamine and serotonin levels. Progressive increase in α-synuclein aggregates and reduced brain volume across numerous regions. Increased ventricular volume. Alterations in cortical and striatal mitochondria. |
Abnormalities in gait, coordination, and strength. As early as 5 weeks of age, KOs had increased foot slips on the tapered balance beam, at 7 weeks they showed hind limb fatigue, which progressed to hind limb dragging, and by 2 months they exhibited alterations in oromotor (lingual) behaviors. |
Thy1-αSyn “Line 61” Mouse
Observed
-
Dopamine Deficiency at 63
Line 61 mice lose striatal dopamine progressively, starting at about 14 months of age. Between approximately 6 and 10 months, however, the neurotransmitter’s extracellular levels are transiently increased.
-
Non-Motor Impairment at 11
Disruptions in olfaction, circadian rhythms, sleep, cognition, social behavior, and autonomic function have been reported. Several are reminiscent of PD non-motor impairments. Alterations in olfaction, circadian rhythms, and the autonomic regulation of heartrate occur as early as 3 months of age.
-
α-synuclein Inclusions at 4
Proteinase K-resistant aggregates are seen at 1 month and increase with age, including the substantia nigra, periqueductal gray, cortex, striatum, vagus, olfactory bulb, thalamus, locus coeruleus, and cerebellum, as well as cholinergic neurons in the colon. Elevated levels of phospho-serine 129 α-synuclein are found in the substantia nigra, striatum, cortex, frontal cortex, and hippocampus.
-
Neuroinflammation at 5
Neuroinflammation markers have been seen in the cortex, striatum, substantia nigra, and hippocampus. The time course and profile vary between brain regions and some features remain subject to debate. However, neuroinflammation appears to affect the striatum first (1 month), and then the substantia nigra (5–6 months). At older ages, particularly in the substantia nigra, inflammation attenuates.
-
Motor Impairment at 4
Impairments in balance, coordination, muscle strength, fine motor skills, vocalizations, and stress-induced defecation arise between 1 and 3 months. Transient hyperactivity is seen between 4 and 9 months of age, followed by hypoactivity and sensorimotor deficits at about 15 months. As the phenotype becomes more severe, the mice develop difficulty eating, akinesia, and hunched posture.
-
Mitochondrial Abnormalities at 18
The functions of mitochondrial respiratory complexes in midbrain and striatum are impaired, with the earliest reported deficit affecting complex I in the midbrain at 4 months. Elevated α-synuclein accumulation was found in mitochondria of the midbrain, striatum, and cortex.
-
Neuronal Loss at 16
Although neuron loss occurs in the neocortex and hippocampal CA3 region as early as 3–4 months of age, there is no reduction in the PD-relevant dopaminergic neurons of the substantia nigra, even at 22 months of age.
Absent
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
SNCA | SNCA: Transgenic | Parkinson's Disease |
Vps35 p.D620N KI Mouse
Observed
-
Dopamine Deficiency at 13
Enhanced peak amplitude in dopamine release and prolonged reuptake kinetics in acute striatal slices from 3-month-old mice; decreased DAT and increased VMAT2. Basal levels of dopamine and metabolites in dorsolateral striatum did not differ, but the DOPAC+HVA/DA ratio was increased. At 16 months, dopamine in striatal homogenates was reduced, but levels of metabolites (DOPAC, HVA) did not differ.
-
α-synuclein Inclusions at 65
No differences in α-synuclein puncta density or distribution in the SNpc at 3 months. No pathological α-synuclein observations seen throughout the brain at 13 months. However, at 15 to 16 months, increased somatic α-synuclein immunoreactivity found in the SNpc, and increased α-synuclein oligomers and aggregated α-synuclein observed in the ventral midbrain.
-
Neuroinflammation at 64
Increased GFAP immunostaining in the SNpc, but not in the striatum, of 15- to 16-month-old VKI mice; no GFAP differences observed at earlier ages. No differences in microgliosis (Iba-1 immunostaining) in the SNpc or the striatum up to 16 months of age.
-
Motor Impairment at 60
Motor deficiencies on the open-field test and the beam walking test appear at 14 months of age, but not earlier from 3 to even 13 months of age. However, performance on other motor tests—Rotarod and grip strength—did not differ at the advanced age (14 months). No deficits seen in the cylinder test (rearing) at 3 months. Amphetamine-induced hyperlocomotion is rescued by LRRK2 kinase inhibition.
-
Mitochondrial Abnormalities at 61
Mitochondrial structure, assesed by EM, was perturbed at 14 months, but not at 3 months, of age. Mitochondrial function—namely, the oxygen consumption rate—was reduced in older (15-month-old) mice.
-
Neuronal Loss at 56
Loss of TH-positive neurons in the SNpc and loss of TH-positive nerve terminals in the striatum at 13-16 months. Widespread axonal degeneration in the brain at 13 months.
Absent
-
Non-Motor Impairment at
No deficits in the buried pellet test, measuring olfactory function, from 6 to 14 months of age. No defects in gastrointestinal function (as measured by stool frequency and water content) up to 14 months of age.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
Vps35 | Vps35: Knock-In | Parkinson's Disease | Loss of TH+ neurons in SNpc and TH+ terminals in striatum at 13-16 mos. Widespread axonal degeneration at 13 mos. Increased somatic α-synuclein in SNpc; α-synuclein oligomers and aggregates in ventral midbrain at 15-16 mos. Increased somatodendritic tau/p-tau with age, but no neurofibrillary pathology. Increased GFAP in SNpc, but not striatum at 15-16 mos. No microgliosis up to 16 mos. |
No deficits in the buried pellet test, measuring olfactory function, from 6-14 months. Deficits in mood (anxiety/apathy) and/or cognition on elevated plus maze, starting at 3 months (unpublished). |
α-synuclein A30P/A53T Mouse (Tg)
Observed
-
Dopamine Deficiency at 9
Striatal dopamine concentrations were lower at all ages tested, including the earliest age, 2-3 months. Dopamine concentrations dropped with age, and levels of metabolites (e.g., DOPAC and HVA) were also lower in HM2 mice than non-Tg by 13-23 months of age.
-
Motor Impairment at 30
At young age 2-3 months, HM2 mice were more active than non-Tg controls, but by middle age (7-9 months) they were less active. At advanced ages (13-23 months), they also exhibited impaired coordination as measured by the time it took to right themselves from an inverted wire screen. However, no deficiencies in Rotarod performance, grip strength, or open-field movements were detected at 6 months.
-
Neuronal Loss at 34
Progressive loss of dopaminergic neurons was reported in the substantia nigra pars compacta (19 percent reduction at 8.5 months and 55 percent at 19 months).
Absent
-
α-synuclein Inclusions at
Inclusions were not observed at any age. Diffuse α- synuclein protein was both cytoplasmic and nuclear.
No Data
-
Non-Motor Impairment at
No data.
-
Neuroinflammation at
No data.
-
Mitochondrial Abnormalities at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
SNCA | SNCA A30P, SNCA A53T | SNCA: Transgenic | Parkinson's Disease | Progressively loss of dopaminergic neurons in the substantia nigra pars compacta, observed by 8.5 months. No α-synuclein inclusions. Morphological abnormalities in the dopaminergic system, including axonal and dendritic abnormalities, reduced dopamine concentration in the striatum. |
More active as young adults, then hypoactive compared to non-Tg. Also reduced motor coordination in old age as measured by the time to right from an inverted wire screen. |
α-synuclein A53T Mouse (Tg)
Observed
-
Dopamine Deficiency at 23
In symptomatic mice, striatal dopamine and metabolites DOPAC and HVA are comparable to wildtype, but at 5 months, striatal tyrosine hydroxylase is reduced. Increased D1 receptors in the substantia nigra and decreased dopamine transporters in the nucleus accumbens and striatum have been reported.
-
Non-Motor Impairment at 50
At 11–12 months, spatial memory was impaired as assessed by the Barnes circular maze.
-
α-synuclein Inclusions at 35
Prior to motor deficits, these mice develop accumulations of α-synuclein in select neuronal populations, including the midbrain, cerebellum, brainstem, and spinal cord. The protein aggregates do not resemble Lewy bodies, but are thioflavin-S-positive, indicating fibrillar structure.
-
Neuroinflammation at 40
In symptomatic mice, increased GFAP immunoreactivity was observed in select brain regions, including the dorsal midbrain, deep cerebellar nuclei, brainstem, and spinal cord. Cortex, hippocampus, and substantia nigra did not have increased reactivity compared with non-Tg controls.
-
Motor Impairment at 32
These mice develop severe motor impairment starting around 9-16 months of age. The deficits start out with mild hyperactivity at 7 months and progress to a wobbling movement, decreased activity, and ultimately paralysis and death.
-
Mitochondrial Abnormalities at 56
At 11–14 months, mitochondria in brainstem neurons were enlarged and their co-localization with the mitochondrial fission protein Drp1 was reduced.
Absent
-
Neuronal Loss at
Overt neuronal loss was not reported in these mice.
No Data
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
SNCA | SNCA A53T | SNCA: Transgenic | Parkinson's Disease | No overt neuronal loss. Alterations in dopaminergic-associated proteins in the striatum, substantia nigra, and nucleus accumbens. Region-specific neuronal accumulation of fibrillar α-synuclein, ubiquitin, and neurofilament-H, and accompanying astrocytosis. |
Early hyperactivity followed by severe motor impairment, manifesting as wobbling, posturing, decreased spontaneous locomotor behavior, inability to navigate the Rotarod, and ultimately paralysis and death. At 11–12 months, spatial memory impaired as assessed by the Barnes circular maze. |
α-synuclein A53T Mouse (Tg) on SNCA KO
Observed
-
Non-Motor Impairment at 14
By 3 months of age, the mice develop gastrointestinal dysfunction.
-
Motor Impairment at 26
By 6 months of age, homozygous mice became hypoactive, traveling less distance. This was not attributed to changes in exploratory behavior caused by anxiety. Also at 6 months, differences in performance on the accelerating Rotarod were seen.
Absent
-
Dopamine Deficiency at
No differences in striatal dopamine concentrations, or dopaminergic metabolites, homovanillic acid (HVA) and 3,4-dihydroxyphenylacetic acid (DOPAC) at 11 and 18 months of age.
-
α-synuclein Inclusions at
No evidence of Lewy body-like inclusions in the brain at any age. Likewise α-synuclein aggregates were not observed in the brain, although they did occur in enteric neurons in the gut.
-
Neuronal Loss at
No evidence of neuronal cell loss in the substantia nigra at 11 and 18 months of age, including dopaminergic neurons (TH-positive neurons) and total neurons.
No Data
-
Neuroinflammation at
No data.
-
Mitochondrial Abnormalities at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
SNCA | SNCA A53T | SNCA: Transgenic; SNCA: Knock-Out | Parkinson's Disease | No loss of dopaminergic neurons in the substantia nigra by 18 months of age. Rare dystrophic synapses in the hippocampus at advanced age, but no Lewy body-like pathology or α-synuclein aggregation in the brain. No change in striatal dopamine concentration. |
Impaired performance on the Rotarod and reduced spontaneous locomotor activity in open-field test. |
α-synuclein E46K Rat (BAC Tg)
Observed
-
α-synuclein Inclusions at 52
By 12 months of age, intracellular aggregates were observed in dopaminergic neurons of the substantia nigra and ventral tegmental area. Aggregates noted to be fairly small compared to those observed in PD brain. In the striatum and cortex α -synuclein accumulation appeared primarily in neuronal processes.
Absent
-
Dopamine Deficiency at
No dopamine deficiency in the striatum at 12 months of age. No serotonin deficiency in the striatum. Dopamine metabolites dihydroxyphenylacetic acid and homovanillic acid were reduced by approximately 25 percent and transmitter turnover was decreased.
-
Motor Impairment at
No overt motor differences out to 12 months of age, unless challenged with low-dose rotenone, upon which the rats exhibit bradykinesia, postural instability, and rigidity.
-
Neuronal Loss at
No overt loss of dopaminergic neurons out to 12 months of age.
No Data
-
Non-Motor Impairment at
No data.
-
Neuroinflammation at
No data.
-
Mitochondrial Abnormalities at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
SNCA | SNCA E46K | SNCA: Transgenic | Parkinson's Disease | No overt neuronal loss. Accumulation of mutant α-synuclein in the brain, in the form of diffuse staining and intracellular aggregates. Aggregates were largely restricted to dopaminergic neurons of the substantia nigra and ventral tegmental area. Elevated nitrotyrosine in dopaminergic neurons. |
No overt behavioral changes until challenged with low-dose rotenone, upon which the rats exhibit bradykinesia, postural instability, and rigidity. |
α-synuclein KO Mouse
Observed
-
Neuroinflammation at 16
Microglia cultured from Snca KO brain were more reactive, ramified. They had vacuole-like structures. Snca KO microglia exhibited exacerbated response to LPS, with greater secretion of pro-inflammatory cytokines.
-
Motor Impairment at 26
Motor function was largely intact. Normal performance on the Rotarod and in total distance travelled in the open field test. Subtle differences only (e.g., less rearing behavior than controls). They also spent less time in the center of the field, suggesting a possible anxiety-related phenotype.
-
Mitochondrial Abnormalities at 39
Mitochondrial abnormalities include reduced levels of the mitochondrial phospholipid cardiolipin and reduced activity of electron transport chain complex I/III.
Absent
-
Non-Motor Impairment at
The mice had normal reflexes and sensory abilities. Also, learning and memory appeared intact at 6-10 months of age, as assesed by the Morris water maze and tests of conditioned fear memory.
No Data
-
Dopamine Deficiency at
A possible modest reduction in striatal dopamine level, but highly variable from mouse to mouse.
-
α-synuclein Inclusions at
No data.
-
Neuronal Loss at
No gross abnormalities in the brain.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
SNCA | SNCA: Knock-Out | Parkinson's Disease | No gross brain abnormalities. Electron microscopy revealed synaptic vesicle abnormalities in hippocampal neurons, i.e., fewer vesicles in the reserve pool. |
Behavior is largely normal. Normal performance on the Rotarod. Subtle differences in locomotor activity (e.g., less rearing) but normal overall distance travelled. Learning and memory appear intact. Possible anxiety-like phenotype. |
α-synuclein KO Mouse (Conditional)
Observed
Absent
No Data
-
Dopamine Deficiency at
No data.
-
Non-Motor Impairment at
No data.
-
α-synuclein Inclusions at
No data.
-
Neuroinflammation at
No data.
-
Motor Impairment at
No data.
-
Mitochondrial Abnormalities at
No data.
-
Neuronal Loss at
No data.
Genes | Mutations | Modification | Disease | Neuropathology | Behavior/Cognition |
---|---|---|---|---|---|
SNCA | SNCA: Conditional Knock-out | Parkinson's Disease | No data. |
No data. |
ALS-related Research Models
- Sex-specific differences
- Cortical Neuron Loss
- Lower Motor Neuron Loss
- Cytoplasmic Inclusions
- Gliosis
- NMJ Abnormalities
- Muscle Atrophy
- Motor Impairment
- Body Weight
- Premature Death
AAV-GFP-(GA)50
Observed
At 6 months, neuron loss observed in cortex, including layer V of motor cortex.
No Data
No data.
Observed
Neuronal poly(GA)- and ubiquitin-positive inclusions already present at 4-6 weeks; rare TDP-43-positive inclusions.
Observed
Astrogliosis, but not microgliosis, in cortex and hippocampus, seen at 6 months.
No Data
No data.
No Data
No data.
Observed
Deficits in tail-suspension and rotarod tests, seen at 6 months.
Observed
Decreased body weight in males, but not females, seen at 6 months.
No Data
No data.
AAV-GFP–(GR)100
Observed
Fewer cortical neurons than controls, observed as early as 6 weeks of age.
Absent
Not observed up to 6 months of age.
Observed
Very rare TDP-43 inclusions, observed at 6 months.
Observed
Astrogliosis and microgliosis in cortex and hippocampus, observed at 6 weeks.
No Data
No data.
No Data
No data.
Observed
Progressive motor deficits, seen in open-field, rotarod, rod-walk, and wire-hang tests.
Absent
Normal at 6 months of age.
No Data
No data.
C9-BAC500 (Brown)
Absent
Not observed.
Absent
Not observed.
Observed
No cytoplasmic mislocalization, or aggregation of TDP-43 in the motor cortex. However, dipeptide repeats accumulated at advanced age and formed small perinuclear inclusion bodies positive for poly-GP.
Absent
No signs of increased activation of microglia or astrocytes in the brain or spinal cord.
Absent
No difference in denervation of neuromuscular junctions at 24 months of age. No difference in motor or sensory spinal nerve root axon number or morphology.
No Data
Muscle histology has not been reported, but no overt muscle atrophy was observed.
Absent
No overt motor deficit as measured by the Rotarod and grip strength.
Absent
Non-significant trend for male C9BAC mice to be heavier than non-Tg controls. Female data have not yet been reported.
Absent
Normal lifespan beyond 2 years in male mice. Female data have not yet been reported.
C9-BACexp (Baloh/Lutz)
Absent
Not observed.
Absent
Not observed.
Observed
RNA foci throughout the nervous system starting at 3 months of age. Foci comprised of RNA transcripts in both the sense and antisense directions. Age-associated formation of dipeptide aggregates, e.g., poly-GP.
Absent
No increase in GFAP staining in the brain and spinal cord compared with non-Tg controls, even at 18 months of age.
Absent
Not observed.
Absent
Not observed.
Absent
No abnormalities in grip strength, Rotarod performance, or open-field testing at a young age (3 months) or advanced age (18 months), compared with non-Tg controls.
Absent
No abnormalities in body weight at a young age (3 months) or advanced age (18 months) compared with non-Tg controls.
Absent
Normal lifespan.
C9orf72 Knock-out
Absent
Not observed.
Absent
Not observed.
Absent
Not observed.
Absent
Not observed.
Absent
Not observed.
Absent
Not observed.
Observed
Reduced activity on open-field test. No abnormalities in grip strength or Rotarod performance.
Absent
Not observed.
Absent
Not observed.
C9ORF72(AAV)(G4C2)149
Observed
Cortical neuron loss by 6 months.
No Data
No data.
Observed
Cytoplasmic inclusions containing dipeptide repeat proteins (DPRs) derived from sense RNA seen in the cortex, hippocampus, cerebellum, and spinal cord; inclusions containing DPRs produced from antisense transcripts seen in the cortex and occasionally in the hippocampus.
Observed
Astrogliosis in the cortex by 3 months.
No Data
No data.
No Data
No data.
Observed
Deficits in the hanging wire test emerge between 3 and 6 months.
No Data
No data.
No Data
No data.
C9ORF72(AAV)(G4C2)66
Observed
Compared with mice expressing 2-repeats, the 66-repeat mice had 17 percent fewer neurons in the cortex at 6 months of age and 11 percent fewer Purkinje cells in the cerebellum. At this age neurons in the hippocampus and thalamus were not affected.
Absent
At 6 months, neuronal loss in the spinal cord was not detected.
Observed
By 6 months, inclusions of C9RAN dipeptides were present in neurons of the cortex and hippocampus, and to a lesser extent in the cerebellum and spinal cord. Inclusions contained polyGA, polyGP, and polyGR dipeptides and were largely ubiquitin-positive.
Observed
Astrogliosis in the cortex by 6 months.
No Data
No data.
No Data
No data.
Observed
At 6 months, 66-repeat mice perform as well as 2-repeat mice on the Rotarod on the first day of testing. However, they fail to improve during subsequent trials, suggesting impairments in coordination and/or motor learning.
Observed
At 6 months females had a lower body weight than mice expressing 2-repeats. Body weight did not differ in males.
No Data
No data.
CamKII;(GR)80
Observed
Neuron loss in the cortex, beginning between 3 and 6 months of age.
No Data
No data, but note that the transgene is not expressed in these neurons.
Absent
No TDP-43 inclusions seen in mice studied up to 8 months of age.
Observed
Microgliosis and astrogliosis evident at 6 and 9 months, respectively.
No Data
No data.
No Data
No data.
Absent
No difference between CamKII;(GR)80 mice and CamKII-tTA control mice up to 9 months of age.
Absent
No difference between CamKII;(GR)80 mice and CamKII-tTA control mice up to 11 months of age.
No Data
No data.
Endogenous Sod1 D83G
Observed
Neuronal numbers comparable to wild-type at 15 weeks, but about 20 percent loss of upper motor neurons by 29 weeks. Neurons in layer V of the motor cortex appeared selectively vulnerable.
Observed
Neuronal numbers comparable to wild-type at 6 weeks, but loss occurred by 15 weeks. The neuronal loss then stabilized; it was not more severe at 52 weeks of age.
Absent
Not observed.
Observed
Gliosis, of both astrocytes and microglia, was evident in the spinal cord by 15 weeks. It was further elevated at 52 weeks.
Observed
Denervation of a hindlimb muscle, the extensor digitorum longus, was detected by 52 weeks of age, and a decreased number of motor units in the EDL muscle.
No Data
Muscle atrophy was not reported, although changes to the muscle composition and histochemistry were observed.
Observed
Both male and female mice develop a variety of progressive motor symptoms. Grip strength was reduced at 6 weeks of age. Tremors developed by about 5 months of age. Rotarod performance was impaired, at 23 weeks in females and 67 weeks in males.
Observed
Homozygous mice, both male and female, showed reduced body weight by 4 weeks of age in contrast to wild-type littermates. Loss of excessive body weight was the primary factor leading to euthanasia.
Observed
Males reach end-stage sooner than females (495 ± 22 versus 588 ± 24 days). Animals were sacrificed when weight loss exceeded 20 percent of maximum weight, in accordance with animal-use guidelines. This is likely explained by the development of hepatocellular carcinomas due to SOD1 loss of function.
FUS-R521C
Absent
No detectable loss of cortical neurons; however, neurons in the sensorimotor cortex show reduced dendritic complexity and reduced synaptic density.
Observed
No detectable difference in spinal motor neurons at P0. At P16, about 20% loss of ChAT-positive neurons in the anterior horn of cervical spinal cord. At P30-P60, about 50% loss of anterior horn neurons. Remaining motor neurons show reduced dendritic complexity and synaptic density.
Observed
Less than 10% of spinal motor neurons have cytoplasmic FUS inclusions.
Observed
Prominent increase in microgliosis and astrogliosis in the anterior horn of the spinal cord by end stage.
Observed
Reduced innervation of neuromuscular junctions in the diaphragm.
Observed
The majority of mice have severe skeletal muscle atrophy in the hindlimb by end stage.
Observed
Early postnatal motor impairment, including abnormal hindlimb clasping when lifted by the tail, gait abnormalities, and impaired Rotarod performance.
Observed
Early postnatal growth is retarded, and the mice experience progressive loss of body weight.
Observed
The majority of mice in the N1F1 generation reached end stage and were sacrificed by postnatal day 100. Mice in subsequent generations live longer: about 40% reach end stage by postnatal day 200.
FUSDelta14 Knock-in
No Data
No data.
Observed
No lower motor neuron loss at 3 months but 14% reduction in the number of motor neurons in lumbar spinal cord at 12 months and 20% reduction by 18 months.
Absent
No increase in the number of spinal motor neurons that contain p62- or ubiquitin-positive inclusions in aged FUSDelta14 mice compared with aged wild-type mice.
No Data
No data.
Observed
Reduction in the number of intact neuromuscular junctions (fully innervated endplates) in hindlimb lumbrical muscles by 18 months of age.
No Data
No data.
Observed
Normal motor activity at 3 months of age but impaired hindlimb function by 15 months.
No Data
No data.
Observed
Reduced survival starting at 19 months of age.
FUSΔ14 (FUSd14)
Absent
Not observed.
Absent
Not observed.
Observed
Neuronal cytoplasmic inclusions were present by 3 months of age in the cerebral cortex. Inclusions occurred in about 20% of neurons and often co-labeled with ubiquitin.
Absent
No obvious astrogliosis or microglial activation at 3 months of age in the cerebral cortex.
No Data
No data.
No Data
No data.
Absent
When the mice were sacrificed at 3 months of age, they appeared healthy and displayed no obvious motor phenotype.
No Data
No data.
No Data
No data.
FusΔNLS
No Data
No data.
Observed
Approximately 30% reduction of motor neuron numbers in the dorsal spinal cord.
Observed
Ubiquitin pathology was observed in motor neurons, but p62 inclusions were not.
Observed
Slight increase in oligodendrocytes in the spinal cord white matter.
Observed
Fibrillation and fasciculation potentials were observed in the gastrocnemius or tibialis anterior muscles. There was also a reduction in compound muscle action potential amplitude.
No Data
No data.
Observed
By 10 months, mice demonstrated irregular walking patterns and reduced hang time on the inverted grid test. No deficits in grip strength or rotarod performance. Paralysis was not observed.
Absent
By 22 months, no weight loss was observed.
Absent
Life span was not altered.
hFUS (+/+) (PrP-hFUS)
Absent
In the brain, overt neuronal loss was absent at end stage (~11 weeks).
Observed
By end stage (~11 weeks), homozygous mice had lost about 60 percent of α-motor neurons in the anterior horn of the lumbar spinal cord.
Observed
By end stage, cytoplasmic FUS inclusions, described as “granular” and “globular,” develop in the spinal cord of homozygous mice. Ubiquitin-positive inclusions also develop, but do not co-localize with FUS inclusions. Neurons in the brain also develop “granular” and “skein-like” FUS inclusions.
Observed
By end stage (~11 weeks), homozygous mice had microgliosis and astrogliosis in the anterior horn of the spinal cord and in the white matter of the dorsal columns. Reactive gliosis was absent in the brain, despite cytoplasmic inclusions of FUS in some neurons.
Observed
Homozygous mice had about 20 percent fewer functional motor units in the extensor digitorum longus muscle as estimated by neurophysiological assessment.
Observed
Muscle histology from end stage (~11 weeks) homozygous mice showed focal muscle atrophy in hindlimb muscles.
Observed
Homozygous mice exhibited motor symptoms at four weeks of age, starting with a tremor and mild signs of hindlimb dysfunction, including gait abnormalities. Motor symptoms progressed quickly, ultimately developing into hindlimb paralysis.
Observed
Homozygous mice fail to gain weight normally starting about four weeks of age. Their body weight declines in subsequent weeks, often precipitating euthanasia.
Observed
Homozygous mice were euthanized between 10 and 13 weeks of age when they developed severe motor impairment or lost 25 percent of their body weight. Average survival was 82 ± 12 days.
hFUS-P525L
No Data
No data.
Observed
Progressive loss of motor neurons at lumbar level 5.
Absent
No FUS inclusions were observed.
Observed
Astrocytosis and microgliosis were observed in the spinal cord.
Observed
Progressive denervation of hindlimb muscles. Decreased density of synaptic vesicles and mitochondria with normal morphologies. Altered electrophysiological properties.
Observed
Reduced fiber diameter in the tibialis anterior muscle.
Observed
Deficits in wire hang test at 360 days.
No Data
No data.
Absent
Not observed by 360 days.
hFUS-R521C
No Data
No data.
Observed
Progressive loss of motor neurons at lumbar level 5.
Absent
No FUS inclusions were observed.
Observed
Astrocytosis and microgliosis were observed in the spinal cord.
Observed
Progressive denervation of hind limb muscles.
No Data
No data.
No Data
No data.
No Data
No data.
Absent
Not observed by 360 days.
hPFN1-G118V
Observed
At 202 days, there was a decrease in the number of corticospinal neurons of the motor cortex.
Observed
Progressive loss of ventral horn neurons from 165 through 202 days of age.
Observed
Spinal cord motor neurons had TDP-43 puncta.
Observed
Astrocytosis and microgliosis were observed in the spinal cord at end stage.
Observed
Denervation of gastrocnemius muscle at end stage. Muscle action potential also had reduced amplitude.
Observed
At 165 days, hind limb muscle atrophy was observed.
Observed
Progressive motor impairments began ~ 120 days. Mice demonstrated tremors, limb clasping, muscle weakness, gait abnormalities, as well as reduced locomotion and decreased performance on the Rotarod.
Observed
Body weight peaked ~ 150 days and then progressively decreased.
Observed
Mice were sacrificed at an average of 202 days when they were unable to right themselves. Females on average reached 191 days while males attained 213 days.
hTDP-43ΔNLS
Observed
Severe neuronal degeneration in the dentate gyrus and deep layers of the neocortex. Other regions, such as the hippocampal CA1 subfield and olfactory bulb, were relatively resistant to neurodegeneration. Approximately 50 percent of dentate gyrus neurons were lost one month after the transgene was activated.
Absent
Not observed.
Absent
High levels of cytosolic TDP-43 but only very rare aggregates (observed in less than 1 percent of cortical neurons and even rarer in other brain regions, such as the hippocampus and striatum).
Observed
Severe astrogliosis and microgliosis in areas affected by neurodegeneration, including cortical and hippocampal regions, as well as the corticospinal tract.
No Data
Unknown.
Absent
Not observed.
Observed
Spastic motor impairment indicated by an abnormal clasping response as early as one week after transgene induction. A variety of motor deficits develop by one month after transgene induction, including impaired coordination on the Rotarod and decreased grip strength.
No Data
Unknown.
Absent
Not observed.
NEFH-tTA x hTDP-43ΔNLS
Observed
Decreased cortical thickness indicative of neuronal degeneration beginning at four weeks off dox. By end stage, rNLS8 mice had significantly smaller brains than non-Tg littermates.
Observed
rNLS8 lost motor neurons in the lumbar spinal cord by six weeks off dox.
Observed
Cytoplasmic inclusions of TDP-43 occur as early as one week off dox in neurons in the brain. Inclusions accumulate over time and are present in many brain regions, including the motor cortex. TDP-43 inclusions are relatively rare in the spinal cord. Ubiquitin-positive inclusions are also seen.
Observed
Astrogliosis develops in many brain regions, including layer V of the motor cortex.
Observed
Denervation of the hindlimb muscle tibialis anterior was detectable by four weeks off dox, that is, two weeks prior to detectable loss of lower motor neurons.
Observed
At end-stage, rNLS8 mice exhibit gross muscle atrophy of the hindlimb muscles tibialis anterior and gastrocnemius.
Observed
rNLS8 mice develop a variety of motor impairments, starting with a deficit in hindlimb clasping and a fine tremor in the forelimb and/or hindlimb. They also develop progressive loss of grip strength (as measured by the wire-hang test) and a progressive decline in coordinated movement and balance (as measured by the accelerating Rotarod).
Observed
Body mass peaked at approximately 7 weeks of age (i.e., two weeks off dox) and then progressively dropped. Excessive loss of body weight (>30% decrease from peak weight) often defined end-stage.
Observed
rNLS8 mice die prematurely. They reach end-stage 8-18 weeks off dox, with a median survival of 10.3 weeks off dox.
PFN1-C71G
Absent
No neuronal loss in the cortex but neurodegeneration in medulla.
Observed
By 4 months there was a loss of cervical motor neurons and an increase in degenerating axons.
Observed
Cytoplasmic inclusions of PFN1, ubiquitin, and p62 in motor neurons around 6 months.
Observed
Microgliosis and astrogliosis observed in the dorsal horn by 5 months.
Observed
Denervation of gastrocnemius muscle occurs by 5 months.
Observed
Muscle atrophy in lower hind limb occurs by 6 months.
Observed
By 4 months, mice began showing minor gate changes and at 5-6 months they began demonstrating progressive deficits in Rotorod performance, vertical behaviors, and grip strength.
Observed
Body weight peaked at 4-6 months and then progressively decreased.
Observed
Mice were sacrificed when they were incapable of locomotion following the paralysis of two or more limbs which occurred around 7 months of age.
PrP-hFUS (R495X)
Absent
Not observed.
Absent
Not observed.
Absent
Despite high levels of cytoplasmic FUS, neuronal inclusions were not observed.
No Data
No data.
Observed
A number of abnormalities were detected in the hindlimb musculature by electromyography (EMG). These phenotypes were detectable by 8-12 months of age and included fibrillation potentials, muscle denervation, and a reduction in the number of motor units.
No Data
No data.
Absent
Not observed.
No Data
No data.
Observed
Hemizygous mice sporadically developed intestinal swelling leading to premature death (mean survival 118 days). Homozygous mice were more severely affected (50 percent of the original cohort died around 59 days of age).
PrP-hFUS (WT)
No Data
No data.
No Data
No data.
Absent
Not observed.
No Data
No data.
Absent
Not observed.
No Data
No data.
Absent
Not observed.
No Data
No data.
Observed
Hemizygous mice die prematurely following a brief illness characterized by poor feeding. Mean survival of hemizygotes ~203 days.
SOD1 (G37R)
Absent
Upper motor neuron loss was not observed, although vacuolization occurred in brainstem neurons.
Observed
Motor neurons in the spinal cord and brainstem degenerated with overt neuronal loss in the ventral horn in some regions of the spinal cord by 19 weeks. The degenerative process involved extensive vacuolization.
Absent
Not observed.
Observed
Astrogliosis occurs in the spinal cord by 11 weeks of age, becoming more severe with age.
Observed
Denervated endplates have been observed.
Observed
Loss of motor axons, denervated endplates, atrophy of muscle fibers, and fiber type grouping observed by end-stage.
Observed
Motor impairment at 4-6 months, beginning with reduced spontaneous movement, then tremors, limb weakness, and poor grooming. Eventual paralysis of the hindlimbs.
Observed
Loss of body weight is observed.
Observed
Mice survive about 6 to 8 months.
SOD1-G85R (hybrid)
No Data
Unknown.
Observed
Extensive degeneration of large spinal axons coincident with the onset of clinical symptoms. By end stage, motor neurons in the ventral horn are lost.
Observed
Astrocytic inclusions occur early, about 6 months of age. The inclusions are immunoreactive for SOD1 and ubiquitin.
Observed
Astrogliosis and microgliosis are observed in the spinal cord starting around 6.5 months of age, and become more severe with age.
Observed
Denervation of muscle fibers is observed.
Observed
Hemizygous mice develop muscle weakness around 9 months of age, coincident with atrophy and denervation of muscle fibers.
Observed
Progressive motor impairment generally starting around 8 months with reduced grip strength in one hindlimb, rapidly spreading to other limbs and leading to paralysis within about two weeks.
Observed
Hemizygous mice start to lose weight at about 9 months of age.
Observed
End stage is characterized by paralysis at about 10 months of age.
SOD1-G93A (hybrid) (G1H)
Observed
Although outright upper motor neuron loss is absent or rare, degenerative signs (e.g., swollen neurites, Gallyas-positive aggregates, vacuoles, and neuritic spheroids) have been shown in motor regions of the cerebral cortex by five months of age.
Observed
Up to 50% loss of motor neurons in the cervical and lumbar segments of the spinal cord at end stage.
Observed
Inclusions accumulate in spinal motor neurons starting around 82 days of age. Inclusions generally take the form of spheroids or Lewy-body-like inclusions and commonly include a variety of neuronal intermediate filament proteins. TDP-43-positive inclusions are not present.
Observed
Gliosis, including the proliferation of reactive microglia and astrocytes, develops in parallel with motor neuron degeneration in the spinal cord.
Observed
Neuromuscular junctions degenerate around 47 days of age; fast-fatiguable motor neurons are affected first.
Observed
Longitudinal MRI has shown reduced muscle volume as early as 8 weeks of age. Atrophy is progressive. Skeletal muscle is affected, including limb and diaphragm.
Observed
Signs of motor impairment begin at about 3 months of age with a shaking tremor that leads to paralysis.
Observed
One of the first signs of illness is a slowing of growth and a plateauing of weight.
Observed
G1H mice reach end-stage disease by 5 months of age. Females typically survive longer than males.
TARDBP (A315T) (congenic)
No Data
These mice lose corticospinal tract axons, but outright loss of cortical neurons has not been reported in the model. When crossed with a Thy-1YFP model to label layer 5 pyramidal neurons, mice expressing TDP-43 (A315T) had fewer neurons at 15 weeks of age than YFP littermate controls (Zhang et al., 2016).
Absent
Most studies reported no lower motor neuron loss. One study observed 20% loss of large ventral horn neurons, possibly dependent on diet and how long the mice live in an individual colony.
Observed
Ubiquitinated inclusions in the cytoplasm of spinal motor neurons and cortical layer V neurons. No evidence for cytoplasmic TDP-43 inclusions.
Observed
Reports of astrocytosis in cortical layer 5 and in the spinal cord, as well as microgliosis in the spinal cord.
Observed
Denervation of neuromuscular junctions at end stage (~11% on normal diet; ~20% loss on a gel diet).
Observed
Atrophy of gastrocnemius muscle (gel diet).
Observed
Deficits have been reported in nonspecific measures of strength and coordination such as the Rotarod (males and females) and hanging-wire test (males). A severely impaired gait (“swimming gait”) was observed in mice fed a gel diet.
Observed
Weight loss is a consistent feature. Potentially confounded by severe gut phenotype.
Observed
Survival is limited by severe gastrointestinal dysfunction and can be prolonged with a gel diet. Lifespan varies, but in general on a standard diet males live about 3 months and females about 6 months.
TARDBP (A315T) (hybrid)
Observed
By end-stage, neuronal numbers in layer 5 of the motor cortex are decreased with about 50 percent loss of corticospinal tract axons.
Observed
By end-stage, ~20% loss of motor neurons in the L3-L5 region of the spinal cord.
Observed
By end-stage, cytoplasmic inclusions of ubiquitinated proteins in layer 5 neurons of motor, sensory, and cingulate cortex. Ubiquitin aggregates in ventral horn neurons. TDP-43 inclusions were rare.
Observed
By end-stage, selective increase in GFAP immunoreactivity in cortical layer 5.
No Data
Unknown.
Observed
By end-stage, atrophic muscle fibers were observed.
Observed
Gait abnormalities around three months of age, developing into a characteristic “swimming gait” by four to five months.
Observed
Weight was comparable to non-Tg mice at birth. By 4.5 months transgenic mice began to lose weight.
Observed
Survival for about 5 months (154 ± 19 days) before dying spontaneously or being euthanized. It was not reported if this analysis includes males, females, or both.
Tardbp Q331K Knock-In
No Data
No data.
No Data
No data.
No Data
No data.
No Data
No data.
No Data
No data.
No Data
No data.
No Data
No data.
No Data
No data.
No Data
No data.
Tardp LCDmut
No Data
No data.
Observed
28 percent reduction in the number of motor neurons in the sciatic motor neuron pool, compared with non-transgenic littermates.
Observed
At 18 months of age, p62- and ubiquitin-positive inclusions are found in the ventral regions of the spinal cord, although these inclusions do not appear to be located in the cytoplasm of motor neurons.
No Data
No data.
Observed
By 24 months, a 15 percent reduction in motor units innervating the extensor digitorum muscle.
No Data
No data.
Observed
Grip strength in both male and female mice begins to decline at 12 months of age. By 24 months, there is a nearly 40 percent reduction in force measured in tibialis anterior muscles.
No Data
No data.
Absent
Do not exhibit premature death, at least until 24 months of age.
Tardp_RRM2mut
No Data
No data.
Absent
Not observed at 2 years.
Absent
Not observed at 2 years.
No Data
No data.
Absent
Not observed at 2 years.
No Data
No data.
Absent
Not observed at 2 years.
No Data
No data.
Absent
Homozygous mutation is embryonic lethal.
TDP-43 (A315T)
Absent
Not observed.
Absent
Not observed.
Observed
Cytoplasmic accumulation of TDP-43 was observed by 10 months of age in the spinal cord. Furthermore, cytoplasmic aggregates were observed and often co-localized with ubiquitin. These inclusions are not detected at three months of age.
Observed
Progressive gliosis of both astrocytes and microglia, starting at a young age (by 3 months) in the brain and spinal cord.
No Data
Unknown.
No Data
Unknown.
Observed
At 38 weeks of age, mice develop impairments on the accelerating Rotarod relative to non-Tg littermates.
No Data
Unknown.
Absent
Not observed.
TDP-43 (A315T) (line 23)
Absent
Not observed.
Absent
Not observed.
Observed
Ubiquitin-positive cytoplasmic inclusions in neurons of the ventral horn and brainstem. Cytoplasmic aggregates of TDP-43 are largely absent, although rare phospho-TDP-43 inclusions were observed, especially at end-stage.
Observed
Mice exhibiting muscle weakness had astrocytosis in the ventral horn of the spinal cord.
No Data
Unknown.
Observed
Atrophy of muscle fibers in the quadriceps muscle of weak mice observed by day 44.
Observed
Progressive motor impairment, characterized by weakness, a decline in grip strength, and reduction in stride length. Weakness was usually more pronounced in the hindlimbs.
Observed
Progressive weight loss.
Observed
Line 23 mice survived about 2.5 months, mean survival 75 days. It was not reported whether this survival analysis includes males, females or both. Colony at Jackson Labs has longer mean survival.
TDP-43 (G348C)
Absent
Not observed.
Absent
Not observed.
Observed
Cytoplasmic accumulation of TDP-43 was observed by 10 months in the spinal cord. Cytoplasmic aggregates occurred and often co-localized with ubiquitin. These inclusions are not detected at 3 months of age.
Observed
Progressive gliosis of both astrocytes and microglia, starting at a young age (by 3 months) in the brain and spinal cord.
Observed
In 10-month-old mice, approximately 10% of NMJs in the gastrocnemius muscle were denervated, with another 20% partially denervated.
No Data
Unknown.
Observed
Performance on the Rotarod was comparable to non-Tg littermates until 36 weeks of age, and became progressively worse with age.
No Data
Unknown.
Absent
Normal lifespan.
TDP-43 (M337V)
Absent
Not observed.
Absent
Not observed.
Observed
TDP-43 protein was largely nuclear, although some cytoplasmic TDP-43 was also observed. Some mild cytoplasmic inclusions were reported.
Observed
Reactive astrocytes and activated microglia proliferate in the spinal cord and brainstem.
No Data
No data.
No Data
No data.
Observed
Body tremors apparent by day 21 and the mice had difficulty recruiting their hindlimbs, leading to an irregular gait pattern, described as “dragging.”
Observed
By one month of age, homozygotes have reduced body weight compared to non-Tg littermates.
Observed
70% mortality of homozygotes by around one month of age.
TDP-43 (M337V) (Mt-TAR6/6)
Observed
Severe neuronal loss in all CA regions of the hippocampus of homozygous mice. Neuronal loss was also observed in layer V cortical neurons and thalamic neurons.
Observed
Neuronal loss was observed in the spinal cords of homozygous mice.
Observed
Some homozygous mice developed cytoplasmic inclusions in layer V cortical neurons. These were often, but not always, ubiquitin–positive. They were not universally observed, even in end-stage mice.
Observed
Elevated astrogliosis and microgliosis compared with non-Tg controls, especially in the motor cortex and spinal cord. Gliosis in the hippocampus was seen at end stage.
No Data
Unknown.
No Data
Unknown.
Observed
Motor impairment developed quickly, by 11 days of age in homozygous mice, starting with an abnormal clasping reflex. They also develop a hunched posture, muscle twitches, and reduced mobility. Paralysis developed within days, leading to death. Hemizygotes do not develop motor symptoms until about one year of age, and impairment varied from mouse to mouse.
Observed
Early postnatal growth retardation in homozygous mice. By day 17 their average body weight is about half that of non-Tg controls.
Observed
Homozygous mice survived an average of just 17 days. In contrast, hemizygous Mt-TAR6 mice lived up to 24 months (average survival ~16.4 months).
TDP-43 (Q331K)
No Data
Unknown.
Observed
Age-dependent loss of lower motor neurons in the lumbar spinal cord. Loss is detectable as early as 2 months of age and is more pronounced by 10 months.
Absent
TDP-43 in the brain and spinal cord was predominantly nuclear. Cytoplasmic TDP-43 aggregates were absent.
Observed
Elevated astrogliosis and microgliosis in the ventral horn of spinal cord by 10-12 months of age compared with non-Tg controls.
Observed
Reduction in neuromuscular junction endplates by 10-12 months of age. Remaining NMJs often had a “bleb-like” appearance.
No Data
Muscle fiber abnormalities including centralized nuclei and damage by 10-12 months of age.
Observed
Tremor, abnormal hindlimb clasping, impaired performance on the Rotarod were detectable starting around 3 months of age. Reduced grip strength occurred later.
No Data
Unknown.
No Data
Unknown.
TDP-43 (Q331K) Knock-In (Line 52)
Observed
25% loss of parvalbumin-positive neurons at 5 months.
Absent
Not observed.
Absent
Not observed.
No Data
Unknown.
Absent
Not observed.
No Data
Unknown.
Absent
Not observed.
Observed
Increased body weight.
Observed
Mutation may be deleterious to male embryos.
TDP-43 (WT) (Elliott)
Absent
Not observed.
Absent
Not observed.
Observed
Cytoplasmic ubiquitin-positive inclusions in skeletal muscle cells. Some TDP-43 inclusions, too.
No Data
No data.
No Data
No data.
Observed
An analysis of the quadriceps muscle, showed signs of myopathy, including variable muscle fiber size and disorganization of the muscle architecture.
Observed
Progressive motor impairment starting with external rotation of one hind limb followed by bilateral weakness and low muscle tone. Variable penetrance of this phenotype.
Observed
Progressive weight loss.
Observed
The mean survival of hemizygous mice was 109 days (it is not clear if this value represents males, females, or both).
TDP-43 (WT) (Julien model)
Absent
Not observed.
Absent
Not observed.
Absent
Primarily nuclear localization of human TDP-43.
Observed
Gliosis, both microgliosis and astrogliosis, occur early in the brain and spinal cord. Reactive glia were detected as early as 3 months of age, with more by 10 months.
Observed
Some NMJ denervation was observed by 10 months of age. About 5% of NMJs at the gastrocnemius muscle were denervated, with another 20 percent partially denervated.
No Data
No data.
Observed
Decreased performance on the accelerating Rotarod at 42 weeks of age. Further impairment at 52 weeks.
No Data
No data.
No Data
No data.
TDP-43 (WT) (Kumar-Singh)
Observed
In homozygous mice, quantitative loss of neurons occurs in the motor cortex compared with non-Tg littermates. Both superficial and deep cortical layers of the anterior cortex are affected.
Observed
By day 18, homozygous mice exhibited about 25 percent loss of motor neurons in the lumbar spinal cord compared with non-Tg littermates.
Observed
Homozygous mice developed cytoplasmic inclusions in the brain and spinal cord, many of which were ubiquitin-positive. A minority of inclusions co-labeled with TDP-43. Ultrastructural analysis revealed ubiquitin–negative cytoplasmic inclusions in anterior horn neurons to be abnormal accumulations of mitochondria.
Observed
Astrogliosis and microgliois especially in cortical layer V of the anterior cortex, including motor and somatosensory cortex, and in the spinal cord.
No Data
No data.
No Data
No data.
Observed
Homozygous mice exhibit an abnormal clasping reflex by postnatal day 14. Other early motor deficits include a shortened stride, a wide stance, and frequent stumbling. By day 18, reduced performance on the Rotarod. Complete paralysis occurs ~10 days after onset.
Observed
Size and weight of homozygous mice lag behind hemizygotes and non-Tg littermates.
Observed
Homozygous mice survive an average of just 24 days. In contrast, hemizygous mice survive to advanced age, although they die more prematurely than non-Tg mice, after 22 to 24 months.
TDP-43 (WT) (Petrucelli)
Absent
Not observed.
Absent
Neuronal loss was not detected in spinal cords of homozygous mice as assessed by TUNEL staining and caspase-3 staining.
Observed
Cytoplasmic eosinophilic aggregates in spinal motor neurons by one month of age in homozygous mice.
Observed
Astrogliosis and microgliosis in the anterior horn of the spinal cord by one month of age.
No Data
Unknown.
Absent
Atrophy of the gastrocnemius muscle was not observed.
Observed
By day 21, homozygous mice displayed body tremors and mild gait impairment which progressed into a “swimming gait” and severe motor impairment.
Observed
Homozygotes diverge early from non-Tg littermates in terms of body weight, showing significantly reduced weight gain.
Observed
Homozygous mice were sacrificed at one to two months of age when they were unable to right themselves.
TDP-43 (Wt-TAR6/6)
Observed
Approximate 15 percent loss of layer V neurons in motor cortex at 6 months.
Observed
Spinal motor neuron loss observed at 3 months, with approximately 10 percent fewer anterior horn neurons in lumbosacral regions at 6 months, compared with non-transgenic mice.
Observed
Inclusions containing phosphorylated TDP-43 rarely observed in the nuclei and cytoplasm of spinal neurons.
Observed
Microgliosis in cortex and spinal cord prominent at 6 months; astrogliosis in cortex and spinal cord apparent at 1.5 months.
No Data
No data.
Observed
Muscle wasting, particularly in flanks.
Observed
Progressive motor impairment; abnormal hind limb reflexes observed as early as 1.5 months.
Observed
TAR6/6 mice had lower body weights than non-transgenic mice between 3.25 and 3.75 months age, but the two genotypes were similar at younger and older ages (at least until 4.25 months).
Observed
Average survival is 6.7 months.
ΔNLS-FUS
Observed
By 1 year, there was neuronal loss in the motor cortex.
Absent
Not observed at 1 year in the L5 anterior horn.
Observed
Ubiquitin- and p62-positive ΔNLS-FUS inclusions in motor cortex neurons.
Observed
Microgliosis and astrocytosis were observed in the motor cortex.
No Data
No data.
No Data
No data.
Observed
Progressive motor impairments by 12 weeks. Mice demonstrated tremors, limb clasping, gait abnormalities, as well as decreased performance on the Rotarod and hanging wire tests.
Observed
Decreased by 48 weeks.
Observed
Approximately 50% mortality by 60 weeks of age.
ΔNLS-FUS x TDP-43(WT)
Observed
By 1 year, there was neuronal loss in the motor cortex.
Absent
Not observed at 1 year in the L5 anterior horn.
Observed
Ubiquitin- and p62-positive ΔNLS-FUS inclusions in motor cortex neurons.
Observed
Microgliosis and astrocytosis were observed in the motor cortex.
No Data
No data.
No Data
No data.
Observed
Progressive motor impairments by 8 weeks. Mice demonstrated tremors, limb clasping, gait abnormalities, as well as decreased performance on the Rotarod and hanging wire test.
Observed
Decreased by 48 weeks.
Observed
Approximately 40% mortality by 60 weeks of age.