. Antibody-Mediated Targeting of Tau In Vivo Does Not Require Effector Function and Microglial Engagement. Cell Rep. 2016 Aug 9;16(6):1690-700. Epub 2016 Jul 28 PubMed.

Recommends

Please login to recommend the paper.

Comments

  1. We read this paper with interest. Because the authors reference our work critically, we would like to clarify the points raised by the authors, specifically neuronal uptake of tau antibodies and the presence of Fc receptors on neurons. 

    We do not dispute the authors’ assertion that their antibodies are effective without being internalized by neurons. Several groups have reported success in preventing the spread of tau using antibodies that do not enter neurons (Castillo-Carranza et al., 2014; d'Abramo et al., 2013; Yanamandra et al., 2013). We have seen this mechanism of action in our own work as well (Congdon et al., 2016; Congdon et al., 2014; Congdon et al., 2015, SFN Abstr. Chicago, IL 579.13/C43). Blockage of neuronal uptake of tau certainly represents a valid avenue for the development of therapeutics. 

    However, we take issue with the suggestion that such uptake detected by us in brain slice cultures may be explained by mechanical damage to cells and membranes during sectioning. As detailed in one article referred to (Congdon et al., 2013), we performed control experiments to ascertain whether uptake was solely on the surface level, or distributed within the tissue. We retained some of the slices that had been incubated with radiolabeled antibody, and further sectioned the tissue. The radioactivity of each section was then assessed using a liquid scintillation counter. We observed similar levels of radioactivity throughout the tissue, indicating that the antibody uptake was not limited to the surface (Congdon et al., 2013). Additionally, when preparing the slice cultures for immunohistochemistry, the tissue was fixed and sectioned (Congdon et al., 2013; Gu et al., 2013). The sections that were stained came from throughout the slice. 

    Furthermore and as reported, we have seen internalization of tau antibodies in multiple model systems. In our initial report on the efficacy of active immunization to target tau pathology, we found that antibodies purified from immunized animals entered the brain and co-localized intraneuronally with tau markers (Asuni et al., 2007). We further confirmed these findings in whole animals given intracarotid or intravenous injections of either whole antibody or its scFv fragment. Both the whole antibody and the fragment were able to cross the blood brain barrier, and co-localized with tau and endosomal markers inside neurons (Krishnaswamy et al., 2014). We have also observed antibody uptake in brain slices, primary neuronal cultures, and human neuroblastoma cells lines, as detected by multiple methodologies including confocal imaging, western blotting, fluorescence measurement/flow cytometry, and radioactivity (Congdon et al., 2013; Gu et al., 2013; Shamir et al., 2016; Krishnamurthy et al., 2011). Other groups have also shown neuronal uptake of antibodies against tau as well as antibodies recognizing Aβ and α-synuclein (Masliah et al., 2005; Masliah et al., 2011; Tampellini et al., 2007; Collin et al., 2014; Kondo et al., 2015; Gustafsson et al., 2016). 

    Collectively, these data clearly demonstrate that antibody uptake into neurons is not dependent on broken cell membranes. It is likely that differences between studies in neuronal uptake of antibodies are primarily related to antibody properties. As we have repeatedly discussed, not all antibodies are taken up into cells to the same extent, which may in large part be explained by differences in antibody charge (for example see review, Pedersen and Sigurdsson, 2015). 

    With respect to Fc receptors on neurons, the authors state that they do not observe detectable levels of expression in neurons, and posit that our findings stem from non-specific antibody reactivity in our experiments. Based on their RT-PCR data (Figure 5A), it appears that detectable neuronal FcR levels were observed in their samples because otherwise ΔCt values could not be calculated. The values presented are relative to housekeeping genes and show that the FcR subtypes have higher expression levels in microglia compared to neurons, which is as expected. Likewise, the confocal images in Figure 5C are qualitative and may just indicate higher expression levels in microglia than in neurons.

    The authors refer to publically available mouse transcriptome as further evidence that these receptors are not present but the database indicates detectable levels of Fc receptor expression in the purified neurons (Zhang et al., 2014), which again is much less than in microglia, as expected. Finally, it is known that expression levels can change depending on various factors such as the developmental stage of cells under study, so it is impossible to generalize what is or is not expressed in different models/tissue. 

    With regard to non-specific binding, we agree that it is always a factor to consider, but ours is not the only report of Fc receptors on neurons. Several other groups have published such findings using a variety of methods (Andoh and Kuraishi, 2004; Andoh and Kuraishi, 2004; Fernandez-Vizarra et al., 2012; Kam et al., 2013; Mohamed et al., 2002; Nakamura et al., 2007; Qu et al., 2011; Suemitsu et al., 2010; van der Kleij et al., 2010; Fuller et al., 2014; Okun et al., 2010). Finally, we obtained similar results with an Fc blocker as we did with an inhibitor of receptor-mediated endocytosis (Congdon et al., 2013). 

    Having addressed these critical points, we do agree with the authors’ overall conclusion that an antibody without an effector function is worthy of further development. Similar findings have been reported in the Aβ field although there are conflicting results, which may be explained by differences between antibodies, models, and experimental design (Bard et al., 2003; Das et al., 2003; Bacskai et al., 2002). Both the authors' antibodies are effective but the two are not always directly compared, and it is not clear at which dose because it is not appropriate to use a t-test for post-hoc analysis of ANOVA data as done throughout the article. Perhaps they would be more efficacious if they could enter neurons where most of pathological tau resides. These investigators are ideally suited to engineer these antibodies to improve their uptake into neurons to assess this possibility, as is routinely done in the cancer immunotherapy field to modulate uptake of antibodies into cells.

    We appreciate the opportunity to address the issues raised in the article, and look forward to seeing additional work from the authors.

    J. Gu, S. Krishnaswamy of the Departments of Neuroscience and Physiology of the New York University School of Medicine and P.K. Krishnamurthy Oligomerix Inc. in New York contributed to this comment.

    References:

    . Passive immunization with Tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles. J Neurosci. 2014 Mar 19;34(12):4260-72. PubMed.

    . Tau passive immunotherapy in mutant P301L mice: antibody affinity versus specificity. PLoS One. 2013;8(4):e62402. PubMed.

    . Anti-Tau Antibodies that Block Tau Aggregate Seeding In Vitro Markedly Decrease Pathology and Improve Cognition In Vivo. Neuron. 2013 Oct 16;80(2):402-14. PubMed.

    . Affinity of Tau antibodies for solubilized pathological Tau species but not their immunogen or insoluble Tau aggregates predicts in vivo and ex vivo efficacy. Mol Neurodegener. 2016 Aug 30;11(1):62. PubMed.

    . Harnessing the immune system for treatment and detection of tau pathology. J Alzheimers Dis. 2014;40 Suppl 1:S113-21. PubMed.

    . Antibody uptake into neurons occurs primarily via clathrin-dependent Fcγ receptor endocytosis and is a prerequisite for acute tau protein clearance. J Biol Chem. 2013 Dec 6;288(49):35452-65. Epub 2013 Oct 25 PubMed.

    . Two novel tau antibodies targeting the 396/404 region are primarily taken up by neurons and reduce tau pathology. J Biol Chem. 2013 Nov 15;288(46):33081-95. PubMed.

    . Immunotherapy targeting pathological tau conformers in a tangle mouse model reduces brain pathology with associated functional improvements. J Neurosci. 2007 Aug 22;27(34):9115-29. PubMed.

    . Antibody-derived in vivo imaging of tau pathology. J Neurosci. 2014 Dec 10;34(50):16835-50. PubMed.

    . Internalization of tau antibody and pathological tau protein detected with a flow cytometry multiplexing approach. Alzheimers Dement. 2016 Oct;12(10):1098-1107. Epub 2016 Mar 23 PubMed.

    . Mechanistic Studies of Antibody-Mediated Clearance of Tau Aggregates Using an ex vivo Brain Slice Model. Front Psychiatry. 2011;2:59. PubMed.

    . Effects of alpha-synuclein immunization in a mouse model of Parkinson's disease. Neuron. 2005 Jun 16;46(6):857-68. PubMed.

    . Passive immunization reduces behavioral and neuropathological deficits in an alpha-synuclein transgenic model of Lewy body disease. PLoS One. 2011;6(4):e19338. PubMed.

    . Internalized antibodies to the Abeta domain of APP reduce neuronal Abeta and protect against synaptic alterations. J Biol Chem. 2007 Jun 29;282(26):18895-906. PubMed.

    . Neuronal uptake of tau/pS422 antibody and reduced progression of tau pathology in a mouse model of Alzheimer's disease. Brain. 2014 Oct;137(Pt 10):2834-46. Epub 2014 Jul 31 PubMed.

    . Antibody against early driver of neurodegeneration cis P-tau blocks brain injury and tauopathy. Nature. 2015 Jul 23;523(7561):431-6. Epub 2015 Jul 15 PubMed.

    . Cellular Uptake of α-Synuclein Oligomer-Selective Antibodies is Enhanced by the Extracellular Presence of α-Synuclein and Mediated via Fcγ Receptors. Cell Mol Neurobiol. 2016 Mar 10; PubMed.

    . Tau immunotherapy for Alzheimer's disease. Trends Mol Med. 2015 Jun;21(6):394-402. Epub 2015 Apr 3 PubMed.

    . An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J Neurosci. 2014 Sep 3;34(36):11929-47. PubMed.

    . Direct action of immunoglobulin G on primary sensory neurons through Fc gamma receptor I. FASEB J. 2004 Jan;18(1):182-4. Epub 2003 Nov 20 PubMed.

    . Expression of Fc epsilon receptor I on primary sensory neurons in mice. Neuroreport. 2004 Sep 15;15(13):2029-31. PubMed.

    . Immunoglobulin G Fc receptor deficiency prevents Alzheimer-like pathology and cognitive impairment in mice. Brain. 2012 Sep;135(Pt 9):2826-37. PubMed.

    . FcγRIIb mediates amyloid-β neurotoxicity and memory impairment in Alzheimer's disease. J Clin Invest. 2013 Jul 1;123(7):2791-802. PubMed.

    . Immunoglobulin Fc gamma receptor promotes immunoglobulin uptake, immunoglobulin-mediated calcium increase, and neurotransmitter release in motor neurons. J Neurosci Res. 2002 Jul 1;69(1):110-6. PubMed.

    . CD3 and immunoglobulin G Fc receptor regulate cerebellar functions. Mol Cell Biol. 2007 Jul;27(14):5128-34. PubMed.

    . Neuronal Fc-gamma receptor I mediated excitatory effects of IgG immune complex on rat dorsal root ganglion neurons. Brain Behav Immun. 2011 Oct;25(7):1399-407. Epub 2011 Apr 18 PubMed.

    . Fcgamma receptors contribute to pyramidal cell death in the mouse hippocampus following local kainic acid injection. Neuroscience. 2010 Mar 31;166(3):819-31. Epub 2010 Jan 13 PubMed.

    . Evidence for neuronal expression of functional Fc (epsilon and gamma) receptors. J Allergy Clin Immunol. 2010 Mar;125(3):757-60. Epub 2010 Feb 4 PubMed.

    . New roles for Fc receptors in neurodegeneration-the impact on Immunotherapy for Alzheimer's Disease. Front Neurosci. 2014;8:235. Epub 2014 Aug 21 PubMed.

    . Involvement of Fc receptors in disorders of the central nervous system. Neuromolecular Med. 2010 Jun;12(2):164-78. PubMed.

    . Epitope and isotype specificities of antibodies to beta -amyloid peptide for protection against Alzheimer's disease-like neuropathology. Proc Natl Acad Sci U S A. 2003 Feb 18;100(4):2023-8. Epub 2003 Feb 3 PubMed.

    . Amyloid-beta immunization effectively reduces amyloid deposition in FcRgamma-/- knock-out mice. J Neurosci. 2003 Sep 17;23(24):8532-8. PubMed.

    . Non-Fc-mediated mechanisms are involved in clearance of amyloid-beta in vivo by immunotherapy. J Neurosci. 2002 Sep 15;22(18):7873-8. PubMed.

Make a Comment

To make a comment you must login or register.