Rather than sequestering Aβ to prevent its clearance from the brain, ApoE may indirectly block the peptide’s path to breakdown, according to results published April 25 in the Proceedings of the National Academy of Sciences. Contrary to earlier reports, David Holtzman and colleagues at the Washington University School of Medicine, St. Louis, Missouri, found little interaction between soluble Aβ and ApoE when measured in physiological conditions. Instead, the two compete for the same astrocyte receptor, low-density lipoprotein receptor-related protein 1 (LRP1), which helps clear Aβ, the authors report. The results provide a new perspective on ApoE and extend previous studies suggesting that lowering brain levels of the protein may be a therapeutic approach for AD. Scientists led by Berislav Zlokovic, University of Southern California, Los Angeles, describe a related strategy in the April 11 Journal of Biological Chemistry. The researchers constructed a fragment of LRP1 that binds more tightly to Aβ. They propose that this could be used to clear amyloid from the brain.

ApoE4 is the strongest genetic risk factor for AD, but its mechanism of action has eluded scientists. Previous research from many labs, including from Holtzman’s, suggested that ApoE binds Aβ directly to influence its clearance (for a review, see Kim et al., 2009). These studies lacked stoichiometric measures of how much of the total peptide ApoE trapped, however. What’s more, researchers often relied on abnormally high ratios of Aβ to ApoE, said Holtzman. Now his group has quantified binding at physiological concentrations of the two proteins.

First author Philip Verghese and colleagues combined synthetic soluble Aβ, or peptide derived from human CSF or neuroglioma cells, with ApoE particles assembled with appropriate lipids, or ApoE extracted from human CSF, or mouse or human astrocytes. With these physiologically relevant sources and ApoE/Aβ ratios (around 50:1), they used ultracentrifugation and size-exclusion chromatography to measure how much Aβ bound to ApoE. They checked in the presence of H4 neuroglioma cells overproducing Aβ, too, to see if ApoE attracted the secreted peptide from cells. In all of these combinations, about 95 percent of Aβ remained free after 24 hours, regardless of ApoE isoform.

If ApoE does not directly bind Aβ in physiological fluids, then how does it modify the peptide’s clearance? The researchers looked to astrocytes, which make most of the ApoE in the central nervous system and help clear Aβ. They cultured astrocytes from ApoE knock-in and ApoE knockout mice with cell-secreted soluble Aβ. Cells without ApoE cleared more Aβ than any of the knock-ins. Of the latter, ApoE2 astrocytes cleared Aβ best, and ApoE4 cells the least. Results suggested that ApoE is unnecessary for Aβ removal by astrocytes, and actually inhibits it. The researchers confirmed this by adding ApoE particles and soluble Aβ to ApoE knockout astrocytes. Regardless of isoform, the more ApoE they added, the less the astrocytes cleared Aβ. Overall, the results imply that ApoE does not bind to Aβ, but competes for clearance somehow, for example, by binding to an Aβ receptor or receptors on the cell surface.

Could LRP1 fit that bill? Recent studies highlight the importance of LRP1 in Aβ metabolism (see Kanekiyo et al., 2011) and AD (see Harris-White and Frautschy, 2005). This receptor rests on astrocyte membranes and binds both Aβ and ApoE. There are even hints it may help clear Aβ from the vasculature (see ARF related news story). Interestingly, though Verghese and colleagues found that LRP1-deficient mouse fibroblasts removed less Aβ than usual, clearance was unaffected by ApoE. An LRP1 antibody also reduced uptake of Aβ by astrocytes. Together, these results suggest that ApoE competes for LRP1, blocking Aβ clearance.

This research says nothing about interaction with aggregated forms, said Holtzman. ApoE is found in Aβ plaques, he pointed out (see Wisniewski and Frangione, 1992). “This study looks at what might lead to Aβ aggregation in the first place,” he told Alzforum. This speaks to the difficulty in studying ApoE/Aβ interactions, since the former comes in three isoforms, different lipoprotein particles, and different lipidation states, while the latter exists as monomeric, oligomeric, and aggregated entities. “The entire problem is more complex than can be discussed in a single paper,” said Zlokovic. Previous work has suggested that ApoE is needed for clearance of aggregated Aβ, and that it facilitates Aβ clearance by astrocytes (see Koistinaho et al., 2004), he added. "With so much conflicting data, this paper suggests the field does not have a final answer," he concluded. In fact, while these new data suggest that ApoE blocks Aβ clearance, Gary Landreth and colleagues at Case Western Reserve University, Cleveland, Ohio, found that bexarotene, which enhances ApoE expression, clears Aβ and improves cognition in mice (see ARF related news story). Using brain microdialysis techniques developed in his lab, Holtzman collaborated with Landreth to show that bexarotene lifts ApoE while suppressing Aβ in interstitial brain fluid. This paper appeared earlier this month (see Ulrich et al., 2013). Bexarotene also boosts ApoE lipidation, which could explain the beneficial effects, Holtzman said.

Any explanation for how ApoE modulates Aβ clearance will have to account for the increased risk for AD that comes with ApoE4. Only further study will shed light on that question, wrote the authors. “It will be interesting to determine whether ApoE4 binds to LRP1 with higher affinity than ApoE2, functioning as a more potent antagonist of Aβ uptake,” said Steven Burden, New York University Medical School, New York. Holtzman said his group plans to investigate that.

“This is a compelling study that will definitely be controversial,” suggested Edwin Weeber, University of South Florida, Tampa. The paper thoroughly examined the ApoE/Aβ binding question, but did not address how this competition affects Aβ uptake into neurons and other cells that express LRP1, he pointed out. Nevertheless, “this study is going to change the way people think about ApoE isoforms and clearance of Aβ,” said Weeber.

Mary Jo LaDu and Leon Tai, University of Illinois at Chicago, cautioned that readers should not discount the importance of binding between ApoE and Aβ based on this paper alone. Given the unique environment of the brain, it is unclear whether in-vitro results are relevant to in vivo, LaDu said. Holtzman’s detection techniques may have missed or disrupted ApoE/Aβ complexes, she cautioned. She wondered if the researchers would find different results using an ELISA method recently developed in her lab that she believes will more likely keep complexes intact. Her group used it to detect soluble ApoE/Aβ binding in the brain tissue of humans and AD transgenic mice, as well as in human CSF (see Tai et al., 2013).

Finally, LRP1 exists in soluble form as well. It circulates in the blood, binds about 70 percent of plasma Aβ, and shepherds it to the liver for clearance, providing a natural peripheral sink. LRP1 becomes oxidized in AD patients, limiting Aβ binding and leaving more free peptide in the plasma. For that reason, Zlokovic and colleagues are working on LRP replacement as a way to reduce Aβ buildup. They previously created a recombinant fragment of one of the protein’s Aβ-binding clusters, LRPIV, and found that it curtailed Aβ pathology in Tg2576 mice and bound free peptide in human plasma (see ARF related news story). Since the protein fragment has multiple ligands, they wanted to boost its affinity for Aβ to clear the peptide more efficiently.

In their JBC paper, co-first authors Abhay Sagare, University of Southern California, Los Angeles; Robert Bell, University of Rochester Medical Center, New York; and Alaka Srivastava at ZZ Alztech, also in Rochester, made a library of LRPIV variants. They picked one, LRPIV-D3674G, that binds tightly to Aβ but weakly to other ligands such as ApoE isoforms. With five days of intravenous treatment, LRPIV-D3674G reduced endogenous brain Aβ40 and Aβ42 in Tg2576 mice about 26 percent better than the wild-type fragment. When given subcutaneously to the same mice for three months, this variant showed no toxicity and cleared Aβ40 and Aβ42 in the hippocampus, cortex, and cerebrospinal fluid by 60-80 percent. Put together with the Verghese study, this suggests that “a higher-affinity LRP fragment could promote cellular Aβ clearance and lessen ApoE-mediated slowing of Aβ removal,” said Holtzman.

With his colleagues at ZZ Alztech, Zlokovic plans to scale up the manufacturing process for this LRP fragment and submit an Investigational New Drug application with the FDA to get it on the road to clinical trials.—Gwyneth Dickey Zakaib

Comments

  1. In this paper, Verghese et al. provide important data confirming earlier studies showing reduced cellular association between Aβ and astrocytes in the presence of ApoE. The current data obtained from studies on rodent astrocytes suggest competing effects of ApoE on the astrocytic uptake of soluble Aβ. These findings are perfectly in line with our previous findings demonstrating a significant reduction in Aβ uptake by primary human adult astrocytes in vitro (1). We suggested that ApoE interferes with astrocytic uptake of oligomeric, but not fibrillar, preparations of Aβ, as astrocytic uptake of fibrillar Aβ appeared unaffected in the presence of ApoE. Verghese et al. now confirm the notion that uptake of soluble forms of Aβ is decreased in the presence of ApoE; however, they did not address whether ApoE also interferes with astrocytic uptake of fibrillar Aβ. In fact, a reduction of soluble Aβ uptake could possibly be due to ApoE chaperone effects on Aβ, yielding more fibrillar Aβ, which decreases astrocytic Aβ uptake as suggested by earlier findings (1).

    Interestingly, uptake of mutant Aβ also appears to be hampered by ApoE. Bruinsma et al. previously showed that ApoE-conditioned culture media from ApoE3/3 primary human pericytes completely abolished the cell association and internalization of Aβ with the Dutch mutation (Aβ Glu22Gln; D-Aβ1-40) by both primary human pericytes and astrocytes (2).

    Last, in regard to the findings presented by Verghese et al., suggesting LRP1 as a receptor candidate mediating astrocytic Aβ internalization in rodent cells, Wilhelmus and coworkers have shown that LRP1 may mediate Aβ uptake and toxicity in primary human brain pericytes, but not in human astrocytes (3). Thus, the contribution of LRP1 to astrocytic Aβ uptake remains elusive before confirming the results of Verghese et al., either in human primary astrocytes or in well-characterized rodent primary astrocyte cultures, both devoid of pericytes. Species-dependent differences in astrocyte functional competence between human and rodent cells have earlier been described (4,5), and these potential differences also need to be ruled out before attributing to LRP1 the role of an astrocytic Aβ receptor with Aβ and ApoE as competing ligands.

    References:

    . Astrocytic A beta 1-42 uptake is determined by A beta-aggregation state and the presence of amyloid-associated proteins. Glia. 2010 Aug;58(10):1235-46. PubMed.

    . Apolipoprotein E protects cultured pericytes and astrocytes from D-Abeta(1-40)-mediated cell death. Brain Res. 2010 Feb 22;1315:169-80. PubMed.

    . Lipoprotein receptor-related protein-1 mediates amyloid-beta-mediated cell death of cerebrovascular cells. Am J Pathol. 2007 Dec;171(6):1989-99. PubMed.

    . Astrocytic complexity distinguishes the human brain. Trends Neurosci. 2006 Oct;29(10):547-53. Epub 2006 Aug 30 PubMed.

    . Uniquely hominid features of adult human astrocytes. J Neurosci. 2009 Mar 11;29(10):3276-87. PubMed.

    View all comments by Henrietta Nielsen

Make a Comment

To make a comment you must login or register.

References

News Citations

  1. Keystone: Symposium Emphasizes Key Aspects of ApoE Biology
  2. Upping Brain ApoE, Drug Treats Alzheimer's Mice
  3. Enhancing Peripheral Sink Scours Brain of Amyloid

Paper Citations

  1. . The role of apolipoprotein E in Alzheimer's disease. Neuron. 2009 Aug 13;63(3):287-303. PubMed.
  2. . Heparan sulphate proteoglycan and the low-density lipoprotein receptor-related protein 1 constitute major pathways for neuronal amyloid-beta uptake. J Neurosci. 2011 Feb 2;31(5):1644-51. PubMed.
  3. . Low density lipoprotein receptor-related proteins (LRPs), Alzheimer's and cognition. Curr Drug Targets CNS Neurol Disord. 2005 Oct;4(5):469-80. PubMed.
  4. . Apolipoprotein E: a pathological chaperone protein in patients with cerebral and systemic amyloid. Neurosci Lett. 1992 Feb 3;135(2):235-8. PubMed.
  5. . Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides. Nat Med. 2004 Jul;10(7):719-26. PubMed.
  6. . In vivo measurement of apolipoprotein E from the brain interstitial fluid using microdialysis. Mol Neurodegener. 2013;8:13. PubMed.
  7. . Levels of soluble apolipoprotein E/amyloid-β (Aβ) complex are reduced and oligomeric Aβ increased with APOE4 and Alzheimer disease in a transgenic mouse model and human samples. J Biol Chem. 2013 Feb 22;288(8):5914-26. PubMed.

Other Citations

  1. Tg2576 mice

Further Reading

Papers

  1. . Targeting ApoE4/ApoE receptor LRP1 in Alzheimer's disease. Expert Opin Ther Targets. 2013 Jul;17(7):781-94. PubMed.
  2. . Low-density lipoprotein receptor-related protein 1: A physiological Aβ homeostatic mechanism with multiple therapeutic opportunities. Pharmacol Ther. 2012 Oct;136(1):94-105. PubMed.
  3. . LRP1 expression in cerebral cortex, choroid plexus and meningeal blood vessels: relationship to cerebral amyloid angiopathy and APOE status. Neurosci Lett. 2012 Sep 13;525(2):123-8. PubMed.
  4. . Blocking the interaction between apolipoprotein E and Aβ reduces intraneuronal accumulation of Aβ and inhibits synaptic degeneration. Am J Pathol. 2013 May;182(5):1750-68. PubMed.

Primary Papers

  1. . A Lipoprotein Receptor Cluster IV Mutant Preferentially Binds Amyloid-β and Regulates Its Clearance from the Mouse Brain. J Biol Chem. 2013 May 24;288(21):15154-66. PubMed.
  2. . ApoE influences amyloid-β (Aβ) clearance despite minimal apoE/Aβ association in physiological conditions. Proc Natl Acad Sci U S A. 2013 May 7;110(19):E1807-16. PubMed.