One study views Aβ production as a normal part of synaptic plasticity, while another starts with the premise that Aβ buildup in neurons is a bad thing. Yet from these divergent starting points, both arrive at a similar conclusion—the endocytic pathway is a critical regulator of AD pathogenesis. In the October 28 Cell, researchers led by Paul Worley at Johns Hopkins University School of Medicine in Baltimore, Maryland, report that generation of Aβ in response to neuronal firing requires the post-synaptic protein Arc, which seems to pull presenilin-1 into endosomes that then process more amyloid precursor protein (APP). And in an October 27 Science paper published online, endocytic proteins—including several linked to sporadic Alzheimer’s disease risk in genomewide association studies—came up in a yeast screen for modifiers of Aβ toxicity. Susan Lindquist of the Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, led that study. Together, the papers suggest that the AD field is getting a leg up on the role of endocytosis in APP metabolism.

Independent research teams led by Worley and coauthor Dietmar Kuhl of the University Medical Center Hamburg-Eppendorf, Germany, identified Arc (aka Arg3.1) in 1995 (Lyford et al., 1995; Link et al., 1995), and today’s Cell paper grew out of a longstanding quest to understand how Arc and other immediate early genes work at synapses to confer plasticity. Arc is dramatically upregulated in neurons engaged in information processing (Guzowski et al., 1999; Guzowski et al., 2000). It associates with endosomes to regulate AMPA receptor trafficking (Chowdhury et al., 2006), and helps these receptors maintain synaptic strength in a process known as homeostatic scaling (see Shepherd et al., 2006 and ARF related news story). Fitting with these roles, mice lacking Arc cannot form long-term memories (Plath et al., 2006).

Conceptually, this line of work converged with AD research when researchers found that increasing synaptic activity drove up Aβ levels in hippocampal slices of AD transgenic mice (Kamenetz et al., 2003) and in the brain interstitial fluid of living mice (Cirrito et al., 2005), and that the latter depended largely on endocytosis (see ARF related news story on Cirrito et al., 2008).

First author Jing Wu and colleagues wondered whether activity-dependent Aβ generation would occur differently in Arc knockout mice. “That was really our starting point,” Worley said. As it turns out, the process was not just different—it was gone. When seizures or drugs were administered to Arc-deficient mice to send hippocampal neurons into overdrive, Aβ levels did not budge.

To explore the mechanism, the researchers put fluorescently tagged Arc into mouse hippocampal neurons and watched the protein make its way through the cell. By immunohistochemistry, they saw Arc co-localize with presenilin-1 (PS1) in dendritic endosomes. Anti-Arc immunoprecipitation of mouse brain extract pulled down the PS1 N-terminal fragment—but not its C-terminus or other γ-secretase components or BACE1—confirming the Arc/PS1 interaction and mapping it to the N-terminal end.

Combined with electron microscopy analysis of rat hippocampus, “the cell biology data make the case that the Arc/PS1 interaction promotes Aβ generation by recruiting γ-secretase to recycling endosomes that process APP,” Worley told ARF. Consistent with this idea, crossing APPswe/PS1deltaE9 transgenic mice onto an Arc-deficient background reduced soluble Aβ40 and plaque load.

Is any of this physiologically relevant? Looking at human brain extracts, the researchers found twice as much Arc in the medial frontal cortex of AD patients (n = 26) compared to that of age-matched controls (n = 14). No Arc differences showed up in the visual cortex, a brain area spared in AD.

All told, the paper suggests that Arc is essential for activity-driven Aβ generation. “Connecting the activity dependence of Aβ formation to the mechanisms controlling plasticity is interesting and should provide new directions of research,” commented Roberto Malinow of the University of California, San Diego. However, he and others said further work will be needed to tease out where Arc is active. “Arc is primarily a post-synaptic protein, whereas Aβ appears to be released both axonally and dendritically,” said John Cirrito at Washington University School of Medicine in St. Louis, Missouri. “Whether Arc impacts both those compartments, or just acts dendritically, remains in question.”

Another question for future studies, Cirrito said, is whether Arc has any relationship with PICALM or BIN1—endocytic factors that were linked to sporadic AD risk in recent genomewide association studies (ARF related news story on Naj et al., 2011 and Hollingworth et al., 2011). Arc does not appear in the AlzGene database of AD risk genes.

Brewing a New Model for Aβ Toxicity
The importance of PICALM and other endocytic trafficking proteins did surface in the Science paper on a yeast model for Aβ toxicity. Though yeast cells are a far cry from the highly specialized neurons involved in AD, they have the basic vesicular trafficking compartments common to eukaryotic cells, senior investigator Lindquist noted. Her yeast models have proven useful for research on other protein aggregation disorders including Parkinson’s and Huntington’s (see ARF related news story on Cooper et al., 2006; ARF related news story on Gitler et al., 2009; ARF related news story on Outeiro and Lindquist, 2003 and Willingham et al., 2003). Yeast prion proteins fold into oligomers shaped like amyloid-β and other pathogenic peptides (Serio et al., 2000; Kayed et al., 2003; Shorter and Lindquist, 2004). “We had good reason to believe these conformational transitions are really an ancient problem,” Lindquist told ARF.

To model Aβ toxicity in yeast, first author Sebastian Treusch and colleagues directed Aβ1-42 to the endoplasmic reticulum sans ER retention signal, then let the peptide ride out of the cell along the secretory pathway. The yeast cell wall keeps the Aβ from diffusing out, and instead allows it to interact with the plasma membrane and transit back into the cell through endocytic compartments. Using stable Aβ1-42 transformants to screen a near-complete yeast genomic library containing some 6,000 genes, the researchers came up with 23 suppressors and 17 enhancers of Aβ toxicity. The former accelerated yeast growth, whereas the latter slowed it. Of the 40 modifiers, 12 had clear human homologs, three with roles in clathrin-mediated endocytosis. One of these is the yeast version of PICALM, one of the top late-onset AD risk genes. The other two have human homologs that interact with BIN1 and CD2AP, which also came up as top hits in recent AD GWAS. Treusch validated all three endocytosis genes in worm glutamatergic neurons and rat cortical neurons, where they modified Aβ toxicity in the same direction as in the yeast.

The research “provides a great system for understanding why PICALM is linked to AD, perhaps pointing to a currently unknown molecular mechanism underlying Aβ toxicity,” noted Sam Gandy of Mount Sinai Medical Center in New York City. As for whether yeast are too simple to model mammalian neurons, he said, “that is a caveat for any model system (yeast, fly, worm, mouse) until you figure out the molecular pathway.” Lindquist’s lab has received funding from the National Institutes of Health, Bethesda, Maryland, to use their yeast system to screen for Aβ toxicity modifiers in the NIH’s ChemIDplus library of 350,000 chemical compounds.—Esther Landhuis

Comments

  1. This is an interesting study by Wu and colleagues. Arc appears to mediate PS1 trafficking through the endosomal pathway which increases γ-secretase cleavage of APP and increases Aβ generation. It will be important to determine where and how Arc is altering PS1 localization. Aβ secretion appears to occur both from axons and dendrites (Wei et al., 2011; Cirrito et al., 2005), though Arc is only found dendritically. Based on Fig 1A and B in this paper, Arc knockouts completely blocked the effect of increased synaptic activity on Aβ levels in brain extracts. It will be important to determine if and how Arc is affecting axonal Aβ generation.

    References:

    . Amyloid beta from axons and dendrites reduces local spine number and plasticity. Nat Neurosci. 2010 Feb;13(2):190-6. PubMed.

    . Synaptic activity regulates interstitial fluid amyloid-beta levels in vivo. Neuron. 2005 Dec 22;48(6):913-22. PubMed.

    View all comments by John Cirrito
  2. This elegant study identifies Arc as a presenilin-1 (PS1) interacting protein that links synaptic activity with γ-secretase processing of APP in neurons. Detailed analysis of subcellular localization of APP, PS1, and Arc by immunofluorescence and immunoelectron microscopy methods suggest a mechanism whereby Arc facilitates association of PS1 with endosomes that contain internalized APP. Thus, it appears that the subset of APP that contributes to activity-dependent Aβ production encounters PS1 in endocytic organelles in dendrites of excitatory neurons.

    Arc is known to associate with endophilin-2/3 and dynamin on endosomes, and mediate AMPA receptor (AMPAR) endocytosis. Whereas the last is markedly reduced in Arc knockout neurons, this does not appear to be the case with PS1 or APP. Whether this indicates Arc regulation of PS1/APP colocalization in tubulovesicular endosomes is distinct from its function in regulating the endocytic machinery responsible for AMPAR endocytosis remains to be investigated.

    View all comments by Gopal Thinakaran

Make a Comment

To make a comment you must login or register.

References

News Citations

  1. Homeostatic Scaling—Presenilin Repertoire Reaches New Heights?
  2. Link Between Synaptic Activity, Aβ Processing Revealed
  3. Large Genetic Analysis Pays Off With New AD Risk Genes
  4. ER-Golgi Traffic Jam Explains α-Synuclein Toxicity
  5. Yeast Screen Implicates PARK9 in Synuclein Toxicity
  6. Yeast Teases Apart Huntington’s and Parkinson’s Protein Aggregation

Paper Citations

  1. . Arc, a growth factor and activity-regulated gene, encodes a novel cytoskeleton-associated protein that is enriched in neuronal dendrites. Neuron. 1995 Feb;14(2):433-45. PubMed.
  2. . Somatodendritic expression of an immediate early gene is regulated by synaptic activity. Proc Natl Acad Sci U S A. 1995 Jun 6;92(12):5734-8. PubMed.
  3. . Environment-specific expression of the immediate-early gene Arc in hippocampal neuronal ensembles. Nat Neurosci. 1999 Dec;2(12):1120-4. PubMed.
  4. . Inhibition of activity-dependent arc protein expression in the rat hippocampus impairs the maintenance of long-term potentiation and the consolidation of long-term memory. J Neurosci. 2000 Jun 1;20(11):3993-4001. PubMed.
  5. . Arc/Arg3.1 interacts with the endocytic machinery to regulate AMPA receptor trafficking. Neuron. 2006 Nov 9;52(3):445-59. PubMed.
  6. . Arc/Arg3.1 mediates homeostatic synaptic scaling of AMPA receptors. Neuron. 2006 Nov 9;52(3):475-84. PubMed.
  7. . Arc/Arg3.1 is essential for the consolidation of synaptic plasticity and memories. Neuron. 2006 Nov 9;52(3):437-44. PubMed.
  8. . APP processing and synaptic function. Neuron. 2003 Mar 27;37(6):925-37. PubMed.
  9. . Synaptic activity regulates interstitial fluid amyloid-beta levels in vivo. Neuron. 2005 Dec 22;48(6):913-22. PubMed.
  10. . Endocytosis is required for synaptic activity-dependent release of amyloid-beta in vivo. Neuron. 2008 Apr 10;58(1):42-51. PubMed.
  11. . Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer's disease. Nat Genet. 2011 May;43(5):436-41. Epub 2011 Apr 3 PubMed.
  12. . Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer's disease. Nat Genet. 2011 May;43(5):429-35. PubMed.
  13. . Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson's models. Science. 2006 Jul 21;313(5785):324-8. PubMed.
  14. . Alpha-synuclein is part of a diverse and highly conserved interaction network that includes PARK9 and manganese toxicity. Nat Genet. 2009 Mar;41(3):308-15. PubMed.
  15. . Yeast cells provide insight into alpha-synuclein biology and pathobiology. Science. 2003 Dec 5;302(5651):1772-5. PubMed.
  16. . Yeast genes that enhance the toxicity of a mutant huntingtin fragment or alpha-synuclein. Science. 2003 Dec 5;302(5651):1769-72. PubMed.
  17. . Nucleated conformational conversion and the replication of conformational information by a prion determinant. Science. 2000 Aug 25;289(5483):1317-21. PubMed.
  18. . Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science. 2003 Apr 18;300(5618):486-9. PubMed.
  19. . Hsp104 catalyzes formation and elimination of self-replicating Sup35 prion conformers. Science. 2004 Jun 18;304(5678):1793-7. PubMed.

External Citations

  1. APPswe/PS1deltaE9
  2. PICALM
  3. BIN1
  4. AlzGene database
  5. CD2AP
  6. ChemIDplus

Further Reading

Papers

  1. . Modelling neurodegeneration in Saccharomyces cerevisiae: why cook with baker's yeast?. Nat Rev Neurosci. 2010 Jun;11(6):436-49. PubMed.

Primary Papers

  1. . Functional links between Aβ toxicity, endocytic trafficking, and Alzheimer's disease risk factors in yeast. Science. 2011 Dec 2;334(6060):1241-5. PubMed.
  2. . Arc/Arg3.1 regulates an endosomal pathway essential for activity-dependent β-amyloid generation. Cell. 2011 Oct 28;147(3):615-28. PubMed.