The Role of Rab GTPases in Alzheimer's Disease - ACS Chemical

Sep 27, 2018 - Alzheimer's disease (AD) comprises two major pathological hallmarks: extraneuronal deposition of β-amyloid (Aβ) peptides (“senile p...
0 downloads 0 Views 509KB Size
Subscriber access provided by Kaohsiung Medical University

Review

The Role of Rab GTPases in Alzheimer’s Disease Xian Zhang, Timothy Y. Huang, Joel Yancey, Hong Luo, and Yunwu Zhang ACS Chem. Neurosci., Just Accepted Manuscript • DOI: 10.1021/acschemneuro.8b00387 • Publication Date (Web): 27 Sep 2018 Downloaded from http://pubs.acs.org on September 28, 2018

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

The Role of Rab GTPases in Alzheimer’s Disease

Xian Zhang,† Timothy Y. Huang,‡ Joel Yancey,‡ Hong Luo,† and Yun-wu Zhang†,*



Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research,

Institute of Neuroscience, Medical College of Xiamen University, Xiamen, Fujian 361102, China ‡

Neuroscience Initiative, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla,

CA 92037, USA

*

Corresponding author: Yun-wu Zhang, Fujian Provincial Key Laboratory of

Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College of Xiamen University, Xiamen, Fujian 361102, China. E-mail: [email protected].

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ABSTRACT: Alzheimer's disease (AD) comprises two major pathological hallmarks: extraneuronal deposition of β-amyloid (Aβ) peptides (“senile plaques”) and intraneuronal aggregation of the microtubule-associated protein tau (“neurofibrillary tangles”). Aβ is derived from sequential cleavage of the β-amyloid precursor protein by β- and γ-secretases, while aggregated tau is hyperphosphorylated in AD. Mounting evidence suggests that dysregulated trafficking of these AD-related proteins contributes to AD pathogenesis. Rab proteins are small GTPases that function as master regulators of vesicular transport and membrane trafficking. Multiple Rab GTPases have been implicated in AD-related protein trafficking and their expression has been observed to be altered in postmortem AD brain. Here we review current implicated roles of Rab GTPase dysregulation in AD pathogenesis. Further elucidation of the pathophysiological role of Rab GTPases will likely reveal novel targets for AD therapeutics.

KEYWORDS: β-amyloid, β-amyloid precursor protein, Alzheimer’s disease, Rab GTPases, tau, trafficking

ACS Paragon Plus Environment

Page 2 of 53

Page 3 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1. INTRODUCTION Alzheimer’s disease (AD) is the most prevalent neurodegenerative disease described thus far, and is clinically characterized by progressive cognitive impairment and memory loss. Two classical pathological hallmarks in AD include the appearance of intraneuronal neurofibrillary tangles (NFTs) and extraneuronal senile plaques.1, 2 NFTs are pathological anomalies consisting of aberrant phosphorylated forms of the microtubule-associated protein tau, which is predominantly hyperphosphorylated by CDK5 and GSK-3 kinases in AD.1,

3

In contrast, senile plaques consist of a heterogeneous population of

proteolytically-generated β-amyloid (Aβ) peptides.4, 5 Results from mounting studies suggest that excessive Aβ in the brain, due either to elevated production or reduced clearance, initiates AD pathogenesis.6, 7 Aβ is derived from amyloidogenic processing of the β-amyloid precursor protein (APP), whereby APP is first cleaved by β-secretase within the ectodomain, generating a soluble sAPPβ fragment and a membrane-anchored β-carboxyl-terminal fragment (βCTF) fragment. The predominant β-secretase enzyme for APP cleavage is BACE1 (beta-site APP-cleaving enzyme 1), which is a type-I transmembrane aspartyl protease.8-11 After β-cleavage, APP βCTF can be cleaved by γ-secretase within the lipid bilayer, leading to the generation of Aβ and APP intracellular domain (AICD). γ-secretase is a high molecular weight complex consisting of at least four transmembrane components: Presenilin (PS1 or PS2), nicastrin, anterior pharynx-defective-1 (APH-1), and presenilin enhancer-2 (PEN2).12, 13 Apart from amyloidogenic processing, APP can also be cleaved by α-secretases within

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

the Aβ domain, releasing an sAPPα fragment. APP processing through this non-amyloidogenic pathway precludes Aβ production and has neurotrophic and neuroprotective effects.14,

15

Several members of the ADAM (a disintegrin and

metalloproteinase) family have been found to possess α-secretase activity, such as ADAM10, ADAM17 and ADAM9.15, 16 Interestingly, α-secretases cleave APP primarily at the cell surface, whereas β- and γ-secretases primarily cleave APP in acidified trans-Golgi and endosomal environments, thereby generating Aβ within the lumen of endosomes, leading to their eventual extracellular secretion.17 Therefore, APP trafficking and its coincident localization with related secretases in endosomes is crucial for Aβ production. This process requires the participation of various trafficking factors including the Rab (Ras-associated binding) family of GTPases; dysregulation of these trafficking factors may affect Aβ production and thus potentially contribute to AD pathogenesis.

2. RAB GTPASES Rab proteins are low molecular weight GTPases that belong to the Ras superfamily.18 By tethering/docking vesicles to their respective compartments, Rab GTPases function as molecular switches, alternating between GTP-bound active and GDP-bound inactive forms which can direct vesicle transport to their appropriate destinations.19 Cycling between GTP- and GDP-bound forms is tightly controlled by protein regulators such as GTPase activating proteins (GAPs), GDP dissociation inhibitors (GDIs) and Guanine exchange factors (GEFs): GAPs inactivate GTP-bound GTPases by promoting GTP

ACS Paragon Plus Environment

Page 4 of 53

Page 5 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

hydrolysis to GDP; GDIs stabilize GDP-bound GTPases; and GEFs activate GTPases by facilitating the exchange of GDP to GTP.18 A single Rab GTPase may exert various trafficking-dependent functions in conjunction with its regulators;20 for example, the well characterized RAB5A/RAB5 protein has been found to interact with over 20 different effectors19 and to participate in endocytic vesicle formation, uncoating, movement,21-23 and endosome fusion.24 More than 60 human Rab GTPases have been identified.25 The overrepresentation of Rab GTPases, in addition to their functional complexity, highlight their importance in vesicle trafficking. Members of the evolutionarily conserved Rab GTPase protein family regulate fundamental vesicle transport pathways and are often ubiquitously expressed. Conversely, Rab family members with poor conservation are typically enriched in various tissues and function in more specific transport pathways.18,

20

For example,

RAB3A, RAB8A/RAB8 and RAB23 are highly expressed in the brain and participate in synaptic vesicle exocytosis, postsynaptic glutamate receptor dynamics, neurite growth, and/or neural development.26 Dysregulation of Rab GTPases may therefore contribute to the pathogenesis of some diseases. Indeed, enlargement of RAB5-positive early endosomes has been observed in AD patient brain, and these endosomal pathological features can be detected in brain regions lacking senile plaques or tau tangles.27, 28 This suggests that early endosomal abnormalities occur prior to the appearance of Aβ or tau pathology. Nevertheless, causal and consequential relationships underlying the association of AD with various Rab GTPases have yet to be determined. One recent

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

review classified the association relevance of Rab GTPases with neurodegenerative diseases into three categories: Rab GTPases whose mutations are the primary cause of neurodegeneration (Class A); Rab GTPases which do not trigger neurodegeneration, but rather affects pathology (Class B); and Rab GTPases which are neither the neurodegenerative triggers nor consequential components to pathology (Class C).29 Through this classification scheme, we similarly categorized Rab GTPases to clarify their respective causal and effectual relationship with AD (Table 1).

3. GENETIC ASSOCIATION OF RAB GTPASES WITH AD Genetic association often provides direct evidence for the causal role of a gene in disease. Mutations in genes encoding APP and the γ-secretase component Presenilin1/2 can enhance the production of total Aβ or the more toxic Aβ42 species and lead to familial, early onset AD (fAD or EOAD), providing strong genetic evidence in support of the “Aβ cascade hypothesis” in AD pathogenesis. However, familial mutations only account for a small minority of AD cases; over 95% of AD cases manifest sporadic, late onset (sAD or LOAD) variants of the disease which lack a well-characterized etiology. The ε4 allele of the APOE gene is the strongest genetic risk factor identified thus far for LOAD.30 Although cholesterol transport is thought to be the primary function of the APOE gene product, apolipoprotein E (apoE), apoE also regulates Aβ metabolism through cellular uptake and degradation.30 More recently, large‐scale genome‐wide association studies (GWAS) and meta‐analyses have identified more than 20 genes associated with

ACS Paragon Plus Environment

Page 6 of 53

Page 7 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

LOAD.31-35 Interestingly, a significant number of the newly identified AD-linked genes encode proteins that play roles in endocytic trafficking, such as PICALM, BIN1, SORL1 and CD2AP.36 Mutations in genes encoding Rab GTPases have been associated with various diseases. For example, RAB27A/RAB27 mutations have been described in Griscelli syndrome, an autosomal recessive disease with defective melanosome transport and deregulated T lymphocyte and macrophage activation.37, 38 RAB23 mutations have been linked to Carpenter syndrome, an autosomal recessive disease which causes cranio-synostosis, obesity and limb malformation.39 RAB7A/RAB7 mutations lead to Charcot‐Marie‐Tooth type 2B disease with sensory loss, distal muscle weakness and high frequency of foot ulcers and infections.40 RAB39B mutations cause X-linked mental retardation and Parkinson’s disease.20, 41-46 Unfortunately, reports describing the genetic association between Rabs and AD are limited: in one study characterizing the genetic association of AD with retromer or retromer-associated proteins in Caucasian samples, RAB7A was identified to be associated with AD.47 The authors also demonstrated that RAB7A interacts with the retromer complex, thereby mediating its association with membranes.47 In another study, "AD resilient" APOE ε4 allele carriers over 75 years of age lacking clinical symptoms of cognitive decline were characterized for genetic factors associated with the lack of AD onset. The authors showed that an SNP (rs142787485) in RAB10 confers significant protection against AD.48 Exome sequencing of LOAD patients and controls also identified significant genetic association of a variant in RAB11A

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(rs117150201) with AD.49 In addition to Rab GTPases, GWAS analyses also implicated a RAB5 guanine nucleotide exchange factor, RIN3 (Ras and Rab interactor 3) 50-52 as a risk factor for AD onset.32-34 Recently, a missense variant in the RIN3 gene was found to be associated with EOAD.53 Despite the genetic implications of these findings, whether these Rab GTPases and regulators have a causal role in AD pathogenesis (Class A) requires further corroboration.

4. ALTERATIONS OF RAB GTPASES IN AD Gene expression and/or protein levels of multiple Rab GTPases have been found to be altered in samples from AD patients, and also in animal and cell models. However, whether such changes contribute to disease progression, or merely signify downstream effects accompanying AD is difficult to ascertain. There is evidence suggesting that Rab GTPases may also be perturbed by AD toxicity. For instance, neuronal internalization of Aβ can trigger neurodegeneration accompanied by increased levels and/or altered distribution of RAB5 and RAB7.54 It is likely that Rab GTPases which regulate Aβ and/or tau and undergo early changes in expression and/or distribution contribute to AD progression (Class B). Rab GTPases that show changes late in AD onset which have no effect on modulating Aβ and/or tau may represent downstream effectors in AD pathogenesis (Class C). RAB5 may be the most extensively characterized Rab GTPase of all family members. By interacting with various effectors, RAB5 regulates tethering and fusion of early

ACS Paragon Plus Environment

Page 8 of 53

Page 9 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

endosomes, endosomal membrane trafficking, and endosomal sorting and fusion.21-24, 55, 56

RAB5-positive early endosomes are significantly enlarged in AD brain and are

associated with elevated levels of the recycling marker RAB4,57 signifying that both endocytic uptake and recycling are activated in AD.27 One study characterizing AD-associated gene expression in CA1 neurons determined that levels of RAB4A/RAB4, RAB5, RAB7 (involved in late endosomes58), and RAB24 (involved in autophagy59) were significantly up-regulated in mild cognitive impairment (MCI) and AD individuals.28 In cholinergic basal forebrain neurons microdissected from postmortem

brains

of

MCI

and

AD

patients,

RAB4,

RAB5,

RAB7,

and RAB27A/RAB27 (involved in exocytosis, endocytosis and phagocytosis60) were found to be upregulated, and their expression correlated with antemortem measures of cognitive decline.61 Selective upregulation of RAB5 and RAB7 levels was observed within basal forebrain, frontal cortex and hippocampus in MCI and AD, which also correlated with advanced Braak staging.62 Additionally, RAB7 levels were found to be elevated in the frontal cortex region in rapid progressive AD forms.63 RAB10 plays an important role in the exocytosis/secretory pathway and may be involved in neurotransmitter release and phagosome maturation.64 In addition to its genetic association with AD, RAB10 expression was also found to be significantly increased in human AD brain; and downregulation of RAB10 resulted in a significant decrease in Aβ42 levels and in the Aβ42/Aβ40 ratio in neuroblastoma cells.48 Another study also determined that the expression of RAB10 as well as its microRNA regulator,

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

miRNA-655, were both altered in AD samples.65 One recent study found that RAB10 phosphorylation at threonine 73 was prominent in NFTs in neurons within the hippocampus of AD cases when compared to controls, suggesting that RAB10 phosphorylation might also be involved in AD.66 Given that Rab GTPases often regulate synaptic function, it is not surprising that levels of RAB3A are enriched in presynaptic vesicles and its levels were found to be markedly decreased in the frontal and parietal cortices of AD specimens, which correlated with cognitive impairment in AD.67 RAB8A/RAB8 is an important regulator of polarized trafficking and is involved in trans-Golgi network (TGN)-basolateral plasma membrane trafficking, protrusion formation and ciliogenesis.68-70 RAB8 levels were significantly increased in the membrane fractions from AD brain tissues compared to controls.71 RAB6A/RAB6 is involved in retrograde Golgi-ER trafficking and was found to be upregulated in the temporal cortex of Braak stage 3/4 AD brains.72 Extensive alterations in Rab family members in AD brain highlight the contribution of Rab GTPase dysregulation to AD and support the idea that endosomal pathology may promote aberrant trafficking, signaling, and neurodegeneration throughout the progression of AD.

5. A ROLE FOR RAB GTPASES IN APP PROCESSING The finding that the majority of Rab GTPase levels are increased in AD is consistent with

ACS Paragon Plus Environment

Page 10 of 53

Page 11 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

the idea that enhanced endocytosis may promote Aβ production and accumulation in early stages of AD onset.27, 73 Aβ is derived from APP through sequential cleavage events by BACE1 and γ-secretase. Amyloidogenic cleavage occurs predominantly in intracellular compartments such as early endosomes, late endosomes and the TGN.74-76 Therefore, sorting APP and its cognate secretase enzymes into coincident compartments to initiate Aβ production involves the participation of endocytic regulators including Rab GTPases. On the other hand, α-secretases cleave APP primarily at the cell surface such that changes in trafficking may also interfere with APP α-cleavage, leading to alterations in β-cleavage.

5.1. Rab-Mediated Regulation of APP Trafficking Nascent APP is transported via the secretory pathway through the endoplasmic reticulum (ER) and the TGN apparatus to the plasma membrane. APP is then either cleaved by α-secretase to produce sAPPα or re-internalized into early endosomes. APP in early endosomes can either be recycled back to the cell surface or be delivered to late endosomes and lysosomes for degradation.77-79 Multiple Rab GTPases have been implicated in regulating APP trafficking. For example, RAB1B regulates vesicular transport between the ER and successive Golgi compartments.80 In a study on APP anterograde transport, RAB1B was found to play a key role in APP trafficking from the ER to Golgi: a dominant-negative RAB1B mutant blocked APP ER/Golgi transport and significantly inhibited Aβ secretion.81 Another study

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 53

found that during APP anterograde transport, APP was present in vesicles containing kinesin-1C (a kinesin heavy chain isoform), ADAM10 (an α-secretase) and RAB3A. Further, RAB3A GTPase was shown to be necessary for the assembly of these vesicles.82 RAB6 is localized in the late Golgi apparatus and is involved in retrograde transport from the Golgi apparatus to the ER, transport of early endosomes and recycling endosomes to TGN, and vesicle exocytosis.83-87 One study found that RAB6 may function as a negative regulator of APP anterograde trafficking through the TGN or as a positive modulator of APP retrograde trafficking back to the TGN. A dominant-negative RAB6 mutant may promote anterograde trafficking of APP through the secretory pathway to promote α-secretase-mediated cleavage.88 The function of RAB6 in modulating APP processing may be mediated through its interaction with Mint family proteins,89, 90 which have been shown to fine-tune APP trafficking and processing by binding to the APP YENPTY motif.91, 92 In addition, RAB6 was found to modulate the unfolded protein response, suggesting that increased RAB6 levels in AD brain may result in a failure to recover from ER stress, thereby contributing to neurodegeneration in AD.72, 93

Rab11

proteins

comprise

related

proteins,

RAB11A,

RAB11B

and

RAB11C/RAB25.94 They direct exocytosis and recycling processes, thereby regulating secretion and plasma membrane composition.94, 95 Interestingly, RAB11B has been found to colocalize to some extent with APP in axons.96 Knockdown of RAB11B led to defects in soma‐to‐axon transcytosis of APP in neurons.97

ACS Paragon Plus Environment

Page 13 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

The amyloid precursor-like protein (APL-1) is the sole APP ortholog in Caenorhabditis elegans. RAB5 knockdown also dramatically decreased APL-1 expression in neurons, suggesting that retrograde trafficking from the plasma membrane to early endosomes is important for APL-1 function.98

5.2. Rab GTPases and BACE1 As the predominant β-secretase for APP, BACE1 initiates APP processing in acidified endosomes, where BACE1 is found to possess maximal enzymatic activity. Imaging analysis revealed that BACE1 was localized and transported along dendrites and axons in RAB11B-positive recycling endosomes. Impairment of RAB11B function led to a diminution of total and endocytosed BACE1 in axons, concomitant with increased BACE1 in the soma. These results suggest that RAB11B function is critical for axonal sorting of BACE1.99 In a human Rab RNAi screening for Aβ modulators, downregulation of RAB3A, RAB11A, RAB17 (involved in phagocytic removal of apoptotic cells100), and RAB36 (involved in late endosome and lysosome distribution101) reduced both Aβ and sAPPβ, implicating a potential role for these factors in modulating BACE1.49 Consequent studies validated a role for RAB11A and its homolog RAB11B in regulating β-cleavage and Aβ generation through controlling BACE1 membrane trafficking and recycling.49

5.3. Rab GTPases and PS1

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

PS1 is the most extensively characterized component of the γ-secretase complex and functions as its catalytic center. PS1 may also exert other γ-secretase-independent functions by interacting with effector proteins. Although PS1 can interact with RAB11A,102 whether PS1 trafficking is regulated by RAB11A or other Rab proteins has yet to be established. Overexpression of RAB11A has no effect on APP processing in HEK293 cells.103 However, the possibility remains that RAB11A may still modulate other aspects of PS1-dependent function. On the other hand, it seems likely that PS1 may have an upstream role in regulating the function of Rab GTPases and Rab-related proteins. It has been reported that membrane association of RAB6 but not RAB1 is dependent on PS1.104 The PS1 amino-terminus was found to interact with Rab GDI, and membrane-associated Rab GDI levels were found to be dramatically decreased in the absence of PS1.105 These findings imply that PS1 may be a membrane receptor for certain Rabs and Rab-related proteins. In cells expressing fAD-associated PS1 mutants (G384A and I143T), and in neurons from 3xTg-AD mice, RAB4 and RAB6 levels were found to be increased.106 In contrast, RAB8 levels were decreased in cells expressing a PS1 mutant (A260V).107 Moreover, in human iPSC-derived neurons carrying a fAD PS1 mutation (∆E9), RAB11B distribution was altered in a manner similar to APP.97 Therefore, PS1 mutations may perturb Rab proteins and membrane vesicle transport, thereby facilitating amyloidogenic APP processing and Aβ generation through γ-cleavage-independent pathways.

ACS Paragon Plus Environment

Page 14 of 53

Page 15 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

5.4. Rab GTPases and α-secretases Several type I transmembrane ADAM family proteins have been found to possess α-secretase activity. Among them, ADAM10 is believed to be the constitutive α-secretase that is active at the cell surface.16, 108 Other α-secretases, such as ADAM17 and ADAM9 affect only regulated α-cleavage in various cell lines.109, 110 RAB14 functions in the endocytic recycling pathway and is involved in trafficking between the Golgi and endosomal compartments.111, 112 It was found that RAB14 can regulate ADAM10 recycling: its depletion resulted in ADAM10 accumulation in a transferrin-positive endocytic compartment, accompanied by reduced cell-surface levels of ADAM10.113, 114 In addition, another study found that downregulation of RAB11A could reduce cell surface expression of ADAM17.115 Hence, aberrations in various Rab GTPase family proteins may indirectly affect Aβ production, implicating their potential contributions in AD.

6. RAB GTPASES AND Aβ DEGRADATION/AGGREGATION Aggregation of Aβ peptides leads to synaptic disruption and neurodegeneration. A major clearance pathway for extracellular Aβ is through cellular uptake and degradation. One study demonstrated that endocytic Aβ internalized in neurons trafficked primarily via early and late endosomes to the lysosome; and lysosomal inhibition resulted in Aβ accumulation and aggregation.116 In addition, endocytosed Aβ caused multivesicular body enlargement in neurons and generated fibril-like structures in endocytic vesicles.117 On

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 53

the other hand, overexpression of either the early endosome protein RAB5 or the late endosome protein RAB7 accelerated endocytic Aβ trafficking to the lysosome.116 Moreover,

a

proportion

of

endocytically

internalized



was

observed

in

RAB11A-positive recycling vesicles. Use of a constitutively active RAB11A mutant to block this recycling significantly accelerated cellular Aβ accumulation.116 Allelic variation in the APOE gene is the strongest risk factor for LOAD. In addition to transporting cholesterol in the brain, apoE also participates in Aβ degradation and the reduction of cellular cholesterol levels by apoE leads to accelerated Aβ delivery to lysosomes to enhance its degradation.118 Moreover, apoE facilitates the recycling of RAB7 from lysosomes to early endosomes, suggesting that efficient RAB7 recycling may accelerate faster endocytic trafficking of Aβ-containing vesicles and enhance Aβ degradation.118 The assembly of soluble Aβ to Aβ fibrils in senile plaques is a critical event in AD. Although abnormalities in the endocytic pathway precede extracellular Aβ fibril deposition in the brain,27 blocking the early endocytic pathway by RAB5 depletion did not lead to fibril formation on the cell surface with exogenous addition of soluble Aβ. In contrast, blocking the late endocytic pathway by RAB7 suppression markedly induced Aβ fibril formation and enlargement of early endosomes. These results suggest that dysfunction of late, rather than early, endocytic mechanisms contribute to Aβ fibril formation, possibly by facilitating the generation of Aβ seeds in AD brain.119

ACS Paragon Plus Environment

Page 17 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

7. RAB GTPASES AND TAU SECRETION The microtubule‐associated protein tau can be released into the extracellular space, possibly via exosomes, and subsequently resorbed by neurons and glial cells, an event that has been proposed to contribute to the propagation of tau pathology.120-123 Recently it was found that RAB7A, involved in the trafficking of endosomes, autophagosomes and lysosomes, can also regulate tau secretion. Deletion of RAB7A and overexpression of a dominant negative form of RAB7A decreased tau secretion, whereas overexpression of a constitutively active form of RAB7A showed converse effects.124 Elevated RAB7A levels in human AD brains and brains from AD mouse models, as well as colocalization between RAB7A and tau, suggests that RAB7A may contribute to tau propagation in AD.63, 124 Neuronal hyperexcitability can promote tau release by neurons and induce Golgi fragmentation. RAB1A is associated with Golgi membranes and its suppression is known to induce Golgi fragmentation. One study found that suppression of RAB1A also increased tau secretion.125 Although the underlying mechanism remains elusive, this indicates that Rab-mediated Golgi dynamics can modulate tau secretion.

8. POTENTIAL USE OF RAB GTPASES IN AD DIAGNOSTICS The success of future AD therapeutics will likely rely on the development of treatment strategies targeting early stages of AD onset, prior to late stage events associated with irreversible brain damage. Neuronal degeneration is observed long before the clinical

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

onset, highlighting the need for biomarkers that can be used for early diagnosis in AD. Since Aβ and tau are major components of AD pathology, they are key indicators of AD pathogenesis. Indeed, it has gradually become accepted that Aβ42 levels are decreased, whereas total tau and phosphorylated-tau levels are increased in cerebrospinal fluid (CSF) of AD patients in early clinical stages when compared to control individuals. This suggests that changes in Aβ42 and total tau/phosphorylated-tau levels may be viable biomarkers in the early diagnosis of AD.126, 127 By using ultra-sensitive techniques, it has recently been noted that both Aβ and phosphorylated tau levels in plasma from AD patients also show differences compared to controls.128, 129 The microglia-enriched innate immune receptor TREM2 has been previously described as a potent genetic risk factor in AD.130-132 Recently, levels of soluble TREM2 (sTREM2) derived from proteolytic TREM2 ectodomain cleavage were found to be increased and correlated with total tau and phosphorylated-tau in CSF from MCI and AD patients. Levels of sTREM2 were also found to be increased in CSF from fAD mutation carriers compared to non-carriers.133, 134 These results suggest that sTREM2 may be a viable early AD biomarker. Changes in the endo-lysosomal network are among the first alterations observed in the AD brain, and characterization of proteins within the endo-lysosomal network in human CSF indicated that RAB3A and RAB7 levels were markedly increased in AD patient samples compared to controls.135 Therefore, further characterization of changes in RAB3 and RAB7 levels in the CSF as well as the plasma from AD patients may determine whether they can be used as viable AD biomarkers and evaluate endo-lysosomal dysfunction during AD

ACS Paragon Plus Environment

Page 18 of 53

Page 19 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

development.

9. THERAPEUTIC POTENTIAL OF RAB GTPASES Dysregulation of the endo‐lysosomal system is an early cellular phenotypic event in AD pathogenesis. Treatment with the lysosomal modulator Z-Phe-Ala-diazomethylketone (PADK) in AD mouse models increased cathepsin B protein levels and enzymatic activity, increased Aβ truncation and reduction, and consequently reversed impairments in animal behaviors and synaptic proteins. Biochemical analysis revealed marked changes to Rab proteins but not to LAMP1 upon PADK treatment, suggesting the role of trafficking modulation during rescue.136 In addition, epidemiological studies suggest that statins may reduce the risk of AD through undetermined mechanisms. One study found that fluvastatin at clinical doses resulted in significant reductions in Aβ and APP βCTF levels, but this effect was abolished with the additive treatment of lysosomal inhibitors. Further analysis indicated that reduced Aβ production was caused by enhanced lysosomal degradation of APP βCTF. This effect arose from enhanced endosomal APP βCTF trafficking to lysosomes, and it was associated with marked changes in Rab proteins.137 Taken together, these findings demonstrate that lysosomal modulation can reduce Aβ accumulation and improve behavioral impairment, suggesting that targeting Rab GTPases may be a viable therapeutic target in AD.

10. CONCLUDING REMARKS

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Rab GTPases are implicated in multiple pathological mechanisms in AD (Table 1). Interestingly, many AD-associated proteins identified thus far play roles in trafficking pathways, some of which are linked to Rab proteins. For instance, PICALM plays an important role in regulating Aβ internalization, trafficking and clearance via the low‐ density lipoprotein receptor‐related protein‐1 and Rab proteins (RAB5 and Rab11 members).138,

139

Dysregulation of AD-associated proteins and Rab GTPases perturbs

endosomal, lysosomal, recycling and autophagic pathways. Further elucidation of the complex physiological function of Rab GTPases and their pathological roles in AD holds great potential for developing novel therapeutics in disease intervention.

ACS Paragon Plus Environment

Page 20 of 53

Page 21 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

AUTHOR INFORMATION Corresponding Author *E-mail: [email protected].

Author Contributions The manuscript was written through contributions of all authors. All authors have given approval to the final version of the manuscript.

Funding This work was supported in part by grants from The National Key Research and Development Program of China (2016YFC1305903), The National Natural Science Foundation of China (81771377 and U1705285 to Y-w.Z.) and Fundamental Research Funds for the Central Universities (20720180049 to Y-w.Z.).

Notes The authors declare no competing financial interest.

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

REFERENCES 1. Lee, V. M., Goedert, M., and Trojanowski, J. Q. (2001) Neurodegenerative tauopathies, Annu. Rev. Neurosci. 24, 1121-1159. 2. Hippius, H., and Neundorfer, G. (2003) The discovery of Alzheimer's disease, Dialogues Clin Neurosci 5, 101-108. 3. Ercan, E., Eid, S., Weber, C., Kowalski, A., Bichmann, M., Behrendt, A., Matthes, F., Krauss, S., Reinhardt, P., Fulle, S., and Ehrnhoefer, D. E. (2017) A validated antibody panel for the characterization of tau post-translational modifications, Mol Neurodegener 12, 87. 4. Glenner, G. G., and Wong, C. W. (1984) Alzheimer's disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein, Biochem. Biophys. Res. Commun. 120, 885-890. 5. Masters, C. L., Simms, G., Weinman, N. A., Multhaup, G., McDonald, B. L., and Beyreuther, K. (1985) Amyloid plaque core protein in Alzheimer disease and Down syndrome, Proc Natl Acad Sci U S A 82, 4245-4249. 6. Hardy, J., and Selkoe, D. J. (2002) The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics, Science 297, 353-356. 7. Hardy, J. A., and Higgins, G. A. (1992) Alzheimer's disease: the amyloid cascade hypothesis, Science 256, 184-185. 8. Sinha, S., Anderson, J. P., Barbour, R., Basi, G. S., Caccavello, R., Davis, D., Doan, M.,

ACS Paragon Plus Environment

Page 22 of 53

Page 23 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Dovey, H. F., Frigon, N., Hong, J., Jacobson-Croak, K., Jewett, N., Keim, P., Knops, J., Lieberburg, I., Power, M., Tan, H., Tatsuno, G., Tung, J., Schenk, D., Seubert, P., Suomensaari, S. M., Wang, S., Walker, D., Zhao, J., McConlogue, L., and John, V. (1999) Purification and cloning of amyloid precursor protein beta-secretase from human brain, Nature 402, 537-540. 9. Vassar, R., Bennett, B. D., Babu-Khan, S., Kahn, S., Mendiaz, E. A., Denis, P., Teplow, D. B., Ross, S., Amarante, P., Loeloff, R., Luo, Y., Fisher, S., Fuller, J., Edenson, S., Lile, J., Jarosinski, M. A., Biere, A. L., Curran, E., Burgess, T., Louis, J. C., Collins, F., Treanor, J., Rogers, G., and Citron, M. (1999) Beta-secretase cleavage of Alzheimer's amyloid precursor protein by the transmembrane aspartic protease BACE, Science 286, 735-741. 10. Yan, R., Bienkowski, M. J., Shuck, M. E., Miao, H., Tory, M. C., Pauley, A. M., Brashier, J. R., Stratman, N. C., Mathews, W. R., Buhl, A. E., Carter, D. B., Tomasselli, A. G., Parodi, L. A., Heinrikson, R. L., and Gurney, M. E. (1999) Membrane-anchored aspartyl protease with Alzheimer's disease beta-secretase activity, Nature 402, 533-537. 11. Lau, K. F., McLoughlin, D. M., Standen, C., and Miller, C. C. (2000) X11 alpha and x11 beta interact with presenilin-1 via their PDZ domains, Mol. Cell. Neurosci. 16, 557-565. 12. Kimberly, W. T., LaVoie, M. J., Ostaszewski, B. L., Ye, W., Wolfe, M. S., and Selkoe, D. J. (2003) Gamma-secretase is a membrane protein complex comprised of presenilin, nicastrin, Aph-1, and Pen-2, Proc Natl Acad Sci U S A 100, 6382-6387.

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

13. Takasugi, N., Tomita, T., Hayashi, I., Tsuruoka, M., Niimura, M., Takahashi, Y., Thinakaran, G., and Iwatsubo, T. (2003) The role of presenilin cofactors in the gamma-secretase complex, Nature 422, 438-441. 14. Zheng, H., and Koo, E. H. (2011) Biology and pathophysiology of the amyloid precursor protein, Mol Neurodegener 6, 27. 15. Zhang, Y. W., Thompson, R., Zhang, H., and Xu, H. (2011) APP processing in Alzheimer's disease, Mol Brain 4, 3. 16. Lammich, S., Kojro, E., Postina, R., Gilbert, S., Pfeiffer, R., Jasionowski, M., Haass, C., and Fahrenholz, F. (1999) Constitutive and regulated alpha-secretase cleavage of Alzheimer's amyloid precursor protein by a disintegrin metalloprotease, Proc Natl Acad Sci U S A 96, 3922-3927. 17. Jiang, S., Li, Y., Zhang, X., Bu, G., Xu, H., and Zhang, Y. W. (2014) Trafficking regulation of proteins in Alzheimer's disease, Mol Neurodegener 9, 6. 18. Corbeel, L., and Freson, K. (2008) Rab proteins and Rab-associated proteins: major actors in the mechanism of protein-trafficking disorders, Eur. J. Pediatr. 167, 723-729. 19. Christoforidis, S., McBride, H. M., Burgoyne, R. D., and Zerial, M. (1999) The Rab5 effector EEA1 is a core component of endosome docking, Nature 397, 621-625. 20. Gao, Y., Wilson, G. R., Stephenson, S. E. M., Bozaoglu, K., Farrer, M. J., and Lockhart, P. J. (2018) The emerging role of Rab GTPases in the pathogenesis of Parkinson's disease, Mov. Disord. 33, 196-207. 21. Semerdjieva, S., Shortt, B., Maxwell, E., Singh, S., Fonarev, P., Hansen, J., Schiavo,

ACS Paragon Plus Environment

Page 24 of 53

Page 25 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

G., Grant, B. D., and Smythe, E. (2008) Coordinated regulation of AP2 uncoating from clathrin-coated vesicles by rab5 and hRME-6, J. Cell Biol. 183, 499-511. 22. McLauchlan, H., Newell, J., Morrice, N., Osborne, A., West, M., and Smythe, E. (1998) A novel role for Rab5-GDI in ligand sequestration into clathrin-coated pits, Curr. Biol. 8, 34-45. 23. Nielsen, E., Severin, F., Backer, J. M., Hyman, A. A., and Zerial, M. (1999) Rab5 regulates motility of early endosomes on microtubules, Nat Cell Biol 1, 376-382. 24. Ohya, T., Miaczynska, M., Coskun, U., Lommer, B., Runge, A., Drechsel, D., Kalaidzidis, Y., and Zerial, M. (2009) Reconstitution of Rab- and SNARE-dependent membrane fusion by synthetic endosomes, Nature 459, 1091-1097. 25. Diekmann, Y., Seixas, E., Gouw, M., Tavares-Cadete, F., Seabra, M. C., and Pereira-Leal, J. B. (2011) Thousands of rab GTPases for the cell biologist, PLoS Comput Biol 7, e1002217. 26. Ng, E. L., and Tang, B. L. (2008) Rab GTPases and their roles in brain neurons and glia, Brain Res Rev 58, 236-246. 27. Cataldo, A. M., Peterhoff, C. M., Troncoso, J. C., Gomez-Isla, T., Hyman, B. T., and Nixon, R. A. (2000) Endocytic pathway abnormalities precede amyloid beta deposition in sporadic Alzheimer's disease and Down syndrome: differential effects of APOE genotype and presenilin mutations, Am. J. Pathol. 157, 277-286. 28. Ginsberg, S. D., Alldred, M. J., Counts, S. E., Cataldo, A. M., Neve, R. L., Jiang, Y., Wuu, J., Chao, M. V., Mufson, E. J., Nixon, R. A., and Che, S. (2010) Microarray

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

analysis of hippocampal CA1 neurons implicates early endosomal dysfunction during Alzheimer's disease progression, Biol. Psychiatry 68, 885-893. 29. Kiral, F. R., Kohrs, F. E., Jin, E. J., and Hiesinger, P. R. (2018) Rab GTPases and Membrane Trafficking in Neurodegeneration, Curr. Biol. 28, R471-R486. 30. Liu, C. C., Kanekiyo, T., Xu, H., and Bu, G. (2013) Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy, Nat Rev Neurol 9, 106-118. 31. Seshadri, S., Fitzpatrick, A. L., Ikram, M. A., DeStefano, A. L., Gudnason, V., Boada, M., Bis, J. C., Smith, A. V., Carassquillo, M. M., Lambert, J. C., Harold, D., Schrijvers, E. M., Ramirez-Lorca, R., Debette, S., Longstreth, W. T., Jr., Janssens, A. C., Pankratz, V. S., Dartigues, J. F., Hollingworth, P., Aspelund, T., Hernandez, I., Beiser, A., Kuller, L. H., Koudstaal, P. J., Dickson, D. W., Tzourio, C., Abraham, R., Antunez, C., Du, Y., Rotter, J. I., Aulchenko, Y. S., Harris, T. B., Petersen, R. C., Berr, C., Owen, M. J., Lopez-Arrieta, J., Varadarajan, B. N., Becker, J. T., Rivadeneira, F., Nalls, M. A., Graff-Radford, N. R., Campion, D., Auerbach, S., Rice, K., Hofman, A., Jonsson, P. V., Schmidt, H., Lathrop, M., Mosley, T. H., Au, R., Psaty, B. M., Uitterlinden, A. G., Farrer, L. A., Lumley, T., Ruiz, A., Williams, J., Amouyel, P., Younkin, S. G., Wolf, P. A., Launer, L. J., Lopez, O. L., van Duijn, C. M., and Breteler, M. M. (2010) Genome-wide analysis of genetic loci associated with Alzheimer disease, Jama 303, 1832-1840. 32. Harold, D., Abraham, R., Hollingworth, P., Sims, R., Gerrish, A., Hamshere, M. L., Pahwa, J. S., Moskvina, V., Dowzell, K., Williams, A., Jones, N., Thomas, C., Stretton, A., Morgan, A. R., Lovestone, S., Powell, J., Proitsi, P., Lupton, M. K., Brayne, C.,

ACS Paragon Plus Environment

Page 26 of 53

Page 27 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Rubinsztein, D. C., Gill, M., Lawlor, B., Lynch, A., Morgan, K., Brown, K. S., Passmore, P. A., Craig, D., McGuinness, B., Todd, S., Holmes, C., Mann, D., Smith, A. D., Love, S., Kehoe, P. G., Hardy, J., Mead, S., Fox, N., Rossor, M., Collinge, J., Maier, W., Jessen, F., Schurmann, B., Heun, R., van den Bussche, H., Heuser, I., Kornhuber, J., Wiltfang, J., Dichgans, M., Frolich, L., Hampel, H., Hull, M., Rujescu, D., Goate, A. M., Kauwe, J. S., Cruchaga, C., Nowotny, P., Morris, J. C., Mayo, K., Sleegers, K., Bettens, K., Engelborghs, S., De Deyn, P. P., Van Broeckhoven, C., Livingston, G., Bass, N. J., Gurling, H., McQuillin, A., Gwilliam, R., Deloukas, P., Al-Chalabi, A., Shaw, C. E., Tsolaki, M., Singleton, A. B., Guerreiro, R., Muhleisen, T. W., Nothen, M. M., Moebus, S., Jockel, K. H., Klopp, N., Wichmann, H. E., Carrasquillo, M. M., Pankratz, V. S., Younkin, S. G., Holmans, P. A., O'Donovan, M., Owen, M. J., and Williams, J. (2009) Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer's disease, Nat Genet 41, 1088-1093. 33. Lambert, J. C., Heath, S., Even, G., Campion, D., Sleegers, K., Hiltunen, M., Combarros, O., Zelenika, D., Bullido, M. J., Tavernier, B., Letenneur, L., Bettens, K., Berr, C., Pasquier, F., Fievet, N., Barberger-Gateau, P., Engelborghs, S., De Deyn, P., Mateo, I., Franck, A., Helisalmi, S., Porcellini, E., Hanon, O., de Pancorbo, M. M., Lendon, C., Dufouil, C., Jaillard, C., Leveillard, T., Alvarez, V., Bosco, P., Mancuso, M., Panza, F., Nacmias, B., Bossu, P., Piccardi, P., Annoni, G., Seripa, D., Galimberti, D., Hannequin, D., Licastro, F., Soininen, H., Ritchie, K., Blanche, H., Dartigues, J. F., Tzourio, C., Gut, I., Van Broeckhoven, C., Alperovitch, A., Lathrop, M., and Amouyel, P.

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(2009) Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer's disease, Nat Genet 41, 1094-1099. 34. Lambert, J. C., and Ibrahim-Verbaas, C. A., and Harold, D., and Naj, A. C., and Sims, R., and Bellenguez, C., and DeStafano, A. L., and Bis, J. C., and Beecham, G. W., and Grenier-Boley, B., and Russo, G., and Thorton-Wells, T. A., and Jones, N., and Smith, A. V., and Chouraki, V., and Thomas, C., and Ikram, M. A., and Zelenika, D., and Vardarajan, B. N., and Kamatani, Y., and Lin, C. F., and Gerrish, A., and Schmidt, H., and Kunkle, B., and Dunstan, M. L., and Ruiz, A., and Bihoreau, M. T., and Choi, S. H., and Reitz, C., and Pasquier, F., and Cruchaga, C., and Craig, D., and Amin, N., and Berr, C., and Lopez, O. L., and De Jager, P. L., and Deramecourt, V., and Johnston, J. A., and Evans, D., and Lovestone, S., and Letenneur, L., and Moron, F. J., and Rubinsztein, D. C., and Eiriksdottir, G., and Sleegers, K., and Goate, A. M., and Fievet, N., and Huentelman, M. W., and Gill, M., and Brown, K., and Kamboh, M. I., and Keller, L., and Barberger-Gateau, P., and McGuiness, B., and Larson, E. B., and Green, R., and Myers, A. J., and Dufouil, C., and Todd, S., and Wallon, D., and Love, S., and Rogaeva, E., and Gallacher, J., and St George-Hyslop, P., and Clarimon, J., and Lleo, A., and Bayer, A., and Tsuang, D. W., and Yu, L., and Tsolaki, M., and Bossu, P., and Spalletta, G., and Proitsi, P., and Collinge, J., and Sorbi, S., and Sanchez-Garcia, F., and Fox, N. C., and Hardy, J., and Deniz Naranjo, M. C., and Bosco, P., and Clarke, R., and Brayne, C., and Galimberti, D., and Mancuso, M., and Matthews, F., and Moebus, S., and Mecocci, P., and Del Zompo, M., and Maier, W., and Hampel, H., and Pilotto, A., and Bullido, M., and

ACS Paragon Plus Environment

Page 28 of 53

Page 29 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Panza, F., and Caffarra, P., and Nacmias, B., and Gilbert, J. R., and Mayhaus, M., and Lannefelt, L., and Hakonarson, H., and Pichler, S., and Carrasquillo, M. M., and Ingelsson, M., and Beekly, D., and Alvarez, V., and Zou, F., and Valladares, O., and Younkin, S. G., and Coto, E., and Hamilton-Nelson, K. L., and Gu, W., and Razquin, C., and Pastor, P., and Mateo, I., and Owen, M. J., and Faber, K. M., and Jonsson, P. V., and Combarros, O., and O'Donovan, M. C., and Cantwell, L. B., and Soininen, H., and Blacker, D., and Mead, S., and Mosley, T. H., Jr., and Bennett, D. A., and Harris, T. B., and Fratiglioni, L., and Holmes, C., and de Bruijn, R. F., and Passmore, P., and Montine, T. J., and Bettens, K., and Rotter, J. I., and Brice, A., and Morgan, K., and Foroud, T. M., and Kukull, W. A., and Hannequin, D., and Powell, J. F., and Nalls, M. A., and Ritchie, K., and Lunetta, K. L., and Kauwe, J. S., and Boerwinkle, E., and Riemenschneider, M., and Boada, M., and Hiltuenen, M., and Martin, E. R., and Schmidt, R., and Rujescu, D., and Wang, L. S., and Dartigues, J. F., and Mayeux, R., and Tzourio, C., and Hofman, A., and Nothen, M. M., and Graff, C., and Psaty, B. M., and Jones, L., and Haines, J. L., and Holmans, P. A., and Lathrop, M., and Pericak-Vance, M. A., and Launer, L. J., and Farrer, L. A., and van Duijn, C. M., and Van Broeckhoven, C., and Moskvina, V., and Seshadri, S., and Williams, J., and Schellenberg, G. D., and Amouyel, P. (2013) Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease, Nat Genet 45, 1452-1458. 35. Efthymiou, A. G., and Goate, A. M. (2017) Late onset Alzheimer's disease genetics implicates microglial pathways in disease risk, Mol Neurodegener 12, 43.

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

36. Karch, C. M., and Goate, A. M. (2015) Alzheimer's disease risk genes and mechanisms of disease pathogenesis, Biol. Psychiatry 77, 43-51. 37. Menasche, G., Pastural, E., Feldmann, J., Certain, S., Ersoy, F., Dupuis, S., Wulffraat, N., Bianchi, D., Fischer, A., Le Deist, F., and de Saint Basile, G. (2000) Mutations in RAB27A cause Griscelli syndrome associated with haemophagocytic syndrome, Nat Genet 25, 173-176. 38. Stinchcombe, J. C., Barral, D. C., Mules, E. H., Booth, S., Hume, A. N., Machesky, L. M., Seabra, M. C., and Griffiths, G. M. (2001) Rab27a is required for regulated secretion in cytotoxic T lymphocytes, J. Cell Biol. 152, 825-834. 39. Jenkins, D., Seelow, D., Jehee, F. S., Perlyn, C. A., Alonso, L. G., Bueno, D. F., Donnai, D., Josifova, D., Mathijssen, I. M., Morton, J. E., Orstavik, K. H., Sweeney, E., Wall, S. A., Marsh, J. L., Nurnberg, P., Passos-Bueno, M. R., and Wilkie, A. O. (2007) RAB23 mutations in Carpenter syndrome imply an unexpected role for hedgehog signaling in cranial-suture development and obesity, Am. J. Hum. Genet. 80, 1162-1170. 40. Zuchner, S., and Vance, J. M. (2006) Molecular genetics of autosomal-dominant axonal Charcot-Marie-Tooth disease, Neuromolecular Med 8, 63-74. 41. Giannandrea, M., Bianchi, V., Mignogna, M. L., Sirri, A., Carrabino, S., D'Elia, E., Vecellio, M., Russo, S., Cogliati, F., Larizza, L., Ropers, H. H., Tzschach, A., Kalscheuer, V., Oehl-Jaschkowitz, B., Skinner, C., Schwartz, C. E., Gecz, J., Van Esch, H., Raynaud, M., Chelly, J., de Brouwer, A. P., Toniolo, D., and D'Adamo, P. (2010) Mutations in the small GTPase gene RAB39B are responsible for X-linked mental retardation associated

ACS Paragon Plus Environment

Page 30 of 53

Page 31 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

with autism, epilepsy, and macrocephaly, Am. J. Hum. Genet. 86, 185-195. 42. Wilson, G. R., Sim, J. C., McLean, C., Giannandrea, M., Galea, C. A., Riseley, J. R., Stephenson, S. E., Fitzpatrick, E., Haas, S. A., Pope, K., Hogan, K. J., Gregg, R. G., Bromhead, C. J., Wargowski, D. S., Lawrence, C. H., James, P. A., Churchyard, A., Gao, Y., Phelan, D. G., Gillies, G., Salce, N., Stanford, L., Marsh, A. P., Mignogna, M. L., Hayflick, S. J., Leventer, R. J., Delatycki, M. B., Mellick, G. D., Kalscheuer, V. M., D'Adamo, P., Bahlo, M., Amor, D. J., and Lockhart, P. J. (2014) Mutations in RAB39B cause X-linked intellectual disability and early-onset Parkinson disease with alpha-synuclein pathology, Am. J. Hum. Genet. 95, 729-735. 43. Mata, I. F., Jang, Y., Kim, C. H., Hanna, D. S., Dorschner, M. O., Samii, A., Agarwal, P., Roberts, J. W., Klepitskaya, O., Shprecher, D. R., Chung, K. A., Factor, S. A., Espay, A. J., Revilla, F. J., Higgins, D. S., Litvan, I., Leverenz, J. B., Yearout, D., Inca-Martinez, M., Martinez, E., Thompson, T. R., Cholerton, B. A., Hu, S. C., Edwards, K. L., Kim, K. S., and Zabetian, C. P. (2015) The RAB39B p.G192R mutation causes X-linked dominant Parkinson's disease, Mol Neurodegener 10, 50. 44. Shi, C. H., Zhang, S. Y., Yang, Z. H., Yang, J., Shang, D. D., Mao, C. Y., Liu, H., Hou, H. M., Shi, M. M., Wu, J., and Xu, Y. M. (2016) A novel RAB39B gene mutation in X-linked juvenile parkinsonism with basal ganglia calcification, Mov. Disord. 31, 1905-1909. 45. Woodbury-Smith, M., Deneault, E., Yuen, R. K. C., Walker, S., Zarrei, M., Pellecchia, G., Howe, J. L., Hoang, N., Uddin, M., Marshall, C. R., Chrysler, C., Thompson, A.,

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Szatmari, P., and Scherer, S. W. (2017) Mutations in RAB39B in individuals with intellectual disability, autism spectrum disorder, and macrocephaly, Mol Autism 8, 59. 46. Ciammola, A., Carrera, P., Di Fonzo, A., Sassone, J., Villa, R., Poletti, B., Ferrari, M., Girotti, F., Monfrini, E., Buongarzone, G., Silani, V., Cinnante, C. M., Mignogna, M. L., D'Adamo, P., and Bonati, M. T. (2017) X-linked Parkinsonism with Intellectual Disability caused by novel mutations and somatic mosaicism in RAB39B gene, Parkinsonism Relat Disord 44, 142-146. 47. Vardarajan, B. N., Bruesegem, S. Y., Harbour, M. E., Inzelberg, R., Friedland, R., St George-Hyslop, P., Seaman, M. N., and Farrer, L. A. (2012) Identification of Alzheimer disease-associated variants in genes that regulate retromer function, Neurobiol. Aging 33, 2231 e2215-2231 e2230. 48. Ridge, P. G., Karch, C. M., Hsu, S., Arano, I., Teerlink, C. C., Ebbert, M. T. W., Gonzalez Murcia, J. D., Farnham, J. M., Damato, A. R., Allen, M., Wang, X., Harari, O., Fernandez, V. M., Guerreiro, R., Bras, J., Hardy, J., Munger, R., Norton, M., Sassi, C., Singleton, A., Younkin, S. G., Dickson, D. W., Golde, T. E., Price, N. D., Ertekin-Taner, N., Cruchaga, C., Goate, A. M., Corcoran, C., Tschanz, J., Cannon-Albright, L. A., and Kauwe, J. S. K. (2017) Linkage, whole genome sequence, and biological data implicate variants in RAB10 in Alzheimer's disease resilience, Genome Med 9, 100. 49. Udayar, V., Buggia-Prevot, V., Guerreiro, R. L., Siegel, G., Rambabu, N., Soohoo, A. L., Ponnusamy, M., Siegenthaler, B., Bali, J., Simons, M., Ries, J., Puthenveedu, M. A., Hardy, J., Thinakaran, G., and Rajendran, L. (2013) A paired RNAi and RabGAP

ACS Paragon Plus Environment

Page 32 of 53

Page 33 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

overexpression screen identifies Rab11 as a regulator of beta-amyloid production, Cell Rep 5, 1536-1551. 50. Kajiho, H., Sakurai, K., Minoda, T., Yoshikawa, M., Nakagawa, S., Fukushima, S., Kontani, K., and Katada, T. (2011) Characterization of RIN3 as a guanine nucleotide exchange factor for the Rab5 subfamily GTPase Rab31, J. Biol. Chem. 286, 24364-24373. 51. Yoshikawa, M., Kajiho, H., Sakurai, K., Minoda, T., Nakagawa, S., Kontani, K., and Katada, T. (2008) Tyr-phosphorylation signals translocate RIN3, the small GTPase Rab5-GEF, to early endocytic vesicles, Biochem. Biophys. Res. Commun. 372, 168-172. 52. Kajiho, H., Saito, K., Tsujita, K., Kontani, K., Araki, Y., Kurosu, H., and Katada, T. (2003) RIN3: a novel Rab5 GEF interacting with amphiphysin II involved in the early endocytic pathway, J. Cell Sci. 116, 4159-4168. 53. Kunkle, B. W., Vardarajan, B. N., Naj, A. C., Whitehead, P. L., Rolati, S., Slifer, S., Carney, R. M., Cuccaro, M. L., Vance, J. M., Gilbert, J. R., Wang, L. S., Farrer, L. A., Reitz, C., Haines, J. L., Beecham, G. W., Martin, E. R., Schellenberg, G. D., Mayeux, R. P., and Pericak-Vance, M. A. (2017) Early-Onset Alzheimer Disease and Candidate Risk Genes Involved in Endolysosomal Transport, JAMA Neurol 74, 1113-1122. 54. Song, M. S., Baker, G. B., Todd, K. G., and Kar, S. (2011) Inhibition of beta-amyloid1-42

internalization

attenuates

neuronal

death

by stabilizing

the

endosomal-lysosomal system in rat cortical cultured neurons, Neuroscience 178, 181-188.

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

55. Gorvel, J. P., Chavrier, P., Zerial, M., and Gruenberg, J. (1991) rab5 controls early endosome fusion in vitro, Cell 64, 915-925. 56. Bucci, C., Parton, R. G., Mather, I. H., Stunnenberg, H., Simons, K., Hoflack, B., and Zerial, M. (1992) The small GTPase rab5 functions as a regulatory factor in the early endocytic pathway, Cell 70, 715-728. 57. Hoogenraad, C. C., Popa, I., Futai, K., Martinez-Sanchez, E., Wulf, P. S., van Vlijmen, T., Dortland, B. R., Oorschot, V., Govers, R., Monti, M., Heck, A. J., Sheng, M., Klumperman, J., Rehmann, H., Jaarsma, D., Kapitein, L. C., and van der Sluijs, P. (2010) Neuron specific Rab4 effector GRASP-1 coordinates membrane specialization and maturation of recycling endosomes, PLoS Biol 8, e1000283. 58. Modica, G., Skorobogata, O., Sauvageau, E., Vissa, A., Yip, C. M., Kim, P. K., Wurtele, H., and Lefrancois, S. (2017) Rab7 palmitoylation is required for efficient endosome-to-TGN trafficking, J. Cell Sci. 130, 2579-2590. 59. Yla-Anttila, P., and Eskelinen, E. L. (2018) Roles for RAB24 in autophagy and disease, Small GTPases 9, 57-65. 60. Catz, S. D. (2013) Regulation of vesicular trafficking and leukocyte function by Rab27 GTPases and their effectors, J. Leukoc. Biol. 94, 613-622. 61. Ginsberg, S. D., Mufson, E. J., Alldred, M. J., Counts, S. E., Wuu, J., Nixon, R. A., and Che, S. (2011) Upregulation of select rab GTPases in cholinergic basal forebrain neurons in mild cognitive impairment and Alzheimer's disease, J. Chem. Neuroanat. 42, 102-110.

ACS Paragon Plus Environment

Page 34 of 53

Page 35 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

62. Ginsberg, S. D., Mufson, E. J., Counts, S. E., Wuu, J., Alldred, M. J., Nixon, R. A., and Che, S. (2010) Regional selectivity of rab5 and rab7 protein upregulation in mild cognitive impairment and Alzheimer's disease, J Alzheimers Dis 22, 631-639. 63. Zafar, S., Younas, N., Correia, S., Shafiq, M., Tahir, W., Schmitz, M., Ferrer, I., Andreoletti, O., and Zerr, I. (2017) Strain-Specific Altered Regulatory Response of Rab7a and Tau in Creutzfeldt-Jakob Disease and Alzheimer's Disease, Mol. Neurobiol. 54, 697-709. 64. Mitra, S., Cheng, K. W., and Mills, G. B. (2010) Rab GTPases implicated in inherited and acquired disorders, Semin. Cell Dev. Biol. 22, 57-68. 65. Zhao, Y., Tan, W., Sheng, W., and Li, X. (2016) Identification of Biomarkers Associated With Alzheimer's Disease by Bioinformatics Analysis, Am J Alzheimers Dis Other Demen 31, 163-168. 66. Yan, T., Wang, L., Gao, J., Siedlak, S. L., Huntley, M. L., Termsarasab, P., Perry, G., Chen, S. G., and Wang, X. (2018) Rab10 Phosphorylation is a Prominent Pathological Feature in Alzheimer's Disease, J Alzheimers Dis 63, 157-165. 67. Reddy, P. H., Mani, G., Park, B. S., Jacques, J., Murdoch, G., Whetsell, W., Jr., Kaye, J., and Manczak, M. (2005) Differential loss of synaptic proteins in Alzheimer's disease: implications for synaptic dysfunction, J Alzheimers Dis 7, 103-117; discussion 173-180. 68. Hattula, K., Furuhjelm, J., Tikkanen, J., Tanhuanpaa, K., Laakkonen, P., and Peranen, J. (2006) Characterization of the Rab8-specific membrane traffic route linked to protrusion formation, J. Cell Sci. 119, 4866-4877.

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

69. Nachury, M. V., Loktev, A. V., Zhang, Q., Westlake, C. J., Peranen, J., Merdes, A., Slusarski, D. C., Scheller, R. H., Bazan, J. F., Sheffield, V. C., and Jackson, P. K. (2007) A core complex of BBS proteins cooperates with the GTPase Rab8 to promote ciliary membrane biogenesis, Cell 129, 1201-1213. 70. Huber, L. A., Pimplikar, S., Parton, R. G., Virta, H., Zerial, M., and Simons, K. (1993) Rab8, a small GTPase involved in vesicular traffic between the TGN and the basolateral plasma membrane, J Cell Biol 123, 35-45. 71. Shimohama, S., Kamiya, S., Taniguchi, T., Sumida, Y., and Fujimoto, S. (1999) Differential involvement of small G proteins in Alzheimer's disease, Int. J. Mol. Med. 3, 597-600. 72. Scheper, W., Hoozemans, J. J., Hoogenraad, C. C., Rozemuller, A. J., Eikelenboom, P., and Baas, F. (2007) Rab6 is increased in Alzheimer's disease brain and correlates with endoplasmic reticulum stress, Neuropathol. Appl. Neurobiol. 33, 523-532. 73. Li, G. (2011) Rab GTPases, membrane trafficking and diseases, Curr Drug Targets 12, 1188-1193. 74. Koo, E. H., and Squazzo, S. L. (1994) Evidence that production and release of amyloid beta-protein involves the endocytic pathway, J. Biol. Chem. 269, 17386-17389. 75. Thinakaran, G., and Koo, E. H. (2008) Amyloid precursor protein trafficking, processing, and function, J. Biol. Chem. 283, 29615-29619. 76. Toh, W. H., and Gleeson, P. A. (2016) Dysregulation of intracellular trafficking and endosomal sorting in Alzheimer's disease: controversies and unanswered questions,

ACS Paragon Plus Environment

Page 36 of 53

Page 37 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Biochem J 473, 1977-1993. 77. Sisodia, S. S. (1992) Beta-amyloid precursor protein cleavage by a membrane-bound protease, Proc Natl Acad Sci U S A 89, 6075-6079. 78. Nordstedt, C., Caporaso, G. L., Thyberg, J., Gandy, S. E., and Greengard, P. (1993) Identification of the Alzheimer beta/A4 amyloid precursor protein in clathrin-coated vesicles purified from PC12 cells, J. Biol. Chem. 268, 608-612. 79. Caporaso, G. L., Takei, K., Gandy, S. E., Matteoli, M., Mundigl, O., Greengard, P., and De Camilli, P. (1994) Morphologic and biochemical analysis of the intracellular trafficking of the Alzheimer beta/A4 amyloid precursor protein, J. Neurosci. 14, 3122-3138. 80. Plutner, H., Cox, A. D., Pind, S., Khosravi-Far, R., Bourne, J. R., Schwaninger, R., Der, C. J., and Balch, W. E. (1991) Rab1b regulates vesicular transport between the endoplasmic reticulum and successive Golgi compartments, J. Cell Biol. 115, 31-43. 81. Dugan, J. M., deWit, C., McConlogue, L., and Maltese, W. A. (1995) The Ras-related GTP-binding protein, Rab1B, regulates early steps in exocytic transport and processing of beta-amyloid precursor protein, J Biol Chem 270, 10982-10989. 82. Szodorai, A., Kuan, Y. H., Hunzelmann, S., Engel, U., Sakane, A., Sasaki, T., Takai, Y., Kirsch, J., Muller, U., Beyreuther, K., Brady, S., Morfini, G., and Kins, S. (2009) APP anterograde transport requires Rab3A GTPase activity for assembly of the transport vesicle, J. Neurosci. 29, 14534-14544. 83. Martinez, O., Schmidt, A., Salamero, J., Hoflack, B., Roa, M., and Goud, B. (1994)

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

The small GTP-binding protein rab6 functions in intra-Golgi transport, J. Cell Biol. 127, 1575-1588. 84. Short, B., Preisinger, C., Schaletzky, J., Kopajtich, R., and Barr, F. A. (2002) The Rab6 GTPase regulates recruitment of the dynactin complex to Golgi membranes, Curr. Biol. 12, 1792-1795. 85. Young, J., Stauber, T., del Nery, E., Vernos, I., Pepperkok, R., and Nilsson, T. (2005) Regulation of microtubule-dependent recycling at the trans-Golgi network by Rab6A and Rab6A', Mol. Biol. Cell 16, 162-177. 86. Mallard, F., Tang, B. L., Galli, T., Tenza, D., Saint-Pol, A., Yue, X., Antony, C., Hong, W., Goud, B., and Johannes, L. (2002) Early/recycling endosomes-to-TGN transport involves two SNARE complexes and a Rab6 isoform, J. Cell Biol. 156, 653-664. 87. Grigoriev, I., Splinter, D., Keijzer, N., Wulf, P. S., Demmers, J., Ohtsuka, T., Modesti, M., Maly, I. V., Grosveld, F., Hoogenraad, C. C., and Akhmanova, A. (2007) Rab6 regulates transport and targeting of exocytotic carriers, Dev Cell 13, 305-314. 88. McConlogue, L., Castellano, F., deWit, C., Schenk, D., and Maltese, W. A. (1996) Differential effects of a Rab6 mutant on secretory versus amyloidogenic processing of Alzheimer's beta-amyloid precursor protein, J Biol Chem 271, 1343-1348. 89. Teber, I., Nagano, F., Kremerskothen, J., Bilbilis, K., Goud, B., and Barnekow, A. (2005) Rab6 interacts with the mint3 adaptor protein, Biol. Chem. 386, 671-677. 90. Thyrock, A., Ossendorf, E., Stehling, M., Kail, M., Kurtz, T., Pohlentz, G., Waschbusch, D., Eggert, S., Formstecher, E., Muthing, J., Dreisewerd, K., Kins, S., Goud,

ACS Paragon Plus Environment

Page 38 of 53

Page 39 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

B., and Barnekow, A. (2013) A new Mint1 isoform, but not the conventional Mint1, interacts with the small GTPase Rab6, PLoS One 8, e64149. 91. Biederer, T., Cao, X., Sudhof, T. C., and Liu, X. (2002) Regulation of APP-dependent transcription complexes by Mint/X11s: differential functions of Mint isoforms, J Neurosci 22, 7340-7351. 92. King, G. D., Perez, R. G., Steinhilb, M. L., Gaut, J. R., and Turner, R. S. (2003) X11alpha modulates secretory and endocytic trafficking and metabolism of amyloid precursor protein: mutational analysis of the YENPTY sequence, Neuroscience 120, 143-154. 93. Elfrink, H. L., Zwart, R., Cavanillas, M. L., Schindler, A. J., Baas, F., and Scheper, W. (2012) Rab6 is a modulator of the unfolded protein response: implications for Alzheimer's disease, J Alzheimers Dis 28, 917-929. 94. Campa, C. C., and Hirsch, E. (2017) Rab11 and phosphoinositides: A synergy of signal transducers in the control of vesicular trafficking, Adv Biol Regul 63, 132-139. 95. Welz, T., Wellbourne-Wood, J., and Kerkhoff, E. (2014) Orchestration of cell surface proteins by Rab11, Trends Cell Biol. 24, 407-415. 96. Niederst, E. D., Reyna, S. M., and Goldstein, L. S. (2015) Axonal amyloid precursor protein and its fragments undergo somatodendritic endocytosis and processing, Mol. Biol. Cell 26, 205-217. 97. Woodruff, G., Reyna, S. M., Dunlap, M., Van Der Kant, R., Callender, J. A., Young, J. E., Roberts, E. A., and Goldstein, L. S. (2016) Defective Transcytosis of APP and

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Lipoproteins in Human iPSC-Derived Neurons with Familial Alzheimer's Disease Mutations, Cell Rep 17, 759-773. 98. Wiese, M., Antebi, A., and Zheng, H. (2010) Intracellular trafficking and synaptic function of APL-1 in Caenorhabditis elegans, PLoS One 5. 99. Buggia-Prevot, V., Fernandez, C. G., Riordan, S., Vetrivel, K. S., Roseman, J., Waters, J., Bindokas, V. P., Vassar, R., and Thinakaran, G. (2014) Axonal BACE1 dynamics and targeting in hippocampal neurons: a role for Rab11 GTPase, Mol Neurodegener 9, 1. 100. Yin, C., Argintaru, D., and Heit, B. (2017) Rab17 mediates intermixing of phagocytosed apoptotic cells with recycling endosomes, Small GTPases, 1-9. 101. Chen, L., Hu, J., Yun, Y., and Wang, T. (2010) Rab36 regulates the spatial distribution of late endosomes and lysosomes through a similar mechanism to Rab34, Mol. Membr. Biol. 27, 23-30. 102. Dumanchin, C., Czech, C., Campion, D., Cuif, M. H., Poyot, T., Martin, C., Charbonnier, F., Goud, B., Pradier, L., and Frebourg, T. (1999) Presenilins interact with Rab11, a small GTPase involved in the regulation of vesicular transport, Hum. Mol. Genet. 8, 1263-1269. 103. Lopez-Perez, E., Dumanchin, C., Czech, C., Campion, D., Goud, B., Pradier, L., Frebourg, T., and Checler, F. (2000) Overexpression of Rab11 or constitutively active Rab11 does not affect sAPPalpha and Abeta secretions by wild-type and Swedish mutated betaAPP-expressing HEK293 cells, Biochem. Biophys. Res. Commun. 275, 910-915. 104. Scheper, W., Zwart, R., and Baas, F. (2004) Rab6 membrane association is

ACS Paragon Plus Environment

Page 40 of 53

Page 41 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

dependent of Presenilin 1 and cellular phosphorylation events, Brain Res. Mol. Brain Res. 122, 17-23. 105. Scheper, W., Zwart, R., Sluijs, P., Annaert, W., Gool, W. A., and Baas, F. (2000) Alzheimer's presenilin 1 is a putative membrane receptor for rab GDP dissociation inhibitor, Hum. Mol. Genet. 9, 303-310. 106. Soejima, N., Ohyagi, Y., Nakamura, N., Himeno, E., Iinuma, K. M., Sakae, N., Yamasaki, R., Tabira, T., Murakami, K., Irie, K., Kinoshita, N., LaFerla, F. M., Kiyohara, Y., Iwaki, T., and Kira, J. (2013) Intracellular accumulation of toxic turn amyloid-beta is associated with endoplasmic reticulum stress in Alzheimer's disease, Curr Alzheimer Res 10, 11-20. 107. Kametani, F., Usami, M., Tanaka, K., Kume, H., and Mori, H. (2004) Mutant presenilin (A260V) affects Rab8 in PC12D cell, Neurochem. Int. 44, 313-320. 108. Kuhn, P. H., Wang, H., Dislich, B., Colombo, A., Zeitschel, U., Ellwart, J. W., Kremmer, E., Rossner, S., and Lichtenthaler, S. F. (2010) ADAM10 is the physiologically relevant, constitutive alpha-secretase of the amyloid precursor protein in primary neurons, EMBO J. 29, 3020-3032. 109. Buxbaum, J. D., Liu, K. N., Luo, Y., Slack, J. L., Stocking, K. L., Peschon, J. J., Johnson, R. S., Castner, B. J., Cerretti, D. P., and Black, R. A. (1998) Evidence that tumor necrosis factor alpha converting enzyme is involved in regulated alpha-secretase cleavage of the Alzheimer amyloid protein precursor, J. Biol. Chem. 273, 27765-27767. 110. Koike, H., Tomioka, S., Sorimachi, H., Saido, T. C., Maruyama, K., Okuyama, A.,

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Fujisawa-Sehara, A., Ohno, S., Suzuki, K., and Ishiura, S. (1999) Membrane-anchored metalloprotease MDC9 has an alpha-secretase activity responsible for processing the amyloid precursor protein, Biochem J 343 Pt 2, 371-375. 111. Junutula, J. R., De Maziere, A. M., Peden, A. A., Ervin, K. E., Advani, R. J., van Dijk, S. M., Klumperman, J., and Scheller, R. H. (2004) Rab14 is involved in membrane trafficking between the Golgi complex and endosomes, Mol. Biol. Cell 15, 2218-2229. 112. Lindsay, A. J., and McCaffrey, M. W. (2015) Rab antibody characterization: comparison of Rab14 antibodies, Methods Mol. Biol. 1298, 161-171. 113. Linford, A., Yoshimura, S., Nunes Bastos, R., Langemeyer, L., Gerondopoulos, A., Rigden, D. J., and Barr, F. A. (2012) Rab14 and its exchange factor FAM116 link endocytic recycling and adherens junction stability in migrating cells, Dev Cell 22, 952-966. 114. Prekeris, R. (2012) The art of "cut and run": the role of Rab14 GTPase in regulating N-cadherin shedding and cell motility, Dev Cell 22, 909-910. 115. Jiang, C., Liu, Z., Hu, R., Bo, L., Minshall, R. D., Malik, A. B., and Hu, G. (2017) Inactivation of Rab11a GTPase in Macrophages Facilitates Phagocytosis of Apoptotic Neutrophils, J. Immunol. 198, 1660-1672. 116. Li, J., Kanekiyo, T., Shinohara, M., Zhang, Y., LaDu, M. J., Xu, H., and Bu, G. (2012) Differential regulation of amyloid-beta endocytic trafficking and lysosomal degradation by apolipoprotein E isoforms, J. Biol. Chem. 287, 44593-44601. 117. Willen, K., Edgar, J. R., Hasegawa, T., Tanaka, N., Futter, C. E., and Gouras, G. K.

ACS Paragon Plus Environment

Page 42 of 53

Page 43 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

(2017) Abeta accumulation causes MVB enlargement and is modelled by dominant negative VPS4A, Mol Neurodegener 12, 61. 118. Lee, C. Y., Tse, W., Smith, J. D., and Landreth, G. E. (2012) Apolipoprotein E promotes beta-amyloid trafficking and degradation by modulating microglial cholesterol levels, J. Biol. Chem. 287, 2032-2044. 119. Yuyama, K., and Yanagisawa, K. (2009) Late endocytic dysfunction as a putative cause of amyloid fibril formation in Alzheimer's disease, J. Neurochem. 109, 1250-1260. 120. Wang, Y., Balaji, V., Kaniyappan, S., Kruger, L., Irsen, S., Tepper, K., Chandupatla, R., Maetzler, W., Schneider, A., Mandelkow, E., and Mandelkow, E. M. (2017) The release and trans-synaptic transmission of Tau via exosomes, Mol Neurodegener 12, 5. 121. Jucker, M., and Walker, L. C. (2013) Self-propagation of pathogenic protein aggregates in neurodegenerative diseases, Nature 501, 45-51. 122. Goedert, M., Eisenberg, D. S., and Crowther, R. A. (2017) Propagation of Tau Aggregates and Neurodegeneration, Annu. Rev. Neurosci. 40, 189-210. 123. Leyns, C. E. G., and Holtzman, D. M. (2017) Glial contributions to neurodegeneration in tauopathies, Mol Neurodegener 12, 50. 124. Rodriguez, L., Mohamed, N. V., Desjardins, A., Lippe, R., Fon, E. A., and Leclerc, N. (2017) Rab7A regulates tau secretion, J. Neurochem. 141, 592-605. 125. Mohamed, N. V., Desjardins, A., and Leclerc, N. (2017) Tau secretion is correlated to an increase of Golgi dynamics, PLoS One 12, e0178288. 126. Kulic, L., and Unschuld, P. G. (2016) Recent advances in cerebrospinal fluid

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

biomarkers for the detection of preclinical Alzheimer's disease, Curr. Opin. Neurol. 29, 749-755. 127. Cicognola, C., Chiasserini, D., Eusebi, P., Andreasson, U., Vanderstichele, H., Zetterberg, H., Parnetti, L., and Blennow, K. (2016) No diurnal variation of classical and candidate biomarkers of Alzheimer's disease in CSF, Mol Neurodegener 11, 65. 128. Nakamura, A., Kaneko, N., Villemagne, V. L., Kato, T., Doecke, J., Dore, V., Fowler, C., Li, Q. X., Martins, R., Rowe, C., Tomita, T., Matsuzaki, K., Ishii, K., Ishii, K., Arahata, Y., Iwamoto, S., Ito, K., Tanaka, K., Masters, C. L., and Yanagisawa, K. (2018) High performance plasma amyloid-beta biomarkers for Alzheimer's disease, Nature 554, 249-254. 129. Tatebe, H., Kasai, T., Ohmichi, T., Kishi, Y., Kakeya, T., Waragai, M., Kondo, M., Allsop, D., and Tokuda, T. (2017) Quantification of plasma phosphorylated tau to use as a biomarker for brain Alzheimer pathology: pilot case-control studies including patients with Alzheimer's disease and down syndrome, Mol Neurodegener 12, 63. 130. Guerreiro, R., Wojtas, A., Bras, J., Carrasquillo, M., Rogaeva, E., Majounie, E., Cruchaga, C., Sassi, C., Kauwe, J. S., Younkin, S., Hazrati, L., Collinge, J., Pocock, J., Lashley, T., Williams, J., Lambert, J. C., Amouyel, P., Goate, A., Rademakers, R., Morgan, K., Powell, J., St George-Hyslop, P., Singleton, A., and Hardy, J. (2013) TREM2 variants in Alzheimer's disease, N. Engl. J. Med. 368, 117-127. 131. Jonsson, T., Stefansson, H., Steinberg, S., Jonsdottir, I., Jonsson, P. V., Snaedal, J., Bjornsson, S., Huttenlocher, J., Levey, A. I., Lah, J. J., Rujescu, D., Hampel, H., Giegling,

ACS Paragon Plus Environment

Page 44 of 53

Page 45 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

I., Andreassen, O. A., Engedal, K., Ulstein, I., Djurovic, S., Ibrahim-Verbaas, C., Hofman, A., Ikram, M. A., van Duijn, C. M., Thorsteinsdottir, U., Kong, A., and Stefansson, K. (2013) Variant of TREM2 associated with the risk of Alzheimer's disease, N. Engl. J. Med. 368, 107-116. 132. Jay, T. R., von Saucken, V. E., and Landreth, G. E. (2017) TREM2 in Neurodegenerative Diseases, Mol Neurodegener 12, 56. 133. Heslegrave, A., Heywood, W., Paterson, R., Magdalinou, N., Svensson, J., Johansson, P., Ohrfelt, A., Blennow, K., Hardy, J., Schott, J., Mills, K., and Zetterberg, H. (2016) Increased cerebrospinal fluid soluble TREM2 concentration in Alzheimer's disease, Mol Neurodegener 11, 3. 134. Suarez-Calvet, M., Kleinberger, G., Araque Caballero, M. A., Brendel, M., Rominger, A., Alcolea, D., Fortea, J., Lleo, A., Blesa, R., Gispert, J. D., Sanchez-Valle, R., Antonell, A., Rami, L., Molinuevo, J. L., Brosseron, F., Traschutz, A., Heneka, M. T., Struyfs, H., Engelborghs, S., Sleegers, K., Van Broeckhoven, C., Zetterberg, H., Nellgard, B., Blennow, K., Crispin, A., Ewers, M., and Haass, C. (2016) sTREM2 cerebrospinal fluid levels are a potential biomarker for microglia activity in early-stage Alzheimer's disease and associate with neuronal injury markers, EMBO Mol Med 8, 466-476. 135. Armstrong, A., Mattsson, N., Appelqvist, H., Janefjord, C., Sandin, L., Agholme, L., Olsson, B., Svensson, S., Blennow, K., Zetterberg, H., and Kagedal, K. (2014) Lysosomal network proteins as potential novel CSF biomarkers for Alzheimer's disease, Neuromolecular Med 16, 150-160.

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

136. Butler, D., Hwang, J., Estick, C., Nishiyama, A., Kumar, S. S., Baveghems, C., Young-Oxendine, H. B., Wisniewski, M. L., Charalambides, A., and Bahr, B. A. (2011) Protective effects of positive lysosomal modulation in Alzheimer's disease transgenic mouse models, PLoS One 6, e20501. 137. Shinohara, M., Sato, N., Kurinami, H., Takeuchi, D., Takeda, S., Shimamura, M., Yamashita, T., Uchiyama, Y., Rakugi, H., and Morishita, R. (2010) Reduction of brain beta-amyloid (Abeta) by fluvastatin, a hydroxymethylglutaryl-CoA reductase inhibitor, through increase in degradation of amyloid precursor protein C-terminal fragments (APP-CTFs) and Abeta clearance, J. Biol. Chem. 285, 22091-22102. 138. Zhao, Z., Sagare, A. P., Ma, Q., Halliday, M. R., Kong, P., Kisler, K., Winkler, E. A., Ramanathan, A., Kanekiyo, T., Bu, G., Owens, N. C., Rege, S. V., Si, G., Ahuja, A., Zhu, D., Miller, C. A., Schneider, J. A., Maeda, M., Maeda, T., Sugawara, T., Ichida, J. K., and Zlokovic, B. V. (2015) Central role for PICALM in amyloid-beta blood-brain barrier transcytosis and clearance, Nat. Neurosci. 18, 978-987. 139. Xu, W., Tan, L., and Yu, J. T. (2015) The Role of PICALM in Alzheimer's Disease, Mol. Neurobiol. 52, 399-413.

ACS Paragon Plus Environment

Page 46 of 53

Page 47 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Table 1. Summary of Rab GTPases associated with AD Rab GTPase Reported/proposed

Categoriesa

Association with AD

functions RAB1A

Mediates Golgi dynamics Affects tau secretion125

RAB1B

Regulates

ER-Golgi Affects APP trafficking and B

vesicular transport RAB3A

B

Aβ generation81

Localizes in presynaptic Decreased in AD brain67; vesicles

and

exocytosis

B, C

regulates Increased in AD CSF135; Affects APP assembly into anterograde

transporting

vesicles82; Affects APP β-cleavage and Aβ generation49 RAB4

Regulates recycling

endosomal Increased in AD brain and C neurons28, 57, 61, in AD mice and

in

cells

expressing

fAD-associated

PS1

mutants106 RAB5A/RAB

Modulates

endosomal Increased in AD brain and B, C

5

membrane

trafficking, neurons28, 57, 61, 62;

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 48 of 53

sorting and endosomal Affects APL-1 expression98; fusion

Accelerates endocytosed Aβ trafficking

to

the

lysosome116; Related

to

PICALM-mediated internalization,



trafficking

and clearance138, 139; Levels/distribution affected by Aβ54 RAB6A/RA

Regulates

B6

Golgi-ER transport

retrograde Increased in AD brain72, in B, C trafficking, AD of

mice

early expressing

and

in

cells

fAD-associated

endosomes and recycling PS1 mutants106; endosomes to the TGN, Affects APP trafficking and and vesicle exocytosis

Aβ generation88; Modulates

AD-related

unfolded protein response72, 93

;

Membrane

ACS Paragon Plus Environment

association

Page 49 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

dependent on PS1104 RAB7A/RAB

Controls transport to late Genetic association47;

7

endosomes lysosomes

A, B, C

and Increased in AD28, 61-63; Increased in AD CSF135; Affects

Aβ-containing

vesicle endocytic trafficking and Aβ degradation116, 118 Affects



fibril

formation119; Levels/distribution affected by Aβ54; Regulates tau secretion124 RAB8A/RA

Modulates

polarized Increased

B8

trafficking

decreased

in

AD71 in

but B, C cells

expressing a PS1 mutant (A260V) 107 Accelerates endocytosed Aβ trafficking

to

the

lysosome116 RAB10

Modulates

Genetic association48;

ACS Paragon Plus Environment

A, B, C

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 50 of 53

exocytosis/secretory

Increased in AD48, 65;

pathway,

Phosphorylated RAB10 (at

neurotransmitter release threonine 73) is prominent and maturation

phagosome in AD neuron NFTs66; Affects

Aβ42

and

Aβ42/Aβ40 ratio48 RAB11A

Regulates both endocytic Genetic association49; and exocytic trafficking Regulates pathways

A, B, C

BACE1

membrane trafficking and recycling

and



generation49, 99; Interacts with PS1102; Affects

endocytosed



trafficking116; Affect cell surface levels of ADAM17115; Related

to

PICALM-mediated internalization,

trafficking

and clearance138, 139

ACS Paragon Plus Environment



Page 51 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

RAB11B

Regulates both endocytic Colocalizes with APP and B and exocytic trafficking affects pathways

soma‐to‐axon

transcytosis

of

APP

in

neurons96, 97; Regulates

BACE1

membrane trafficking and recycling

and



generation49, 99; Related

to

PICALM-mediated internalization,



trafficking

and clearance138, 139 RAB14

Involved

in

endocytic Affects

ADAM10 B, C

and recycling113, 114

recycling Golgi-endosome trafficking RAB17

Involved in phagocytic Affects APP β-cleavage and B removal

of

apoptotic Aβ generation49

cells RAB24

Involved in autophagy

Increased in AD28

ACS Paragon Plus Environment

C

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

RAB27A/RA Regulates B27

Page 52 of 53

exocytosis, Increased in AD61

endocytosis

C

and

phagocytosis RAB36

involved

in

late Affects APP β-cleavage and B

endosome and lysosome Aβ generation49 distribution RIN3

A

guanine

exchange

nucleotide Genetic association32-34, 53 factor

A

for

RAB5 Rab GDI

Promotes GTP hydrolysis Membrane

association B, C

to Inactivate GTP-bound affected by PS1105 GTPases a

Classification based on Kiral et al.29

ACS Paragon Plus Environment

Page 53 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Rab GTPases and AD 80x39mm (300 x 300 DPI)

ACS Paragon Plus Environment