Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jul 12.
Published in final edited form as: Semin Cell Dev Biol. 2010 Dec 13;22(1):57–68. doi: 10.1016/j.semcdb.2010.12.005

Rab GTPases implicated in inherited and acquired disorders

Shreya Mitra 1,*, Kwai W Cheng 1, Gordon B Mills 1
PMCID: PMC3395236  NIHMSID: NIHMS384559  PMID: 21147240

Abstract

Endocytotic pathways ensure that internalized receptor complexes are routed in a highly orchestrated manner to the correct subcellular destinations. This, in turn, determines the consequences of receptor activation through targeting receptors and ligand for recycling or degradation as well as by the formation of signaling complexes within the cell that alter the kinetics and magnitude of activation of specific downstream signal transduction cascades. Thus the control of the fate of activated receptor complexes has profound physiologic and pathophysiologic implications. Rab GTPases, the largest subfamily of the RAS small GTPase superfamily, are the key regulators of endocytosis through decorating and targeting intracellular vesicles and cargoes to appropriate subcellular compartments. The six-fold increase in Rab family members from yeast to man correlates closely with the evolutionary increase in endomembrane complexity compatible with a rapid diversification of function of Rab GTPase decorated vesicles. As the vesicular cargo includes growth factors, nutrients, cytokines, integrins and even pathogens, aberrations in the pathway are likely to exhibit dire consequences. Several genetic diseases driven by mutations in Rab GTPases or their interacting proteins have been identified [1], [2], [3]. Aberrant Rab GTPase function has been implicated in diverse pathophysiologies including loss of hair, skin and eye pigmentation, loss of vision, loss of renal function, mental retardation, muscle skeletal degeneration, immune deficiency, infection, obesity, diabetes and cancer.

1. Introduction

Vesicular trafficking

We are only beginning to comprehend the far-reaching consequences of vesicular trafficking in health and disease. The endocytotic pathway regulates overall cell signaling and protein stability, cell polarity, motility, differentiation and survival of cells, as well as the integration of cellular processes into normal physiological function of tissues and organs [4], [5], [2], [6]. A dynamic endomembrane system compartmentalizes the eukaryotic cell into chemically distinct subcellular domains. Each domain has a signature lipid bilayer composition decorated by a unique set of Rab GTPases, optimized for seamless import, trafficking and export of biomolecular cargoes. The vesicular trafficking machinery ensures correct localization and routing of diverse protein complexes from their sites of synthesis or uptake to specific destinations, without any promiscuous interactions or deleterious mixing of organelle constituents. Dysregulation of vesicle trafficking leads to diverse pathological outcomes, some of which are life threatening while others significantly impair quality of life [2], [7]. A number of comprehensive reviews summarizing the molecular, physiological and pathological aspects of vesicular trafficking have been published [2, 5],[8],[9]. In this review, we update the role of the Rab GTPases as the “master organizers” of vesicular trafficking. We also use an interactive process based on experiments of nature such as monogenic inherited disorders, to identify functions of Rab GTPases and elucidate how disruption of vesicular trafficking can contribute to such monogeneic inherited disorders as well as multigenic conditions such as diabetes and cancer [10]. The concept that unraveling the role of derailed endocytosis in disease could increase the rate of development of useful therapies and biomarkers is driving current research [11], [12],[13].

Rab GTPases

The small GTPases (20–25 kDa) comprise the Ras superfamily members which includes the Ras, Rho, Rab, Ran and Arf families [14]. The processes controlling their dynamic interaction and hydrolysis of guanosine diphosphate (GDP; inactive) or guanosine triphosphate (GTP; active) makes the small GTPases effective molecular switches controlling diverse cellular processes. This review specifically focuses on the Rab GTPases, which constitute the largest subgroup of small GTPases [15].

Originally described as Ras-like proteins in brain (Rab), about 70 putative Rab family members have been identified in humans [1, 16]. Their conserved GTP binding sites and variable C-terminals impart the necessary specificity to act as molecular switches controlling downstream effector interactions. As byproducts of recent gene duplication, however, many Rab proteins have likely retained a degree of functional redundancy [17]. Overall, the Rab GTPases regulate various steps of dynamic assembly and disassembly of multiprotein scaffolds involved in vesicular traffic in both endocytic and secretory pathways [18] [19], [20]. Rab proteins have evolved to regulate endocytosis, recycling and degradation as well as exocytosis of various cargos within the cell, providing appropriate raw material as well as signals for cellular and overall tissue homeostasis. Importantly, their physical and functional links to signal transduction pathways provides the key to spatiotemporal regulation of signaling [6, 21]. Membranous vesicles with characteristic Rab GTPase/effector domains comprise the basic infrastructure of the cellular trafficking machinery. Rab proteins and their effectors couple endomembranes to motor proteins and the cytoskeleton [22] facilitating transport of vesicles and their cargoes to subsequent compartments. Thus, in a multistep process, Rab proteins facilitate vesicle formation via budding from the donor compartment, transport to the acceptor compartment, vesicle fusion and release of the vesicle content into the appropriate acceptor compartment [2327].

Biochemistry and Regulation of Rab GTPases

A. GTP/GDP Cycle

Rab proteins have two highly conserved motifs housing the effector-interacting switch I and II regions. Binding of GDP or GTP in the switch region, results in marked conformational changes in Rab structure. The intrinsic GTP hydrolysis capacity of Rab GTPase proteins as well as GDP/GTP exchange rates is low. GDP/GTP cycling is controlled by two interrelated processes. The guanine-nucleotide-exchange factors (GEFs) promote formation of the active, GTP-bound form [28], whereas GTPase-activating proteins (GAPs) accelerate the intrinsic GTPase activity to promote formation of the inactive GDP-bound form (Fig 1). Amongst the two nucleotide-bound states, only the GTP-bound conformation has high affinity for effector molecules.

Figure 1. RabGTPases Mediated Vesicular Trafficking.

Figure 1

Rab GTpases are involved in trafficking of cell membrane receptors including RTKs, GLUT4 and integrins.

Rab5 facilitates fusion of clathrin-coated vesicles containing activated receptor to EE. The cargo is then sorted either to Rab4 decorated vesicles for fast-track recycling or to Rab11 decorated long loop slow recyling endosomes. Alternatively, for attenuation of signal transduction, the cargo laden vesicles are targetted to the lysosome via Rab7 and Rab9 coated late endosomes. The Endosomal Sorting Complex (ESCRT) recognizes ubiquitinated proteins and routes them for degradation. Rab GTPases such as Rab3, Rab10 and Rab27 play critical role in the exocytosis/secretory pathway, while Rab1 and 2 are necessary for ER-Golgi transport. Receptor ligand complexes are differentially sensitive to pH, a fact that is utilized by the vesicular trafficking machinery to uncouple the signalosome.

B. Membrane Insertion Cycle

Rab proteins undergo a membrane insertion and extraction cycle, partially in tandem with the GDP/GTP cycle, to allow exchange of cargo between specific membrane domains (Fig 1). Posttranslational lipidation (prenylation), dictated by divergent C-terminal sequences, is critical for membrane insertion into the appropriate compartments [29, 30]. De novo, Rab, as synthesized by cytosolic ribosomes, is GDP bound and inactive, with very low affinity for enzymes that mediate lipidation, such as geranylgeranyltransferase (RabGGTase or GGTaseII) [29, 31]. In contrast to other GTPases, the GDP-Rab requires an additional escort protein, referred to as the Rab escort protein (Rep) [32], to be presented to the prenylating enzyme. This posttranslational modification occurs at the C-terminal by addition of the 20-carbon isoprenoid geranylgeranyl, mediated by RabGGTase. Next, the REP sequesters the prenyl groups attached to the C-terminus of Rab to form a soluble complex until the prenylated protein is delivered to its appropriate membrane compartment. GDP dissociation inhibitor (GDI) binds to prenylated GDP-Rab, however, masking its isoprenyl anchor and thereby maintaining the Rab protein in the cytosol. Membrane attachment of Rab proteins therefore requires the function of a GDI displacement factor [33]. REP is also required to release the GDI from the Rab molecule so it can be delivered to its specific membrane. Once dissociated from GDI and associated with the appropriate membrane compartment, the Rab is available for GEF-stimulated GTP binding. Thus the importance of Rep is underscored by the fact that lethal phenotypes or pathological conditions occur where Rep activity is aberrant [34]. The active, membrane-bound Rab participates in various functions in membrane traffic by binding to one or more effector molecules. After inactivation by its specific GAP, the GDP-bound Rab is extracted from the membrane by GDI and recycled back to the cytosol [29], [35]. This allows for a dynamic change in the Rabs decorating particular vesicles, allowing maturation of the vesicle and sorting to different compartments.

Rab Effectors

Using a variety of approaches, such as the yeast two-hybrid system, genetic screens and affinity purification, a plethora of potential Rab effectors have been implicated in diverse aspects of membrane transport. Rab effectors are a highly heterogeneous group of proteins with broad Rab-binding specificity. Some are coiled-coil proteins involved in membrane tethering or docking, while others are enzymes or cytoskeleton-associated proteins regulating molecular events at restricted membrane locations and microtubule-dependent motility of endocytic structures [36] [37]. Thus in addition to the GTP switch and delivery of Rab proteins to specific compartments by lipidation, the coordinate interactions of Rab proteins with specific effector molecules adds an additional level of complexity allowing Rab proteins to regulate an even more diverse set of cellular processes and functional outcomes.

Regulation of Cargo Delivery by Rab GTPAses

Rab GTPases are not mere transporters of cargoes; they also facilitate the arrival of cargoes at the appropriate vesicle (Fig 2). As an example, mannose 6-phosphate receptors recycle between trans-Golgi networks via late endosomes, carrying lysosomal hydrolases laden with mannose 6-phosphate. The late endosomal GTP-Rab9 effector protein, TIP 47 by binding to the cytoplasmic tail of the mannose 6-phosphate receptor captures and enriches these receptors within Rab9-containing vesicles, thus ensuring their transport back to the Golgi [38], [39]. Thus the Rab proteins ensure the delivery of the mannose 6-phosphate receptors and lysosomal hydrolases to the correct compartments. Rab GTPases also influence vesicle tethering and fusion through their effects on members of the family of soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE), which are upstream of various Rab effectors [40], [41], [23],[42]. An example of a more direct mechanism, the coiled-coiled EEA1, a Rab5 effector, bridges Rab5-bearing donor and acceptor membranes [43], [44]. Thus the Rab proteins and their effectors act in a coordinate manner to ensure delivery of vesicles and cargoes to the correct compartment as well as to ensure that inappropriate mixing of vesicle and cargoes does not occur.

Figure 2. GTP Exchange and Membrane Association Cycles.

Figure 2

The newly translated GDP bound Rab in the ER is recognized by REP, and presented to GTT enzyme for prenylation. In many cases, following prenylation, a GDP dissociation inhibitor (GDI) associates with the prenylation group, allowing the Rab to remain in the cytosol, prior to being translocated to the correct endosomal compartment. At this compartment, GDI is removed by GDF allowing a guanine exchange factor (GEF) to facilitate GDP-GTP exchange.

The GTP bound Rab can now interact with its effectors and perform various trafficking functions. The membrane associated active GTP Rab is inactivated by GTPase activating proteins (GAPs), thus returning back to the inactive GDP bound state. In some cases, the GDI extracts the GDP bound Rab back into the cytosol, allowing the process to reinitiate.

Vesicles are often actively transported through the cytoplasm toward their target membrane by either actin-dependent motors (myosins) or microtubule-dependent motors (kinesins or dyneins). During evolution, Rabs became functionally connected to motors of the actin cytoskeleton. This collaboration facilitates tethering of vesicle membranes to molecular motors, which can then efficiently transport the cargo to specific cellular addresses. For example, Rab27A uses its effector melanophilin as a linker to Myosin 5a [45] and active Rab6 binds the microtubule motor Rabkinesin-6 as a means to deliver vesicles from the Golgi to the endoplasmic reticulum [46],[47].

Multiple Rab cascades feed into each other to relay their cargo to the correct subcellular compartment. Rab domains and effectors can be structurally and functionally linked allowing coordination of transfer of cargoes between specific compartments. As an example, Rab4 and Rab5 share a common set of effectors, namely, Rabaptin4, Rabaptin5, Rabenosyn5 and Rabip4 [48], [49], [50] coordinating endocytic traffic mediated via Rab5 with recycling traffic mediated via Rab4. First Rab5 is recruited to the membrane where it is activated by the GEF Rabex5, facilitating interaction with its effector, Rabaptin5. Subsequently, Rabaptin5 binds to Rabex5, which increases the guanine nucleotide exchange activity of Rabex5 for Rab5, generating a positive feedback loop which outcompetes the GAP inactivation and GDI-mediated membrane extraction effects on Rab5 [51]. As the enlarging Rab5-bearing early endosomes migrate toward the center of the cell, they progressively lose the Rab5/EEA1 complex. Rab7 begins to replace Rab5 in these vesicles along with late endosome-specific cargo [51]. Such Rab exchange also alters the endosome qualitatively as degradative properties typical of late endosomes develop. These interrelated mechanisms where several aspects of a process are regulated by a single molecule such as the GEF of a downstream Rab GTPase (Rabex5) also serving as an effector of an upstream Rab protein (Rab4) suggests that it will be necessary to implement systems biology and modeling approaches to determine the consequences of mutation or change in levels of specific Rab GTPases and their regulators and effectors to capture and understand the complexity of the process.

Distribution and Localization of Rabs

Some Rabs are expressed ubiquitously in human tissues, whereas others demonstrate tissue type specificity [52]. Despite the variability among different tissues, Rabs appear to cluster in unique groups based on similarities in their expression patterns across different tissues [53], [18], [54] [55],[15],[56] suggesting a coordinated action within the groups. In addition to coordinate expression across tissues, the function of a specific Rab is governed by their subcellular distribution combined with the unique tissue/subcellular distribution of requisite GDIs, Reps, GEFs, GAPs, and effector proteins. It is thus likely that Rab proteins demonstrate markedly different functions based on the cellular context combined with the formation of Rab-regulated protein hubs.

A. Housekeeping Rabs

Gurkan et al., used a hierarchical clustering approach to propose a functional organization for Rab GTPases [53]. They proposed that a class of Rabs delegated to maintenance of constitutive exocytic and endocytic pathways includes Rab1 isoforms (involved in endoplasmic reticulum-to-Golgi transport in the exocytic pathway), and isoforms of Rab 4, 5, 7 and 11 involved in early and late endosome function [55] be designated housekeeping Rabs. However as several of these Rab proteins are aberrant in human disease resulting in discrete phenotypes and the nature of cargoes and functions are altered by levels of these Rabs and their effectors, they likely subserve important cell specific functions independent from the proposed “housekeeping” role [53].

B. Specialized Rabs

The abundance of Rabs in higher eukaryotes suggests that they have evolved to contribute to specific subcellular trafficking pathways, forming a class of specialized Rabs. For example, Rab3A, which is expressed predominantly in brain and synaptic vesicles, specifically tethers and docks synaptic vesicles prior to fusion, a critical step in regulated release of neurotransmitters [57]. In contrast, Rab27, a phylogenetically distinct protein, functions in the secretory lysosome pathway in a variety of specialized subcellular organelles such as cytotoxic T lymphocyte granules, antigen-processing compartments involved in major histocompatibility complex class II presentation, platelet granules and melanosomes in melanocytes [58], [59]. These Rabs however, do not appear to exhibit functions in other tissue types.

2. Rab GTPase Associated Diseases

A summary of the various monogenic and multigenic diseases associated with Rab GTPases are listed in Table 1 and broadly discussed in the following paragraphs.

Table 1.

Rab GTPases implicated in monogenic and multigenic diseases

RAB Isoforms Cellular Locations Functions Diseases
Rab1
A, B
ER, Golgi ER-Golgi trafficking Elevated in tongue squamous cell carcinoma
Oculocerebrorenal Syndrome
Viral infections
Rab2
A,B
ER, Golgi
Plasma membrane
Melanosomes
ER-Golgi trafficking Elevated in various cancers
Rab3
A, B, C, D
Plasma membrane Neurotransmitter release
Fusion of synaptic vesicle
Exocytosis (insulin)
Protein transport
Cancers of nervous system, Neuro-endocrine related cancers
Pituitary adenomas

Warburg Micro Syndrome
Rab4
A,B
Plasma membrane Protein transport Obesity, Diabetes
Rab5
A, B, C
Plasma membrane, Early endosome, Melanosomes Fusion of PM to EE
Protein transport
Rab5A elevated in lung adenocarcinoma, hepatocellular carcinoma, thyroid carcinoma 5B decreased in metastatic melanoma

Tuberous Sclerosis Complex
Oculocerebrorenal Syndrome
Neurodegenerative Diseases
Huntington’s Disease
Bacterial infection
Diabetes
Rab6
A, A′, B
Golgi membrane Golgi-ER trafficking Oculocerebrorenal Syndrome
Rab7
A, B
Late endosome, Lysosome
Phagosome
Melanosomes
Late endocytic transport, Maturation of phagosome Hereditary Sensory and Autonomic Neuropathies
Charcot-Marie-Tooth Types 2B
Niemann-Picktype CPC
Alzheimer’s Disease
Chagas Disease
Thyroid carcinoma
Rab8
A, B
Plasma membrane Vesicular trafficking
Neurotransmitter release
Dendrite Extension
Ciliogenesis
Melanoma
Retinal degeneration in mice/Glaucoma
Microvillous Inclusion Disease
Bardet-Biedel Syndrome
Rab9
A, B
Late endosome Endosome-TGN trafficking Niemann-Picktype C Disease
Rab10 Plasma membrane Neurotransmitter release
Vesicular trafficking
Phagosome Maturation
Glut4 translocation
Type 2 Diabetes
Rab11 family
A,B,C
Rab11C synonymous with Rab25
Recycling endosome
Rab25 in Apical Recycling compartments
Log loop recycling
Rab25 implicated in integrin recycling
Cell motility
Ciliogenesis
Breast, lung, colon, ovarian endometrial, prostrate, bladder carcinomas.

Rab11 implicated in Chagas Disease
Rab11A in Diabetes
Rab25 in various Cancers
Rab13 Tight junction Polarized transport, Tight junction activity Crohn’s disease
Upregulated in Tuberculosis
Rab23 Plasma membrane Negative regulator of sonic
hedgehog signaling
Vesicular Trafficking
Carpenters Syndrome
Craniosynostosis
Hepatocellular carcinoma
Gastric cancer
Rab24 ER-Golgi, Late Endosome Autophagy Tuberculosis
Rab27
A, B
Plasma membrane, Melanosomes Exocytosis Choroidermia
Griscelli Syndrome 2
Type 2 Diabetes
Breast cancer
Rab31 Plasma membrane
Trans-Golgi network
Anterograde transport Metastatic breast cancer
Rab33
A, B
Golgi Autophagosome formation Rab33A downregulated in Tuberculosis
Rab38 Melanosomes Melanosomal transport, docking, sorting TYRP1 Hermansky Pudlak Syndrome

Monogenic Diseases Resulting from Rab GTPase Aberrations

Many of the monogenic disease driven by aberrations in Rab GTPase and their effectors are rare orphan diseases, affecting only a limited number of families. However, these monogenic diseases have the potential to provide important information on the function of specific Rab GTPases as well as on the integration of vesicle trafficking into cellular and organ functions. Thus understanding these diseases is not only important for members of the affected families and their caregivers but also of great importance based on the application of lessons from these “experiments of nature” to more common multigenic diseases.

A. Choroidermia

Mutations in the Rab escort protein 1 (Rep1) are the cause of choroidermia, a rare X-linked disorder that manifests as degeneration of the choriocapillaris, the retinal pigment epithelium and the photoreceptor layer in the eye, resulting in night blindness and loss of peripheral vision [60]. Although Rep1 is phylogenetically similar to GDI, the latter cannot compensate for the loss of Rep function [34]. Interestingly, despite Rep2 being able to substitute for Rep1 for several Rabs, REP2 is unable to interact with Rab27A, the predominant Rab in retinal pigment epithelium cells. Loss of Rep1 thus results in accumulation of unprenylated Rab27A in the eye, leading to specific deficits due to an essential role for Rab27A. Although there is a diagnostic test for this disorder there are no current interventions.

B. X-Linked Mental Retardation

Mutations in neuronal Rab GDIa, one of the three members of a mammalian GDI family bearing close resemblance to Rep, are the cause of one form of X-linked mental retardation. Rab GDIa, a potential negative regulator of synaptic function, is essential for neurotransmission (along with Rab3A) as evinced by onset of neuronal hypersensitivity and epileptic seizures in Rab GDI-deficient mice [61]. More recently, Rab39B has also been linked to X-linked mental retardation that is associated with autism, epilepsy, and macrocephaly [62] suggesting a major role for Rab proteins in maintenance of normal neuronal function.

C. Griscelli Syndrome

Griscelli syndrome is a rare autosomal recessive disorder characterized by pigment dilution in hair and skin and variable immune deficiency that can lead to a life-threatening hemophagocytic condition. Griscelli syndrome is classified into three subtypes based on genetic and molecular features. GS3 is restricted to hypopigmentation, while the two other subtypes are additionally characterized by immunological defects (GS2) or neurological dysfunction (GS1). All three types demonstrate impaired vesicular trafficking manifest as perinuclear accumulation of melanosomes [63, 64]. GS2 is attributed to mutations in Rab27A gene [65].

Skin and hair melanocytes store melanin pigment in specialized lysosome-related melanosomes. Mature melanosomes traverse along microtubule tracks from the cell center to the cell periphery. Their bidirectional transport engages dynein motors to reach the minus end of the microtubule [66] and kinesin motors to arrive at the plus end before they reach their final peripheral destination [67]. Finally, from the microtubule, they are transported and trapped into the distal actin-rich regions of dendritic filaments [68, 69]. Rab27A and the actin motor Myo5A [70], co-localize with melanosomes [71] with the help of linker melanophilin (Rab27 effector), forming a tripartite complex that leads to attachment to the actin network [28],[72], [45], [33], [32]. Thus aberration in any component of this process could lead to a common phenotypic outcome, Griscelli syndrome.

The recurring mutations in Rab27A are homozygous nonsense or frame shift mutations, with only a few reported cases of missense mutations. Typically a missense mutation results in either a nonfunctional protein or complete loss of the protein through altered stability of the mRNA or protein, thus diminishing the available cellular pool of Rab27A [73]. If the mutation results in a premature truncation of the C-terminal geranylgeranylation motif (an early stop codon insertion), the Rab27A proteins cannot be targeted to the correct vesicle, resulting in loss of functionality.

D. Hermansky-Pudlak Syndrome

Hermansky-Pudlak syndrome similar to Griscelli syndrome is characterized by deficiency of the melanin pigment in the eyes, skin and hair; other features include bleeding diathesis and lung disease. This disease is associated with alterations in Rab38, which is highly expressed in melanocytes of the skin and alveolar type II cells in the lungs. In rodents, two different mutations causing inactivation of Rab38 reduced membrane binding or enhanced intracellular degradation, which resulted in oculocutaneous albinism and reduced clotting. Alteration in Rab38 also affected surfactant homeostasis and alveolar function in rodent lungs. Thus a genetic abnormality of Rab38 can affect multiple lysosome-related organelles, resulting in lung disease in addition to oculocutaneous albinism [74, 75].

E. Charcot-Marie-Tooth Type 2B

Charcot-Marie-Tooth disease type 2B (CMT2B) disease is a genetically and clinically heterogeneous group of autosomal-dominant axonal disorders affecting specifically peripheral neurons with a median motor conduction velocity of >38 m/s [76]. Manifestations include severe sensory loss, distal muscle weakness and a high frequency of foot ulcers, infections and even amputations because of recurrent infections. CMT2B maps to chromosome 3q13-q2, in a 2.5-cM region with four missense mutations in the small GTPase late endosomal protein Rab7. Rab7 plays a role in vesicular transport to late endosomes and lysosomes in the endocytic pathway in most cell types [77], [78]. As with many other inherited disorders, it is unknown how mutations in a ubiquitously expressed Rab GTPase with such a generalized functions result in defects in a specific cell population. The most likely explanation is based on another Rab protein demonstrating redundancy with Rab7 either not being expressed in the target neurons or by analogy a particular effector of Rab7 being rate limiting for neuronal function but not for functions of other tissues.

F. Hereditary Sensory and Autonomic Neuropathy

Hereditary sensory and autonomic neuropathy (HSAN) is an autosomal recessive disorder, more common in Jewish individuals [79], affecting the autonomous nervous system. Some clinical symptoms include loss of pain and temperature sensation, alacrima, excessive sweating and absence of fungiform tongue papillae. Some patients exhibit piercing pain. The HSAN type 1 subtype is associated with mutations in Rab7 [79] and in serine palmitoyl transferase, a rate-limiting step in sphingolipid synthesis [80], [81]. Rab7 plays a role in the transport of synthesized lipid from early to late endosomes and late endosomes to lysosomes in many cell lineages and mutations in the broadly expressed serine palmitoyl transferase result in selective disorders of cells in the autonomous nervous system. Why Rab7 mutations cause Charcot-Marie-Tooth and HSAN in different families is unknown. Interestingly, HSAN type II, which shares common traits with Charcot-Marie-Tooth Type 2B suggesting that HSAN and Charcot-Marie-Tooth Type may represent different manifestations of the same syndrome potentially due to the presence of important modifier genes.

G. Warburg Micro Syndrome and Martsolf Syndrome

The Warburg Micro and Martsolf syndromes are autosomal recessive disorders with developmental abnormalities of the eye, central nervous system including severe mental retardation, and microgenitalia [82]. Presynaptic vesicle trafficking and priming are important steps in regulating synaptic transmission and plasticity. Isoforms of Rab3A, the most abundant Rab in the brain, and its effectors are involved in Ca2+-dependent exocytosis during neurotransmitter release from nerve terminals [83], [84]. The Warburg Micro and Martsolf syndromes results from failure of exocytic release of ocular and neuro-developmental trophic factors, implicating Rab3. Indeed, mutations in the catalytic subunit p130 and the noncatalytic subunit p150 of Rab3 GAP were recently found to cause Warburg Micro syndrome and Martsolf syndrome, respectively [84] [85]. As predicted above, aberrations in Rab proteins as well as their regulators and effectors alter vesicle function.

H. Tuberous Sclerosis Complex (TSC)

TSC is characterized by the presence of benign hamartomas in multiple organs, including the central nervous system, lung, kidney, heart and skin, as well as learning and behavioral difficulties and renal complications (hematuria, renal cysts). TSC2 functions as a GAP for Rab5 and also for Rheb. Both Rab5 and Rheb interact in the mTOR pathway [86, 87]. How aberrations in the ubiquitously expressed Rab5 “housekeeping” Rab or in Rheb lead to the development of benign hamartomas in particular tissues remains unknown. However, the observation that inhibition of mTOR, a downstream target of Rab5 and Rheb, has marked clinical activity in Tuberous Sclerosis supports the contention that an understanding of the physiology and pathophysiology of vesicle function could have marked clinical relevance [88, 89].

I. Carpenter Syndrome

Carpenter syndrome is pleiotropic disorder manifested by craniosynostosis, polysyndactyly, obesity and cardiac defects. This autosomal recessive disorder is caused by truncation or missense mutations in the Rab23 gene. How aberrations in Rab23 that regulates trafficking of a plethora of proteins, including members of the intraflagellar transport complex and associated motor proteins leads to Carpenter syndrome remains to be elucidated. However, as Rab23 plays a major role in sonic hedgehog (SHH) signaling [90], [91] and drugs targeting this pathway are under development, they may alleviate some of the problems associated with this disease.

J. Niemann-Pick Type C Disease

Niemann-Pick type C (NPC) disease, which is associated with aberrant cholesterol trafficking at endosomes, is characterized by progressive neurological degeneration associated with hepatosplenomegaly. Symptoms first appear at age 2 to 4 years, resulting from accumulation of abnormal amounts of cholesterol and other lipids (including glycosphingolipids, sphingomyelin, lysobisphosphatidic acid and phospholipids) in the late endosomal/lysosomal compartment. In neurons, abnormal distribution of cholesterol in the cytoplasm rather than the endosome appears as the key lesion.

NPC is caused by mutations in the NPC1 or NPC2 transmembrane proteins that localize in late endosomes, facilitating relocation of free cholesterol to other cell compartments. It is possible that activating the Rab-mediated late endosomal recycling machinery, such as that regulated by Rab7 or Rab9 [92], [93] could reverse the effects of NPC.

At the cellular level, NPC shares some traits with Alzheimer disease including endosome enlargement, elevated hydrolase content and accumulation of p-cleaved amyloid precursor protein and amyloid precursor peptides within endosomes [94]. Although mutations in Rab proteins, regulators or targets have not be identified in Alzheimer disease, upregulation of Rab5 and Rab7 mRNA have been observed, implicating early endosomal dysfunction in Alzheimer’s disease progression [95]. Thus an exploration of endosomal trafficking could provide potential insight leading to effective treatments for this common and devastating disease.

K. Oculocerebrorenal Syndrome of Lowe

The Oculocerebrorenal Syndrome of Lowe, first described in 1952, is an X-linked multi-organ disease caused by mutation of OCRL1 an inositol polyphosphate 5-phosphatase. OCRL1 catalyzes the conversion of phosphatidylinositol (4, 5)-biphosphate to phosphatidylinositol 4-phosphate. The disease is characterized by mental retardation, congenital cataracts and reduced ammonia production by the kidney. Upon growth factor stimulation, OCRL1 translocates from the Golgi apparatus and early endosomes to the lamellipodia, a journey intimately associated with several members of the Rab family of small GTPases [96], [97],[98]. Overexpression or depletion of OCRL1 inhibits trafficking of proteins from early endosomes to the trans-Golgi networks, suggesting a role in clathrin-mediated trafficking between these compartments [98]. Since clathrin adaptors mediate transport of different proteins, including lysosomal hydrolases from trans-Golgi networks to endosomes, defects in OCRL1 could disrupt normal endosomal trafficking. The Golgi-associated Rab1 and Rab6 and endosomal Rab5 have been proposed to play a dual role in targeting OCRL1 to both the Golgi apparatus and endosomes and in directly stimulating the 5-phosphatase activity of OCRL1 following membrane recruitment.

Multigenic Diseases Associated with Rab GTPases

A. Neurodegenerative Diseases

Neuronal Rab5 is essential for synaptic plasticity and neuroprotection [99], playing an important role in AMPA receptor internalization and overall neurotransmission. Not surprisingly, Rab5 defects are detected in various neurodegenerative syndromes such as autosomal recessive motor neuron disease caused by loss of function of Alsin (encoded by ALS2), which is a GEF for Rab5 [100]. Rab5-alsin interaction modulates insulin-like growth factor 1 and other neurotrophic signaling factors likely contributing to the progression of the ALS syndrome [101].

In one study, Huntingtin and Huntingtin-associated protein (HAP40) were identified as Rab5 effectors that regulate endosome motility [102]. Increased HAP40, as observed in Huntington’s disease, reduces mobility of Rab5 endosomes and derails endosomal transport, contributing to neurodegeneration [103]. Moreover, in a recent report, aberrant Rab11 mediated trafficking of a neuronal glutamate transporter was linked to oxidative stress and cell death leading to neurodegeneration in Huntington’s disease [104].

During juvenile onset of Parkinson disease, ER-Golgi transport is disrupted because of mutations in the α-synuclein gene. Overexpression of Rab1GTPase can reverse the toxic effects of α-synuclein and protect against neurodegeneration [105107] suggesting a potential therapeutic approach.

B. Cystic Fibrosis

CTFR is a chlorine-selective ion channel belonging to the ATP-binding cassette transporter superfamily, which is often mutated in cystic fibrosis and other genetic diseases. The clinical symptoms include exocrine pancreatic insufficiency, increase in sweat concentration of sodium chloride, male infertility and airway disease. Ion channel transporters travel along the canonical endosomal recycling pathway regulated by Rab GTPases. Transport of the cystic fibrosis transmembrane conductance regulator (CTFR) from the plasma membrane to early endosomes is regulated by Rab5; Rab11 recycles it back to the plasma membrane, while Rab7 and Rab9 control its trafficking from early endosomes to late endosomes and finally to the trans-Golgi networks, respectively. As Rab GTPases act as molecular switches that regulate CTFR, they could demonstrate therapeutic utility [108, 109].

Rab Proteins in Immune Response and Infection

A. Crohn’s Disease

Crohn’s disease is associated with deregulated immunological responses and chronic inflammation in the intestinal mucosa. The underlying cause is increased permeability of the intestine due to dislocation of tight junction proteins. Rab13, along with vasodilator-stimulated phosphoprotein and zonula occludin-1, an apical tight junction protein, are often dislocated to the basolateral position in patients with inactive Crohn’s disease, with little alteration in F-actin. The immune response is altered when tight junctions are compromised, allowing invasion of gut antigens [110]. Whether mutations in Rab13, its effectors, or regulators contribute to Crohn’s disease is as yet unknown but clearly warrants investigation.

B. Infection

Rab proteins regulate the entry and exit of a number of bacteria and virus cargoes in the cell. Rab5 is essential for routing internalized biomolecules from the plasma membrane to the endosomal trafficking pathway, which eventually directs bacteria contained in internalized phagosomes to the lysosome for degradation of the pathogen. A number of bacterial pathogens have developed survival mechanisms allowing escape from the phagosome into the cytoplasm by disrupting Rab function, and thus eluding lysosomal degradation. Immune responses initiate with cytokine stimulation of natural killer cells and T-helper cells, which upregulates expression of Rab5a. Subsequently Rab5a facilitates intracellular degradation of the pathogen by recruiting Rac2 to phagolysosomes [111]. The effect of Rab5 is effectively countered by Listeria monocytogenes by preventing extraction of Rab5 from the membrane. In another example, Salmonella type III uses secretory proteins such as SopE with GEF activity to stabilize GTP-Rab5 on the phagosome, thus preventing extraction by RabGDI. These strategies ensure that the phagosome fails to reach lysosome, allowing the bacteria avoid degradation. In another study, gene expression arrays showed a signature of downregulated Rab33A, a T-cell regulator, and upregulated Rab33 and Rab14 in patients with tuberculosis (Mycobacterium tuberculosis) [112, 113]. In sexually transmitted Chlamydia infection, a yeast two-hybrid system linked a Rab4A effector, a Chlamydia inclusion membrane protein Cpn0585, to pathogenesis of the disease [114, 115]. In Chagas disease, the causative protozoan (Trypanosoma cruzi) impairs endocytosis in the host by altering the recycling Rab7 and Rab11 [116].

Viruses enter the host cells via receptor-mediated endocytosis. However, they must escape endosomal vesicles to avoid degradation. In the case of the hepatitis C virus, the host Rab1 GAP (TBC1D20), implicated in regulation of anterograde traffic between the endoplasmic reticulum and the Golgi complex, is hijacked by a viral nonstructural protein to promote viral membrane-associated RNA replication [117] and propagation.

Rab Trafficking and Metabolic Diseases

A. Obesity and Impaired Lipid Trafficking

Obesity is a major health concern in the modern world, as it is an underlying contributor to multiple diseases including diabetes, hypertension, atherosclerosis and cancer. Rab GTPases are not merely transporters of proteins and carbohydrates; one of their most critical functions is proper lipid trafficking. Cells regulate the levels and distribution of lipids such as cholesterol, a key structural component of membranes. In adipose tissue, cholesterol homeostasis is regulated via lipogenesis and lipolysis. Impairment of the lipolytic pathways in adipose tissue and skeletal muscle are important factors in increased triglyceride storage, contributing to obesity, insulin resistance and type-2 diabetes. Small GTPases, especially Rab7 and Rab5 GTPases, by virtue of their roles in trafficking to and from the endosome/lysosome organelles, are implicated in the pathology of these diseases.

Loss of the recycling Rab4b, possibly as a compensatory modification to counter faulty glucose transport [118] in adipocytes, increased obesity in rodents implicating Rab4 proteins in obesity. In neurons, the Rab7-mediated endocytic pathway is under the regulation of Tub1, and mutations in Tub1 were shown to lead to increased fat storage in mice [119]. Whether the regulation of Rab7 contributes to this function of Tub1 remains to be elucidated.

B. Type 2 Diabetes

Cellular uptake of glucose from the circulation occurs through the translocation of glucose transporters to the plasma membrane in response to insulin. In type 2 diabetes, insulin resistance results in insufficient amounts of GLUT4 being translocated from the endosomal recycling compartment, the Golgi complex and the trans-Golgi networks [120], [121] to the plasma membrane despite normal GLUT4 expression. The AKT substrate AS160, also a Rab GAP, was shown recently to connect AKT signaling downstream of insulin to GLUT4 trafficking [122]. Interestingly, in a cochlear hair cell model, downregulation of another Rab, the recycling Rab11a, and two of its effectors, Rip11 and Rab11FIP2, reduced the plasma membrane abundance of GLUT4, providing further evidence of a potential role of Rab GTPases in pathogenesis of metabolic diseases such as diabetes [123, 124].

Rab27 mutant (Ashen) mice demonstrate glucose intolerance of insulin resistance in peripheral tissues or insulin deficiency in the pancreas. This appears to be due to a role for the Rab27a in F-actin mediated exocytosis of insulin containing granules in pancreatic beta cells [125],[121], [126]. Similar mutations in human diabetes have not yet been identified.

C. Cancer

Dynamic genomic changes drive tumorigenesis by deregulating cell proliferation and homeostasis. Signals emanating from membrane-bound growth factor receptors and integrins regulate these critical proliferation and survival pathways. Derailed endocytosis imparts growth factor and extracellular matrix autonomy to cancer cells by shunting degradation and promoting recycling of growth factors as well as integrins (see [4, 5] for recent reviews ). Thus aberrant endocytosis, vesicle targeting and receptor recycling, which are instrumental in altering cell adhesion, migration, proliferation, polarity, asymmetrical division and overall survival, represent emerging hallmarks of cancer.

I. Aberrant Rab GTPase expression in Cancer

Integrative genomic studies have implicated aberrations in Rab pathways as contributing to the initiation and progression of multiple cancer lineages. Elevated levels of Rab23 are associated with invasive gastric cancer [127], Rab38 with melanoma [128], Rab2B with colon carcinoma and Rab1A with tongue cancer [129, 130]. Upregulation of Rab5a and Rab7b occurs in thyroid-associated adenomas [131], while the Rab27A effector JFC1/Slp1 is an important regulator of exocytotic processes in prostate carcinoma cells [132, 133]. Hepatocellular carcinomas may be particularly sensitive to Rab deregulation, as Rab1B, Rab4B, Rab10, Rab22A, Rab24 and Rab25 were shown to be commonly upregulated in these cancers [134]. Rab27 isoforms, which are secretory GTPases that control vesicle exocytosis and deliver critical proinvasive growth regulators into the tumor microenvironment, are associated with poor outcome in invasive breast cancers. Rab27A promotes invasion and metastasis of human breast cancer cells through regulation of insulin-like growth factor II secretion [135]. Rab27B, on the other hand, activates heat shock protein 90 (Hsp90) and matrix metalloproteinase 2 (MMP2), thus increasing G1-S transition, proliferation, invasive tumor growth and lymph node metastasis in an estrogen receptor–positive mammary carcinoma xenograft model [26]. Rab31 is also associated with poor outcome in invasive breast cancer [136]. Systematic analysis of ovarian cancers demonstrated upregulation of almost half of known RAB or RAB-associated genes [2].

The phosphatidylinositol 3 kinase pathway has been implicated in a broad variety of cancers. Intriguingly, PIK3R1, which encodes p85alpha and is mutated in a number of cancers, exhibits GAP activity towards early endosomal Rabs Rab4 and Rab5 [137]. A GAP-defective mutant p85R274A induces sustained levels of activated platelet-derived growth factor receptors and enhanced downstream signaling [137]. Rab4 interacts with the N-myc–downregulated gene protein 1 (NDRG1) altering the kinetics of E-cadherin recycling and stability. In metastatic breast and prostate cancers, downregulation of NDRG1 is associated with poor prognosis [138] Rab5-mediates trafficking of Rac to the cell membrane during Rac-mediated actin assembly and cell migration as well as the internalization and recycling of β1 integrins increasing Rac-dependent cell migration [139, 140]. Thus the consequences of mutation of PIK3R1 in cancer may be mediated through aberrations in Rab function.

II. Special Focus on the Rab11 Family

Evidence suggests that long loop recycling members (Rab11A, Rab11B, Rab11C/Rab25 and its effectors) play a particularly important role in cancers of multiple lineages, including breast, colon, lung, ovarian, renal, endometrial, prostate, bladder and carcinoid types [5]. Tumor cells maintain high levels of incessant growth (RTK) and survival (integrins) signals by blocking steps of endocytosis that lead to degradation and favoring loops that promote recycling. Further, endosomes provide a compartment for the formation of active signaling complexes that does not occur at the cell surface [11, 141]. These complexes can signal within the cell or on return of the endosome to the cell surface. This can also be ligand specific with transforming growth factor alpha (TGFα) dissociating from the epidermal growth factor (EGF) receptor (EGFR) in the acidic environment of late endosomes allowing its prompt return to the plasma membrane by recycling endosomes, while the more stable EGF/EGFR complex is delivered to the lysosome compartment for degradation and attenuation of signaling [142]. Thus TGFα, by virtue of it recycling status, becomes a more potent mitogen that EGF. Detailed genomic and transcriptomic analysis of the frequent 1q22 amplicon in ovarian and breast cancer implicated the apical recycling Rab25 (aka Rab11c) as a likely driver of the amplicon [2, 4, 12]. Ectopic expression of Rab25 markedly increases tumorigenicity and metastatic potential in ovarian cancer cell models [12]. Rab25/Rab11 is also associated with aberrant growth factor and integrin recycling in 3D culture of ovarian cancer cells [141]. Subsequent to these studies, elevated levels of Rab25 have been observed in hepatocellular carcinoma, prostate cancer [143], transitional cell carcinoma of the bladder and testicular germ cell tumors [144].

Rab-coupling protein (RCP; also known as RAB11FIP1), an effector of Rab11 and Rab25, is a “driver” of the 8p11–12 amplicon in human breast cancer [145]. In mouse xenograft models of mammary carcinogenesis, RCP conferred aggressiveness to mammary tumors. Further overexpression of RCP in normal human mammary epithelial cells (MCF10A) conferred tumorigenic properties such as loss of contact inhibition, growth-factor independence and anchorage-independent growth [145], further supporting a role for Rab25 and its effectors in tumor initiation and progression.

Rab25 facilitates cell survival during stress including that induced by chemotherapy, hypoxia or anoikis at least in part by activation of the phosphoinositide-3 kinase pathway leading to AKT activation. Hypoxia can also stabilize microtubules by diminishing phosphorylation of Tau proteins facilitating Rab11–mediated trafficking of α5β6 via AKT activation and subsequent inhibition of glycogen syntheses kinase 3 beta [146]. Rab25 and Rab11 regulate α5β6 recycling and subsequent association with the EGFR leading to directed motility and invasion [11]. Overall, Rab25 through an interaction with AKT and integrins contributes prosurvival cues that coordinately allow cytoskeletal changes required for aggressive invasion through the extracellular matrix.

The effects of Rab25 may be conditional dependent on the cell lineage and the expression of effectors such as Rab11FIP1/RCP. For example, loss of Rab25 appears to confers aggressiveness to claudin low breast cancers [147] and Rab25 is present at low levels in Barrett esophagus [148] and in colorectal adenocarcinomas [149]. Further, deletion of Rab25 in mice promotes the development of intestinal neoplasia [150]. Together the data is most consistent with the function of Rab25 being context and cell type dependent and determined by the relative levels and activities of effectors molecules.

Why is Rab25, which is closely related to Rab11, more frequently implicated in tumorigenesis than Rab11? One possibility is that the sequence of GTP-binding site of Rab25 is DTAGLE instead of the DTAGQE found in Rab11 and most other Rab family members. This sequence is similar to the oncogenic Ras mutation (Q61L) that maintains a RAS in constitutively active form, which binds more efficiently to effectors.

III. Altered Developmental Pathways

The diverse roles that SHH, WNT and Notch signals play during the development and maintenance of normal tissues are recapitulated in different forms of cancer, and evidence supports an interesting role for Rab GTPases in their function and regulation. In Xenopus, the noncanonical WNT pathway requires Rab function, while in Drosophila a Rab6 homolog is essential for Notch signaling. In mammalian systems, Rab11-mediated recycling of the Notch effector Delta is critical for its activity, abnormalities in the function of Rab11 are implicated in both developmental defects and cancer [151].

In basal cell carcinoma and skin cancer [152] cancers of the prostate [153], stomach [154] and pancreas [155]; and juvenile medulloblastomas, SHH signaling is aberrant. Interestingly, Rab23 appears to be a key inhibitor of SHH signaling during development [156158] affecting an as yet undefined step between Smoothened and Gli-transcription factors [159]. Analysis of Gli3 protein suggests that Rab23 also has a role in promoting production of the Gli3 repressor. Blockade of SHH signaling leads to tumor shrinkage and remission in preclinical tumor xenograft models. Thus Rab GTPases present a future therapeutic biomarker for various cancers with alterations in this pathway.

3. Concluding Remarks

The role of aberrations in the Rab family in monogenic as well as complex diseases is just beginning to be elucidated. We have provided examples of where aberrant Rab function is both a target for therapy and a biomarker predicting clinical outcomes. We expect that a concerted systems approach to the role of vesicle function including endosome recycling will extend the relevance of derailed endocytosis to the pathophysiology of many diseases including complex diseases such as diabetes, obesity and cancer. While it has proven difficult to selectively target RAS superfamily members with current therapeutic approaches, a number of new methods to “target the untargetable” such as siRNA and stapled peptides as well as the potential to selectively target Rab effectors raises the exciting potential that targeting the function of Rab proteins in regulating vesicle function will eventually enter the therapeutic lexicon.

Acknowledgments

This study is supported by the Ovarian Cancer Research Fund, Cancer Center Support Grant P30CA16672, PO1CA099031 to GBM and by Susan G. Komen Post Doctoral Fellowship for Breast Cancer Research.

Abbreviations

ER

Endoplasmic Reticulum

EEA

Early Endosome Antigen

GDP

Guanosine Diphosphate

GDI

GDP Dissociation Inhibitor

GEF

Guanine Exchange Factor

GAP

GTPase Activating Protein

Glut4

Glucose Transporter 4

GTP

Guanosine Triphosphate

MAPK

Mitogen Activated Protein Kinase

PI3K

Phosphoinositide-3 Kinase

RAB

Ras in the Brain

REP

Rab Escort Protein

RTK

Receptor Tyrosine Kinase

SNARE

Soluble NSF Attachment Receptors

SHH

Sonic Hedgehog

TGN

Trans-Golgi Network

References

  • 1.Pereira-Leal JB, Seabra MC. Evolution of the rab family of small GTP-binding proteins. Journal of Molecular Biology. 2001;313(4):889–901. doi: 10.1006/jmbi.2001.5072. [DOI] [PubMed] [Google Scholar]
  • 2.Cheng KW, et al. Emerging Role of RAB GTPases in Cancer and Human Disease. Cancer Research. 2005;65(7):2516–2519. doi: 10.1158/0008-5472.CAN-05-0573. [DOI] [PubMed] [Google Scholar]
  • 3.Stein MP, Dong J, Wandinger-Ness A. Rab proteins and endocytic trafficking: potential targets for therapeutic intervention. Advanced Drug Delivery Reviews. 2003;55(11):1421–1437. doi: 10.1016/j.addr.2003.07.009. [DOI] [PubMed] [Google Scholar]
  • 4.Mills GB, et al. Genomic amplicons target vesicle recycling in breast cancer. The Journal of Clinical Investigation. 2009;119(8):2123–2127. doi: 10.1172/JCI40256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Mosesson Y, Mills GB, Yarden Y. Derailed endocytosis: an emerging feature of cancer. Nat Rev Cancer. 2008;8(11):835–850. doi: 10.1038/nrc2521. [DOI] [PubMed] [Google Scholar]
  • 6.Sorkin A, von Zastrow M. Endocytosis and signalling: intertwining molecular networks. Nat Rev Mol Cell Biol. 2009;10(9):609–622. doi: 10.1038/nrm2748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Agarwal R, et al. The Emerging Role of the RAB25 Small GTPase in Cancer. Traffic. 2009;10(11):1561–1568. doi: 10.1111/j.1600-0854.2009.00969.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Gissen P, Maher ER. Cargos and genes: insights into vesicular transport from inherited human disease. Journal of Medical Genetics. 2007;44:545–555. doi: 10.1136/jmg.2007.050294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Novick P, Brennwald P. Friends and family: The role of the rab GTPases in vesicular traffic. Cell. 1993;75(4):597–601. doi: 10.1016/0092-8674(93)90478-9. [DOI] [PubMed] [Google Scholar]
  • 10.Polo S, Pece S, Di Fiore PP. Endocytosis and cancer. Current Opinion in Cell Biology. 2004;16(2):156–161. doi: 10.1016/j.ceb.2004.02.003. [DOI] [PubMed] [Google Scholar]
  • 11.Caswell PT, et al. Rab25 Associates with α5β1 Integrin to Promote Invasive Migration in 3D Microenvironments. 2007;13(4):496–510. doi: 10.1016/j.devcel.2007.08.012. [DOI] [PubMed] [Google Scholar]
  • 12.Cheng KW, et al. The RAB25 small GTPase determines aggressiveness of ovarian and breast cancers. Nat Med. 2004;10(11):1251–6. doi: 10.1038/nm1125. [DOI] [PubMed] [Google Scholar]
  • 13.Lanzetti L, Fiore PPD. Endocytosis and Cancer: an ‘Insider’ Network with Dangerous Liaisons. Traffic. 2008;9(12):2011–2021. doi: 10.1111/j.1600-0854.2008.00816.x. [DOI] [PubMed] [Google Scholar]
  • 14.Wennerberg K, Rossman KL, Der CJ. The Ras superfamily at a glance. J Cell Sci. 2005;118(5):843–846. doi: 10.1242/jcs.01660. [DOI] [PubMed] [Google Scholar]
  • 15.Pereira-Leal JB, Seabra MC. The mammalian Rab family of small GTPases: definition of family and subfamily sequence motifs suggests a mechanism for functional specificity in the Ras superfamily. Journal of Molecular Biology. 2000;301(4):1077–1087. doi: 10.1006/jmbi.2000.4010. [DOI] [PubMed] [Google Scholar]
  • 16.Jiang SY, Ramachandran S. Comparative and evolutionary analysis of genes encoding small GTPases and their activating proteins in eukaryotic genomes. Physiol Genomics. 2006;24(3):235–251. doi: 10.1152/physiolgenomics.00210.2005. [DOI] [PubMed] [Google Scholar]
  • 17.Alory C, Balch WE, Edward AD. Handbook of Cell Signaling. Academic Press; Burlington: 2003. Molecular and Structural Organization of Rab GTPase Trafficking Networks; pp. 689–693. [Google Scholar]
  • 18.Zerial M, McBride H. Rab proteins as membrane organizers. Nat Rev Mol Cell Biol. 2001;2(2):107–117. doi: 10.1038/35052055. [DOI] [PubMed] [Google Scholar]
  • 19.Somsel Rodman J, Wandinger-Ness A. Rab GTPases coordinate endocytosis. J Cell Sci. 2000;113(2):183–192. doi: 10.1242/jcs.113.2.183. [DOI] [PubMed] [Google Scholar]
  • 20.Stenmark H, Olkkonen VM. The Rab GTPase family. Genome Biol. 2001;2(5):REVIEWS3007. doi: 10.1186/gb-2001-2-5-reviews3007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fehrenbacher N, Bar-Sagi D, Philips M. Ras/MAPK signaling from endomembranes. Molecular Oncology. 2009;3(4):297–307. doi: 10.1016/j.molonc.2009.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhang J, et al. Rab35 Controls Actin Bundling by Recruiting Fascin as an Effector Protein. Science. 2009;325(5945):1250–1254. doi: 10.1126/science.1174921. [DOI] [PubMed] [Google Scholar]
  • 23.Angers CG, Merz AJ. New links between vesicle coats and Rab-mediated vesicle targeting. Seminars in Cell & Developmental Biology. 2010 doi: 10.1016/j.semcdb.2010.07.003. In Press, Uncorrected Proof. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Barysch SV, et al. Sorting in early endosomes reveals connections to docking- and fusion-associated factors. Proceedings of the National Academy of Sciences. 2009;106(24):9697–9702. doi: 10.1073/pnas.0901444106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Luzio JP, et al. The delivery of endocytosed cargo to lysosomes. Biochemical Society Transactions. 2009;037(5):1019–1021. doi: 10.1042/BST0371019. [DOI] [PubMed] [Google Scholar]
  • 26.Hendrix A, et al. Effect of the Secretory Small GTPase Rab27B on Breast Cancer Growth, Invasion, and Metastasis. J Natl Cancer Inst. 2010;102(12):866–880. doi: 10.1093/jnci/djq153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Collins RN. Rab and ARF GTPase regulation of exocytosis (Review) Molecular Membrane Biology. 2003;20(2):105–115. doi: 10.1080/0968768031000085892. [DOI] [PubMed] [Google Scholar]
  • 28.Seabra MC, Wasmeier C. Controlling the location and activation of Rab GTPases. Current Opinion in Cell Biology. 2004;16(4):451–457. doi: 10.1016/j.ceb.2004.06.014. [DOI] [PubMed] [Google Scholar]
  • 29.Gomes AQ, et al. Membrane Targeting of Rab GTPases Is Influenced by the Prenylation Motif. Mol Biol Cell. 2003;14(5):1882–1899. doi: 10.1091/mbc.E02-10-0639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wu YW, et al. Membrane targeting mechanism of Rab GTPases elucidated by semisynthetic protein probes. Nat Chem Biol. 2010;6(7):534–540. doi: 10.1038/nchembio.386. [DOI] [PubMed] [Google Scholar]
  • 31.Leung KF, et al. Rab GTPases Containing a CAAX Motif Are Processed Post-geranylgeranylation by Proteolysis and Methylation. Journal of Biological Chemistry. 2007;282(2):1487–1497. doi: 10.1074/jbc.M605557200. [DOI] [PubMed] [Google Scholar]
  • 32.Wu YW, et al. Interaction analysis of prenylated Rab GTPase with Rab escort protein and GDP dissociation inhibitor explains the need for both regulators. Proceedings of the National Academy of Sciences. 2007;104(30):12294–12299. doi: 10.1073/pnas.0701817104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Pereira-Leal JB, et al. Structural determinants of Rab and Rab Escort Protein interaction: Rab family motifs define a conserved binding surface. Biochemical and Biophysical Research Communications. 2003;301(1):92–97. doi: 10.1016/s0006-291x(02)02963-7. [DOI] [PubMed] [Google Scholar]
  • 34.Alory C, Balch WE. Organization of the Rab-GDI/CHM Superfamily: The Functional Basis for Choroideremia Disease. Traffic. 2001;2(8):532–543. doi: 10.1034/j.1600-0854.2001.20803.x. [DOI] [PubMed] [Google Scholar]
  • 35.Ullrich O, et al. Membrane association of Rab5 mediated by GDP-dissociation inhibitor and accompanied by GDP/GTP exchange. Nature. 1994;368(6467):157–60. doi: 10.1038/368157a0. [DOI] [PubMed] [Google Scholar]
  • 36.Barnekow A, et al. International Review of Cell and Molecular Biology. Academic Press; 2009. Chapter 5 Rab Proteins and Their Interaction Partners; pp. 235–274. [DOI] [PubMed] [Google Scholar]
  • 37.Olchowik M, Miaczynska M. Effectors of GTPase Rab5 in endocytosis and signal transduction. Postepy Biochem. 2009;55(2):171–80. [PubMed] [Google Scholar]
  • 38.Díaz E, Pfeffer SR. TIP47: A Cargo Selection Device for Mannose 6-Phosphate Receptor Trafficking. Cell. 1998;93(3):433–443. doi: 10.1016/s0092-8674(00)81171-x. [DOI] [PubMed] [Google Scholar]
  • 39.Carroll KS, et al. Role of Rab9 GTPase in Facilitating Receptor Recruitment by TIP47. Science. 2001;292(5520):1373–1376. doi: 10.1126/science.1056791. [DOI] [PubMed] [Google Scholar]
  • 40.Ohya T, et al. Reconstitution of Rab- and SNARE-dependent membrane fusion by synthetic endosomes. Nature. 2009;459(7250):1091–1097. doi: 10.1038/nature08107. [DOI] [PubMed] [Google Scholar]
  • 41.Wickner W. Membrane Fusion: Five Lipids, Four SNAREs, Three Chaperones, Two Nucleotides, and a Rab, All Dancing in a Ring on Yeast Vacuoles. Annual Review of Cell and Developmental Biology. 2010;26(1) doi: 10.1146/annurev-cellbio-100109-104131. [DOI] [PubMed] [Google Scholar]
  • 42.Kwok MCM, et al. Proteomics of Photoreceptor Outer Segments Identifies a Subset of SNARE and Rab Proteins Implicated in Membrane Vesicle Trafficking and Fusion. Molecular & Cellular Proteomics. 2008;7(6):1053–1066. doi: 10.1074/mcp.M700571-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Woodman PG. Biogenesis of the Sorting Endosome: The Role of Rab5. Traffic. 2000;1(9):695–701. doi: 10.1034/j.1600-0854.2000.010902.x. [DOI] [PubMed] [Google Scholar]
  • 44.Christoforidis S, et al. The Rab5 effector EEA1 is a core component of endosome docking. Nature. 1999;397(6720):621–625. doi: 10.1038/17618. [DOI] [PubMed] [Google Scholar]
  • 45.Fukuda M, Kuroda TS, Mikoshiba K. Slac2-a/Melanophilin, the Missing Link between Rab27 and Myosin Va. Journal of Biological Chemistry. 2002;277(14):12432–12436. doi: 10.1074/jbc.C200005200. [DOI] [PubMed] [Google Scholar]
  • 46.Hill E, Clarke M, Barr FA. The Rab6-binding kinesin, Rab6-KIFL, is required for cytokinesis. EMBO J. 2000;19(21):5711–5719. doi: 10.1093/emboj/19.21.5711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Opdam F, et al. The small GTPase Rab6B, a novel Rab6 subfamily member, is cell-type specifically expressed and localised to the Golgi apparatus. J Cell Sci. 2000;113(15):2725–2735. doi: 10.1242/jcs.113.15.2725. [DOI] [PubMed] [Google Scholar]
  • 48.De Renzis S, Sonnichsen B, Zerial M. Divalent Rab effectors regulate the sub-compartmental organization and sorting of early endosomes. Nat Cell Biol. 2002;4(2):124–133. doi: 10.1038/ncb744. [DOI] [PubMed] [Google Scholar]
  • 49.Deneka M, van der Sluijs P. ‘Rab’ing up endosomal membrane transport. Nat Cell Biol. 2002;4(2):E33–E35. doi: 10.1038/ncb0202-e33. [DOI] [PubMed] [Google Scholar]
  • 50.Fouraux MA, et al. rabip4′ Is an Effector of rab5 and rab4 and Regulates Transport through Early Endosomes. Mol Biol Cell. 2004;15(2):611–624. doi: 10.1091/mbc.E03-05-0343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Rink J, et al. Rab Conversion as a Mechanism of Progression from Early to Late Endosomes. Cell. 2005;122(5):735–749. doi: 10.1016/j.cell.2005.06.043. [DOI] [PubMed] [Google Scholar]
  • 52.Lee MTG, Mishra A, Lambright DG. Structural Mechanisms for Regulation of Membrane Traffic by Rab GTPases. Traffic. 2009;10(10):1377–1389. doi: 10.1111/j.1600-0854.2009.00942.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gurkan C, et al. Large-Scale Profiling of Rab GTPase Trafficking Networks: The Membrome. Mol Biol Cell. 2005;16(8):3847–3864. doi: 10.1091/mbc.E05-01-0062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Seabra MC, Mules EH, Hume AN. Rab GTPases, intracellular traffic and disease. Trends in Molecular Medicine. 2002;8(1):23–30. doi: 10.1016/s1471-4914(01)02227-4. [DOI] [PubMed] [Google Scholar]
  • 55.Pfeffer SR. Rab GTPases: specifying and deciphering organelle identity and function. Trends in Cell Biology. 2001;11(12):487–491. doi: 10.1016/s0962-8924(01)02147-x. [DOI] [PubMed] [Google Scholar]
  • 56.Spang A. Vesicle Transport: A Close Collaboration of Rabs and Effectors. Current Biology. 2004;14(1):R33–R34. doi: 10.1016/j.cub.2003.12.021. [DOI] [PubMed] [Google Scholar]
  • 57.Schluter OM, et al. A Complete Genetic Analysis of Neuronal Rab3 Function. J Neurosci. 2004;24(29):6629–6637. doi: 10.1523/JNEUROSCI.1610-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Izumi T. Physiological roles of Rab27 effectors in regulated exocytosis. Endocr J. 2007;54(5):649–57. doi: 10.1507/endocrj.kr-78. [DOI] [PubMed] [Google Scholar]
  • 59.Mizuno K, et al. Rab27b Regulates Mast Cell Granule Dynamics and Secretion. Traffic. 2007;8(7):883–892. doi: 10.1111/j.1600-0854.2007.00571.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hurk Jvd, et al. Molecular basis of choroideremia (CHM): Mutations involving the rab escort protein-1 (REP-1</I>) gene. Human Mutation. 1997;9(2):110–117. doi: 10.1002/(SICI)1098-1004(1997)9:2<110::AID-HUMU2>3.0.CO;2-D. [DOI] [PubMed] [Google Scholar]
  • 61.Bianchi V, et al. Cognitive impairment in Gdi1-deficient mice is associated with altered synaptic vesicle pools and short-term synaptic plasticity, and can be corrected by appropriate learning training. Hum Mol Genet. 2009;18(1):105–117. doi: 10.1093/hmg/ddn321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Giannandrea M, et al. Mutations in the Small GTPase Gene RAB39B Are Responsible for X-linked Mental Retardation Associated with Autism, Epilepsy, and Macrocephaly. The American Journal of Human Genetics. 2010;86(2):185–195. doi: 10.1016/j.ajhg.2010.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Anikster Y, et al. Evidence that Griscelli Syndrome with Neurological Involvement Is Caused by Mutations in RAB27A, Not MYO5A. The American Journal of Human Genetics. 2002;71(2):407–414. doi: 10.1086/341606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Vincent LM, et al. Novel 47.5-kb deletion in RAB27A results in severe Griscelli Syndrome Type 2. Molecular Genetics and Metabolism. 2010 doi: 10.1016/j.ymgme.2010.05.015. In Press, Corrected Proof. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Gele MV, Dynoodt P, Lambert J. Griscelli syndrome: a model system to study vesicular trafficking. Pigment Cell & Melanoma Research. 2009;22(3):268–282. doi: 10.1111/j.1755-148X.2009.00558.x. [DOI] [PubMed] [Google Scholar]
  • 66.Vancoillie G, et al. Cytoplasmic dynein colocalizes with melanosomes in normal human melanocytes. British Journal of Dermatology. 2000;143(2):298–306. doi: 10.1046/j.1365-2133.2000.03654.x. [DOI] [PubMed] [Google Scholar]
  • 67.VANCOILLIE G, et al. Colocalization of Dynactin Subunits P150, Glued</i> and P50 with Melasomes inrmal Human Melacytes. Pigment Cell Research. 2000;13(6):449–457. doi: 10.1034/j.1600-0749.2000.130607.x. [DOI] [PubMed] [Google Scholar]
  • 68.Goud B. How Rab proteins link motors to membranes. Nat Cell Biol. 2002;4(4):E77–E78. doi: 10.1038/ncb0402-e77. [DOI] [PubMed] [Google Scholar]
  • 69.Westbroek W, et al. Interactions of Human Myosin Va Isoforms, Endogenously Expressed in Human Melanocytes, Are Tightly Regulated by the Tail Domain. J Investig Dermatol. 2003;120(3):465–475. doi: 10.1046/j.1523-1747.2003.12068.x. [DOI] [PubMed] [Google Scholar]
  • 70.Coudrier E. Myosins in melanocytes: to move or not to move? Pigment Cell Research. 2007;20(3):153–160. doi: 10.1111/j.1600-0749.2007.00376.x. [DOI] [PubMed] [Google Scholar]
  • 71.Bahadoran P, et al. Rab27a: A key to melanosome transport in human melanocytes. J Cell Biol. 2001;152(4):843–50. doi: 10.1083/jcb.152.4.843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Nascimento AAC, et al. Subcellular Localization of Myosin-V in the B16 Melanoma Cells, a Wild-type Cell Line for the dilute Gene. Mol Biol Cell. 1997;8(10):1971–1988. doi: 10.1091/mbc.8.10.1971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Menasche G, et al. Mutations in RAB27A cause Griscelli syndrome associated with haemophagocytic syndrome. Nat Genet. 2000;25:173–176. doi: 10.1038/76024. [DOI] [PubMed] [Google Scholar]
  • 74.Osanai K, et al. Altered lung surfactant system in a Rab38-deficient rat model of Hermansky-Pudlak syndrome. Am J Physiol Lung Cell Mol Physiol. 2010;298(2):L243–251. doi: 10.1152/ajplung.00242.2009. [DOI] [PubMed] [Google Scholar]
  • 75.Osanai K, et al. Methods in Enzymology. Academic Press; 2008. Analysis and Expression of Rab38 in Oculocutaneous Lung Disease; pp. 203–215. [DOI] [PubMed] [Google Scholar]
  • 76.Corbeel L, Freson K. Rab proteins and Rab-associated proteins: major actors in the mechanism of protein-trafficking disorders. Eur J Pediatr. 2008;167(7):723–9. doi: 10.1007/s00431-008-0740-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Spinosa MR, et al. Functional Characterization of Rab7 Mutant Proteins Associated with Charcot-Marie-Tooth Type 2B Disease. J Neurosci. 2008;28(7):1640–1648. doi: 10.1523/JNEUROSCI.3677-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Cogli L, Piro F, Bucci C. Rab7 and the CMT2B disease. Biochemical Society Transactions. 2009;037(5):1027–1031. doi: 10.1042/BST0371027. [DOI] [PubMed] [Google Scholar]
  • 79.Klein CJ, et al. SPTLC1 and RAB7 mutation analysis in dominantly inherited and idiopathic sensory neuropathies. Journal of Neurology, Neurosurgery & Psychiatry. 2005;76:1022–1024. doi: 10.1136/jnnp.2004.050062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Dawkins JL, et al. Exclusion of serine palmitoyltransferase long chain base subunit 2 (SPTLC2) as a common cause for hereditary sensory neuropathy. Neuromuscular Disorders. 2002;12(7–8):656–658. doi: 10.1016/s0960-8966(02)00015-9. [DOI] [PubMed] [Google Scholar]
  • 81.Rotthier A, et al. Genes for hereditary sensory and autonomic neuropathies: a genotype-phenotype correlation. Brain. 2009;132(10):2699–2711. doi: 10.1093/brain/awp198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Sakane A, et al. Methods in Enzymology. Academic Press; 2008. Analysis on the Emerging Role of Rab3 GTPase[hyphen (true graphic)]Activating Protein in Warburg Micro and Martsolf Syndrome; pp. 131–139. [DOI] [PubMed] [Google Scholar]
  • 83.Oishi H, Sasaki T, Takai Y. Interaction of Both the C2A and C2B Domains of Rabphilin3 with Ca2+and Phospholipid. Biochemical and Biophysical Research Communications. 1996;229(2):498–503. doi: 10.1006/bbrc.1996.1833. [DOI] [PubMed] [Google Scholar]
  • 84.Schluter OM, et al. Rab3 Superprimes Synaptic Vesicles for Release: Implications for Short-Term Synaptic Plasticity. J Neurosci. 2006;26(4):1239–1246. doi: 10.1523/JNEUROSCI.3553-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Aligianis IA, et al. Mutations of the catalytic subunit of RAB3GAP cause Warburg Micro syndrome. Nat Genet. 2005;37(3):221–3. doi: 10.1038/ng1517. [DOI] [PubMed] [Google Scholar]
  • 86.Flinn RJ, Backer JM. mTORC1 signals from late endosomes: Taking a TOR of the endocytic system. Cell Cycle. 9(10) doi: 10.4161/cc.9.10.11679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Flinn RJ, et al. The late endosome is essential for mTORC1 signaling. Mol Biol Cell. 2010;21(5):833–41. doi: 10.1091/mbc.E09-09-0756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Davies DM, et al. Sirolimus therapy in tuberous sclerosis or sporadic lymphangioleiomyomatosis. N Engl J Med. 2008;358(2):200–3. doi: 10.1056/NEJMc072500. [DOI] [PubMed] [Google Scholar]
  • 89.Bissler JJ, et al. Sirolimus for angiomyolipoma in tuberous sclerosis complex or lymphangioleiomyomatosis. N Engl J Med. 2008;358(2):140–51. doi: 10.1056/NEJMoa063564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Alessandri JL, et al. RAB2 mutation in a large family from Comoros Islands with Carpenter syndrome. American Journal of Medical Genetics Part A. 2010;152A(4):982–986. doi: 10.1002/ajmg.a.33327. [DOI] [PubMed] [Google Scholar]
  • 91.Wang Y, Ng EL, Tang BL. Rab23: What Exactly Does it Traffic? Traffic. 2006;7(6):746–750. doi: 10.1111/j.1600-0854.2006.00416.x. [DOI] [PubMed] [Google Scholar]
  • 92.Sobo K, et al. Late Endosomal Cholesterol Accumulation Leads to Impaired Intra-Endosomal Trafficking. PLoS ONE. 2007;2(9):e851. doi: 10.1371/journal.pone.0000851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Karten B, Peake KB, Vance JE. Mechanisms and consequences of impaired lipid trafficking in Niemann-Pick type C1-deficient mammalian cells. Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 2009;1791(7):659–670. doi: 10.1016/j.bbalip.2009.01.025. [DOI] [PubMed] [Google Scholar]
  • 94.Kodam A, et al. Altered levels and distribution of amyloid precursor protein and its processing enzymes in Niemann-Pick type C1-deficient mouse brains. Glia. 2010;58(11):1267–1281. doi: 10.1002/glia.21001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Ginsberg SD, et al. Microarray Analysis of Hippocampal CA1 Neurons Implicates Early Endosomal Dysfunction During Alzheimer’s Disease Progression. Biological Psychiatry. 2010 doi: 10.1016/j.biopsych.2010.05.030. In Press, Corrected Proof. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Hyvola N, et al. Membrane targeting and activation of the Lowe syndrome protein OCRL1 by rab GTPases. EMBO J. 2006;25(16):3750–3761. doi: 10.1038/sj.emboj.7601274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ungewickell A, et al. The inositol polyphosphate 5-phosphatase Ocrl associates with endosomes that are partially coated with clathrin. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(37):13501–13506. doi: 10.1073/pnas.0405664101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Choudhury R, et al. Lowe Syndrome Protein OCRL1 Interacts with Clathrin and Regulates Protein Trafficking between Endosomes and the Trans-Golgi Network. Mol Biol Cell. 2005;16(8):3467–3479. doi: 10.1091/mbc.E05-02-0120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.BASKYS A, et al. Rab-Mediated Endocytosis. Annals of the New York Academy of Sciences, 2007; Neuroprotective Agents: Eighth International Neuroprotection Society Meeting; pp. 313–329. [DOI] [PubMed] [Google Scholar]
  • 100.Topp JD, et al. Alsin Is a Rab5 and Rac1 Guanine Nucleotide Exchange Factor. Journal of Biological Chemistry. 2004;279(23):24612–24623. doi: 10.1074/jbc.M313504200. [DOI] [PubMed] [Google Scholar]
  • 101.Kunita R, et al. The Rab5 Activator ALS2/alsin Acts as a Novel Rac1 Effector through Rac1-activated Endocytosis. Journal of Biological Chemistry. 2007;282(22):16599–16611. doi: 10.1074/jbc.M610682200. [DOI] [PubMed] [Google Scholar]
  • 102.Pal A, et al. Huntingtin-HAP40 complex is a novel Rab5 effector that regulates early endosome motility and is up-regulated in Huntington’s disease. The Journal of Cell Biology. 2006;172(4):605–618. doi: 10.1083/jcb.200509091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Pal A, et al. Methods in Enzymology. Academic Press; 2008. Regulation of Endosome Dynamics by Rab5 and Huntingtin[hyphen (true graphic)]HAP40 Effector Complex in Physiological versus Pathological Conditions; pp. 239–257. [DOI] [PubMed] [Google Scholar]
  • 104.Li X, et al. Aberrant Rab11-Dependent Trafficking of the Neuronal Glutamate Transporter EAAC1 Causes Oxidative Stress and Cell Death in Huntington’s Disease. J Neurosci. 30(13):4552–4561. doi: 10.1523/JNEUROSCI.5865-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Cooper AA, et al. {alpha}-Synuclein Blocks ER-Golgi Traffic and Rab1 Rescues Neuron Loss in Parkinson’s Models. Science. 2006;313(5785):324–328. doi: 10.1126/science.1129462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lashuel HA, Hirling H. Rescuing Defective Vesicular Trafficking Protects against α-Synuclein Toxicity in Cellular and Animal Models of Parkinson’s Disease. ACS Chemical Biology. 2006;1(7):420–424. doi: 10.1021/cb600331e. [DOI] [PubMed] [Google Scholar]
  • 107.Gitler AD, et al. The Parkinson’s disease protein α-synuclein disrupts cellular Rab homeostasis. Proceedings of the National Academy of Sciences. 2008;105(1):145–150. doi: 10.1073/pnas.0710685105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Saxena SK, Kaur S. Regulation of epithelial ion channels by Rab GTPases. Biochemical and Biophysical Research Communications. 2006;351(3):582–587. doi: 10.1016/j.bbrc.2006.10.087. [DOI] [PubMed] [Google Scholar]
  • 109.Silvis MR, et al. Rab11b Regulates the Apical Recycling of the Cystic Fibrosis Transmembrane Conductance Regulator in Polarized Intestinal Epithelial Cells. Mol Biol Cell. 2009;20(8):2337–2350. doi: 10.1091/mbc.E08-01-0084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Ohira M, et al. Dislocation of Rab13 and vasodilator-stimulated phosphoprotein in inactive colon epithelium in patients with Crohn’s disease. Int J Mol Med. 2009;24(6):829–35. doi: 10.3892/ijmm_00000300. [DOI] [PubMed] [Google Scholar]
  • 111.Prada-Delgado A, et al. Inhibition of Rab5a Exchange Activity Is a Key Step for Listeria monocytogenes Survival. Traffic. 2005;6(3):252–265. doi: 10.1111/j.1600-0854.2005.00265.x. [DOI] [PubMed] [Google Scholar]
  • 112.Brumell JH, Scidmore MA. Manipulation of Rab GTPase Function by Intracellular Bacterial Pathogens. Microbiol Mol Biol Rev. 2007;71(4):636–652. doi: 10.1128/MMBR.00023-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Kyei GB, et al. Rab14 is critical for maintenance of Mycobacterium tuberculosis phagosome maturation arrest. EMBO J. 2006;25(22):5250–5259. doi: 10.1038/sj.emboj.7601407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Rzomp KA, et al. Rab GTPases Are Recruited to Chlamydial Inclusions in Both a Species-Dependent and Species-Independent Manner. Infect Immun. 2003;71(10):5855–5870. doi: 10.1128/IAI.71.10.5855-5870.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Cortes C, et al. Chlamydia pneumoniae Inclusion Membrane Protein Cpn0585 Interacts with Multiple Rab GTPases. Infect Immun. 2007;75(12):5586–5596. doi: 10.1128/IAI.01020-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Batista DGJ, et al. Trypanosoma cruzi Modulates the Expression of Rabs and Alters the Endocytosis in Mouse Cardiomyocytes In Vitro. J Histochem Cytochem. 2006;54(6):605–614. doi: 10.1369/jhc.5A6654.2005. [DOI] [PubMed] [Google Scholar]
  • 117.Sklan EH, et al. A Rab-GAP TBC Domain Protein Binds Hepatitis C Virus NS5A and Mediates Viral Replication. J Virol. 2007;81(20):11096–11105. doi: 10.1128/JVI.01249-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kaddai V, et al. Rab4b Is a Small GTPase Involved in the Control of the Glucose Transporter GLUT4 Localization in Adipocyte. PLoS ONE. 2009;4(4):e5257. doi: 10.1371/journal.pone.0005257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Mukhopadhyay A, et al. An endocytic pathway as a target of tubby for regulation of fat storage. EMBO Rep. 2007;8(10):931–938. doi: 10.1038/sj.embor.7401055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Bryant NJ, Govers R, James DE. Regulated transport of the glucose transporter GLUT4. Nat Rev Mol Cell Biol. 2002;3(4):267–277. doi: 10.1038/nrm782. [DOI] [PubMed] [Google Scholar]
  • 121.Ishikura S, Koshkina A, Klip A. Small G proteins in insulin action: Rab and Rho families at the crossroads of signal transduction and GLUT4 vesicle traffic. Acta Physiologica. 2008;192(1):61–74. doi: 10.1111/j.1748-1716.2007.01778.x. [DOI] [PubMed] [Google Scholar]
  • 122.Zaid H, et al. Insulin action on glucose transporters through molecular switches, tracks and tethers. Biochem J. 2008;413(2):201–215. doi: 10.1042/BJ20080723. [DOI] [PubMed] [Google Scholar]
  • 123.Schwenk RW, Eckel J. A novel method to monitor insulin-stimulated GTP-loading of Rab11a in cardiomyocytes. Cellular Signalling. 2007;19(4):825–830. doi: 10.1016/j.cellsig.2006.10.008. [DOI] [PubMed] [Google Scholar]
  • 124.Schwenk RW, Luiken JJFP, Eckel J. FIP2 and Rip11 specify Rab11a-mediated cellular distribution of GLUT4 and FAT/CD36 in H9c2-hIR cells. Biochemical and Biophysical Research Communications. 2007;363(1):119–125. doi: 10.1016/j.bbrc.2007.08.111. [DOI] [PubMed] [Google Scholar]
  • 125.Wang Z, Thurmond DC. Mechanisms of biphasic insulin-granule exocytosis - roles of the cytoskeleton, small GTPases and SNARE proteins. J Cell Sci. 2009;122(7):893–903. doi: 10.1242/jcs.034355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Kasai K, et al. Rab27a mediates the tight docking of insulin granules onto the plasma membrane during glucose stimulation. The Journal of Clinical Investigation. 2005;115(2):388–396. doi: 10.1172/JCI22955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Hou Q, et al. Integrative Genomics Identifies RAB23 as an Invasion Mediator Gene in Diffuse-Type Gastric Cancer. Cancer Research. 2008;68(12):4623–4630. doi: 10.1158/0008-5472.CAN-07-5870. [DOI] [PubMed] [Google Scholar]
  • 128.Jäger D, et al. Serological Cloning of a Melanocyte rab Guanosine 5′-Triphosphate-binding Protein and a Chromosome Condensation Protein from a Melanoma Complementary DNA Library. Cancer Research. 2000;60(13):3584–3591. [PubMed] [Google Scholar]
  • 129.Shimada K, et al. Aberrant expression of RAB1A in human tongue cancer. Br J Cancer. 2005;92(10):1915–1921. doi: 10.1038/sj.bjc.6602594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Ni X, et al. Molecular cloning and characterization of a novel human Rab (Rab2B) gene. J Hum Genet. 2002;47(10):548–551. doi: 10.1007/s100380200083. [DOI] [PubMed] [Google Scholar]
  • 131.Laezza C, et al. Control of Rab5 and Rab7 Expression by the Isoprenoid Pathway. Biochemical and Biophysical Research Communications. 1998;248(3):469–472. doi: 10.1006/bbrc.1998.9007. [DOI] [PubMed] [Google Scholar]
  • 132.Johnson JL, et al. The Rab27a-binding protein, JFC1, regulates androgen-dependent secretion of prostate-specific antigen and prostatic-specific acid phosphatase. Biochem J. 2005;391(3):699–710. doi: 10.1042/BJ20050380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Catz SD, William CJD, Balch E, Alan H. Methods in Enzymology. Academic Press; 2008. Characterization of Rab27a and JFC1 as Constituents of the Secretory Machinery of Prostate[hyphen (true graphic)]Specific Antigen in Prostate Carcinoma Cells; pp. 25–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.He H, et al. Identification and characterization of nine novel human small GTPases showing variable expressions in liver cancer tissues. Gene Expr. 2002;10(5–6):231–42. doi: 10.3727/000000002783992406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Wang JS, et al. Enhanced Expression of Rab27A Gene by Breast Cancer Cells Promoting Invasiveness and the Metastasis Potential by Secretion of Insulin-Like Growth Factor-II. Molecular Cancer Research. 2008;6(3):372–382. doi: 10.1158/1541-7786.MCR-07-0162. [DOI] [PubMed] [Google Scholar]
  • 136.Kotzsch M, et al. Urokinase receptor splice variant uPAR-del4/5-associated gene expression in breast cancer: identification of rab31 as an independent prognostic factor. Breast Cancer Res Treat. 2008;111(2):229–40. doi: 10.1007/s10549-007-9782-6. [DOI] [PubMed] [Google Scholar]
  • 137.Chamberlain MD, et al. Deregulation of Rab5 and Rab4 proteins in p85R274A-expressing cells alters PDGFR trafficking. Cellular Signalling. 2010;22(10):1562–1575. doi: 10.1016/j.cellsig.2010.05.025. [DOI] [PubMed] [Google Scholar]
  • 138.Kachhap SK, et al. The N-Myc Down Regulated Gene1 (NDRG1) Is a Rab4a Effector Involved in Vesicular Recycling of E-Cadherin. PLoS ONE. 2007;2(9):e844. doi: 10.1371/journal.pone.0000844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Zech T, Machesky L. Rab5 and Rac Team Up in Cell Motility. Cell. 2008;134(1):18–20. doi: 10.1016/j.cell.2008.06.039. [DOI] [PubMed] [Google Scholar]
  • 140.Torres VA, et al. Caspase 8 Promotes Peripheral Localization and Activation of Rab5. Journal of Biological Chemistry. 2008;283(52):36280–36289. doi: 10.1074/jbc.M805878200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Caswell PT, et al. Rab-coupling protein coordinates recycling of α5β1 integrin and EGFR1 to promote cell migration in 3D microenvironments. The Journal of Cell Biology. 2008;183(1):143–155. doi: 10.1083/jcb.200804140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Roepstorff K, et al. Differential Effects of EGFR Ligands on Endocytic Sorting of the Receptor. Traffic. 2009;10(8):1115–1127. doi: 10.1111/j.1600-0854.2009.00943.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Calvo A, et al. Alterations in Gene Expression Profiles during Prostate Cancer Progression. Cancer Research. 2002;62(18):5325–5335. [PubMed] [Google Scholar]
  • 144.Korkola JE, et al. In vivo differentiation and genomic evolution in adult male germ cell tumors. Genes Chromosomes and Cancer. 2008;47(1):43–55. doi: 10.1002/gcc.20504. [DOI] [PubMed] [Google Scholar]
  • 145.Zhang J, et al. RCP is a human breast cancer-promoting gene with Ras-activating function. The Journal of Clinical Investigation. 2009;119(8):2171–2183. doi: 10.1172/JCI37622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Yoon SO, Shin S, Mercurio AM. Hypoxia Stimulates Carcinoma Invasion by Stabilizing Microtubules and Promoting the Rab11 Trafficking of the α6β4 Integrin. Cancer Research. 2005;65(7):2761–2769. doi: 10.1158/0008-5472.CAN-04-4122. [DOI] [PubMed] [Google Scholar]
  • 147.Cheng J-M, et al. Tumor suppressor function of Rab25 in triple-negative breast cancer. International Journal of Cancer. 126(12):2799–2812. doi: 10.1002/ijc.24900. [DOI] [PubMed] [Google Scholar]
  • 148.Goldenring JR, Ray GS, Lee JR. Rab11 in dysplasia of Barrett’s epithelia. Yale J Biol Med. 1999;72(2–3):113–20. [PMC free article] [PubMed] [Google Scholar]
  • 149.Nam KT, et al. Loss of Rab25 promotes the development of intestinal neoplasia in mice and is associated with human colorectal adenocarcinomas. The Journal of Clinical Investigation. 120(3):840–849. doi: 10.1172/JCI40728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Nam KT, et al. Loss of Rab25 promotes the development of intestinal neoplasia in mice and is associated with human colorectal adenocarcinomas. The Journal of Clinical Investigation. 2010;120(3):840–849. doi: 10.1172/JCI40728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Emery G, et al. Asymmetric Rab11 Endosomes Regulate Delta Recycling and Specify Cell Fate in the Drosophila Nervous System. Cell. 2005;122(5):763–773. doi: 10.1016/j.cell.2005.08.017. [DOI] [PubMed] [Google Scholar]
  • 152.Athar M, et al. Hedgehog signalling in skin development and cancer. Experimental Dermatology. 2006;15(9):667–677. doi: 10.1111/j.1600-0625.2006.00473.x. [DOI] [PubMed] [Google Scholar]
  • 153.Datta S, Datta MW. Sonic Hedgehog signaling in advanced prostate cancer. Cell Mol Life Sci. 2006;63(4):435–48. doi: 10.1007/s00018-005-5389-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Katoh Y, Katoh M. Hedgehog signaling pathway and gastrointestinal stem cell signaling network (review) Int J Mol Med. 2006;18(6):1019–23. [PubMed] [Google Scholar]
  • 155.Lau J, Kawahira H, Hebrok M. Hedgehog signaling in pancreas development and disease. Cell Mol Life Sci. 2006;63(6):642–52. doi: 10.1007/s00018-005-5357-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Evans TM, et al. Methods in Enzymology. Academic Press; 2005. Characterization of Rab23, a Negative Regulator of Sonic Hedgehog Signaling; pp. 759–777. [DOI] [PubMed] [Google Scholar]
  • 157.Evans TM, et al. Rab23, a Negative Regulator of Hedgehog Signaling, Localizes to the Plasma Membrane and the Endocytic Pathway. Traffic. 2003;4(12):869–884. doi: 10.1046/j.1600-0854.2003.00141.x. [DOI] [PubMed] [Google Scholar]
  • 158.Eggenschwiler JT, Espinoza E, Anderson KV. Rab23 is an essential negative regulator of the mouse Sonic hedgehog signalling pathway. Nature. 2001;412(6843):194–198. doi: 10.1038/35084089. [DOI] [PubMed] [Google Scholar]
  • 159.Eggenschwiler JT, et al. Mouse Rab23 regulates Hedgehog signaling from Smoothened to Gli proteins. Developmental Biology. 2006;290(1):1–12. doi: 10.1016/j.ydbio.2005.09.022. [DOI] [PubMed] [Google Scholar]

RESOURCES