Rab GTPases control intracellular membrane traffic by recruiting specific effector proteins to restricted membranes in a GTP-dependent manner. In this Cell Science at a Glance and the accompanying poster, we highlight the regulation of Rab GTPases by proteins that control their membrane association and activation state, and provide an overview of the cellular processes that are regulated by Rab GTPases and their effectors, including protein sorting, vesicle motility and vesicle tethering. We also discuss the physiological importance of Rab GTPases and provide examples of diseases caused by their dysfunctions.

Rab GTPases constitute the largest family of small GTPases (almost 70 members in humans) and are known as master regulators of intracellular membrane traffic (Stenmark, 2009; Wandinger-Ness and Zerial, 2014). Distinct Rab GTPases localize to different membrane compartments in order to control the specificity and directionality of membrane trafficking pathways, mostly related to vesicular transport. In doing so, they contribute to confer membrane identity (Pfeffer, 2013) and to ensuring that membrane-bound cargoes are transported to their correct destinations within the cell. For many human Rab GTPases several isoforms (structurally related proteins encoded by different genes) exist that perform partially redundant functions, either by working in slightly different ways or by being expressed differentially in different cell types. A useful web-based tool has recently been developed to identify Rab GTPases and classify them into subfamilies based on sequence (Diekmann et al., 2011). In this Cell Science at a Glance and the accompanying poster, we will provide a brief overview of the cellular functions of Rab GTPases with a focus on their regulators and effectors, and the pathways they control.

Like other small GTPases, Rab GTPases principally function as molecular switches that are ‘on’ when GTP is bound and ‘off’ when GDP is bound. Conformational differences between the GDP- and GTP-bound forms mainly involve two regions, termed switch I and switch II, which specifically interact with effector proteins when GTP is bound (Eathiraj et al., 2005). In the GDP-bound state, the switch regions appear to be unfolded, whereas they adopt well-defined conformations when GTP is bound, and this allows effector binding (Lee et al., 2009). Examples of structurally well-characterized Rab-effector interactions include those of Rab3a with rabphilin-3A (Ostermeier and Brunger, 1999), Rab5a with rabaptin-5 (Zhu et al., 2004), Rab4 and Rab22 with rabenosyn-5 (Eathiraj et al., 2005), Rab27a with Slp2-a (also known as Sytl2-a) (Chavas et al., 2008), Rab6a with Rab6IP1 (also known as DENND5A) (Recacha et al., 2009), and Rab11a with PI4KIIIβ (Burke et al., 2014).

As is the case for other small GTPases, the nucleotide cycle of Rab GTPases is tightly controlled by guanine-nucleotide-exchange factors (GEFs) and GTPase-activating proteins (GAPs) that are specific for single Rab GTPases or Rab subfamilies (see poster) (Barr and Lambright, 2010). GEFs mediate the activation of Rab GTPases by promoting the exchange of bound GDP with GTP, which is in large excess over GDP in cytosol. Different types of GEFs function in slightly different ways and have different catalytic domains, such as DENN and Vps9 domains. Whereas DENN domains appear to promote GDP release by forcing contacts between a lysine residue in the phosphate-binding ‘P loop’ and glutamine and aspartate residues in the switch II region, Vps9 domains (which are found in GEFs for the Rab5 subfamily) contribute an acidic residue that interacts with the P-loop lysine (Langemeyer et al., 2014). Rab GAPs turn Rab GTPases off by stimulating their ability to hydrolyze GTP into GDP. Most (but not all) Rab GAPs are distinguished by the presence of a TBC1 domain. Because many Rab GTPases, including Rab5, hydrolyze GTP at a relatively high intrinsic rate, Rab GAPs might be less crucial than GEFs in general (Barr and Lambright, 2010).

Rab effectors, defined as proteins that interact specifically with the GTP-bound from of a Rab GTPase, come in many flavours and include molecular tethers, fusion regulators, motors, sorting adaptors, kinases, phosphatases, components of membrane contact sites and Rab regulators (Gillingham et al., 2014). The recruitment of such effectors in a spatiotemporally controlled manner contributes strongly to the fidelity and specificity of intracellular membrane traffic. There are also a few examples of proteins that are regulated by GDP-bound Rabs or that interact with Rabs in a nucleotide-independent fashion, including the interactions between Rab21 and β1-integrin, Rab11 and protrudin, Rab7 and VPS34, and Rab27a and Coronin3 (Kimura et al., 2008; Pellinen et al., 2006; Shirane and Nakayama, 2006; Stein et al., 2003); however, the term ‘effector’ should be reserved for those proteins that interact exclusively with the GTP-bound form of a Rab GTPase. In some cases, different Rab GTPases bind to overlapping or non-overlapping sites on the same effector, such as the interaction of Rab4, Rab5 and Rab22 with rabenosyn-5, Rab4, Rab5 and Rab33 with rabaptin-5, Rab5 and Rab22 with EEA1, and Rab2, Rab6 and Rab39 with bicaudal-D (Eathiraj et al., 2005; Gillingham et al., 2014; Valsdottir et al., 2001; Vitale et al., 1998). There are also examples of Rab effectors that have Rab GAP or GEF activity, such as RUTBC1 and RUTBC2, which are effectors for Rab9 and GAPs for Rab32, Rab33 and Rab36 (Nottingham et al., 2011, 2012), the rabaptin-5–Rabex5 complex, which is an effector and GEF for Rab5 (Horiuchi et al., 1997), and the HOPS complex, an effector of Rab5 and a GEF for Rab7 (Nottingham et al., 2011, 2012; Rink et al., 2005). The resulting feed-forward loops in GEF activation are likely to promote the rapid membrane accumulation of Rab GTPases whereas feedback loops involving GAPs likewise promote their rapid removal. Together, these systems enable the promotion of time-limited membrane domains with unique compositions (Barr, 2013; Stenmark, 2009; Wandinger-Ness and Zerial, 2014).

Rab GTPases exist in both soluble and membrane-bound pools. Strong membrane association is ensured by posttranslational modification of C-terminal cysteine residues with one or (in most cases) two lipophilic geranylgeranyl groups (20-carbon isoprenoid groups; see poster). Geranylgeranylation is mediated by Rab geranylgeranyl transferase (GGTase II, also known as RABGGTB) in cooperation with Rab escort protein (REP, for which there are several isoforms) (Leung et al., 2006). The latter protein chaperones the newly geranylgeranylated Rab GTPase to its correct cellular membrane. A related protein, Rab GDP dissociation inhibitor (GDI, for which there are several isoforms), mediates dissociation of geranylgeranylated Rab GTPases from membranes and chaperones the hydrophobic conjugates in the cytosol. Rab GDI specifically recognizes Rabs in their GDP-bound form and thereby serves to solubilize Rabs from membranes once GTP hydrolysis has been completed (Goody et al., 2005). GDI also serves to present Rab GTPases to specific membranes (Soldati et al., 1994; Ullrich et al., 1994). This has been proposed to occur through a membrane-associated GDI displacement factor (GDF) that recognizes the Rab–GDI complex (Sivars et al., 2003). However, there is also evidence that a membrane-bound GEF is sufficient to lead to the accumulation of a Rab GTPase on a specific membrane (Schoebel et al., 2009), and in budding yeast, GEFs but not GDFs are required for membrane-targeting of Rab GTPases (Cabrera and Ungermann, 2013). This indicates that compartment-specific GEFs play a central role in defining the precise localization of Rab GTPases.

The fact that Rab effectors are highly diverse illustrates that Rab GTPases control multiple biochemical events (see poster). Most functions of Rab GTPases and their effectors are related to vesicular traffic between a donor and an acceptor compartment, and it is interesting to note that distinct Rab effectors are involved in the sorting of cargo into budding vesicles, vesicle uncoating or vesicle motility along actin filaments or microtubules, as well as vesicle tethering to acceptor membranes (Stenmark, 2009). Through these activities, Rab GTPases control compartment maturation, as well as vesicle shuttling between different membrane compartments. The first Rab GTPases identified through yeast genetics, Sec4 (Rab8) and Ypt1 (Rab1), were shown to have crucial roles in vesicle transport in the exocytic pathway (Hutagalung and Novick, 2011), but studies in multiple organisms have identified a number of Rab GTPases that also control endocytic membrane traffic (Wandinger-Ness and Zerial, 2014). Furthermore, mostly through their roles in vesicular transport, Rab GTPases also control specialized structures, such as primary cilia, lipid droplets, autophagosomes, focal adhesions, tight junctions and interorganeller membrane contact sites (see poster) (Deretic, 2013; Martin et al., 2005; Martin and Parton, 2008; Marzesco et al., 2002; Munafo and Colombo, 2002; Pellinen et al., 2006; Raiborg et al., 2015). Because membrane traffic impacts on numerous cellular functions, Rab GTPases are not only regulators of membrane traffic as such, but, indirectly they also control cell signaling, polarity, migration and division (De Franceschi et al., 2015; Militello and Colombo, 2013; Numrich and Ungermann, 2014; van IJzendoorn et al., 2003). This illustrates the wide functional repertoire and great importance of this GTPase family.

Given the importance of Rab GTPases in membrane traffic, and the importance of membrane traffic for human health (Olkkonen and Ikonen, 2000), it is perhaps surprising that only a limited number of genetic diseases are associated with Rab dysfunctions. The existence of multiple Rab isoforms and trafficking pathways presumably makes humans less vulnerable to mutations in individual Rab-encoding genes. Nevertheless, several mutations in Rab GTPases or their effectors or regulators are associated with genetic diseases (Seixas et al., 2013). Examples of genetic diseases associated with mutations in Rab GTPases include albinism, immunodeficiences, neuropathies, mental retardation and ciliopathies (see Table 1). Rab27a provides a good illustration of how mutations in a Rab GTPase or its effectors can cause genetic disease. This GTPase controls exocytosis of lysosome-related organelles, such as melanosomes (in melanocytes) or lytic granules (in natural killer and T cells), and mutations in the RAB27A gene cause a combined albinism and immunodeficiency syndrome known at Griscelli syndrome type 2 (Menasche et al., 2001). Interestingly, mutations in the gene encoding the Rab27a effector Munc13-4 (also known as UNC13D) in natural killer and T cells cause an immunodeficiency disease, familial hemophagocytic lymphohistiocytosis type 3 (Feldmann et al., 2003), and mutations in RAB27A that selectively affect binding to this effector cause a form of Griscelli syndrome without an albinism phenotype (Cetica et al., 2015). In melanocytes, the Rab27a effector melanophilin (also known as Slac2-a) functions as a motor adaptor that connects Rab27a-positive melanosomes to the actin motor myosin-5a to promote melanosome exocytosis, and mutations in the genes encoding melanophilin or Myosin-5a cause Griscelli syndrome variants with an albinism phenotype but not immunodeficiency (Menasche et al., 2003).

Table 1.

Rab GTPases and their regulators/effectors whose genes are mutated in genetic diseases

Rab GTPases and their regulators/effectors whose genes are mutated in genetic diseases
Rab GTPases and their regulators/effectors whose genes are mutated in genetic diseases

In addition to genetic mutations, dysregulation of Rab GTPases is also observed in cancers (Table 2). Typically, increased expression levels of certain Rab GTPases, such as Rab1a, Rab3a, Rab5a or Rab7a, are associated with progression of specific cancer types, although there are also examples suggesting that loss of Rab expression can drive cancers, as shown for Rab25 in colon cancer (Goldenring, 2013). An example of how increased Rab expression might drive cancer progression is provided by Rab5a, which is overexpressed in aggressive breast cancers. Together with Rab4 and the Rab5 and Rab4 effector rabenosyn-5, Rab5a drives an endocytic–exocytic cycle that is crucial for the formation of invadopodia, cancer cell protrusions that promote tissue invasions and metastasis (Frittoli et al., 2014).

Table 2.

Rab GTPases and cancer

Rab GTPases and cancer
Rab GTPases and cancer

Rab GTPases play a crucial role in phagocytosis and phagosome maturation, and as such they are important components of innate immunity (Flannagan et al., 2012). Many intracellular pathogens target Rab GTPases in order to interfere with the ability of the host cell to phagocytose and degrade pathogens. Frequent targets for bacterial effectors are Rab GTPases that reside on endosomes and phagosomes, such as Rab4, Rab5, Rab9, Rab11 and Rab22, but a number of Rab GTPases found in the endoplasmic reticulum (ER) and Golgi are also targeted by pathogenesis factors, including Rab1, Rab2, Rab6 and Rab8 (Sherwood and Roy, 2013). These bacterial pathogenesis factors can either act as Rab GEFs or GAPs that activate or inactivate specific Rab GTPases, or they can interfere with Rab functions through enzymatic modifications, such as lipidation, AMPylation or proteolytic cleavage, or by antagonizing Rab effectors. A typical outcome of such interference with Rab functions is that the pathogen achieves a remodeling of the endomembranes of the host, which enables it to evade destruction in the phagolysosome and establish a replicative niche within the host.

The specific localization of different Rab GTPases to defined membrane compartments, and their ability to regulate specific trafficking pathways, have made them attractive as research tools to study intracellular membrane transport. It must be noted that overexpression of various Rab GTPases frequently produces profound cellular phenotypes, such as changes in organelle and cell morphology, or cytoskeletal rearrangements (Bucci et al., 1992; Peranen et al., 1996); therefore, the use of transfected Rab GTPases as membrane markers is only advisable when expression levels are kept low. Likewise, dominant-negative and constitutively active Rab mutants have been frequently used to dissect membrane trafficking, but the interpretations of experiments that employ Rab mutants are complicated by the fact that similar mutations affect different Rab GTPases differentially. For instance, whereas an ‘activating’ mutation of the conserved glutamine residue in switch II in Rab5 inhibits its intrinsic but not GAP-stimulated GTP hydrolysis, a similar mutation in Rab1 and Rab35 inhibits GAP-stimulated but not intrinsic GTP hydrolysis. The same mutation also prevents GEF-mediated activation of Rab35 but not of Rab1 (Langemeyer et al., 2014). Thus, although Rab mutants are very useful research tools, conclusions that are based on soley their use should be made with caution.

Together with SNARE proteins that mediate specificity of vesicle docking and fusion (Sudhof and Rothman, 2009), Rab GTPases are central regulators of intracellular membrane traffic. The diversity of Rab GTPases and their effectors is consistent with the view that intracellular trafficking pathways are complex, and such complexity indeed started to evolve early in eukaryotic history (Diekmann et al., 2011; Klopper et al., 2012). In contrast to canonical SNARE proteins, which are irreversibly anchored to membranes by transmembrane segments, Rab GTPases shuttle between the cytosol and membranes, and this makes them well suited to define the directionality of vesicle transport processes (Stenmark, 2009). Our appreciation of the importance of Rab GTPases in cell biology and biomedicine is continuously increasing as we learn more about these proteins, their regulators and effectors.

Funding

The work in our laboratory is supported by the Norwegian Cancer Society, the South-Eastern Norway Regional Health Authority, and by the Research Council of Norway through its Centres of Excellence funding scheme [project number 179571].

Alonso-Curbelo
,
D.
,
Riveiro-Falkenbach
,
E.
,
Pérez-Guijarro
,
E.
,
Cifdaloz
,
M.
,
Karras
,
P.
,
Osterloh
,
L.
,
Megías
,
D.
,
Cañón
,
E.
,
Calvo
,
T. G.
,
Olmeda
,
D.
, et al. 
(
2014
).
RAB7 controls melanoma progression by exploiting a lineage-specific wiring of the endolysosomal pathway
.
Cancer Cell
26
,
61
-
76
.
Andres
,
D. A.
,
Seabra
,
M. C.
,
Brown
,
M. S.
,
Armstrong
,
S. A.
,
Smeland
,
T. E.
,
Cremers
,
F. P. M.
and
Goldstein
,
J. L.
(
1993
).
cDNA cloning of component A of Rab geranylgeranyl transferase and demonstration of its role as a Rab escort protein
.
Cell
73
,
1091
-
1099
.
Barr
,
F. A.
(
2013
).
Rab GTPases and membrane identity: causal or inconsequential?
J. Cell Biol.
202
,
191
-
199
.
Barr
,
F.
and
Lambright
,
D. G.
(
2010
).
Rab GEFs and GAPs
.
Curr. Opin. Cell Biol.
22
,
461
-
470
.
Bem
,
D.
,
Yoshimura
,
S.-I.
,
Nunes-Bastos
,
R.
,
Bond
,
F. F.
,
Kurian
,
M. A.
,
Rahman
,
F.
,
Handley
,
M. T. W.
,
Hadzhiev
,
Y.
,
Masood
,
I.
,
Straatman-Iwanowska
,
A. A.
, et al. 
(
2011
).
Loss-of-function mutations in RAB18 cause Warburg micro syndrome
.
Am. J. Hum. Genet.
88
,
499
-
507
.
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
.
Burke
,
J. E.
,
Inglis
,
A. J.
,
Perisic
,
O.
,
Masson
,
G. R.
,
McLaughlin
,
S. H.
,
Rutaganira
,
F.
,
Shokat
,
K. M.
and
Williams
,
R. L.
(
2014
).
Structures of PI4KIIIbeta complexes show simultaneous recruitment of Rab11 and its effectors
.
Science
344
,
1035
-
1038
.
Cabrera
,
M.
and
Ungermann
,
C.
(
2013
).
Guanine nucleotide exchange factors (GEFs) have a critical but not exclusive role in organelle localization of Rab GTPases
.
J. Biol. Chem.
288
,
28704
-
28712
.
Cetica
,
V.
,
Hackmann
,
Y.
,
Grieve
,
S.
,
Sieni
,
E.
,
Ciambotti
,
B.
,
Coniglio
,
M. L.
,
Pende
,
D.
,
Gilmour
,
K.
,
Romagnoli
,
P.
,
Griffiths
,
G. M.
, et al. 
(
2015
).
Patients with Griscelli syndrome and normal pigmentation identify RAB27A mutations that selectively disrupt MUNC13-4 binding
.
J. Allergy Clin. Immunol.
135
,
1310
-
1318.e1
.
Chavas
,
L. M. G.
,
Ihara
,
K.
,
Kawasaki
,
M.
,
Torii
,
S.
,
Uejima
,
T.
,
Kato
,
R.
,
Izumi
,
T.
and
Wakatsuki
,
S.
(
2008
).
Elucidation of Rab27 recruitment by its effectors: structure of Rab27a bound to Exophilin4/Slp2-a
.
Structure
16
,
1468
-
1477
.
Cheng
,
K. W.
,
Lahad
,
J. P.
,
Kuo
,
W.-L.
,
Lapuk
,
A.
,
Yamada
,
K.
,
Auersperg
,
N.
,
Liu
,
J.
,
Smith-McCune
,
K.
,
Lu
,
K. H.
,
Fishman
,
D.
, et al. 
(
2004
).
The RAB25 small GTPase determines aggressiveness of ovarian and breast cancers
.
Nat. Med.
10
,
1251
-
1256
.
Cheng
,
J.-M.
,
Ding
,
M.
,
Aribi
,
A.
,
Shah
,
P.
and
Rao
,
K.
(
2006
).
Loss of RAB25 expression in breast cancer
.
Int. J. Cancer
118
,
2957
-
2964
.
D'Adamo
,
P.
,
Menegon
,
A.
,
Lo Nigro
,
C.
,
Grasso
,
M.
,
Gulisano
,
M.
,
Tamanini
,
F.
,
Bienvenu
,
T.
,
Gedeon
,
A. K.
,
Oostra
,
B.
,
Wu
,
S.-K.
, et al. 
(
1998
).
Mutations in GDI1 are responsible for X-linked non-specific mental retardation
.
Nat. Genet.
19
,
134
-
139
.
De Franceschi
,
N.
,
Hamidi
,
H.
,
Alanko
,
J.
,
Sahgal
,
P.
and
Ivaska
,
J.
(
2015
).
Integrin traffic - the update
.
J. Cell Sci.
128
,
839
-
852
.
Deretic
,
D.
(
2013
).
Crosstalk of Arf and Rab GTPases en route to cilia
.
Small GTPases.
4
,
70
-
77
.
Detter
,
J. C.
,
Zhang
,
Q.
,
Mules
,
E. H.
,
Novak
,
E. K.
,
Mishra
,
V. S.
,
Li
,
W.
,
McMurtrie
,
E. B.
,
Tchernev
,
V. T.
,
Wallace
,
M. R.
,
Seabra
,
M. C.
, et al. 
(
2000
).
Rab geranylgeranyl transferase alpha mutation in the gunmetal mouse reduces Rab prenylation and platelet synthesis
.
Proc. Natl. Acad. Sci. USA
97
,
4144
-
4149
.
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
.
Eathiraj
,
S.
,
Pan
,
X.
,
Ritacco
,
C.
and
Lambright
,
D. G.
(
2005
).
Structural basis of family-wide Rab GTPase recognition by rabenosyn-5
.
Nature
436
,
415
-
419
.
Feldmann
,
J.
,
Callebaut
,
I.
,
Raposo
,
G.
,
Certain
,
S.
,
Bacq
,
D.
,
Dumont
,
C.
,
Lambert
,
N.
,
Ouachée-Chardin
,
M.
,
Chedeville
,
G.
,
Tamary
,
H.
, et al. 
(
2003
).
Munc13-4 is essential for cytolytic granules fusion and is mutated in a form of familial hemophagocytic lymphohistiocytosis (FHL3)
.
Cell
115
,
461
-
473
.
Flannagan
,
R. S.
,
Jaumouillé
,
V.
and
Grinstein
,
S.
(
2012
).
The cell biology of phagocytosis
.
Annu. Rev. Pathol.
7
,
61
-
98
.
Frittoli
,
E.
,
Palamidessi
,
A.
,
Marighetti
,
P.
,
Confalonieri
,
S.
,
Bianchi
,
F.
,
Malinverno
,
C.
,
Mazzarol
,
G.
,
Viale
,
G.
,
Martin-Padura
,
I.
,
Garre
,
M.
, et al. 
(
2014
).
A RAB5/RAB4 recycling circuitry induces a proteolytic invasive program and promotes tumor dissemination
.
J. Cell Biol.
206
,
307
-
328
.
Gerondopoulos
,
A.
,
Langemeyer
,
L.
,
Liang
,
J.-R.
,
Linford
,
A.
and
Barr
,
F. A.
(
2012
).
BLOC-3 mutated in Hermansky-Pudlak syndrome is a Rab32/38 guanine nucleotide exchange factor
.
Curr. Biol.
22
,
2135
-
2139
.
Gillingham
,
A. K.
,
Sinka
,
R.
,
Torres
,
I. L.
,
Lilley
,
K. S.
and
Munro
,
S.
(
2014
).
Toward a comprehensive map of the effectors of rab GTPases
.
Dev. Cell
31
,
358
-
373
.
Goldenring
,
J. R.
(
2013
).
A central role for vesicle trafficking in epithelial neoplasia: intracellular highways to carcinogenesis
.
Nat. Rev. Cancer
13
,
813
-
820
.
Goody
,
R. S.
,
Rak
,
A.
and
Alexandrov
,
K.
(
2005
).
The structural and mechanistic basis for recycling of Rab proteins between membrane compartments
.
Cell. Mol. Life Sci.
62
,
1657
-
1670
.
Handley
,
M. T.
,
Morris-Rosendahl
,
D. J.
,
Brown
,
S.
,
Macdonald
,
F.
,
Hardy
,
C.
,
Bem
,
D.
,
Carpanini
,
S. M.
,
Borck
,
G.
,
Martorell
,
L.
,
Izzi
,
C.
, et al. 
(
2013
).
Mutation spectrum in RAB3 GAP1, RAB3 GAP2, and RAB18 and genotype-phenotype correlations in warburg micro syndrome and Martsolf syndrome
.
Hum. Mutat.
34
,
686
-
696
.
Ho
,
J. R.
,
Chapeaublanc
,
E.
,
Kirkwood
,
L.
,
Nicolle
,
R.
,
Benhamou
,
S.
,
Lebret
,
T.
,
Allory
,
Y.
,
Southgate
,
J.
,
Radvanyi
,
F.
and
Goud
,
B.
(
2012
).
Deregulation of Rab and Rab effector genes in bladder cancer
.
PLoS. ONE
7
,
e39469
.
Horiuchi
,
H.
,
Lippé
,
R.
,
McBride
,
H. M.
,
Rubino
,
M.
,
Woodman
,
P.
,
Stenmark
,
H.
,
Rybin
,
V.
,
Wilm
,
M.
,
Ashman
,
K.
,
Mann
,
M.
, et al. 
(
1997
).
A novel Rab5 GDP/GTP exchange factor complexed to Rabaptin-5 links nucleotide exchange to effector recruitment and function
.
Cell
90
,
1149
-
1159
.
Hutagalung
,
A. H.
and
Novick
,
P. J.
(
2011
).
Role of Rab GTPases in membrane traffic and cell physiology
.
Physiol. Rev.
91
,
119
-
149
.
Jacob
,
A.
,
Jing
,
J.
,
Lee
,
J.
,
Schedin
,
P.
,
Gilbert
,
S. M.
,
Peden
,
A. A.
,
Junutula
,
J. R.
and
Prekeris
,
R.
(
2013
).
Rab40b regulates trafficking of MMP2 and MMP9 during invadopodia formation and invasion of breast cancer cells
.
J. Cell Sci.
126
,
4647
-
4658
.
Jenkins
,
D.
,
Seelow
,
D.
,
Jehee
,
F. S.
,
Perlyn
,
C. A.
,
Alonso
,
L. G.
,
Bueno
,
D. F.
,
Donnai
,
D.
,
Josifiova
,
D.
,
Mathijssen
,
I. M. J.
,
Morton
,
J. E. V.
, et al. 
(
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
.
Kim
,
J.-K.
,
Lee
,
S.-Y.
,
Park
,
C.-W.
,
Park
,
S.-H.
,
Yin
,
J.
,
Kim
,
J.
,
Park
,
J.-B.
,
Lee
,
J.-Y.
,
Kim
,
H.
and
Kim
,
S.-C.
(
2014
).
Rab3a promotes brain tumor initiation and progression
.
Mol. Biol. Rep.
41
,
5903
-
5911
.
Kimura
,
T.
,
Kaneko
,
Y.
,
Yamada
,
S.
,
Ishihara
,
H.
,
Senda
,
T.
,
Iwamatsu
,
A.
and
Niki
,
I.
(
2008
).
The GDP-dependent Rab27a effector coronin 3 controls endocytosis of secretory membrane in insulin-secreting cell lines
.
J. Cell Sci.
121
,
3092
-
3098
.
Klopper
,
T. H.
,
Kienle
,
N.
,
Fasshauer
,
D.
and
Munro
,
S.
(
2012
).
Untangling the evolution of Rab G proteins: implications of a comprehensive genomic analysis
.
BMC Biol.
10
,
71
.
Knowles
,
B. C.
,
Roland
,
J. T.
,
Krishnan
,
M.
,
Tyska
,
M. J.
,
Lapierre
,
L. A.
,
Dickman
,
P. S.
,
Goldenring
,
J. R.
and
Shub
,
M. D.
(
2014
).
Myosin Vb uncoupling from RAB8A and RAB11A elicits microvillus inclusion disease
.
J. Clin. Invest
124
,
2947
-
2962
.
Kotzsch
,
M.
,
Sieuwerts
,
A. M.
,
Grosser
,
M.
,
Meye
,
A.
,
Fuessel
,
S.
,
Meijer-van Gelder
,
M. E.
,
Smid
,
M.
,
Schmitt
,
M.
,
Baretton
,
G.
,
Luther
,
T.
, et al. 
(
2008
).
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.
111
,
229
-
240
.
Langemeyer
,
L.
,
Nunes Bastos
,
R.
,
Cai
,
Y.
,
Itzen
,
A.
,
Reinisch
,
K. M.
and
Barr
,
F. A.
(
2014
).
Diversity and plasticity in Rab GTPase nucleotide release mechanism has consequences for Rab activation and inactivation
.
Elife
3
,
e01623
.
Lee
,
M.-T. G.
,
Mishra
,
A.
and
Lambright
,
D. G.
(
2009
).
Structural mechanisms for regulation of membrane traffic by rab GTPases
.
Traffic
10
,
1377
-
1389
.
Leung
,
K. F.
,
Baron
,
R.
and
Seabra
,
M. C.
(
2006
).
Lipid posttranslational modifications: geranylgeranylation of Rab GTPases
.
J. Lipid Res.
47
,
467
-
475
.
Li
,
Y.
,
Jia
,
Q.
,
Wang
,
Y.
,
Li
,
F.
,
Jia
,
Z.
and
Wan
,
Y.
(
2015
).
Rab40b upregulation correlates with the prognosis of gastric cancer by promoting migration, invasion, and metastasis
.
Med. Oncol.
32
,
126
.
Martin
,
S.
and
Parton
,
R. G.
(
2008
).
Characterization of Rab18, a lipid droplet-associated small GTPase
.
Methods Enzymol.
438
,
109
-
129
.
Martin
,
S.
,
Driessen
,
K.
,
Nixon
,
S. J.
,
Zerial
,
M.
and
Parton
,
R. G.
(
2005
).
Regulated localization of Rab18 to lipid droplets: effects of lipolytic stimulation and inhibition of lipid droplet catabolism
.
J. Biol. Chem.
280
,
42325
-
42335
.
Marzesco
,
A.-M.
,
Dunia
,
I.
,
Pandjaitan
,
R.
,
Recouvreur
,
M.
,
Dauzonne
,
D.
,
Benedetti
,
E. L.
,
Louvard
,
D.
and
Zahraoui
,
A.
(
2002
).
The small GTPase Rab13 regulates assembly of functional tight junctions in epithelial cells
.
Mol. Biol. Cell
13
,
1819
-
1831
.
Menasche
,
G.
,
Pastural
,
E.
,
Feldmann
,
J.
,
Certain
,
S.
,
Ersoy
,
F.
,
Dupuis
,
S.
,
Wulfraat
,
N.
,
Bianchi
,
D.
,
Fischer
,
A.
,
Le Deist
,
F.
, et al. 
(
2001
).
Mutations in RAB27A cause Griscelli syndrome associated with haemophagocytic syndrome
.
Nat. Genet.
25
,
173
-
176
.
Menasche
,
G.
,
Ho
,
C. H.
,
Sanal
,
O.
,
Feldmann
,
J.
,
Tezcan
,
I.
,
Ersoy
,
F.
,
Houdusse
,
A.
,
Fischer
,
A.
and
de Saint Basile
,
G.
(
2003
).
Griscelli syndrome restricted to hypopigmentation results from a melanophilin defect (GS3) or a MYO5A F-exon deletion (GS1)
.
J. Clin. Invest.
112
,
450
-
456
.
Militello
,
R.
and
Colombo
,
M. I.
(
2013
).
Small GTPases as regulators of cell division
.
Commun. Integr. Biol.
6
,
e25460
.
Munafo
,
D. B.
and
Colombo
,
M. I.
(
2002
).
Induction of autophagy causes dramatic changes in the subcellular distribution of GFP-Rab24
.
Traffic
3
,
472
-
482
.
Nakano
,
T.
,
Shimizu
,
K.
,
Kawashima
,
O.
,
Kamiyoshihara
,
M.
,
Kakegawa
,
S.
,
Sugano
,
M.
,
Ibe
,
T.
,
Nagashima
,
T.
,
Kaira
,
K.
,
Sunaga
,
N.
, et al. 
(
2012
).
Establishment of a human lung cancer cell line with high metastatic potential to multiple organs: gene expression associated with metastatic potential in human lung cancer
.
Oncol. Rep.
28
,
1727
-
1735
.
Nottingham
,
R. M.
,
Ganley
,
I. G.
,
Barr
,
F. A.
,
Lambright
,
D. G.
and
Pfeffer
,
S. R.
(
2011
).
RUTBC1 protein, a Rab9A effector that activates GTP hydrolysis by Rab32 and Rab33B proteins
.
J. Biol. Chem.
286
,
33213
-
33222
.
Nottingham
,
R. M.
,
Pusapati
,
G. V.
,
Ganley
,
I. G.
,
Barr
,
F. A.
,
Lambright
,
D. G.
and
Pfeffer
,
S. R.
(
2012
).
RUTBC2 protein, a Rab9A effector and GTPase-activating protein for Rab36
.
J. Biol. Chem.
287
,
22740
-
22748
.
Numrich
,
J.
and
Ungermann
,
C.
(
2014
).
Endocytic Rabs in membrane trafficking and signaling
.
Biol. Chem.
395
,
327
-
333
.
Olkkonen
,
V. M.
and
Ikonen
,
E.
(
2000
).
Genetic defects of intracellular-membrane transport
.
N. Eng. J. Med.
343
,
1095
-
1104
.
Ostermeier
,
C.
and
Brunger
,
A. T.
(
1999
).
Structural basis of Rab effector specificity: crystal structure of the small G protein Rab3A complexed with the effector domain of Rabphilin-3A
.
Cell
96
,
363
-
374
.
Pellinen
,
T.
,
Arjonen
,
A.
,
Vuoriluoto
,
K.
,
Kallio
,
K.
,
Fransen
,
J. A. M.
and
Ivaska
,
J.
(
2006
).
Small GTPase Rab21 regulates cell adhesion and controls endosomal traffic of beta1-integrins
.
J. Cell Biol.
173
,
767
-
780
.
Peranen
,
J.
,
Auvinen
,
P.
,
Virta
,
H.
,
Wepf
,
R.
and
Simons
,
K.
(
1996
).
Rab8 promotes polarized membrane transport through reorganization of actin and microtubules in fibroblasts
.
J. Cell Biol.
135
,
153
-
167
.
Pfeffer
,
S. R.
(
2013
).
Rab GTPase regulation of membrane identity
.
Curr. Opin. Cell Biol.
25
,
414
-
419
.
Raiborg
,
C.
,
Wenzel
,
E. M.
and
Stenmark
,
H.
(
2015
).
ER-endosome contact sites: molecular compositions and functions
.
EMBO J.
34
,
1848
-
1858
.
Recacha
,
R.
,
Boulet
,
A.
,
Jollivet
,
F.
,
Monier
,
S.
,
Houdusse
,
A.
,
Goud
,
B.
and
Khan
,
A. R.
(
2009
).
Structural basis for recruitment of Rab6-interacting protein 1 to Golgi via a RUN domain
.
Structure
17
,
21
-
30
.
Rink
,
J.
,
Ghigo
,
E.
,
Kalaidzidis
,
Y.
and
Zerial
,
M.
(
2005
).
Rab conversion as a mechanism of progression from early to late endosomes
.
Cell
122
,
735
-
749
.
Roosing
,
S.
,
Rohrschneider
,
K.
,
Beryozkin
,
A.
,
Sharon
,
D.
,
Weisschuh
,
N.
,
Staller
,
J.
,
Kohl
,
S.
,
Zelinger
,
L.
,
Peters
,
T. A.
,
Neveling
,
K.
, et al. 
(
2013
).
Mutations in RAB28, encoding a farnesylated small GTPase, are associated with autosomal-recessive cone-rod dystrophy
.
Am. J. Hum. Genet.
93
,
110
-
117
.
Schoebel
,
S.
,
Oesterlin
,
L. K.
,
Blankenfeldt
,
W.
,
Goody
,
R. S.
and
Itzen
,
A.
(
2009
).
RabGDI displacement by DrrA from Legionella is a consequence of its guanine nucleotide exchange activity
.
Mol. Cell
36
,
1060
-
1072
.
Seifert
,
W.
,
Kühnisch
,
J.
,
Maritzen
,
T.
,
Lommatzsch
,
S.
,
Hennies
,
H. C.
,
Bachmann
,
S.
,
Horn
,
D.
and
Haucke
,
V.
(
2015
).
Cohen syndrome-associated protein COH1 physically and functionally interacts with the small GTPase RAB6 at the Golgi complex and directs neurite outgrowth
.
J. Biol. Chem.
290
,
3349
-
3358
.
Seixas
,
E.
,
Barros
,
M.
,
Seabra
,
M. C.
and
Barral
,
D. C.
(
2013
).
Rab and Arf proteins in genetic diseases
.
Traffic
14
,
871
-
885
.
Sherwood
,
R. K.
and
Roy
,
C. R.
(
2013
).
A Rab-centric perspective of bacterial pathogen-occupied vacuoles
.
Cell Host. Microbe
14
,
256
-
268
.
Shimada
,
K.
,
Uzawa
,
K.
,
Kato
,
M.
,
Endo
,
Y.
,
Shiiba
,
M.
,
Bukawa
,
H.
,
Yokoe
,
H.
,
Seki
,
N.
and
Tanzawa
,
H.
(
2005
).
Aberrant expression of RAB1A in human tongue cancer
.
Br. J. Cancer
92
,
1915
-
1921
.
Shirane
,
M.
and
Nakayama
,
K. I.
(
2006
).
Protrudin induces neurite formation by directional membrane trafficking
.
Science
314
,
818
-
821
.
Sivars
,
U.
,
Aivazian
,
D.
and
Pfeffer
,
S. R.
(
2003
).
Yip3 catalyses the dissociation of endosomal Rab-GDI complexes
.
Nature
425
,
856
-
859
.
Soldati
,
T.
,
Shapiro
,
A. D.
,
Svejstrup
,
A. B. D.
and
Pfefffer
,
S. R.
(
1994
).
Membrane targeting of the small GTPase Rab9 is accompanied by nucleotide exchange
.
Nature
369
,
76
-
78
.
Stein
,
M.-P.
,
Feng
,
Y.
,
Cooper
,
K. L.
,
Welford
,
A. M.
and
Wandinger-Ness
,
A.
(
2003
).
Human VPS34 and p150 are Rab7 interacting partners
.
Traffic
4
,
754
-
771
.
Stenmark
,
H.
(
2009
).
Rab GTPases as coordinators of vesicle traffic
.
Nat. Rev. Mol. Cell Biol.
10
,
513
-
525
.
Sudhof
,
T. C.
and
Rothman
,
J. E.
(
2009
).
Membrane fusion: grappling with SNARE and SM proteins
.
Science
323
,
474
-
477
.
Thomas
,
J. D.
,
Zhang
,
Y.-J.
,
Wei
,
Y.-H.
,
Cho
,
J.-H.
,
Morris
,
L. E.
,
Wang
,
H.-Y.
and
Zheng
,
X. F. S.
(
2014
).
Rab1A is an mTORC1 activator and a colorectal oncogene
.
Cancer Cell
26
,
754
-
769
.
Ullrich
,
O.
,
Horiuchi
,
H.
,
Bucci
,
C.
and
Zerial
,
M.
(
1994
).
Membrane association of Rab5 mediated by GDP-dissociation inhibitor and accompanied by GDP/GTP exchange
.
Nature
368
,
157
-
160
.
Valsdottir
,
R.
,
Hashimoto
,
H.
,
Ashman
,
K.
,
Koda
,
T.
,
Storrie
,
B.
and
Nilsson
,
T.
(
2001
).
Identification of rabaptin-5, rabex-5, and GM130 as putative effectors of rab33b, a regulator of retrograde traffic between the Golgi apparatus and ER
.
FEBS Lett.
508
,
201
-
209
.
van IJzendoorn
,
S. C. D.
,
Mostov
,
K. E.
and
Hoekstra
,
D.
(
2003
).
Role of rab proteins in epithelial membrane traffic
.
Int. Rev. Cytol.
232
,
59
-
88
.
Verhoeven
,
K.
,
De Jonghe
,
P.
,
Coen
,
K.
,
Verpoorten
,
N.
,
Auer-Grumbach
,
M.
,
Kwon
,
J. M.
,
FitzPatrick
,
D.
,
Schmedding
,
E.
,
De Vriendt
,
E.
,
Jacobs
,
A.
, et al. 
(
2003
).
Mutations in the small GTP-ase late endosomal protein RAB7 cause Charcot-Marie-Tooth type 2B neuropathy
.
Am. J. Hum. Genet.
72
,
722
-
727
.
Vitale
,
G.
,
Rybin
,
V.
,
Christoforidis
,
S.
,
Thornqvist
,
P.-Ö.
,
McCaffrey
,
M.
,
Stenmark
,
H.
and
Zerial
,
M.
(
1998
).
Distinct Rab-binding domains mediate the interaction of rabaptin-5 with GTP-bound rab4 and rab5
.
EMBO J.
17
,
1941
-
1951
.
Wandinger-Ness
,
A.
and
Zerial
,
M.
(
2014
).
Rab proteins and the compartmentalization of the endosomal system
.
Cold Spring Harb. Perspect. Biol.
6
,
a022616
.
Wang
,
H.
and
Jiang
,
C.
(
2013
).
RAB38 confers a poor prognosis, associated with malignant progression and subtype preference in glioma
.
Oncol. Rep.
30
,
2350
-
2356
.
Wang
,
R.
,
Wang
,
Z.-X.
,
Yang
,
J.-S.
,
Pan
,
X.
,
De
,
W.
and
Chen
,
L.-B.
(
2011
).
MicroRNA-451 functions as a tumor suppressor in human non-small cell lung cancer by targeting ras-related protein 14 (RAB14)
.
Oncogene
30
,
2644
-
2658
.
Wilson
,
G. R.
,
Sim
,
J. C. H.
,
McLean
,
C.
,
Giannandrea
,
M.
,
Galea
,
C. A.
,
Riseley
,
J. R.
,
Stephenson
,
S. E. M.
,
Fitzpatrick
,
E.
,
Haas
,
S. A.
,
Pope
,
K.
, et al. 
(
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
.
Yang
,
P.-S.
,
Yin
,
P.-H.
,
Tseng
,
L.-M.
,
Yang
,
C.-H.
,
Hsu
,
C.-Y.
,
Lee
,
M.-Y.
,
Horng
,
C.-F.
and
Chi
,
C.-W.
(
2011
).
Rab5A is associated with axillary lymph node metastasis in breast cancer patients
.
Cancer Sci.
102
,
2172
-
2178
.
Zhu
,
G.
,
Zhai
,
P.
,
Liu
,
J.
,
Terzyan
,
S.
,
Li
,
G.
and
Zhang
,
X. C.
(
2004
).
Structural basis of Rab5-Rabaptin5 interaction in endocytosis
.
Nat. Struct. Mol. Biol.
11
,
975
-
983
.

Competing interests

The authors declare no competing or financial interests.