Next Article in Journal
The Cardiovascular Therapeutic Potential of Propolis—A Comprehensive Review
Previous Article in Journal
Stem and Cancer Stem Cell Identities, Cellular Markers, Niche Environment and Response to Treatments to Unravel New Therapeutic Targets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

RAB11-Mediated Trafficking and Human Cancers: An Updated Review

1
Department of Applied Science and Technology, Politecnico di Torino, 24 Corso Duca degli Abruzzi, 10129 Turin, Italy
2
Italian Institute for Genomic Medicine, c/o IRCCS, Str. Prov. le 142, km 3.95, 10060 Candiolo, Italy
*
Author to whom correspondence should be addressed.
Biology 2021, 10(1), 26; https://doi.org/10.3390/biology10010026
Submission received: 15 November 2020 / Revised: 15 December 2020 / Accepted: 30 December 2020 / Published: 4 January 2021
(This article belongs to the Section Cell Biology)

Abstract

:

Simple Summary

The small GTPase RAB11 is a master regulator of both vesicular trafficking and membrane dynamic defining the surface proteome of cellular membranes. As a consequence, the alteration of RAB11 activity induces changes in both the sensory and the transduction apparatuses of cancer cells leading to tumor progression and invasion. Here, we show that this strictly depends on RAB11′s ability to control the sorting of signaling receptors from endosomes. Therefore, RAB11 is a potential therapeutic target over which to develop future therapies aimed at dampening the acquisition of aggressive traits by cancer cells.

Abstract

Many disorders block and subvert basic cellular processes in order to boost their progression. One protein family that is prone to be altered in human cancers is the small GTPase RAB11 family, the master regulator of vesicular trafficking. RAB11 isoforms function as membrane organizers connecting the transport of cargoes towards the plasma membrane with the assembly of autophagic precursors and the generation of cellular protrusions. These processes dramatically impact normal cell physiology and their alteration significantly affects the survival, progression and metastatization as well as the accumulation of toxic materials of cancer cells. In this review, we discuss biological mechanisms ensuring cargo recognition and sorting through a RAB11-dependent pathway, a prerequisite to understand the effect of RAB11 alterations in human cancers.

1. Introduction

To manage pathogenic alterations induced by genetic/epigenetic/transcriptomic changes, cells evolved various strategies to exploit cellular machinery and guarantee their own survival. Specific genetic alterations modify key elements controlling the molecular composition of the cell surface, the assembly of internal organelles and the efficacy of intracellular and extracellular transmission. RAB GTPases are key players in membrane transport events and are fundamental to ensure the efficacy of intracellular logistics [1,2,3].
RAB GTPases are members of the Ras superfamily of monomeric G proteins, a large protein family which includes among its members RAS, RHO–RAC, ARF and RAN as well as the more recently characterized RHEB, RAD and RIT subfamilies [4]. Similarly to other small GTPases, Rab proteins are molecular switches that cycle between an active and an inactive state through the association and the subsequent hydrolysis of guanosine-5′-triphosphate (GTP). In particular, association with GTP is stimulated by Guanine nucleotide exchange factors (GEFs), whereas hydrolysis of GTP to GDP is promoted by GTPase-activating proteins (GAPs) [5]. This activation cycle is pivotal for the binding of multiple effector proteins which mediate the delivery of cellular material to various intracellular compartments such as—the plasma membrane, endosomes, autophagosomes, leading edge of migrating cells, midbodies, primary cilium and centrioles [6]. As a matter of fact, alterations of RAB-mediated trafficking are pivotal in various aspects of both progression and tumorgenicity of cancer cells including the sustainment of proliferative signaling, the evasion of growth suppression, the induction of receptor recycling, the activation of invasion and metastasis as well as the reprogramming of tumor metabolism and the evasion of immune destruction [7,8].

2. Different RAB11 Isoforms for Distinct Cellular Functions

One prominent Rab-mediated transport pathway that cancer cells exploit to adapt their internal states to fluctuations of both their gene expression and microenvironmental status is the RAB11-mediated trafficking pathway. This transport route relies on members of the RAB11 protein family which control the delivery of both proteins and lipids toward several organelles and therefore are involved in many cellular processes (Figure 1A,B).
The RAB11 protein family includes three different isoforms named RAB11A, RAB11B and RAB25, each one encoded by a different gene located in a distinct chromosome (Table 1). All RAB11 isoforms are widely distributed in human tissues. In particular, while RAB11A is ubiquitously expressed, both RAB11B and RAB25 are enriched in specific organs (Table 1) [9]. Notably, RAB11 proteins differ in their percentage of sequence identity. While RAB11A and RAB11B share 89% of sequence similarity, the identity between RAB25 with either RAB11A or RAB11B is lower than 70% (Table 1) [10].
Although minimal, such differences in protein sequence identity significantly impact both protein structure and specificity of effector binding, providing an explanation for the different roles of RAB11 isoforms in cells [11,12]. All RAB11 proteins present several post-translational modifications, including prenylation, phosphorylation and ubiquitination. In particular, while prenylation at RAB11 C-termini allows Rab membrane association, both phosphorylation and ubiquitination modulate either activity or degradation of RAB11 isoforms, respectively [9].
RAB11A is the first discovered and best characterized member of the RAB11 subfamily. Homozygous depletion of Rab11a in mice is embryo lethal; whereas the loss of RAB11A protein in neurons does not significantly impact both brain development and functionality. In contrast, depletion of Rab11a in mice intestine increases intracellular accumulation of apical proteins and it induces shortening of microvilli [13,14].
In cells, RAB11A localizes to the recycling endosome (RE) a perinuclear positioned membrane-bound compartment implicated in the control of both proteins and lipids trafficking through and from the RE to the plasma membrane (Figure 1B) [15,16]. In addition, RAB11A mediates transport of vesicular cargoes from peripheral sorting endosomes (SEs) to the RE (Figure 1A) [17]. Furthermore, RAB11A localizes at the trans-Golgi network (TGN), where it controls the transport of material towards both the apical and the basolateral membrane in polarized cells (Figure 1B) [18]. Based on both its wide distribution and functions aimed at mediating the delivery of cellular material to both plasma membrane, endosomes, autophagosome and midbody, RAB11A influences several cellular processes such as cytokinesis, phagocytosis, cell migration and proliferation, immunological synapse assembly, ciliogenesis, influenza virus replication, and autophagy [19,20,21,22,23,24,25].
The RAB11B isoform is highly enriched in the brain and, consistently, homozygous depletion of Rab11b in mice leads to neurological dysfunction (data from International Mouse Phenotyping Consortium, https://www.mousephenotype.org/). Although, RAB11B localizes in both the RE and SE similarly to RAB11A, RAB11B depletion does not alter accumulation of the Transferrin receptor (TfR) at the perinuclear RE whereas downregulation of RAB11A does, thereby indicating a non-overlapping function for these small GTPases in endocytic recycling [26]. In parallel, accumulation of fibroblast growth factor receptor 4 (FGFR4) was observed after downregulation of RAB11B, but not RAB11A [27]. Consistently, RAB11B localizes to an apical pericentrosomal membrane-bound compartment distinct from RAB11A [28]. In this context, recent findings indicate that RAB11A and RAB11B perform opposing roles in the same cells. In particular, RAB11B was found to control recycling of the PAR1 receptor from the endosome to plasma membrane while RAB11A defines the rate of PAR1 degradation by delivering such signaling receptor into the autophagic pathway [29]. Lastly, in polarized epithelial cells, RAB11B specifically mediates the apical transport of the cystic fibrosis transmembrane conductance regulator (CFTR) [30] while it regulates exocytosis in neurons and neuroendocrine cells [31].
RAB25-deficient mice do not exhibit gross pathology [32]. However, when Rab25 depletion is associated with a specific genetic background, as found in 129/J mouse strain, it induces vaginal cancer in virgin females and distal esophageal stricture in both males and females [32]. The localization of RAB25 in cells partially overlap with those of RAB11A (Figure 1B) [33], whereas its role has been associated both in transport of oncogenic signaling receptors and in cellular bioenergetics [9,34].
Through both the presence of various isoforms, the wide distribution amongst distinct cellular compartments and the occurrence of several post-translational mechanisms of regulation, they confer to RAB11 and its trafficking route an elevated level of plasticity. Consequently, RAB11-mediated trafficking pathways provide essential support to various aspects of cellular processes.
In the sections below, we will discuss the implications of RAB11′s role in the planning, implementation and control of the efficient and effective flow and storage of cellular materials, with the aim at understanding the basic principles governing cellular processes underlying tumor progression.

3. RAB11-Mediated Trafficking Sustains Proliferative Signaling

One of the fundamental traits of cancer cells is their ability to sustain chronic proliferation. Growth-promoting signals are tightly regulated in normal cells and they are exploited to instruct cells about both their microenvironment, their duties and their fate. As a consequence, both growth factors, signaling receptors and intracellular signaling cascades are essential to ensure normal tissue homeostasis. Therefore, it is not surprising that by deregulating these signals, cancer cells become independent of extracellular messages that maintain normal tissue homeostasis and thus turn into masters of their own destiny [35].
Typically, binding of extracellular growth factors to surface receptors such as tyrosine kinases provides the key event that induces activation of proliferative signaling cascades in cells. Cancer cells acquire the capability to sustain such a mitogenic response in a number of alternative ways such as inducing autocrine signaling, increasing the number of mitogenic receptors on the cell surface, or extending the mitogenic stimulation by altering the information processing of downstream signaling cascades [36]. RAB11 isoforms were described to influence these processes by controlling the intracellular distribution of cargoes such as growth factor receptors, transmembrane receptors, cell adhesion molecules and signaling lipids [36]. The function of RAB11 isoforms in the cellular logistic implies a role for this small GTPase family in the organization and implementation of operations ensuring the flow of information between distinct cellular districts with spatiotemporal precision. Such procedures direct both the generation, packaging, transportation and stocking of membrane cargoes at the right place and at the right time [5,37]. Therefore, it is conceivable that RAB11 isoforms serve multiple roles impacting various cellular processes when dysfunctional. However, it remains still difficult to predict RAB11 functions in human diseases.
This is exemplified by the dual character of RAB11 isoforms in human cancers. RAB11 isoforms both enhance and diminish cancer progression by functioning either as oncogenes or as tumor suppressors (Table 2, Table 3 and Table 4).
Loss of RAB11 isoforms (i.e., RAB11A and RAB25) is not tumorigenic per se and results in both hyperplasia and dysplasia of colorectal tissues [32,60], whereas their overexpression is not sufficient to transform nontumorigenic/immortalized cells, rather they promote the acquisition of both metastatic and invasive properties in already transformed cells [34,43,51]. Conversely, in cells characterized by a compromised genetic background, alterations in the expression of RAB11 isoforms promote cancer progression. Notably, this occurs both when RAB11 isoforms are downregulated and when they are overexpressed (Table 2, Table 3 and Table 4). As an example, in epithelial cancer cells characterized by loss of either APC or SMAD3 proteins, RAB11 isoforms display a tumor suppressor function promoting both proliferation and aggressiveness of cancer cells when downregulated [32,61]. On the contrary, RAB11 family members show oncogenic potential by promoting proliferation and invasiveness cancer cells when overexpressed [51,62].
Notably, recent evidence suggests that alteration of RAB11 gene expression is part of the adaptive programs that cells exploit to match environmental changes. In particular, RAB11B is up-regulated during adaptation of breast cancer cells to the brain microenvironment, a process that induces the massive modification of the cell surface proteome [43]. Intriguingly, the finding that, in breast cancer, other RAB11 isoforms (i.e., RAB25) are either not altered or epigenetically silenced, strongly support the idea that RAB11 proteins perform non-overlapping functions in cancer [43,63,64].
Despite the fact that these oncogenic phenotypes have been mechanistically linked to an alteration in both proliferative and migratory cellular capacity through either recycling of receptor tyrosine kinases, E-Cadherin, α5β1 Integrins, or to activation of oncogenic signaling and metabolic stress, the molecular underpinnings of these tumorigenic events are still poorly understood [33,34,51,65,66,67,68]. However, we are still struggling to understand how RAB11-mediated trafficking is orchestrated, the evidence that RAB11 acts in concert with an elevated number of effectors and interactors, to ensure the fidelity of intracellular trafficking, provides a novel framework that allows to interpret RAB11-induced cellular dysfunction.

4. RAB11-Mediated Sorting Controls Intracellular Accumulation of Surface Proteins

Most growth factor receptors and transmembrane proteins are transported to the cell surface by RAB11 isoforms. This event confers excitability to mitogenic stimulation. By deregulating this transport route, cancer cells increase the number of growth factor receptors on the plasma membrane prompting the amplification of mitogenic stimulation.
An elevated number of reports confirms the significance of the RAB11-mediated trafficking pathway in transport of pro-tumorigenic signaling receptors to cell surface. In particular, RAB11A as well as RAB11B and RAB25 enhance receptor recycling to the cell surface, and, they concomitantly decrease degradation in cytoplasm [12,43,61]. As a consequence, RAB11-mediated trafficking supports both the amplification and duration of signaling cascades by increasing receptor half-life in the plasma membrane. In addition, the directional flux of membrane cargoes from internal organelles towards the cell surface promotes directional cell migration of cancer cells and therefore their ability to invade healthy tissue and metastasize to distant sites. Accordingly, RAB11 proteins were found to recycle β1-Integrin to the plasma membrane [32,43] and in parallel their depletion blocks receptors inside cells for a longer time, with major consequences in terms of amplification of oncogenic signaling cascades [43,69,70].
However, the evidence that RAB11 isoforms function both as oncogenes and tumor suppressors challenges this view, raising the possibility that in some cellular contexts, such as in the presence of massive alteration in both signaling and metabolic pathways, RAB11 might misdirect cargoes assigned to the cell surface to a different destination.
In this context, the analysis of both specific and overlapping RAB11 roles might provide novel insights into mechanisms controlling intracellular accumulation of surface proteins. As an example, RAB11A and RAB11B perform divergent roles in the regulation of PAR1 receptor trafficking by controlling either its degradation or its recycling, respectively. In the absence of RAB11B-mediated recycling, the PAR1 receptor is trafficked to autophagic organelles for degradation, a process that requires RAB11A. Accordingly, RAB11A knockdown restores PAR1 expression in silenced RAB11B cells [29].

5. RAB11 Balances Distinct Trafficking Routes with Opposing Functions

Balance between either recycling, intracellular accumulation or degradation of surface receptors is ensured by a vast number of intracellular membrane compartments that both evolve and mature dynamically to connect both the endocytic and the exocytic pathway. In particular, at the crossroads between recycling and degradation routes resides the early endosome, the sorting station of the endocytic system. The role of early endosomes is to accept endocytosed proteins and lipids and to redirect them either back to plasma membrane or to lysosomes. Serving these two intracellular highways, two opposing sorting machineries operate at the early endosome and both require the assembly of multiprotein complexes. In particular, targeted degradation of surface receptors is controlled by the endosomal sorting complex required for transport (ESCRT), while the recycling to the cell surface of endocytosed cargo is ensured by distinct molecular assemblies such as the retromer/SNX27/WASH complex, the Retriever complex, and the FERARI complex [71]. Notably, both retromer/SNX27/WASH complex and Retriever share common regulators such as the Vacuolar protein sorting-associated protein 29 (VPS29) and the WASH complex. In particular, VPS29 is essential for cargo selection, transcytosis of the polymeric immunoglobulin receptor (pIgR-pIgA) and recycling of NxxY-motif-containing cargo proteins by coupling to SNX17 [72,73,74,75], whereas the WASH complex functions as a nucleation-promoting factor (NPF) at the surface of endosomes, where it induces actin polymerization and fission of recycling tubules containing either transferrin receptor (TfR), EGFR or α5β1-Integrin [71,76,77,78,79,80]. Remarkably, despite the similarities between the retromer/SNX27/WASH and Retriever complexes, it is not known whether Rab proteins participate in the endosomal recruitment of these recycling machineries.
Recently, the FERARI complex has emerged as a critical component in RAB11-mediated trafficking. In particular, it promotes membrane fission and fusion at the recycling compartment of early/sorting endosomes [81]. The FERARI complex consists of three interacting units—the Rab-module, the SNARE module, and the membrane-fission module. These elements coordinate respectively—(i) tethering of RAB11 vesicle to RAB5 positive early endosome, (ii) fusion of incoming RAB11 vesicle and, (iii) fission of the newly generated RAB11 vesicle from the endosome. Notably, these modules act synergistically, thereby coordinating kiss-and-run vesicle release for cargo delivery at the recycling pathways [81].
The important role of the FERARI complex in controlling RAB11-mediated trafficking relies on the Rab-module. This functional unit includes the Rab effectors Rabenosyn-5 (RBSN) and RAB11FIP5, two proteins connecting the endocytic and recycling transport machinery [81]. In particular, the RAB5 effector RBSN controls both membrane fusion and membrane trafficking of recycling endosomes generated at the early endosome and, interestingly, it is considered a tumor-suppressive gene [82]. In parallel, RAB11FIP5 belongs to the RAB11 family interacting protein (RAB11FIP), a protein family that comprises five distinct genes (i.e., RAB11FIP1, RAB11FIP2, RAB11FIP3, RAB11FIP4, RAB11FIP5) which are characterized by both their binding to RAB11 isoforms and their tumorigenic potential when overexpressed [65,83,84,85,86,87,88,89,90]. Although RAB11FIPs are associated with the recycling of pro-tumorigenic receptors, they do not directly bind surface oncogenic proteins even if both molecular species were found in the same macromolecular complexes [65,91]. Therefore, the RAB11-RAB11FIPs interaction has the purpose of linking recycling vesicles to cytoskeletal transport and tethering machineries rather than regulating receptor sorting.
In FERARI, the receptor sorting function is executed by SNX1, a component of the FERARI’s fission module. SNX1 is a member of the sorting nexin (SNX) protein family that includes 33 distinct genes which share the ability to associate with intracellular membranes containing phosphatidylinositol 3-phosphate (PtdIns(3P)), through their lipid-binding PX domain [92,93]. Moreover, recent evidences indicate that SNX’s PX domain interacts with specific aminoacidic motifs present in plasma membrane receptors [94]. In addition, the presence of a BAR domain gives to SNX1 the ability to sense membrane curvature [95]. As a consequence, SNX1 coordinates sequence-dependent cargo recognition with membrane remodeling to generate cargo-enriched tubulo-vesicular transport carriers. Therefore, SNX1 provides an essential element to control both export and transport of numerous cargoes to cell surface. However, despite these biochemical indications, the role of this SNX1 in human cancers remains debated.
The activity of SNX1 in RAB11-mediated trafficking suggests that such sorting nexin functions as an oncogene by increasing the amount of transmembrane proteins at the plasma membrane when overexpressed. In marked contrast, SNX1 targets EGFR to the lysosome for degradation, thereby decreasing the amount of EGFR available for ligand binding at the cell surface when overexpressed [96]. This designates SNX1 as a potential tumor suppressor involved in transport from the early endosome to the lysosomal compartment [96]. Accordingly, SNX1 downmodulation increases receptor accumulation in endosomes thereby amplifying the mitogenic response downstream of EGFR [97]. Although experimental data appear incompatible with one another, as they link SNX1 with transport of membranes towards both the plasma membrane and lysosome, recent reports address this apparent contradiction.
SNX-1 was found to sort cargo away from degradation. In particular, SNX1 generates specific regions on endosome, termed microdomains, in which the degradative and recycling pathways occupy distinct portions of membrane surface [98]. The depletion of SNX1 mediates cargo degradation by concentrating ubiquitinated cargoes and organizing the activities for its degradation [98]. As a consequence, loss of SNX1 function might result in a decreased endocytic recycling and a parallel accumulation of cargoes into the degradative organelle. The ability of SNX1 to organize trafficking of signaling receptors by controlling the juxtaposition of adjacent microdomains highlights the relevance of sub-compartmental organization of the sorting endosome in membrane transport and, in particular, in RAB11-mediated trafficking. In theory, to preserve the ability of the early endosome to sort cargoes, the two distinct molecular machineries that direct incoming molecules either to the degradative or to the recycling pathway need to be maintained spatially segregated. If one takes over the entire endosome, sorting might fail and cargo will be misdirected [99]. In such a “winner takes all” process, mechanisms which promote the functional association between distinct membrane domains within the same endosome, will provide a novel theoretical framework over which to interpret the impact of genetic alterations in endocytic genes. In this context, it appears to be essential how RAB11 might cross-regulate assembly or disassembly of opposing microdomains.

6. RAB11 Sets the Time for PtdIns Conversion on Endosomal Membrane Microdomain

Endocytic Rabs including RAB4, RAB5 and RAB11 were among the first proteins to be recognized as markers of endosomal membrane microdomains [100]. These small GTPases mark, at various degrees, distinct portions of the early endosome’s surface, thereby revealing extensive compartmentalization within the same continuous membrane [19,101]. Such spatial segregation is ensured by the RAB “GTPase switch” which connects the localization of RAB proteins to their biological function with spatiotemporal precision. Therefore, by controlling the RAB activation cycle, distinct complexes with both different and mutually exclusive function can be connected, thus providing a strategy to control the biochemical identity of membrane both over time and across space. In this context, the direct visualization that RAB11 marks tiny regions of early endosome surface where recycling cargoes accumulate hint at a role for the RAB11 “GTPase switch” in endosomal sorting.
Inactivation of RAB11 reduces the number of recycling tubules emerging from the early endosome, thereby providing evidence that RAB11 activity determines the efficacy of receptor transport to the cell surface [17]. Interestingly, RAB11 activation is coupled with generation of PtdIns(3P), the early endosome resident lipid, a process that requires the phosphorylation of the PtdIns inositol moieties by lipid kinase enzymes [102]. In particular, several reports link the activation of RAB11 to the production of PtdIns(3P) by PI3KC2A, a member of the well-known oncogene PI3K [17,103,104]. Notably, PtdIns(3P) activates RAB11 and, as a consequence, the PI3KC2A depletion reduces RAB11-GTP level in cells. This, in turn, decreases the release of recycling vesicles from the early endosome [17].
Although PtdIns(3P) generation and the subsequent RAB11 activation are essential to induce the production of recycling vesicles from early endosomes, the parallel conversion of PtdIns(3P) to PtdIns defines the timing for fission of such recycling carries [17]. As an example, PtdIns(3P) accumulation blocks the recycling of receptors such as β1-integrin [105].
In cells, PtdIns(3P) accumulation necessitates the depletion of PtdIns(3P)-phosphatases, a protein family that is responsible for the conversion of PtdIns(3P) to PtdIns. The depletion of PtdIns(3P)-phosphatases, such as MTM1, increases PtdIns(3P) in endosomes, thereby resulting in delayed transport of RAB11-positive vesicles. Notably, active RAB11 interacts with MTM1 promoting the endosomal localization of MTM1 [17]. This event leads to failure in localizing SNX1 on the early endosome membrane preventing TfR recycling and delaying EGFR trafficking [106].
The functional interplay between RAB11, PtdIns(3P) and components of the endocytic recycling machinery is underlined by recent works on the COMMD/CCDC22/CCDC93 (CCC) complex. The CCC complex is required for both retromer- and retriever-dependent protein trafficking and hence it defines the transport of β1-Integrin, LDLR, and LRP1. Moreover, the CCC complex regulates the phosphorylation and endosomal recruitment of the PtdIns(3P) phosphatase MTMR2 and then it primes membrane for the subsequent hydrolysis of PtdIns(3P). Therefore, the CCC complex–MTMR2 interaction is pivotal for activation of the WASH complex which drives the generation of branched F-actin filaments on endosomal membranes and thus the fission of recycling tubules [75,107,108,109,110,111].
The ability of RAB11 to set the time for PtdIns conversion through changes in its nucleotide binding status links RAB11 activation cycle with both endosome dynamics and membrane surface proteome remodeling. Therefore, perturbation of RAB11 activity might be a strategy to alter the biochemical composition of cellular membranes paving the way for fine control of complex cellular processes (Figure 1A).
In the sections below, we will discuss recent attempts aimed at supporting the therapeutic potential of RAB11 pharmacological targeting in human cancers.

7. RAB11 Targeting for Human Cancer Treatment

The significant role played by RAB11-mediated trafficking pathways in human cancers indicate that RAB11 isoforms are important targets for drug development. However, given the presence of conserved structural features shared by the over 60 members composing the RAB protein family, it makes it difficult to develop RAB11 selective inhibitors. Therefore, current pharmacological approaches are designed to block either the localization of RAB11 at membranes or the binding of specific effector molecules rather than to inhibit RAB11 nucleotide binding ability (Table 5).
In particular, hypolipidemic agents such as Pitavastatin, Simvastatin, Fluvastatin block the generation of isoprenoid chains by 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMG-CoA reductase) and therefore both prenylation and membrane localization of Rab GTPase proteins including the RAB11 subfamily. Notably, both Pitavastatin and Simvastatin were found effective to reduce brain metastatic adaptation and outgrowth of breast cancer cells [43,114,115].
On the contrary, CDKI-73, a known cyclin-dependent kinase inhibitor, does not affect RAB11 membrane localization despite the fact that it was found to alter both endosome morphology and RAB11-mediated trafficking. Although the mechanism of action of this compound is currently unknown, it may be possible that CDKI-73 acts indirectly by perturbing endosome dynamics rather than activation/localization/effector binding of RAB11 isoforms [113].
Recently, a first generation of RAB11-mediated trafficking pathway inhibitors was developed targeting the RAB11:FIP binding interface, thereby enabling pharmacologic disruption of RAB25 and/or RAB11 signaling in cells. In particular, these inhibitors named RFP14, RFP24 and RFP26 were developed by engineering the RAB11 binding domain of RAB11FIP proteins and improving both their stability, cell-permeability and isoform selectivity (Table 5). Notably, these molecules were proven effective to block RAB11-mediated trafficking phenotypes in several cancer cell lines derived from breast and ovarian tissues [62].

8. Conclusions

RAB11 isoforms play crucial functions in cellular physiology by ensuring the maintenance of the biochemical composition of cellular membranes. A broad range of knowledge supports a role for these small GTPases in cancer progression. However, we are still struggling to understand the pathological implications of dysfunctional RAB11-mediated transport in tumors.
Novel ways to interpret RAB11 dysfunction comes from basic research studies. These reveal that proper RAB11 function requires—(i) assembly of large multiprotein complexes, (ii) cross-regulation between endosomal membrane domains with opposing functions, (iii) interlinked control of both RAB11 GTPase switch cycle and PtdIns(3)P metabolism.
We considered that research aimed at expanding and connecting these aspects will provide novel exciting mechanistic insights to understand how RAB11-mediated trafficking pathways sustain replenishment of the cell surface and hence they will offer novel insights into the role of RAB11 dysfunction in human cancers.

Author Contributions

All authors equally contributed to this work. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the internal grant program of the Italian Institute for Genomic Medicine.

Institutional Review Board Statement

This study did not involve humans or animals.

Informed Consent Statement

This study did not involve humans.

Data Availability Statement

This study did not report any data.

Conflicts of Interest

C.C.C. is inventor of a granted patent related to RAB11 activity assays (TO2014A000264).

References

  1. Mellman, I.; Yarden, Y. Endocytosis and cancer. Cold Spring Harb. Perspect. Biol. 2013, 5, a016949. [Google Scholar] [CrossRef] [Green Version]
  2. Schmid, S.L. Reciprocal regulation of signaling and endocytosis: Implications for the evolving cancer cell. J. Cell Biol. 2017, 216, 2623–2632. [Google Scholar] [CrossRef] [Green Version]
  3. Gopal Krishnan, P.D.; Golden, E.; Woodward, E.A.; Pavlos, N.J.; Blancafort, P. Rab GTPases: Emerging Oncogenes and Tumor Suppressive Regulators for the Editing of Survival Pathways in Cancer. Cancers 2020, 12, 259. [Google Scholar] [CrossRef] [Green Version]
  4. Goitre, L.; Trapani, E.; Trabalzini, L.; Retta, S.F. The Ras superfamily of small GTPases: The unlocked secrets. Methods Mol. Biol. 2014, 1120, 1–18. [Google Scholar] [CrossRef] [PubMed]
  5. Stenmark, H. Rab GTPases as coordinators of vesicle traffic. Nat. Rev. Mol. Cell Biol. 2009, 10, 513–525. [Google Scholar] [CrossRef] [PubMed]
  6. Hutagalung, A.H.; Novick, P.J. Role of Rab GTPases in membrane traffic and cell physiology. Physiol. Rev. 2011, 91, 119–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Tzeng, H.T.; Wang, Y.C. Rab-mediated vesicle trafficking in cancer. J. Biomed. Sci. 2016, 23, 70. [Google Scholar] [CrossRef] [Green Version]
  8. Wang, Y.; Wang, H.; Yao, H.; Li, C.; Fang, J.Y.; Xu, J. Regulation of PD-L1: Emerging Routes for Targeting Tumor Immune Evasion. Front. Pharmacol. 2018, 9, 536. [Google Scholar] [CrossRef]
  9. Welz, T.; Wellbourne-Wood, J.; Kerkhoff, E. Orchestration of cell surface proteins by Rab11. Trends Cell Biol. 2014, 24, 407–415. [Google Scholar] [CrossRef]
  10. Kumar, A.P.; Lukman, S. Allosteric binding sites in Rab11 for potential drug candidates. PLoS ONE 2018, 13, e0198632. [Google Scholar] [CrossRef]
  11. Scapin, S.M.; Carneiro, F.R.; Alves, A.C.; Medrano, F.J.; Guimaraes, B.G.; Zanchin, N.I. The crystal structure of the small GTPase Rab11b reveals critical differences relative to the Rab11a isoform. J. Struct. Biol. 2006, 154, 260–268. [Google Scholar] [CrossRef] [PubMed]
  12. Kelly, E.E.; Horgan, C.P.; McCaffrey, M.W. Rab11 proteins in health and disease. Biochem. Soc. Trans. 2012, 40, 1360–1367. [Google Scholar] [CrossRef] [PubMed]
  13. Sobajima, T.; Yoshimura, S.; Iwano, T.; Kunii, M.; Watanabe, M.; Atik, N.; Mushiake, S.; Morii, E.; Koyama, Y.; Miyoshi, E.; et al. Rab11a is required for apical protein localisation in the intestine. Biol. Open 2014, 4, 86–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Yu, S.; Yehia, G.; Wang, J.; Stypulkowski, E.; Sakamori, R.; Jiang, P.; Hernandez-Enriquez, B.; Tran, T.S.; Bonder, E.M.; Guo, W.; et al. Global ablation of the mouse Rab11a gene impairs early embryogenesis and matrix metalloproteinase secretion. J. Biol. Chem. 2014, 289, 32030–32043. [Google Scholar] [CrossRef] [Green Version]
  15. Maxfield, F.R.; McGraw, T.E. Endocytic recycling. Nat. Rev. Mol. Cell Biol. 2004, 5, 121–132. [Google Scholar] [CrossRef]
  16. Ullrich, O.; Reinsch, S.; Urbe, S.; Zerial, M.; Parton, R.G. Rab11 regulates recycling through the pericentriolar recycling endosome. J. Cell Biol. 1996, 135, 913–924. [Google Scholar] [CrossRef]
  17. Campa, C.C.; Margaria, J.P.; Derle, A.; Del Giudice, M.; De Santis, M.C.; Gozzelino, L.; Copperi, F.; Bosia, C.; Hirsch, E. Rab11 activity and PtdIns(3)P turnover removes recycling cargo from endosomes. Nat. Chem. Biol. 2018, 14, 801–810. [Google Scholar] [CrossRef]
  18. Wang, X.; Kumar, R.; Navarre, J.; Casanova, J.E.; Goldenring, J.R. Regulation of vesicle trafficking in madin-darby canine kidney cells by Rab11a and Rab25. J. Biol. Chem. 2000, 275, 29138–29146. [Google Scholar] [CrossRef] [Green Version]
  19. Redpath, G.M.I.; Ecker, M.; Kapoor-Kaushik, N.; Vartoukian, H.; Carnell, M.; Kempe, D.; Biro, M.; Ariotti, N.; Rossy, J. Flotillins promote T cell receptor sorting through a fast Rab5-Rab11 endocytic recycling axis. Nat. Commun. 2019, 10, 4392. [Google Scholar] [CrossRef] [Green Version]
  20. Puri, C.; Vicinanza, M.; Ashkenazi, A.; Gratian, M.J.; Zhang, Q.; Bento, C.F.; Renna, M.; Menzies, F.M.; Rubinsztein, D.C. The RAB11A-Positive Compartment Is a Primary Platform for Autophagosome Assembly Mediated by WIPI2 Recognition of PI3P-RAB11A. Dev. Cell 2018, 45, 114–131.e118. [Google Scholar] [CrossRef] [Green Version]
  21. Walia, V.; Cuenca, A.; Vetter, M.; Insinna, C.; Perera, S.; Lu, Q.; Ritt, D.A.; Semler, E.; Specht, S.; Stauffer, J.; et al. Akt Regulates a Rab11-Effector Switch Required for Ciliogenesis. Dev. Cell 2019, 50, 229–246.e227. [Google Scholar] [CrossRef] [PubMed]
  22. Yeo, J.C.; Wall, A.A.; Luo, L.; Stow, J.L. Sequential recruitment of Rab GTPases during early stages of phagocytosis. Cell. Logist. 2016, 6, e1140615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Jiang, C.; Liu, Z.; Hu, R.; Bo, L.; Minshall, R.D.; Malik, A.B.; Hu, G. Inactivation of Rab11a GTPase in Macrophages Facilitates Phagocytosis of Apoptotic Neutrophils. J. Immunol. 2017, 198, 1660–1672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Niu, F.; Sun, K.; Wei, W.; Yu, C.; Wei, Z. F-actin disassembly factor MICAL1 binding to Myosin Va mediates cargo unloading during cytokinesis. Sci. Adv. 2020, 6. [Google Scholar] [CrossRef] [PubMed]
  25. Assaker, G.; Ramel, D.; Wculek, S.K.; Gonzalez-Gaitan, M.; Emery, G. Spatial restriction of receptor tyrosine kinase activity through a polarized endocytic cycle controls border cell migration. Proc. Natl. Acad. Sci. USA 2010, 107, 22558–22563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Takahashi, S.; Kubo, K.; Waguri, S.; Yabashi, A.; Shin, H.W.; Katoh, Y.; Nakayama, K. Rab11 regulates exocytosis of recycling vesicles at the plasma membrane. J. Cell Sci. 2012, 125, 4049–4057. [Google Scholar] [CrossRef] [Green Version]
  27. Haugsten, E.M.; Brech, A.; Liestol, K.; Norman, J.C.; Wesche, J. Photoactivation approaches reveal a role for Rab11 in FGFR4 recycling and signalling. Traffic 2014, 15, 665–683. [Google Scholar] [CrossRef] [Green Version]
  28. Lapierre, L.A.; Dorn, M.C.; Zimmerman, C.F.; Navarre, J.; Burnette, J.O.; Goldenring, J.R. Rab11b resides in a vesicular compartment distinct from Rab11a in parietal cells and other epithelial cells. Exp. Cell Res. 2003, 290, 322–331. [Google Scholar] [CrossRef]
  29. Grimsey, N.J.; Coronel, L.J.; Cordova, I.C.; Trejo, J. Recycling and Endosomal Sorting of Protease-activated Receptor-1 Is Distinctly Regulated by Rab11A and Rab11B Proteins. J. Biol. Chem. 2016, 291, 2223–2236. [Google Scholar] [CrossRef] [Green Version]
  30. Silvis, M.R.; Bertrand, C.A.; Ameen, N.; Golin-Bisello, F.; Butterworth, M.B.; Frizzell, R.A.; Bradbury, N.A. Rab11b regulates the apical recycling of the cystic fibrosis transmembrane conductance regulator in polarized intestinal epithelial cells. Mol. Biol. Cell 2009, 20, 2337–2350. [Google Scholar] [CrossRef] [Green Version]
  31. Khvotchev, M.V.; Ren, M.; Takamori, S.; Jahn, R.; Sudhof, T.C. Divergent functions of neuronal Rab11b in Ca2+-regulated versus constitutive exocytosis. J. Neurosci. 2003, 23, 10531–10539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Nam, K.T.; Lee, H.J.; Smith, J.J.; Lapierre, L.A.; Kamath, V.P.; Chen, X.; Aronow, B.J.; Yeatman, T.J.; Bhartur, S.G.; Calhoun, B.C.; et al. Loss of Rab25 promotes the development of intestinal neoplasia in mice and is associated with human colorectal adenocarcinomas. J. Clin. Investig. 2010, 120, 840–849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Dozynkiewicz, M.A.; Jamieson, N.B.; Macpherson, I.; Grindlay, J.; van den Berghe, P.V.; von Thun, A.; Morton, J.P.; Gourley, C.; Timpson, P.; Nixon, C.; et al. Rab25 and CLIC3 collaborate to promote integrin recycling from late endosomes/lysosomes and drive cancer progression. Dev. Cell 2012, 22, 131–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Cheng, K.W.; Agarwal, R.; Mitra, S.; Lee, J.S.; Carey, M.; Gray, J.W.; Mills, G.B. Rab25 increases cellular ATP and glycogen stores protecting cancer cells from bioenergetic stress. Embo Mol. Med. 2012, 4, 125–141. [Google Scholar] [CrossRef]
  35. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  36. Porther, N.; Barbieri, M.A. The role of endocytic Rab GTPases in regulation of growth factor signaling and the migration and invasion of tumor cells. Small Gtpases 2015, 6, 135–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Wandinger-Ness, A.; Zerial, M. Rab proteins and the compartmentalization of the endosomal system. Cold Spring Harb. Perspect. Biol. 2014, 6, a022616. [Google Scholar] [CrossRef]
  38. Palmieri, D.; Bouadis, A.; Ronchetti, R.; Merino, M.J.; Steeg, P.S. Rab11a differentially modulates epidermal growth factor-induced proliferation and motility in immortal breast cells. Breast Cancer Res. Treat. 2006, 100, 127–137. [Google Scholar] [CrossRef] [PubMed]
  39. Dong, Q.; Fu, L.; Zhao, Y.; Du, Y.; Li, Q.; Qiu, X.; Wang, E. Rab11a promotes proliferation and invasion through regulation of YAP in non-small cell lung cancer. Oncotarget 2017, 8, 27800–27811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Zhang, Z.Y.; Lu, M.; Liu, Z.K.; Li, H.; Yong, Y.L.; Zhang, R.Y.; Chen, Z.N.; Bian, H. Rab11a regulates MMP2 expression by activating the PI3K/AKT pathway in human hepatocellular carcinoma cells. Pathol. Res. Pract. 2020, 216, 153046. [Google Scholar] [CrossRef]
  41. Yu, L.; Li, X.; Li, H.; Chen, H.; Liu, H. Rab11a sustains GSK3beta/Wnt/beta-catenin signaling to enhance cancer progression in pancreatic cancer. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2016, 37, 13821–13829. [Google Scholar] [CrossRef] [PubMed]
  42. D’Agostino, L.; Nie, Y.; Goswami, S.; Tong, K.; Yu, S.; Bandyopadhyay, S.; Flores, J.; Zhang, X.; Balasubramanian, I.; Joseph, I.; et al. Recycling Endosomes in Mature Epithelia Restrain Tumorigenic Signaling. Cancer Res. 2019, 79, 4099–4112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Howe, E.N.; Burnette, M.D.; Justice, M.E.; Schnepp, P.M.; Hedrick, V.; Clancy, J.W.; Guldner, I.H.; Lamere, A.T.; Li, J.; Aryal, U.K.; et al. Rab11b-mediated integrin recycling promotes brain metastatic adaptation and outgrowth. Nat. Commun. 2020, 11, 3017. [Google Scholar] [CrossRef] [PubMed]
  44. Cho, K.H.; Lee, H.Y. Rab25 and RCP in cancer progression. Arch. Pharmacal Res. 2019, 42, 101–112. [Google Scholar] [CrossRef] [PubMed]
  45. Mor, O.; Nativ, O.; Stein, A.; Novak, L.; Lehavi, D.; Shiboleth, Y.; Rozen, A.; Berent, E.; Brodsky, L.; Feinstein, E.; et al. Molecular analysis of transitional cell carcinoma using cDNA microarray. Oncogene 2003, 22, 7702–7710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Zhang, J.; Wei, J.; Lu, J.; Tong, Z.; Liao, B.; Yu, B.; Zheng, F.; Huang, X.; Chen, Z.; Fang, Y.; et al. Overexpression of Rab25 contributes to metastasis of bladder cancer through induction of epithelial-mesenchymal transition and activation of Akt/GSK-3beta/Snail signaling. Carcinogenesis 2013, 34, 2401–2408. [Google Scholar] [CrossRef] [Green Version]
  47. Cao, C.; Lu, C.; Xu, J.; Zhang, J.; Zhang, J.; Li, M. Expression of Rab25 correlates with the invasion and metastasis of gastric cancer. Chin. J. Cancer Res. Chung Kuo Yen Cheng Yen Chiu 2013, 25, 192–199. [Google Scholar] [CrossRef]
  48. Jeong, B.Y.; Cho, K.H.; Jeong, K.J.; Park, Y.Y.; Kim, J.M.; Rha, S.Y.; Park, C.G.; Mills, G.B.; Cheong, J.H.; Lee, H.Y. Rab25 augments cancer cell invasiveness through a beta1 integrin/EGFR/VEGF-A/Snail signaling axis and expression of fascin. Exp. Mol. Med. 2018, 50, e435. [Google Scholar] [CrossRef]
  49. Ding, B.; Cui, B.; Gao, M.; Li, Z.; Xu, C.; Fan, S.; He, W. Knockdown of Ras-Related Protein 25 (Rab25) Inhibits the In Vitro Cytotoxicity and In Vivo Antitumor Activity of Human Glioblastoma Multiforme Cells. Oncol. Res. 2017, 25, 331–340. [Google Scholar] [CrossRef]
  50. Geng, D.; Zhao, W.; Feng, Y.; Liu, J. Overexpression of Rab25 promotes hepatocellular carcinoma cell proliferation and invasion. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2016, 37, 7713–7718. [Google Scholar] [CrossRef]
  51. 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. The RAB25 small GTPase determines aggressiveness of ovarian and breast cancers. Nat. Med. 2004, 10, 1251–1256. [Google Scholar] [CrossRef] [PubMed]
  52. Ma, Y.F.; Yang, B.; Li, J.; Zhang, T.; Guo, J.T.; Chen, L.; Li, M.; Chu, J.; Liang, C.Y.; Liu, Y. Expression of Ras-related protein 25 predicts chemotherapy resistance and prognosis in advanced non-small cell lung cancer. Genet. Mol. Res. 2015, 14, 13998–14008. [Google Scholar] [CrossRef]
  53. Jo, U.; Park, K.H.; Whang, Y.M.; Sung, J.S.; Won, N.H.; Park, J.K.; Kim, Y.H. EGFR endocytosis is a novel therapeutic target in lung cancer with wild-type EGFR. Oncotarget 2014, 5, 1265–1278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kessler, D.; Gruen, G.C.; Heider, D.; Morgner, J.; Reis, H.; Schmid, K.W.; Jendrossek, V. The action of small GTPases Rab11 and Rab25 in vesicle trafficking during cell migration. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2012, 29, 647–656. [Google Scholar] [CrossRef]
  55. Hu, C.; Chen, B.; Zhou, Y.; Shan, Y. High expression of Rab25 contributes to malignant phenotypes and biochemical recurrence in patients with prostate cancer after radical prostatectomy. Cancer Cell Int. 2017, 17, 45. [Google Scholar] [CrossRef] [PubMed]
  56. Natrajan, R.; Williams, R.D.; Hing, S.N.; Mackay, A.; Reis-Filho, J.S.; Fenwick, K.; Iravani, M.; Valgeirsson, H.; Grigoriadis, A.; Langford, C.F.; et al. Array CGH profiling of favourable histology Wilms tumours reveals novel gains and losses associated with relapse. J. Pathol. 2006, 210, 49–58. [Google Scholar] [CrossRef]
  57. Tong, M.; Chan, K.W.; Bao, J.Y.; Wong, K.Y.; Chen, J.N.; Kwan, P.S.; Tang, K.H.; Fu, L.; Qin, Y.R.; Lok, S.; et al. Rab25 is a tumor suppressor gene with antiangiogenic and anti-invasive activities in esophageal squamous cell carcinoma. Cancer Res. 2012, 72, 6024–6035. [Google Scholar] [CrossRef] [Green Version]
  58. Amornphimoltham, P.; Rechache, K.; Thompson, J.; Masedunskas, A.; Leelahavanichkul, K.; Patel, V.; Molinolo, A.; Gutkind, J.S.; Weigert, R. Rab25 regulates invasion and metastasis in head and neck cancer. Clin. Cancer Res. 2013, 19, 1375–1388. [Google Scholar] [CrossRef] [Green Version]
  59. Cheng, J.M.; Ding, M.; Aribi, A.; Shah, P.; Rao, K. Loss of RAB25 expression in breast cancer. Int. J. Cancer 2006, 118, 2957–2964. [Google Scholar] [CrossRef]
  60. Yu, S.; Nie, Y.; Knowles, B.; Sakamori, R.; Stypulkowski, E.; Patel, C.; Das, S.; Douard, V.; Ferraris, R.P.; Bonder, E.M.; et al. TLR sorting by Rab11 endosomes maintains intestinal epithelial-microbial homeostasis. Embo J. 2014, 33, 1882–1895. [Google Scholar] [CrossRef]
  61. Zhang, L.; Xie, B.; Qiu, Y.; Jing, D.; Zhang, J.; Duan, Y.; Li, Z.; Fan, M.; He, J.; Qiu, Y.; et al. Rab25-Mediated EGFR Recycling Causes Tumor Acquired Radioresistance. iScience 2020, 23, 100997. [Google Scholar] [CrossRef] [PubMed]
  62. Mitra, S.; Montgomery, J.E.; Kolar, M.J.; Li, G.; Jeong, K.J.; Peng, B.; Verdine, G.L.; Mills, G.B.; Moellering, R.E. Stapled peptide inhibitors of RAB25 target context-specific phenotypes in cancer. Nat. Commun. 2017, 8, 660. [Google Scholar] [CrossRef] [PubMed]
  63. Mitra, S.; Federico, L.; Zhao, W.; Dennison, J.; Sarkar, T.R.; Zhang, F.; Takiar, V.; Cheng, K.W.; Mani, S.; Lee, J.S.; et al. Rab25 acts as an oncogene in luminal B breast cancer and is causally associated with Snail driven EMT. Oncotarget 2016, 7, 40252–40265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Cheng, J.M.; Volk, L.; Janaki, D.K.; Vyakaranam, S.; Ran, S.; Rao, K.A. Tumor suppressor function of Rab25 in triple-negative breast cancer. Int. J. Cancer 2010, 126, 2799–2812. [Google Scholar] [CrossRef]
  65. Caswell, P.T.; Chan, M.; Lindsay, A.J.; McCaffrey, M.W.; Boettiger, D.; Norman, J.C. Rab-coupling protein coordinates recycling of alpha5beta1 integrin and EGFR1 to promote cell migration in 3D microenvironments. J. Cell Biol. 2008, 183, 143–155. [Google Scholar] [CrossRef] [Green Version]
  66. Muller, P.A.; Caswell, P.T.; Doyle, B.; Iwanicki, M.P.; Tan, E.H.; Karim, S.; Lukashchuk, N.; Gillespie, D.A.; Ludwig, R.L.; Gosselin, P.; et al. Mutant p53 drives invasion by promoting integrin recycling. Cell 2009, 139, 1327–1341. [Google Scholar] [CrossRef]
  67. Caswell, P.T.; Spence, H.J.; Parsons, M.; White, D.P.; Clark, K.; Cheng, K.W.; Mills, G.B.; Humphries, M.J.; Messent, A.J.; Anderson, K.I.; et al. Rab25 associates with alpha5beta1 integrin to promote invasive migration in 3D microenvironments. Dev. Cell 2007, 13, 496–510. [Google Scholar] [CrossRef]
  68. Chung, Y.C.; Wei, W.C.; Hung, C.N.; Kuo, J.F.; Hsu, C.P.; Chang, K.J.; Chao, W.T. Rab11 collaborates E-cadherin to promote collective cell migration and indicates a poor prognosis in colorectal carcinoma. Eur. J. Clin. Investig. 2016, 46, 1002–1011. [Google Scholar] [CrossRef]
  69. Koseska, A.; Bastiaens, P.I.H. Processing Temporal Growth Factor Patterns by an Epidermal Growth Factor Receptor Network Dynamically Established in Space. Annu. Rev. Cell Dev. Biol. 2020, 36, 359–383. [Google Scholar] [CrossRef]
  70. York, H.M.; Coyle, J.; Arumugam, S. To be more precise: The role of intracellular trafficking in development and pattern formation. Biochem. Soc. Trans. 2020, 48, 2051–2066. [Google Scholar] [CrossRef]
  71. Wang, J.; Fedoseienko, A.; Chen, B.; Burstein, E.; Jia, D.; Billadeau, D.D. Endosomal receptor trafficking: Retromer and beyond. Traffic 2018, 19, 578–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Vergés, M.; Luton, F.; Gruber, C.; Tiemann, F.; Reinders, L.G.; Huang, L.; Burlingame, A.L.; Haft, C.R.; Mostov, K.E. The mammalian retromer regulates transcytosis of the polymeric immunoglobulin receptor. Nat. Cell Biol. 2004, 6, 763–769. [Google Scholar] [CrossRef] [PubMed]
  73. Harterink, M.; Port, F.; Lorenowicz, M.J.; McGough, I.J.; Silhankova, M.; Betist, M.C.; van Weering, J.R.T.; van Heesbeen, R.; Middelkoop, T.C.; Basler, K.; et al. A SNX3-dependent retromer pathway mediates retrograde transport of the Wnt sorting receptor Wntless and is required for Wnt secretion. Nat. Cell Biol. 2011, 13, 914–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Steinberg, F.; Gallon, M.; Winfield, M.; Thomas, E.C.; Bell, A.J.; Heesom, K.J.; Tavaré, J.M.; Cullen, P.J. A global analysis of SNX27-retromer assembly and cargo specificity reveals a function in glucose and metal ion transport. Nat. Cell Biol. 2013, 15, 461–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. McNally, K.E.; Faulkner, R.; Steinberg, F.; Gallon, M.; Ghai, R.; Pim, D.; Langton, P.; Pearson, N.; Danson, C.M.; Nagele, H.; et al. Retriever is a multiprotein complex for retromer-independent endosomal cargo recycling. Nat. Cell Biol. 2017, 19, 1214–1225. [Google Scholar] [CrossRef]
  76. Zech, T.; Calaminus, S.D.; Caswell, P.; Spence, H.J.; Carnell, M.; Insall, R.H.; Norman, J.; Machesky, L.M. The Arp2/3 activator WASH regulates α5β1-integrin-mediated invasive migration. J. Cell Sci. 2011, 124, 3753–3759. [Google Scholar] [CrossRef] [Green Version]
  77. Gomez, T.S.; Billadeau, D.D. A FAM21-containing WASH complex regulates retromer-dependent sorting. Dev. Cell 2009, 17, 699–711. [Google Scholar] [CrossRef] [Green Version]
  78. Derivery, E.; Sousa, C.; Gautier, J.J.; Lombard, B.; Loew, D.; Gautreau, A. The Arp2/3 activator WASH controls the fission of endosomes through a large multiprotein complex. Dev. Cell 2009, 17, 712–723. [Google Scholar] [CrossRef] [Green Version]
  79. Jia, D.; Gomez, T.S.; Metlagel, Z.; Umetani, J.; Otwinowski, Z.; Rosen, M.K.; Billadeau, D.D. WASH and WAVE actin regulators of the Wiskott-Aldrich syndrome protein (WASP) family are controlled by analogous structurally related complexes. Proc. Natl. Acad. Sci. USA 2010, 107, 10442–10447. [Google Scholar] [CrossRef] [Green Version]
  80. Hao, Y.H.; Doyle, J.M.; Ramanathan, S.; Gomez, T.S.; Jia, D.; Xu, M.; Chen, Z.J.; Billadeau, D.D.; Rosen, M.K.; Potts, P.R. Regulation of WASH-dependent actin polymerization and protein trafficking by ubiquitination. Cell 2013, 152, 1051–1064. [Google Scholar] [CrossRef] [Green Version]
  81. Solinger, J.A.; Rashid, H.O.; Prescianotto-Baschong, C.; Spang, A. FERARI is required for Rab11-dependent endocytic recycling. Nat. Cell Biol. 2020, 22, 213–224. [Google Scholar] [CrossRef] [PubMed]
  82. Morrison, H.A.; Dionne, H.; Rusten, T.E.; Brech, A.; Fisher, W.W.; Pfeiffer, B.D.; Celniker, S.E.; Stenmark, H.; Bilder, D. Regulation of early endosomal entry by the Drosophila tumor suppressors Rabenosyn and Vps45. Mol. Biol. Cell 2008, 19, 4167–4176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Hwang, M.H.; Cho, K.H.; Jeong, K.J.; Park, Y.Y.; Kim, J.M.; Yu, S.L.; Park, C.G.; Mills, G.B.; Lee, H.Y. RCP induces Slug expression and cancer cell invasion by stabilizing β1 integrin. Oncogene 2017, 36, 1102–1111. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, J.; Liu, X.; Datta, A.; Govindarajan, K.; Tam, W.L.; Han, J.; George, J.; Wong, C.; Ramnarayanan, K.; Phua, T.Y.; et al. RCP is a human breast cancer-promoting gene with Ras-activating function. J. Clin. Investig. 2009, 119, 2171–2183. [Google Scholar] [CrossRef]
  85. Gundry, C.; Marco, S.; Rainero, E.; Miller, B.; Dornier, E.; Mitchell, L.; Caswell, P.T. Phosphorylation of Rab-coupling protein by LMTK3 controls Rab14-dependent EphA2 trafficking to promote cell:cell repulsion. Nat. Commun. 2017, 8, 14646. [Google Scholar] [CrossRef]
  86. Tong, D.; Liang, Y.N.; Stepanova, A.A.; Liu, Y.; Li, X.; Wang, L.; Zhang, F.; Vasilyeva, N.V. Increased Eps15 homology domain 1 and RAB11FIP3 expression regulate breast cancer progression via promoting epithelial growth factor receptor recycling. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2017, 39, 1010428317691010. [Google Scholar] [CrossRef] [Green Version]
  87. March, H.N.; Rust, A.G.; Wright, N.A.; ten Hoeve, J.; de Ridder, J.; Eldridge, M.; van der Weyden, L.; Berns, A.; Gadiot, J.; Uren, A.; et al. Insertional mutagenesis identifies multiple networks of cooperating genes driving intestinal tumorigenesis. Nat. Genet. 2011, 43, 1202–1209. [Google Scholar] [CrossRef] [Green Version]
  88. Takeda, H.; Rust, A.G.; Ward, J.M.; Yew, C.C.; Jenkins, N.A.; Copeland, N.G. Sleeping Beauty transposon mutagenesis identifies genes that cooperate with mutant Smad4 in gastric cancer development. Proc. Natl. Acad. Sci. USA 2016, 113, E2057–E2065. [Google Scholar] [CrossRef] [Green Version]
  89. Das, L.; Gard, J.M.C.; Prekeris, R.; Nagle, R.B.; Morrissey, C.; Knudsen, B.S.; Miranti, C.K.; Cress, A.E. Novel Regulation of Integrin Trafficking by Rab11-FIP5 in Aggressive Prostate Cancer. Mol. Cancer Res. 2018, 16, 1319–1331. [Google Scholar] [CrossRef] [Green Version]
  90. Hu, F.; Deng, X.; Yang, X.; Jin, H.; Gu, D.; Lv, X.; Wang, C.; Zhang, Y.; Huo, X.; Shen, Q.; et al. Hypoxia upregulates Rab11-family interacting protein 4 through HIF-1α to promote the metastasis of hepatocellular carcinoma. Oncogene 2015, 34, 6007–6017. [Google Scholar] [CrossRef]
  91. Muller, P.A.; Trinidad, A.G.; Timpson, P.; Morton, J.P.; Zanivan, S.; van den Berghe, P.V.; Nixon, C.; Karim, S.A.; Caswell, P.T.; Noll, J.E.; et al. Mutant p53 enhances MET trafficking and signalling to drive cell scattering and invasion. Oncogene 2013, 32, 1252–1265. [Google Scholar] [CrossRef] [Green Version]
  92. Worby, C.A.; Dixon, J.E. Sorting out the cellular functions of sorting nexins. Nat. Rev. Mol. Cell Biol. 2002, 3, 919–931. [Google Scholar] [CrossRef] [PubMed]
  93. Cozier, G.E.; Carlton, J.; McGregor, A.H.; Gleeson, P.A.; Teasdale, R.D.; Mellor, H.; Cullen, P.J. The phox homology (PX) domain-dependent, 3-phosphoinositide-mediated association of sorting nexin-1 with an early sorting endosomal compartment is required for its ability to regulate epidermal growth factor receptor degradation. J. Biol. Chem. 2002, 277, 48730–48736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Simonetti, B.; Paul, B.; Chaudhari, K.; Weeratunga, S.; Steinberg, F.; Gorla, M.; Heesom, K.J.; Bashaw, G.J.; Collins, B.M.; Cullen, P.J. Molecular identification of a BAR domain-containing coat complex for endosomal recycling of transmembrane proteins. Nat. Cell Biol. 2019, 21, 1219–1233. [Google Scholar] [CrossRef] [PubMed]
  95. Carlton, J.; Bujny, M.; Peter, B.J.; Oorschot, V.M.; Rutherford, A.; Mellor, H.; Klumperman, J.; McMahon, H.T.; Cullen, P.J. Sorting nexin-1 mediates tubular endosome-to-TGN transport through coincidence sensing of high- curvature membranes and 3-phosphoinositides. Curr. Biol. 2004, 14, 1791–1800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Kurten, R.C.; Cadena, D.L.; Gill, G.N. Enhanced degradation of EGF receptors by a sorting nexin, SNX1. Science 1996, 272, 1008–1010. [Google Scholar] [CrossRef]
  97. Nguyen, L.N.; Holdren, M.S.; Nguyen, A.P.; Furuya, M.H.; Bianchini, M.; Levy, E.; Mordoh, J.; Liu, A.; Guncay, G.D.; Campbell, J.S.; et al. Sorting nexin 1 down-regulation promotes colon tumorigenesis. Clin. Cancer Res. 2006, 12, 6952–6959. [Google Scholar] [CrossRef] [Green Version]
  98. Norris, A.; Tammineni, P.; Wang, S.; Gerdes, J.; Murr, A.; Kwan, K.Y.; Cai, Q.; Grant, B.D. SNX-1 and RME-8 oppose the assembly of HGRS-1/ESCRT-0 degradative microdomains on endosomes. Proc. Natl. Acad. Sci. USA 2017, 114, E307–E316. [Google Scholar] [CrossRef] [Green Version]
  99. Norris, A.; Grant, B.D. Endosomal microdomains: Formation and function. Curr. Opin. Cell Biol. 2020, 65, 86–95. [Google Scholar] [CrossRef]
  100. Sönnichsen, B.; De Renzis, S.; Nielsen, E.; Rietdorf, J.; Zerial, M. Distinct membrane domains on endosomes in the recycling pathway visualized by multicolor imaging of Rab4, Rab5, and Rab11. J. Cell Biol. 2000, 149, 901–914. [Google Scholar] [CrossRef]
  101. Franke, C.; Repnik, U.; Segeletz, S.; Brouilly, N.; Kalaidzidis, Y.; Verbavatz, J.M.; Zerial, M. Correlative single-molecule localization microscopy and electron tomography reveals endosome nanoscale domains. Traffic 2019, 20, 601–617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Campa, C.C.; Franco, I.; Hirsch, E. PI3K-C2α: One enzyme for two products coupling vesicle trafficking and signal transduction. FEBS Lett. 2015, 589, 1552–1558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Franco, I.; Gulluni, F.; Campa, C.C.; Costa, C.; Margaria, J.P.; Ciraolo, E.; Martini, M.; Monteyne, D.; De Luca, E.; Germena, G.; et al. PI3K class II α controls spatially restricted endosomal PtdIns3P and Rab11 activation to promote primary cilium function. Dev. Cell 2014, 28, 647–658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Franco, I.; Margaria, J.P.; De Santis, M.C.; Ranghino, A.; Monteyne, D.; Chiaravalli, M.; Pema, M.; Campa, C.C.; Ratto, E.; Gulluni, F.; et al. Phosphoinositide 3-Kinase-C2α Regulates Polycystin-2 Ciliary Entry and Protects against Kidney Cyst Formation. J. Am. Soc. Nephrol. JASN 2016, 27, 1135–1144. [Google Scholar] [CrossRef] [Green Version]
  105. Ketel, K.; Krauss, M.; Nicot, A.S.; Puchkov, D.; Wieffer, M.; Müller, R.; Subramanian, D.; Schultz, C.; Laporte, J.; Haucke, V. A phosphoinositide conversion mechanism for exit from endosomes. Nature 2016, 529, 408–412. [Google Scholar] [CrossRef] [PubMed]
  106. Fili, N.; Calleja, V.; Woscholski, R.; Parker, P.J.; Larijani, B. Compartmental signal modulation: Endosomal phosphatidylinositol 3-phosphate controls endosome morphology and selective cargo sorting. Proc. Natl. Acad. Sci. USA 2006, 103, 15473–15478. [Google Scholar] [CrossRef] [Green Version]
  107. Phillips-Krawczak, C.A.; Singla, A.; Starokadomskyy, P.; Deng, Z.; Osborne, D.G.; Li, H.; Dick, C.J.; Gomez, T.S.; Koenecke, M.; Zhang, J.S.; et al. COMMD1 is linked to the WASH complex and regulates endosomal trafficking of the copper transporter ATP7A. Mol. Biol. Cell 2015, 26, 91–103. [Google Scholar] [CrossRef] [Green Version]
  108. Starokadomskyy, P.; Gluck, N.; Li, H.; Chen, B.; Wallis, M.; Maine, G.N.; Mao, X.; Zaidi, I.W.; Hein, M.Y.; McDonald, F.J.; et al. CCDC22 deficiency in humans blunts activation of proinflammatory NF-κB signaling. J. Clin. Investig. 2013, 123, 2244–2256. [Google Scholar] [CrossRef] [Green Version]
  109. Bartuzi, P.; Billadeau, D.D.; Favier, R.; Rong, S.; Dekker, D.; Fedoseienko, A.; Fieten, H.; Wijers, M.; Levels, J.H.; Huijkman, N.; et al. CCC- and WASH-mediated endosomal sorting of LDLR is required for normal clearance of circulating LDL. Nat. Commun. 2016, 7, 10961. [Google Scholar] [CrossRef]
  110. Fedoseienko, A.; Wijers, M.; Wolters, J.C.; Dekker, D.; Smit, M.; Huijkman, N.; Kloosterhuis, N.; Klug, H.; Schepers, A.; Willems van Dijk, K.; et al. The COMMD Family Regulates Plasma LDL Levels and Attenuates Atherosclerosis Through Stabilizing the CCC Complex in Endosomal LDLR Trafficking. Circ. Res. 2018, 122, 1648–1660. [Google Scholar] [CrossRef]
  111. Singla, A.; Fedoseienko, A.; Giridharan, S.S.P.; Overlee, B.L.; Lopez, A.; Jia, D.; Song, J.; Huff-Hardy, K.; Weisman, L.; Burstein, E.; et al. Endosomal PI(3)P regulation by the COMMD/CCDC22/CCDC93 (CCC) complex controls membrane protein recycling. Nat. Commun. 2019, 10, 4271. [Google Scholar] [CrossRef] [Green Version]
  112. Ronzier, E.; Parks, X.X.; Qudsi, H.; Lopes, C.M. Statin-specific inhibition of Rab-GTPase regulates cPKC-mediated IKs internalization. Sci. Rep. 2019, 9, 17747. [Google Scholar] [CrossRef] [Green Version]
  113. Sorvina, A.; Shandala, T.; Wang, S.; Sharkey, D.J.; Parkinson-Lawrence, E.; Selemidis, S.; Brooks, D.A. CDKI-73 is a Novel Pharmacological Inhibitor of Rab11 Cargo Delivery and Innate Immune Secretion. Cells 2020, 9, 372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Borgquist, S.; Broberg, P.; Tojjar, J.; Olsson, H. Statin use and breast cancer survival—A Swedish nationwide study. BMC Cancer 2019, 19, 54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Nielsen, S.F.; Nordestgaard, B.G.; Bojesen, S.E. Statin use and reduced cancer-related mortality. N. Engl. J. Med. 2012, 367, 1792–1802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Overview of the main cellular processes and trafficking pathways regulated by RAB11 isoforms. (A) Schematic representation of cellular processes controlled by RAB11. Black lines represent cells’ contour, green arrows represent directions of RAB11-mediated trafficking pathways. (B) Schematic representation of RAB11 isoform localization in both endocytic (left panel) and exocytic pathways (right panel). Green circles represent RAB11-positive compartments, yellow, blue, red lines represent RAB11A, RAB11B and RAB25, respectively.
Figure 1. Overview of the main cellular processes and trafficking pathways regulated by RAB11 isoforms. (A) Schematic representation of cellular processes controlled by RAB11. Black lines represent cells’ contour, green arrows represent directions of RAB11-mediated trafficking pathways. (B) Schematic representation of RAB11 isoform localization in both endocytic (left panel) and exocytic pathways (right panel). Green circles represent RAB11-positive compartments, yellow, blue, red lines represent RAB11A, RAB11B and RAB25, respectively.
Biology 10 00026 g001
Table 1. Structure of RAB11 gene isoforms.
Table 1. Structure of RAB11 gene isoforms.
Gene NameChromosome
Position
Number of Coding ExonNumber of Amino AcidsPercentage of IdentityTissue
Expression
RAB11A15q22.315216100%
(with RAB11A)
Ubiquitous
RAB11B19p13.2521889%
(with RAB11A)
Wide,
enriched in brain testis, heart
RAB251q22521361% and 66%
(with RAB11A and RAB11B)
Wide,
enriched in lung kidney, gastric tract
Table 2. Roles of RAB11A in cancer.
Table 2. Roles of RAB11A in cancer.
RolesExpression StatusCancer TypeMechanism of ActionFunctionReferences
OncogenicOver expressionBreast cancerIncreased signaling (ERK)
Increased expression
(EGFR)
Proliferation[38]
Non-small cell lung cancerDecreased signaling
(Hippo)
Tumorigenesis
invasion
[39]
Hepatocellular carcinomaIncreased signaling
(PI3K/AKT)
Increased expression (MMP2)
Tumorigenesis
invasion
[40]
Pancreatic cancerIncreased signaling
(GSK3β/Wnt/β-catenin)
Proliferation
invasion
[41]
Tumor suppressiveUnder expressionColon cancerDecreased signaling
(Hippo/YAP/IL6)
Tumorigenesis[42]
Table 3. Roles of RAB11B in cancer.
Table 3. Roles of RAB11B in cancer.
RolesExpression StatusCancer TypeMechanism of ActionFunctionReferences
OncogenicOver expressionBreast cancerIncreased expression (β1-Integrin)Metastasis[43]
Tumor suppressiveNot yet
defined
Not yet
defined
Not yet
defined
Not yet definedNot yet defined
Table 4. Roles of RAB25 in cancer (adapted from [44]).
Table 4. Roles of RAB25 in cancer (adapted from [44]).
RolesExpression StatusCancer TypeMechanism of ActionFunctionReferences
OncogenicOver expressionBladder cancerIncreased signaling (AKT/GSK3β /SNAIL)Tumorigenesis metastasis[45,46]
Gastric cancerIncreased signaling (β1-Integrin/EGFR/SNAIL)Invasion
metastasis
[47,48]
GlioblastomaIncreased signaling
(AKT)
Tumorigenesis[49]
Liver cancerIncreased signaling
(AKT/WNT)
Invasion[50]
Luminal Breast cancerIncreased signaling (β1-Integrin/EGFR/SNAIL)Tumorigenesis
metastasis
[48,51]
Lung CancerIncreased signaling
(EGFR)
Tumorigenesis
invasion
[52,53]
Ovarian cancerIncreased signaling (β1-Integrin/EGFR/SNAIL)Tumorigenesis
metastasis
[48,51]
Prostate cancerNot yet definedTumorigenesis
invasion
[54,55]
WilmsNot yet definedTumorigenesis[56]
Tumor suppressiveUnder expressionColon cancerIncreased expression
(EGFR)
Tumorigenesis[32]
Oesophageal cancerReduced signaling
(ERK/FAK)
Tumorigenesis
invasion
[57]
Head and Neck cancerReduced cytoskeleton
(F-actin)
Metastasis[58]
Triple negative breast cancerReduced expression
(VEGF/VEGFR)
Tumorigenesis[34,59]
Table 5. Inhibitors of RAB11-mediated trafficking pathways.
Table 5. Inhibitors of RAB11-mediated trafficking pathways.
Molecule NameMechanism of ActionSpecificityReference
PitavastatinHMG-CoA reductase inhibitorNot yet determined[43]
SimvastatinHMG-CoA reductase inhibitorNot yet determined[43,112]
FluvastatinHMG-CoA reductase inhibitorNot yet determined[112]
CDKI-73Not yet determinedNot yet determined[113]
RFP14RAB11:FIP interaction inhibitorRAB25[62]
RFP24RAB11:FIP interaction inhibitorRAB25[62]
RFP26RAB11:FIP interaction inhibitorRAB11A, RAB25[62]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ferro, E.; Bosia, C.; Campa, C.C. RAB11-Mediated Trafficking and Human Cancers: An Updated Review. Biology 2021, 10, 26. https://doi.org/10.3390/biology10010026

AMA Style

Ferro E, Bosia C, Campa CC. RAB11-Mediated Trafficking and Human Cancers: An Updated Review. Biology. 2021; 10(1):26. https://doi.org/10.3390/biology10010026

Chicago/Turabian Style

Ferro, Elsi, Carla Bosia, and Carlo C. Campa. 2021. "RAB11-Mediated Trafficking and Human Cancers: An Updated Review" Biology 10, no. 1: 26. https://doi.org/10.3390/biology10010026

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop