Next Article in Journal
Preparation of a Paeonol-Containing Temperature-Sensitive In Situ Gel and Its Preliminary Efficacy on Allergic Rhinitis
Next Article in Special Issue
Roles of Rho GTPases in Intracellular Transport and Cellular Transformation
Previous Article in Journal
Differential Expression of Genes Associated with the Progression of Renal Disease in the Kidneys of Liver-Specific Glucokinase Gene Knockout Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

RUFY, Rab and Rap Family Proteins Involved in a Regulation of Cell Polarity and Membrane Trafficking

Department of Environmental Health Science, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2013, 14(3), 6487-6498; https://doi.org/10.3390/ijms14036487
Submission received: 31 December 2012 / Revised: 11 March 2013 / Accepted: 15 March 2013 / Published: 21 March 2013
(This article belongs to the Special Issue Regulation of Membrane Trafficking and Its Potential Implications)

Abstract

:
Cell survival, homeostasis and cell polarity rely on the control of membrane trafficking pathways. The RUN domain (comprised of the RPIP8, UNC-14, and NESCA proteins) has been suggested to be implicated in small GTPase-mediated membrane trafficking and cell polarity. Accumulating evidence supports the hypothesis that the RUN domain-containing proteins might be responsible for an interaction with a filamentous network linked to actin cytoskeleton and/or microtubules. In addition, several downstream molecules of PI3K are involved in regulation of the membrane trafficking by interacting with vesicle-associated RUN proteins such as RUFY family proteins. In this review, we summarize the background of RUN domain research with an emphasis on the interaction between RUN domain proteins including RUFY proteins (designated as RUN and FYVE domain-containing proteins) and several small GTPases with respect to the regulation of cell polarity and membrane trafficking on filamentous network.

Abbreviations

CC
Coiled Coil
FYVE
Fab-1, YGL023, Vps27, and EEA1 proteins
GAP
GTPase-activating protein
GDF
GDI displacement factor
GDI
guanine nucleotide dissociation inhibitor
GEF
guanine nucleotide-exchange factor Glut4: glucose transporter 4
PH
plekstrin homology
PIP
phosphoinositidephosphate
PIP2
phosphatidylinositol 4,5-bisphosphate
PIP3
phosphatidylinositol 3,4,5-triphosphate
PI3K
phosphatidylinositol-3 kinase
RUFY
RUN and FYVE domain-containing proteins; RUN, RPIP8, UNC-14, and NESCA proteins

1. Introduction

Asymmetric organization of cellular components and structures is called cell polarity. Establishment of the cell polarity involves many processes including signaling cascade [1], membrane trafficking events, [2] and cytoskeletal dynamics [3], which are implicated in differentiation, proliferation and morphogenesis of various cellular organisms [4]. Cell polarization is required in order for a cell to function properly. For example, the presence of axon in neuronal cells determines the directional flow of the signal (Figure 1). It is well known that initial trafficking of down-regulated protein is performed by clathrin-mediated internalization and sorting into the lumen of the endosomes [5] (Figure 2). Dysregulation of cell polarity can cause developmental disorder and cancer [6]. Studies have revealed links between cell polarity establishment and cellular membrane traffic [2,7], and the function of endosome-associated proteins has been implicated in cell polarity [8].
The direction and specificity of endosomal membrane trafficking is definitely regulated by various membrane-bound factors including protein receptors, small GTPases, and phosphoinositides [9,10]. While many components of the trafficking pathway have been ascribed, there may be additional factors that remain unknown, which precisely control the direction and specificity of membrane trafficking. In the present brief review, we summarize the function of RUN domain proteins, specifically RUFY family proteins, and their binding partners such as Rab proteins from the viewpoint of endosomal membrane trafficking. We will thus cover what is currently known about the function of protein signaling, which is also important for membrane trafficking.

2. RUN Domain Binds Several Signaling Molecules

The RUN domains, named after RPIP8, UNC-14, and NESCA proteins [11], might function as effectors of the small GTPase superfamily [12,13]. Because many RUN domain-containing proteins are involved in the signaling of small GTPases, including members of the Rap and Rab family, the RUN domain has been suggested to be involved in membrane trafficking [13]. The RUN domain encloses hydrophobic amino acids in the conserved positions, which is a conserved protein motif that consists of approximately 200 amino acids with binding activity to small GTP-binding proteins [11]. The sequence analysis has predicted that the RUN domain is composed of several conserved blocks, which constitute the core of a globular structure. The overall crystal structure of the RUN domain adopts a single globular fold consisting of eight alpha-helices [14]. However, it is not the only function of the RUN domain to bind a GTPase. The RUN domain binds to some molecules, including motor proteins, and the RUN domain might be responsible for an interaction with a filamentous network [13]. NESCA, a signaling adapter protein in the NGF-mediated signaling pathway, contains a RUN domain at the N-terminus [12]. The RUN domain of NESCA comprises nine helices, resembling the other RUN domain-containing proteins [15]. Mutational analyses have demonstrated that the RUN domain is an important structural determinant for the nuclear translocation of NESCA and that the nuclear redistribution of NESCA is essential to its neurite outgrowth-promoting properties [12]. However, the RUN domain of NESCA has different surface electrostatic distributions at the putative GTPase-interacting interface compared to the other RUN domains [15]. The RUN domain of NESCA can bind H-Ras, a downstream signaling molecule of TrkA, as well as TrkA itself, suggesting that the NESCA participates in the NGF-TrkA signaling pathway [15].
The RUN domain-containing proteins have been shown to promote endosomal fusion and are important for vesicular transport. In addition, the RUN domains appear to be required for localization to detergent-insoluble endosomal microdomains [12,13]. The physical interaction between RUN proteins and filamentous materials has been confirmed by several biochemical experiments using wild type and mutant proteins [13]. The association among small GTPases, RUN proteins, and motor proteins might reflect a novel function for these proteins in the transport of vesicular cargoes in cells. It has been reported that FYCO1 functions as an adapter linking autophagosomes to microtubule molecular motors and the Rab7, which is implicated in the phagosomal transport and fusion [16]. Kinesin-1 is a heterotetramer composed of kinesin heavy chain and kinesin light chain. UNC-14, a RUN domain protein binds to the kinesin-1 and regulates synaptic vesicle localization [17]. UNC-14 is also predicted to play an important role in multiple Ras-like GTPase signaling pathways [18]. Because RUN domains are often found in proteins involved in the regulation of Rab family small GTPases, the RUN domain has been suggested to be involved in the Rab-mediated membrane trafficking. It seems there is a common function underlying the mechanism for association of RUN domain to small GTPases and motor proteins.

3. Function of RUFY Family Proteins with the RUN Domain

The RUFY, designated as the RUN and FYVE domain-containing protein family, contains an amino-terminal RUN domain, and a carboxyl-terminal FYVE domain, which associate with phosphatidylinositol 3-phosphate in membranes of early endosomes [19]. Actually, RUFY proteins are localized predominantly to the early endosomes. RUFY proteins are often tyrosine-phosphorylated and the mutant lacking the phosphorylation sites fails to go to the endosomes [19]. It has been suggested Rab10, Rab11, Rab14, and RUFY proteins play an important role in the Glut4 trafficking in adipocytes and in skeletal muscle [20]. Sequence and genome analysis has revealed that a RUFY family consists of 4 members of proteins (Figure 3).
RUFY1, also known as RABIP4 or ZFYVE12, is a 708-amino acids protein that localizes to the cytoplasm and early endosome membrane [19,21]. RUFY1 has been identified as downstream effector of Etk protein kinase, which is highly expressed in testis, lung, brain and kidney. RUFY1 functions to bind PIP3-containing phospholipid vesicles and participates in early endosomal membrane trafficking [19]. The downstream effects of PI3K signaling are mediated by proteins containing a PIP3-binding module indicated by the FYVE finger domain. Many FYVE domain-containing proteins are localized at the endosomes and play an important role in endocytosis [22]. Through the SH3 and SH2 domains of Etk, the Etk interacts with RUFY1, then phosphorylates Tyr-281 and Tyr-292 of RUFY1, which is essential for the endosomal localization [19]. The Etk plays an important role in the regulation of endocytosis as a downstream effector of PI3K. Two coiled coil domains also determine endosomal localization of RUFY1 [23]. The PI3K inhibitor wortmannin blocks the endosomal localization of RUFY1 [23]. Rab14 engages in a GTP-dependent interaction with RUFY1 [24]. The active Rab14 regulates RUFY1 recruitment onto endosomal membranes, and Rab4 allows endosomal fusion [24]. The Rab14 seems to be the primary determinant of RUFY1 recruitment to the endosomes, and the FYVE domain may assist RUFY1 targeting to PIP3-enriched early endosomes [24]. Both Rab14 and RUFY1 are involved in Rab4-dependent recycling endosome, and enlargement of the early endosomes mediated by RUFY1 needs the interaction with Rab4 [25]. The RUFY1 is present in the sorting endosomes, while Rab4 is present both on sorting and recycling endosomes [26]. This would provide directional trafficking away from the recycling endosomes to the sorting endosomes. RUFY1 can also modify the kinetic parameters of Glut1 protein recycling [26].
RUFY2, also known as RABIP4R or ZFYVE13, contains a RUN domain and a carboxy terminal FYVE zinc finger, separated by two coiled coil domains [27]. Localizing to nucleus, RUFY2 is expressed in brain, lung and testis. RUFY2 as well as RUFY1 interacts with the Etk that is a tyrosine kinase involved in regulation of various cellular processes [19]. The carboxyl domain of RUFY2 binds to a negative form of Rab33A [28]. RUFY3, also known as RIPX or SINGAR1, is diffusely localized in hippocampal neurons and accumulated in the growth cones and axons [29]. RUFY3 ensures the robustness of neuronal polarity by suppressing formation of surplus axons. RUFY3 also contains the RUN domain and seems to play important roles in multiple Ras-like GTPase signaling pathways. Rab5 engages in a GTP-dependent interaction with RUFY3. RUFY3 can bind to the active Rab5 and weakly associates to Rap2 [30]. RUFY3 may function as a docking protein for distinct two small GTPases. It has been reported that oxidized LDL-containing immune complexes affect the gene expression of RUFY3 in human U937 monocytic cells [31]. RUFY4 is a 571-amino acids protein that contains a RUN domain and a FYVE zinc finger domain [32]. RUFY4 is also believed to be involved with zinc ion binding. As poorly characterized for RUFY2 and for RUFY4, little is known about the precise intracellular functions for these proteins.

4. Regulation of Cell Polarity and Membrane Trafficking

Cell polarity and vesicle sorting are significant processes that influence normal cell function including cell adhesion, migration, and neurotransmission. Endosomal membrane trafficking is a spatiotemporally regulated process that confirms suitable delivery of cargo via the pathway. Endosomes can bud inwardly from the membranes to form vesicles, which receive cargo from the cell surface via endocytosis and biosynthetic cargo from the late Golgi complex [33]. The endocytic trafficking has been shown to be a critical component of many signaling pathways, which is indispensable for a wide range of developmental processes [34]. In addition, the endosomal trafficking is regulated by sequential recruitment of a variety of cytosolic and membrane-bound proteins [35]. Studies have elucidated many of the key components of the events that take place during the membrane trafficking. For example, small GTPases of Rab family, its effectors, Ca2+ levels, and phosphoinositides may be all important. Previous studies have demonstrated that Ca2+ influx triggers the final step in exocytotic membrane fusion events during neurotransmission [36].
Similarly, phosphoinositides have been shown to be important for the recruitment of Rab family proteins and the binding factors [37]. Plasma membrane channels are known to be regulated by the Rab proteins [38] and phosphoinositides, which are molecules that determine the vesicular identity and direction of membrane trafficking. In particular, PIP3 is essential for the membrane trafficking of early endosome [39]. The PIP3 has lots of effector proteins in mammalian cells, all of which contain PIP3-binding motifs such as FYVE and/or PH domains [40]. Selective recruitment of these effectors by PIP3 may provide a mechanism by which the directionality for incoming vesicles and endosomes may be established. The PIP3 is thus critical for the maturation of endosomes, and for fusion events with intracellular organelles [41]. Correspondingly, disruption of PIP3 synthesis by wortmannin, a PI3K inhibitor, affects the formation of internal vesicles and the maturation of endosomes [42]. While phosphoinositides can recruit phosphoinositide-binding proteins to regulate the activity of GTPases, the GTPases can, in turn, control the activity of PIP-metabolizing enzymes.

5. Several Small GTPases Involved in the Membrane Trafficking

Eukaryotic cells have developed a diverse family of small GTPases to regulate the membrane trafficking pathways. Together with their effector proteins, Rab and Rap proteins mediate various aspects of vesicle formation, docking and fusion [43,44]. Some of Rab proteins recruit effectors to promote membrane fusion and vesicle formation and play a key role in early endocytic pathways. Rab proteins use the guanine nucleotide-dependent alteration mechanism to regulate the membrane traffic [45]. By binding to the guanine nucleotide exchange proteins that activate the Rab, certain effectors act to establish a feedback loop that helps to define and maintain localized domains of activated Rab proteins, which then serve to recruit the other effector molecules. Recent advances have extended the number of known Rab effectors. The Rab effectors include sorting adaptors, binding factors, lipid kinases, and lipid phosphatases, which are present on the surface of the acceptor compartment. These different tasks are carried out by a diverse collection of effector molecules that bind to specific Rab family proteins in their GTP-bound state [45]. Binding factors mediate vesicle fusion by interacting with molecules on the acceptor membrane, while lipid kinases produce phosphoinositides to define vesicular identity and the direction of membrane trafficking. In addition, the Rab cascades appear to confer directionality to membrane traffic [45].
Rab proteins are classified in specific organelle membrane proteins, establishing the basic tags to define organelle identity. Endosomal Rab proteins such as Rab 4, 5 and 9 are characterized by their localization and functional interactions with the effector proteins [46]. Rab4 and Rab11 regulate recycling of receptors from early endosomes to the cell surface via distinct pathways [47]. Rab4 functions at the level of early endosomes, and Rab11 is involved in the trafficking of cargo through recycling endosomes [48]. Rab4 is also an important player in Glut4 trafficking in adipocytes, skeletal muscle, and cardiocytes [49]. Rab5 plays important roles in clathrin-mediated endocytosis [50] and in endosome to endosome fusion. EEA1, a Rab5 effector, is recruited selectively onto early endosomes, whereas Rab5 is symmetrically distributed between the clathrin-coated vesicles and early endosomes [51]. Rab5 is localized in early endosome. In contrast, Rab7 is localized to late endosome and lysosomes [52]. Endsomal trafficking is also controlled by small GTPase Rab6, which regulates vesicle trafficking at the level of Golgi via recruitment of numerous and unrelated effectors. The crystal structure of Rab6-GTP in complex with a 378-residue internal fragment of the effector Rab6IP1 has been solved [53]. Flexibility in Rab6 mediates recognition of compositionally distinct alpha-helical coiled coils, thereby contributing to Rab6 promiscuity in effector recruitment [53]. Rab8 plays an important role in exocytic membrane trafficking from Golgi complex to plasma membrane [54]. The Rab8 is associated with myosinVI via optineurin, and the Rab8-optineurin-myosin VI complex might be involved in presenting the secretory vesicle to plasma membrane [55]. Rab9 is associated with trafficking to the Golgi network [56]. Rab11 and Rab11 family-interacting protein 3, which plays a role in membrane trafficking and regulation of actin dynamics, are both required to support the formation of filamentous virions [57]. Rab11 is also involved in recycling endosomes back to the plasma membrane. Rab14 has been implicated in phagosome and early endosome fusion. Rab14 also localizes to the Golgi complex. The Rab27A indirectly recognizes myosin Va on melanosomes via Slac2-a [58]. The Rab27B, a closely related isoform of Rab27A, is also associated with Myosin Va/VIIa via Alac2c/MyRIP [59]. Rab27 and Slac2c/MyRIP are part of a complex mediating the interaction of secretory granules with actin cytoskeleton and participate to the regulation of exocytosis. Rab35 regulates neurite-outgrowth, which is due to its direct influence on actin dynamics [60].
Rap proteins (Rap1a, -1b, -2a, and -2b) are small GTPases closely related to Ras. Rap1 is involved in various cellular processes, such as regulation of integrin-mediated cell adhesion and cadherin-mediated cell junction formation. It has been demonstrated that interaction between Rap family proteins and profilin II, an important activator of actin polymerization, is mediated via Rgl3, a RalGDS-related protein [61]. RAPL, a Rap1-associating molecule, localizes on microtubules and the activated Rap1 and RAPL control the directional migration of vascular endothelial cells [62]. Rap2 interacts with the platelet cytoskeleton by direct binding to the actin filament, which is the polymerized but not the monomeric form of actin [63]. The interaction of Rap2 with actin filaments is independent of the bound nucleotide.
Rab6 and the binding partner DENND5, a RUN domain protein, are involved in the trafficking of vesicles at the level of Golgi [64]. A RUN domain of the DENND5 plays an important role in polarized transport in Golgi through the association with sorting nexins [65], which are a large family of phosphoinositide-binding proteins that have fundamental roles in orchestrating cargo sorting [66]. In this way, small GTPases, which are related to RUN domain proteins, also associate with motor proteins and/or filamentous molecules. While small GTPases are essential for membrane trafficking, actin remodeling is the most critical. Actin is polymerized at the site of particle attachment and directs membrane extension. Many actin-associated proteins are enriched at the phagocytic cup and play important roles in phagocytosis [67]. Importantly, it is the interactions of these small GTPase proteins to the effector molecules that form a functional interaction to regulate the membrane trafficking. However, details of the biological roles elicited by effectors are largely unknown.

6. Perspective

Neurodegenerative diseases, including Alzheimer’s, sometimes exhibit defective endsomal trafficking. The transport would be necessary to ensure organelle homeostasis. The functional significance of membrane trafficking in the signaling pathways remains to be more established. Indeed, the RUN domain-containing proteins will be an active focus for investigation of this field. The RUFY proteins may be activated in endosomal microdomains enriched in some unidentified cytoskeletal elements. The localization of RUFY protein during the membrane trafficking seems to be dynamic. Although specific kinetic information would be required, RUFY and Rab proteins may have an inhibitory function in endocytosis, which is similar to the case of Rab15 in neuronal system [68]. This inhibitory effect could result from a negative regulation of endosome recycling. Likewise, RUFY3 may ensure the strength of neuronal polarity by suppressing formation of surplus axons. The molecular details of how RUFY3 inhibits the formation of surplus axons remain important issues for future investigation [29,69]. Deciphering the precise mechanisms will provide new insight into the physiological roles of these interesting molecules in regulating membrane traffic [13]. It is expected that future studies will address these issues in order to achiee a better understanding of the potential partners and roles of the RUFY and Rab/Rap proteins.

Acknowledgments

This work was supported by grants-in-aid from the Ministry of Education, Culture, Sports, Science and Technology in Japan. In addition, this work was supported in part by the grant from SHIN-EI Pharmaceutical Co., Ltd.

Conflict of Interest

The authors declare no conflict of interest.

References

  1. Happé, H.; de Heer, E.; Peters, D.J. Polycystic kidney disease: The complexity of planar cell polarity and signaling during tissue regeneration and cyst formation. Biochim. Biophys. Acta 2011, 1812, 1249–1255. [Google Scholar]
  2. Santiago-Tirado, F.H.; Bretscher, A. Membrane-trafficking sorting hubs: Cooperation between PI4P and small GTPases at the trans-Golgi network. Trends Cell Biol 2011, 21, 515–525. [Google Scholar]
  3. Baum, B.; Georgiou, M. Dynamics of adherens junctions in epithelial establishment, maintenance, and remodeling. J. Cell Biol 2011, 192, 907–917. [Google Scholar]
  4. Wu, G.; Ge, J.; Huang, X.; Hua, Y.; Mu, D. Planar cell polarity signaling pathway in congenital heart diseases. J. Biomed. Biotechnol 2011, 2011, 589414. [Google Scholar]
  5. Traub, L.M. Tickets to ride: Selecting cargo for clathrin-regulated internalization. Nat. Rev. Mol. Cell Biol 2009, 10, 583–596. [Google Scholar]
  6. Martin-Belmonte, F.; Perez-Moreno, M. Epithelial cell polarity, stem cells and cancer. Nat. Rev. Cancer 2011, 12, 23–38. [Google Scholar]
  7. Layton, A.; Savage, N.S.; Howell, A.S.; Carroll, S.Y.; Drubin, D.G.; Lew, D.J. Modeling vesicle traffic reveals unexpected consequences for Cdc42p-mediated polarity establishment. Curr. Biol 2011, 21, 184–194. [Google Scholar]
  8. Golachowska, M.R.; Hoekstra, D.; van IJzendoorn, S.C. Recycling endosomes in apical plasma membrane domain formation and epithelial cell polarity. Trends Cell Biol 2010, 20, 618–626. [Google Scholar]
  9. Tóth, D.J.; Tóth, J.T.; Gulyá, S.G.; Balla, A.; Balla, T.; Hunyady, L.; Várnai, P. Acute depletion of plasma membrane phosphatidylinositol 4,5-bisphosphate impairs specific steps in endocytosis of the G-protein-coupled receptor. J. Cell Sci. 2012, 125, 2185–2197. [Google Scholar]
  10. Horgan, C.P.; McCaffrey, M.W. Endosomal trafficking in animal cytokinesis. Front. Biosci 2012, 4, 547–555. [Google Scholar]
  11. Callebaut, I.; de Gunzburg, J.; Goud, B.; Mornon, J.P. RUN domains: A new family of domains involved in Ras-like GTPase signaling. Trends Biochem. Sci 2001, 26, 79–83. [Google Scholar]
  12. MacDonald, J.I.; Kubu, C.J.; Meakin, S.O. Nesca, a novel adapter, translocates to the nuclear envelope and regulates neurotrophin-induced neurite outgrowth. Cell Biol 2004, 164, 851–862. [Google Scholar]
  13. Yoshida, H.; Kitagishi, Y.; Okumura, N.; Murakami, M.; Nishimura, Y.; Matsuda, S. How do you RUN on? FEBS Lett 2011, 585, 1707–1710. [Google Scholar]
  14. Kukimoto-Niino, M.; Takagi, T.; Akasaka, R.; Murayama, K.; Uchikubo-Kamo, T.; Terada, T.; Inoue, M.; Watanabe, S.; Tanaka, A.; Hayashizaki, Y.; et al. Crystal structure of the RUN domain of the RAP2-interacting protein x. J. Biol. Chem 2006, 281, 31843–31853. [Google Scholar]
  15. Sun, Q.; Han, C.; Liu, L.; Wang, Y.; Deng, H.; Bai, L.; Jiang, T. Crystal structure and functional implication of the RUN domain of human NESCA. Protein Cell 2012, 3, 609–617. [Google Scholar]
  16. Pankiv, S.; Alemu, E.A.; Brech, A.; Bruun, J.A.; Lamark, T.; Overvatn, A.; Bjørkøy, G.; Johansen, T. FYCO1 is a Rab7 effector that binds to LC3 and PI3P to mediate microtubule plus end-directed vesicle transport. J. Cell Biol 2010, 188, 253–269. [Google Scholar]
  17. Sakamoto, R.; Byrd, D.T.; Brown, H.M.; Hisamoto, N.; Matsumoto, K.; Jin, Y. The Caenorhabditis elegans UNC-14 RUN domain protein binds to the kinesin-1 and UNC-16 complex and regulates synaptic vesicle localization. Mol. Biol. Cell 2005, 16, 483–496. [Google Scholar]
  18. Ogura, K.; Goshima, Y. The autophagy-related kinase UNC-51 and its binding partner UNC-14 regulate the subcellular localization of the Netrin receptor UNC-5 in Caenorhabditis elegans. Development 2006, 133, 3441–3450. [Google Scholar]
  19. Yang, J.; Kim, O.; Wu, J.; Qiu, Y. Interaction between tyrosine kinase Etk and a RUN domain- and FYVE domain-containing protein RUFY1. A possible role of ETK in regulation of vesicle trafficking. J. Biol. Chem 2002, 277, 30219–30226. [Google Scholar]
  20. Larance, M.; Ramm, G.; Stöckli, J.; van Dam, E.M.; Winata, S.; Wasinger, V.; Simpson, F.; Graham, M.; Junutula, J.R.; Guilhaus, M.; James, D.E. Characterization of the role of the Rab GTPase-activating protein AS160 in insulin-regulated GLUT4 trafficking. J. Biol. Chem 2005, 280, 37803–37813. [Google Scholar]
  21. Fouraux, M.A.; Deneka, M.; Ivan, V.; van der Heijden, A.; Raymackers, J.; van Suylekom, D.; van Venrooij, W.J.; van der Sluijs, P.; Pruijn, G.J. Rabip4′ is an effector of rab5 and rab4 and regulates transport through early endosomes. Mol. Biol. Cell 2004, 15, 611–624. [Google Scholar]
  22. Simonsen, A.; Wurmser, A.E.; Emr, S.D.; Stenmark, H. The role of phosphoinositides in membrane transport. Curr. Opin. Cell Biol 2001, 13, 485–492. [Google Scholar]
  23. Mari, M.; Macia, E.; le Marchand-Brustel, Y.; Cormont, M. Role of the FYVE finger and the RUN domain for the subcellular localization of Rabip4. J. Biol. Chem 2001, 276, 42501–42508. [Google Scholar]
  24. Yamamoto, H.; Koga, H.; Katoh, Y.; Takahashi, S.; Nakayama, K.; Shin, H.W. Functional cross-talk between Rab14 and Rab4 through a dual effector, RUFY1/Rabip4. Mol. Biol. Cell 2010, 21, 2746–2755. [Google Scholar]
  25. Mari, M.; Monzo, P.; Kaddai, V.; Keslair, F.; Gonzalez, T.; le Marchand-Brustel, Y.; Cormont, M. The Rab4 effector Rabip4 plays a role in the endocytotic trafficking of Glut 4 in 3T3-L1 adipocytes. J. Cell Sci 2006, 119, 1297–1306. [Google Scholar]
  26. Cormont, M.; Mari, M.; Galmiche, A.; Hofman, P.; le Marchand-Brustel, Y. A FYVE-finger-containing protein, Rabip4, is a Rab4 effector involved in early endosomal traffic. Proc. Natl. Acad. Sci. USA 2001, 98, 1637–1642. [Google Scholar]
  27. Barbe, L.; Lundberg, E.; Oksvold, P.; Stenius, A.; Lewin, E.; Björling, E.; Asplund, A.; Pontén, F.; Brismar, H.; Uhlén, M.; Andersson-Svahn, H. Toward a confocal subcellular atlas of the human proteome. Mol. Cell Proteomics 2008, 7, 499–508. [Google Scholar]
  28. Fukuda, M.; Kobayashi, H.; Ishibashi, K.; Ohbayashi, N. Genome-wide investigation of the Rab binding activity of RUN domains: Development of a novel tool that specifically traps GTP-Rab35. Cell Struct. Funct 2011, 36, 155–170. [Google Scholar]
  29. Mori, T.; Wada, T.; Suzuki, T.; Kubota, Y.; Inagaki, N. Singar1, a novel RUN domain-containing protein, suppresses formation of surplus axons for neuronal polarity. J. Biol. Chem 2007, 282, 19884–19893. [Google Scholar]
  30. Yoshida, H.; Okumura, N.; Kitagishi, Y.; Shirafuji, N.; Matsuda, S. Rab5(Q79L) interacts with the carboxyl terminus of RUFY3. Int. J. Biol. Sci 2010, 6, 187–189. [Google Scholar]
  31. Hammad, S.M.; Twal, W.O.; Barth, J.L.; Smith, K.J.; Saad, A.F.; Virella, G.; Argraves, W.S.; Lopes-Virella, M.F. Oxidized LDL immune complexes and oxidized LDL differentially affect the expression of genes involved with inflammation and survival in human U937 monocytic cells. Atherosclerosis 2009, 202, 394–404. [Google Scholar]
  32. Kimura, K.; Wakamatsu, A.; Suzuki, Y.; Ota, T.; Nishikawa, T.; Yamashita, R.; Yamamoto, J.; Sekine, M.; Tsuritani, K.; Wakaguri, H.; et al. Diversification of transcriptional modulation: Large-scale identification and characterization of putative alternative promoters of human genes. Genome Res 2006, 16, 55–65. [Google Scholar]
  33. Pfeffer, S.R. Multiple routes of protein transport from endosomes to the trans Golgi network. FEBS Lett 2009, 583, 3811–3816. [Google Scholar]
  34. González-Gaitán, M. Endocytic trafficking during Drosophila development. Mech. Dev 2003, 120, 1265–1282. [Google Scholar]
  35. Bakhru, S.H.; Altiok, E.; Highley, C.; Delubac, D.; Suhan, J.; Hitchens, T.K.; Ho, C.; Zappe, S. Enhanced cellular uptake and long-term retention of chitosan-modified iron-oxide nanoparticles for MRI-based cell tracking. Int. J. Nanomed 2012, 7, 4613–4623. [Google Scholar]
  36. Monck, J.R.; Fernandez, J.M. The exocytotic fusion pore and neurotransmitter release. Neuron 1994, 12, 707–716. [Google Scholar]
  37. Zaid, H.; Antonescu, C.N.; Randhawa, V.K.; Klip, A. Insulin action on glucose transporters through molecular switches, tracks and tethers. Biochem. J 2008, 413, 201–215. [Google Scholar]
  38. Saxena, S.K.; Kaur, S. Regulation of epithelial ion channels by Rab GTPases. Biochem. Biophys. Res. Commun 2006, 351, 582–587. [Google Scholar]
  39. Kanamarlapudi, V. Centaurin-alpha1 and KIF13B kinesin motor protein interaction in ARF6 signalling. Biochem. Soc. Trans 2005, 33, 1279–1281. [Google Scholar]
  40. Saito, K.; Tautz, L.; Mustelin, T. The lipid-binding SEC14 domain. Biochim. Biophys. Acta 2007, 1771, 719–726. [Google Scholar]
  41. Stenmark, H.; Gillooly, D.J. Intracellular trafficking and turnover of phosphatidylinositol 3-phosphate. Semin. Cell Dev. Biol 2001, 12, 193–199. [Google Scholar]
  42. Shisheva, A. Phosphoinositides in insulin action on GLUT4 dynamics: Not just PtdIns(3,4,5)P3. Am. J. Physiol. Endocrinol. Metab 2008, 295, E536–E544. [Google Scholar]
  43. Lundquist, E.A. Small GTPases. WormBook 2006, 17, 1–18. [Google Scholar]
  44. Yang, H.; Sasaki, T.; Minoshima, S.; Shimizu, N. Identification of three novel proteins (SGSM1, 2, 3) which modulate small G protein (RAP and RAB)-mediated signaling pathway. Genomics 2007, 90, 249–260. [Google Scholar]
  45. Grosshans, B.L.; Ortiz, D.; Novick, P. Rabs and their effectors: Achieving specificity in membrane traffic. Proc. Natl. Acad. Sci. USA 2006, 103, 11821–11827. [Google Scholar]
  46. Mohrmann, K.; van der Sluijs, P. Regulation of membrane transport through the endocytic pathway by rabGTPases. Mol. Membr. Biol 1999, 16, 81–87. [Google Scholar]
  47. Jones, M.C.; Caswell, P.T.; Norman, J.C. Endocytic recycling pathways: Emerging regulators of cell migration. Curr. Opin. Cell Biol 2006, 18, 549–557. [Google Scholar]
  48. Nachury, M.V.; Seeley, E.S.; Jin, H. Trafficking to the ciliary membrane: How to get across the periciliary diffusion barrier? Annu. Rev. Cell Dev. Biol 2010, 26, 59–87. [Google Scholar]
  49. Foster, L.J.; Klip, A. Mechanism and regulation of GLUT-4 vesicle fusion in muscle and fat cells. Am. J. Physiol. Cell Physiol 2000, 279, C877–C890. [Google Scholar]
  50. Cavalli, V.; Corti, M.; Gruenberg, J. Endocytosis and signaling cascades: A close encounter. FEBS Lett 2001, 498, 190–196. [Google Scholar]
  51. Hayakawa, A.; Hayes, S.; Leonard, D.; Lambright, D.; Corvera, S. Evolutionarily conserved structural and functional roles of the FYVE domain. Biochem. Soc. Symp 2007, 74, 95–105. [Google Scholar]
  52. Wang, T.; Ming, Z.; Xiaochun, W.; Hong, W. Rab7: Role of its protein interaction cascades in endo-lysosomal traffic. Cell Signal 2011, 23, 516–521. [Google Scholar]
  53. Recacha, R.; Boulet, A.; Jollivet, F.; Monier, S.; Houdusse, A.; Goud, B.; Khan, A.R. Structural basis for recruitment of Rab6-interacting protein 1 to Golgi via a RUN domain. Structure 2009, 17, 21–30. [Google Scholar]
  54. Dugani, C.B.; Klip, A. Glucose transporter 4: Cycling, compartments and controversies. EMBO Rep 2005, 6, 1137–1142. [Google Scholar]
  55. Chibalina, M.V.; Roberts, R.C.; Arden, S.D.; Kendrick-Jones, J.; Buss, F. Rab8-optineurin-myosin VI: Analysis of interactions and functions in the secretory pathway. Methods Enzymol 2008, 438, 11–24. [Google Scholar]
  56. Chia, P.Z.; Gasnereau, I.; Lieu, Z.Z.; Gleeson, P.A. Rab9-dependent retrograde transport and endosomal sorting of the endopeptidase furin. J. Cell Sci 2011, 124, 2401–2413. [Google Scholar]
  57. Bruce, E.A.; Digard, P.; Stuart, A.D. The Rab11 pathway is required for influenza A virus budding and filament formation. J. Virol 2010, 84, 5848–5859. [Google Scholar]
  58. Kuroda, T.S.; Itoh, T.; Fukuda, M. Functional analysis of slac2-a/melanophilin as a linker protein between Rab27A and myosin Va in melanosome transport. Methods Enzymol 2005, 403, 419–431. [Google Scholar]
  59. Westbroek, W.; Lambert, J.; de Schepper, S.; Kleta, R.; van den Bossche, K.; Seabra, M.C.; Huizing, M.; Mommaas, M.; Naeyaert, J.M. Rab27b is up-regulated in human Griscelli syndrome type II melanocytes and linked to the actin cytoskeleton via exon F-Myosin Va transcripts. Pigment Cell Res 2004, 17, 498–505. [Google Scholar]
  60. Chua, C.E.; Lim, Y.S.; Tang, B.L. Rab35—A vesicular traffic-regulating small GTPase with actin modulating roles. FEBS Lett 2010, 584, 1–6. [Google Scholar]
  61. Xu, J.; Shi, S.; Matsumoto, N.; Noda, M.; Kitayama, H. Identification of Rgl3 as a potential binding partner for Rap-family small G-proteins and profilin II. Cell Signal 2007, 19, 1575–1582. [Google Scholar]
  62. Kinashi, T.; Katagiri, K. Regulation of lymphocyte adhesion and migration by the small GTPase Rap1 and its effector molecule, RAPL. Immunol Lett 2004, 93, 1–5. [Google Scholar]
  63. Kardassis, D.; Murphy, C.; Fotsis, T.; Moustakas, A.; Stournaras, C. Control of transforming growth factor beta signal transduction by small GTPases. FEBS J 2009, 276, 2947–2965. [Google Scholar]
  64. Fernandes, H.; Franklin, E.; Jollivet, F.; Bliedtner, K.; Khan, A.R. Mapping the interactions between a RUN domain from DENND5/Rab6IP1 and sorting nexin 1. PLoS One 2012, 7, e35637. [Google Scholar]
  65. Wassmer, T.; Attar, N.; Harterink, M.; van Weering, J.R.; Traer, C.J.; Oakley, J.; Goud, B.; Stephens, D.J.; Verkade, P.; Korswagen, H.C.; Cullen, P.J. The retromer coat complex coordinates endosomal sorting and dynein-mediated transport, with carrier recognition by the trans-Golgi network. Dev. Cell 2009, 17, 110–122. [Google Scholar]
  66. Cullen, P.J.; Korswagen, H.C. Sorting nexins provide diversity for retromer-dependent trafficking events. Nat. Cell Biol 2011, 14, 29–37. [Google Scholar]
  67. Fehrenbacher, K.; Huckaba, T.; Yang, H.C.; Boldogh, I.; Pon, L. Actin comet tails, endosomes and endosymbionts. J. Exp. Biol 2003, 206, 1977–1984. [Google Scholar]
  68. Strick, D.J.; Elferink, L.A. Rab15 effector protein: A novel protein for receptor recycling from the endocytic recycling compartment. Mol. Biol. Cell 2005, 16, 5699–5709. [Google Scholar]
  69. Maher-Laporte, M.; Berthiaume, F.; Moreau, M.; Julien, L.A.; Lapointe, G.; Mourez, M.; DesGroseillers, L. Molecular composition of staufen2-containing ribonucleoproteins in embryonic rat brain. PLoS One 2010, 5, e11350. [Google Scholar]
Figure 1. Cell polarity is observed in many cases. For example, neuronal cells are asymmetric with discrete regions responsible for different roles that may underlie the generation of specific compartments within cells, which is distinct in biochemical composition, function, and structure. An arrow indicates the direction of polarity.
Figure 1. Cell polarity is observed in many cases. For example, neuronal cells are asymmetric with discrete regions responsible for different roles that may underlie the generation of specific compartments within cells, which is distinct in biochemical composition, function, and structure. An arrow indicates the direction of polarity.
Ijms 14 06487f1
Figure 2. Schematic illustration of intracellular vesicle transport. The model shows several pathways used by biosynthetic secretory and internalized endocytotic cargoes to reach their destinations. Note that some critical trafficking routes have been omitted for clarity.
Figure 2. Schematic illustration of intracellular vesicle transport. The model shows several pathways used by biosynthetic secretory and internalized endocytotic cargoes to reach their destinations. Note that some critical trafficking routes have been omitted for clarity.
Ijms 14 06487f2
Figure 3. Schematic diagram indicating the domain structures of the RUFY1, RUFY2, RUFY3, and RUFY4 proteins. The functionally important sites and their interaction proteins are shown. Genomic locations of each the genes and approximate molecular mass of each the proteins are also shown at the both ends. RUN = RPIP8, UNC-14, and NESCA proteins, FYVE = Fab-1, YGL023, Vps27, and EEA1 proteins, CC = Coiled Coil.
Figure 3. Schematic diagram indicating the domain structures of the RUFY1, RUFY2, RUFY3, and RUFY4 proteins. The functionally important sites and their interaction proteins are shown. Genomic locations of each the genes and approximate molecular mass of each the proteins are also shown at the both ends. RUN = RPIP8, UNC-14, and NESCA proteins, FYVE = Fab-1, YGL023, Vps27, and EEA1 proteins, CC = Coiled Coil.
Ijms 14 06487f3

Share and Cite

MDPI and ACS Style

Kitagishi, Y.; Matsuda, S. RUFY, Rab and Rap Family Proteins Involved in a Regulation of Cell Polarity and Membrane Trafficking. Int. J. Mol. Sci. 2013, 14, 6487-6498. https://doi.org/10.3390/ijms14036487

AMA Style

Kitagishi Y, Matsuda S. RUFY, Rab and Rap Family Proteins Involved in a Regulation of Cell Polarity and Membrane Trafficking. International Journal of Molecular Sciences. 2013; 14(3):6487-6498. https://doi.org/10.3390/ijms14036487

Chicago/Turabian Style

Kitagishi, Yasuko, and Satoru Matsuda. 2013. "RUFY, Rab and Rap Family Proteins Involved in a Regulation of Cell Polarity and Membrane Trafficking" International Journal of Molecular Sciences 14, no. 3: 6487-6498. https://doi.org/10.3390/ijms14036487

Article Metrics

Back to TopTop