1
|
Fukaya M, Ibuchi K, Sugawara T, Itakura M, Ito A, Shiroshima T, Hara Y, Okamoto H, Luton F, Sakagami H. EFA6A, an Exchange Factor for Arf6, Regulates NGF-Dependent TrkA Recycling From Early Endosomes and Neurite Outgrowth in PC12 Cells. Traffic 2024; 25:e12936. [PMID: 38725127 DOI: 10.1111/tra.12936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 12/18/2023] [Accepted: 12/26/2023] [Indexed: 06/03/2024]
Abstract
Endosomal trafficking of TrkA is a critical process for nerve growth factor (NGF)-dependent neuronal cell survival and differentiation. The small GTPase ADP-ribosylation factor 6 (Arf6) is implicated in NGF-dependent processes in PC12 cells through endosomal trafficking and actin cytoskeleton reorganization. However, the regulatory mechanism for Arf6 in NGF signaling is largely unknown. In this study, we demonstrated that EFA6A, an Arf6-specific guanine nucleotide exchange factor, was abundantly expressed in PC12 cells and that knockdown of EFA6A significantly inhibited NGF-dependent Arf6 activation, TrkA recycling from early endosomes to the cell surface, prolonged ERK1/2 phosphorylation, and neurite outgrowth. We also demonstrated that EFA6A forms a protein complex with TrkA through its N-terminal region, thereby enhancing its catalytic activity for Arf6. Similarly, we demonstrated that EFA6A forms a protein complex with TrkA in cultured dorsal root ganglion (DRG) neurons. Furthermore, cultured DRG neurons from EFA6A knockout mice exhibited disturbed NGF-dependent TrkA trafficking compared with wild-type neurons. These findings provide the first evidence for EFA6A as a key regulator of NGF-dependent TrkA trafficking and signaling.
Collapse
Affiliation(s)
- Masahiro Fukaya
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kanta Ibuchi
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Takeyuki Sugawara
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Makoto Itakura
- Department of Biochemistry, Kitasato University School of Medicine, Sagamihara, Japan
| | - Akiko Ito
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Tomoko Shiroshima
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yoshinobu Hara
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hirotsugu Okamoto
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Frédéric Luton
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Université Côte d'Azur, Valbonne, France
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| |
Collapse
|
2
|
Glover KKM, Zahedi-Amiri A, Lao Y, Spicer V, Klonisch T, Coombs KM. Zika Infection Disrupts Proteins Involved in the Neurosensory System. Front Cell Dev Biol 2020; 8:571. [PMID: 32850779 PMCID: PMC7403212 DOI: 10.3389/fcell.2020.00571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/15/2020] [Indexed: 11/13/2022] Open
Abstract
Newly re-emerging viruses are of great global concern, especially when there are no therapeutic interventions available during the time of an outbreak. There are still no therapeutic interventions for the prevention of Zika virus (ZIKV) infections despite its resurgence more than a decade ago. Newborns infected with ZIKV suffer from microcephaly and delayed neurodevelopment, but the underlying causes are largely unknown. All viruses hijack the host cellular machinery to undergo successful replication. Our tandem mass tag mass spectrometry-based proteomic monitoring of cells infected with ZIKV revealed that among the thousands of host proteins dysregulated over time, many protein candidates were linked to neurodevelopmental processes, including the development of the auditory and visual/retinal system. The role of these dysregulated neurodevelopmental-associated host proteins for ZIKV propagation in eukaryotic cells remains elusive. For the first time, we present temporal neurodevelopmental proteomic responses in cells undergoing ZIKV infection. The future goal is to identify host proteins whose dysregulation results in neurosensory alterations reported in children born to ZIKV-infected mothers.
Collapse
Affiliation(s)
- Kathleen K. M. Glover
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB, Canada
| | - Ali Zahedi-Amiri
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB, Canada
| | - Ying Lao
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB, Canada
| | - Victor Spicer
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB, Canada
| | - Thomas Klonisch
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
- John Buhler Research Centre, Children’s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Kevin M. Coombs
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB, Canada
- John Buhler Research Centre, Children’s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
3
|
Oguchi ME, Okuyama K, Homma Y, Fukuda M. A comprehensive analysis of Rab GTPases reveals a role for Rab34 in serum starvation-induced primary ciliogenesis. J Biol Chem 2020; 295:12674-12685. [PMID: 32669361 DOI: 10.1074/jbc.ra119.012233] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 07/14/2020] [Indexed: 12/17/2022] Open
Abstract
Primary cilia are sensors of chemical and mechanical signals in the extracellular environment. The formation of primary cilia (i.e. ciliogenesis) requires dynamic membrane trafficking events, and several Rab small GTPases, key regulators of membrane trafficking, have recently been reported to participate in ciliogenesis. However, the precise mechanisms of Rab-mediated membrane trafficking during ciliogenesis remain largely unknown. In the present study, we used a collection of siRNAs against 62 human Rabs to perform a comprehensive knockdown screening for Rabs that regulate serum starvation-induced ciliogenesis in human telomerase reverse transcriptase retinal pigment epithelium 1 (hTERT-RPE1) cells and succeeded in identifying Rab34 as an essential Rab. Knockout (KO) of Rab34, but not of Rabs previously reported to regulate ciliogenesis (e.g. Rab8 and Rab10) in hTERT-RPE1 cells, drastically impaired serum starvation-induced ciliogenesis. Rab34 was also required for serum starvation-induced ciliogenesis in NIH/3T3 cells and MCF10A cells but not for ciliogenesis in Madin-Darby canine kidney (MDCK)-II cysts. We then attempted to identify a specific region(s) of Rab34 that is essential for ciliogenesis by performing deletion and mutation analyses of Rab34. Unexpectedly, instead of a specific sequence in the switch II region, which is generally important for recognizing effector proteins (e.g. Rab interacting lysosomal protein [RILP]), a unique long N-terminal region of Rab34 before the conserved GTPase domain was found to be essential. These findings suggest that Rab34 is an atypical Rab that regulates serum starvation-induced ciliogenesis through its unique N-terminal region.
Collapse
Affiliation(s)
- Mai E Oguchi
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi, Japan
| | - Koki Okuyama
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi, Japan
| | - Yuta Homma
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi, Japan
| |
Collapse
|
4
|
Abstract
The mechanism of GTPase specificity remains incompletely understood. In this issue of Structure, Lin et al. (2019) present crystal structures of Rab35 in complex with two effectors, ACAP2 and RUSC2, respectively, unraveling the molecular basis for the exquisite mutual specificity between Rab35 and these effectors.
Collapse
Affiliation(s)
- Yi-Chun Kuo
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390
| | - Xuewu Zhang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390.
| |
Collapse
|
5
|
Lin L, Shi Y, Wang M, Wang C, Zhu J, Zhang R. Rab35/ACAP2 and Rab35/RUSC2 Complex Structures Reveal Molecular Basis for Effector Recognition by Rab35 GTPase. Structure 2019; 27:729-740.e3. [PMID: 30905672 DOI: 10.1016/j.str.2019.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/03/2019] [Accepted: 02/24/2019] [Indexed: 11/16/2022]
Abstract
Rab35, a master regulator of membrane trafficking, regulates diverse cellular processes and is associated with various human diseases. Although a number of effectors have been identified, the molecular basis of Rab35-effector interactions remains unclear. Here, we provide the high-resolution crystal structures of Rab35 in complex with its two specific effectors ACAP2 and RUSC2, respectively. In the Rab35/ACAP2 complex structure, Rab35 binds to the terminal ankyrin repeat and a C-terminal extended α helix of ACAP2, revealing a previously uncharacterized binding mode both for Rabs and ankyrin repeats. In the Rab35/RUSC2 complex structure, Arg1015 of RUSC2 functions as a "pseudo-arginine finger" that stabilizes the GTP-bound Rab35, thus facilitating the assembly of Rab35/RUSC2 complex. The structural analysis allows us to design specific Rab35 mutants capable of eliminating Rab35/ACAP2 and Rab35/RUSC2 interactions, but not interfering with other effector bindings. The atomic structures also offer possible explanations to disease-associated mutants identified at the Rab35-effector interfaces.
Collapse
Affiliation(s)
- Lin Lin
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai Science Research Center, 333 Haike Road, Shanghai 201210, China
| | - Yingdong Shi
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Mengli Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Chao Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Jinwei Zhu
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai Science Research Center, 333 Haike Road, Shanghai 201210, China.
| | - Rongguang Zhang
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai Science Research Center, 333 Haike Road, Shanghai 201210, China.
| |
Collapse
|
6
|
Tanna CE, Goss LB, Ludwig CG, Chen PW. Arf GAPs as Regulators of the Actin Cytoskeleton-An Update. Int J Mol Sci 2019; 20:ijms20020442. [PMID: 30669557 PMCID: PMC6358971 DOI: 10.3390/ijms20020442] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/25/2022] Open
Abstract
Arf GTPase-activating proteins (Arf GAPs) control the activity of ADP-ribosylation factors (Arfs) by inducing GTP hydrolysis and participate in a diverse array of cellular functions both through mechanisms that are dependent on and independent of their Arf GAP activity. A number of these functions hinge on the remodeling of actin filaments. Accordingly, some of the effects exerted by Arf GAPs involve proteins known to engage in regulation of the actin dynamics and architecture, such as Rho family proteins and nonmuscle myosin 2. Circular dorsal ruffles (CDRs), podosomes, invadopodia, lamellipodia, stress fibers and focal adhesions are among the actin-based structures regulated by Arf GAPs. Arf GAPs are thus important actors in broad functions like adhesion and motility, as well as the specialized functions of bone resorption, neurite outgrowth, and pathogen internalization by immune cells. Arf GAPs, with their multiple protein-protein interactions, membrane-binding domains and sites for post-translational modification, are good candidates for linking the changes in actin to the membrane. The findings discussed depict a family of proteins with a critical role in regulating actin dynamics to enable proper cell function.
Collapse
Affiliation(s)
- Christine E Tanna
- Department of Biology, Williams College, Williamstown, MA 01267, USA.
| | - Louisa B Goss
- Department of Biology, Williams College, Williamstown, MA 01267, USA.
| | - Calvin G Ludwig
- Department of Biology, Williams College, Williamstown, MA 01267, USA.
| | - Pei-Wen Chen
- Department of Biology, Williams College, Williamstown, MA 01267, USA.
| |
Collapse
|
7
|
Chiu CC, Yeh TH, Lai SC, Weng YH, Huang YC, Cheng YC, Chen RS, Huang YZ, Hung J, Chen CC, Lin WY, Chang HC, Chen YJ, Chen CL, Chen HY, Lin YW, Wu-Chou YH, Wang HL, Lu CS. Increased Rab35 expression is a potential biomarker and implicated in the pathogenesis of Parkinson's disease. Oncotarget 2018; 7:54215-54227. [PMID: 27509057 PMCID: PMC5342336 DOI: 10.18632/oncotarget.11090] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/29/2016] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease (PD) is the second common neurodegenerative disease. Identification of biomarkers for early diagnosis and prediction of disease progression is important. The present comparative proteomic study of serum samples using two-dimensional fluorescence differential gel electrophoresis followed by ELISA confirmation demonstrated that protein expression of Rab35 was increased in PD patients compared with matched control subjects and other parkinsonian disorders, progressive supranuclear palsy (PSP) and multiple system atrophy (MSA). The serum level of Rab35 was significantly correlated with the age at onset of PD. The median age of onset in patients with higher Rab35 serum level was 5 years younger than those with lower Rab35 serum level. There was a positive correlation between the Rab35 level and disease duration of PD. Moreover, the protein expression of Rab35 was increased in the substantia nigra but not in the striatum of mouse models of PD, including MPTP-treated mice, rotenone-treated mice, (R1441C) LRRK2 or (G2019S) LRRK2 transgenic mice. Furthermore, overexpression of Rab35 increased the aggregation and secretion of mutant A53T α-synuclein in dopaminergic SH-SY5Y cells. Co-expression of Rab35 with wild-type or A53T α-synuclein in SH-SY5Y cells deteriorated cell death. Our results suggest that Rab35 is potentially useful in the differential diagnosis of parkinsonian disorders and is implicated in the pathogenesis of PD.
Collapse
Affiliation(s)
- Ching-Chi Chiu
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan.,Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Tu-Hsueh Yeh
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Szu-Chia Lai
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Hsin Weng
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yin-Cheng Huang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Neurosurgery, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yi-Chuan Cheng
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Biomedical Sciences, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Rou-Shayn Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ying-Zu Huang
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Institute of Cognitive Neuroscience, National Central University, Taoyuan,Taiwan
| | - June Hung
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chiung-Chu Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wey-Yil Lin
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsiu-Chen Chang
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Yu-Jie Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chao-Lang Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Hsin-Yi Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yan-Wei Lin
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yah-Huei Wu-Chou
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Hung-Li Wang
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Department of Physiology and Pharmacology, Chang Gung University School of Medicine, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Chin-Song Lu
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
8
|
Rab20, a novel Rab small GTPase that negatively regulates neurite outgrowth of PC12 cells. Neurosci Lett 2017; 662:324-330. [PMID: 29107708 DOI: 10.1016/j.neulet.2017.10.056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/17/2017] [Accepted: 10/27/2017] [Indexed: 11/20/2022]
Abstract
The Rab family small GTPases are key players in the membrane traffic that underlies various cellular phenomena. Neurite outgrowth, which is a prerequisite for neuronal network formation, also requires membrane traffic from the cell body to the tips of neurites. Although several Rabs have been shown to promote neurite outgrowth, very little is known about Rab involvement in the negative regulation of neurite outgrowth. Here we used nerve growth factor-stimulated PC12 cells to perform siRNA-based comprehensive knockdown screenings for Rabs that negatively regulate neurite outgrowth and succeeded in identifying Rab20 as a novel negative regulator of neurite outgrowth. Our findings showed that knockdown of endogenous Rab20 in PC12 cells promoted neurite outgrowth, whereas overexpression of active Rab20 inhibited it. We also found that the presence of Gly-64 and Cys-70, both of which are conserved only in the switch II region, a putative effector binding domain, of Rab20 is required for the inhibitory effect of Rab20 on neurite outgrowth. These findings indicated that active Rab20 suppresses neurite outgrowth of PC12 cells, possibly through interaction with an unidentified effector molecule that specifically recognizes certain amino acids in the switch II region of Rab20.
Collapse
|
9
|
Rab35 Functions in Axon Elongation Are Regulated by P53-Related Protein Kinase in a Mechanism That Involves Rab35 Protein Degradation and the Microtubule-Associated Protein 1B. J Neurosci 2017; 36:7298-313. [PMID: 27383602 DOI: 10.1523/jneurosci.4064-15.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 05/30/2016] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Rab35 is a key protein for cargo loading in the recycling endosome. In neuronal immortalized cells, Rab35 promotes neurite differentiation. Here we describe that Rab35 favors axon elongation in rat primary neurons in an activity-dependent manner. In addition, we show that the p53-related protein kinase (PRPK) negatively regulates axonal elongation by reducing Rab35 protein levels through the ubiquitin-proteasome degradation pathway. PRPK-induced Rab35 degradation is regulated by its interaction with microtubule-associated protein 1B (MAP1B), a microtubule stabilizing binding protein essential for axon elongation. Consistently, axon defects found in MAP1B knock-out neurons were reversed by Rab35 overexpression or PRPK inactivation suggesting an epistatic relationship among these proteins. These results define a novel mechanism to support axonal elongation, by which MAP1B prevents PRPK-induced Rab35 degradation. Such a mechanism allows Rab35-mediated axonal elongation and connects the regulation of actin dynamics with membrane trafficking. In addition, our study reveals for the first time that the ubiquitin-proteasome degradation pathway regulates a Rab GTPase. SIGNIFICANCE STATEMENT Rab35 is required for axonal outgrowth. We define that its protein levels are negatively regulated by p53-related protein kinase (PRPK). We show that microtubule-associated protein 1B (MAP1B) interacts with PRPK, preventing PRPK-dependent Rab35 proteasome degradation. We demonstrate that Rab35 regulates Cdc42 activity in neurons. This is the first evidence showing that a Rab protein is regulated by degradation dependent on the ubiquitin-proteasome system.
Collapse
|
10
|
Abstract
Cell polarity refers to the asymmetric localization of cellular components that allows cells to carry out their specialized functions, be they epithelial barrier function, transmission of action potentials in nerve cells, or modulation of the immune response. The establishment and maintenance of cell polarity requires the directed trafficking of membrane proteins and lipids - essential processes that are mediated by Rab GTPases. Interestingly, several of the Rabs that impact polarity are present in the earliest eukaryotes, and the Rab polarity repertoire has expanded as cells have become more complex. There is a substantial conservation of Rab function across diverse cell types. Rabs act through an assortment of effector proteins that include scaffolding proteins, cytoskeletal motors, and other small GTPases. In this review we highlight the similarities and differences in Rab function for the instruction of polarity in diverse cell types.
Collapse
Affiliation(s)
- Sara S Parker
- a Department of Cellular and Molecular Medicine , University of Arizona , Tucson , AZ , USA
| | - Christopher Cox
- a Department of Cellular and Molecular Medicine , University of Arizona , Tucson , AZ , USA
| | - Jean M Wilson
- a Department of Cellular and Molecular Medicine , University of Arizona , Tucson , AZ , USA
| |
Collapse
|
11
|
Klinkert K, Echard A. Rab35 GTPase: A Central Regulator of Phosphoinositides and F-actin in Endocytic Recycling and Beyond. Traffic 2016; 17:1063-77. [PMID: 27329675 DOI: 10.1111/tra.12422] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/12/2016] [Accepted: 06/12/2016] [Indexed: 12/11/2022]
Abstract
Rab35 is one of the first discovered members of the large Rab GTPase family, yet it received little attention for 10 years being considered merely as a Rab1-like GTPase. In 2006, Rab35 was recognized as a unique Rab GTPase localized both at the plasma membrane and on endosomes, playing essential roles in endocytic recycling and cytokinesis. Since then, Rab35 has become one of the most studied Rabs involved in a growing number of cellular functions, including endosomal trafficking, exosome release, phagocytosis, cell migration, immunological synapse formation and neurite outgrowth. Recently, Rab35 has been acknowledged as an oncogenic GTPase with activating mutations being found in cancer patients. In this review, we provide a comprehensive summary of known Rab35-dependent cellular functions and detail the few Rab35 effectors characterized so far. We also review how the Rab35 GTP/GDP cycle is regulated, and emphasize a newly discovered mechanism that controls its tight activation on newborn endosomes. We propose that the involvement of Rab35 in such diverse and apparently unrelated cellular functions can be explained by the central role of this GTPase in regulating phosphoinositides and F-actin, both on endosomes and at the plasma membrane.
Collapse
Affiliation(s)
- Kerstin Klinkert
- Membrane Traffic and Cell Division Lab, Cell Biology and Infection Department, Institut Pasteur, 25-28 rue du Dr Roux, 75724, Paris, France.,Centre National de la Recherche Scientifique, UMR3691, 75015, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie, Université Paris 06, Institut de formation doctorale, 75252, Paris, France
| | - Arnaud Echard
- Membrane Traffic and Cell Division Lab, Cell Biology and Infection Department, Institut Pasteur, 25-28 rue du Dr Roux, 75724, Paris, France. .,Centre National de la Recherche Scientifique, UMR3691, 75015, Paris, France.
| |
Collapse
|
12
|
Mrozowska PS, Fukuda M. Regulation of podocalyxin trafficking by Rab small GTPases in 2D and 3D epithelial cell cultures. J Cell Biol 2016; 213:355-69. [PMID: 27138252 PMCID: PMC4862332 DOI: 10.1083/jcb.201512024] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/18/2016] [Indexed: 11/22/2022] Open
Abstract
MDCK II cells, a widely used model of polarized epithelia, develop into different structures depending on culture conditions: two-dimensional (2D) monolayers when grown on synthetic supports or three-dimensional (3D) cysts when surrounded by an extracellular matrix. The establishment of epithelial polarity is accompanied by transcytosis of the apical marker podocalyxin from the outer plasma membrane to the newly formed apical domain, but its exact route and regulation remain poorly understood. Here, through comprehensive colocalization and knockdown screenings, we identified the Rab GTPases mediating podocalyxin transcytosis and showed that different sets of Rabs coordinate its transport during cell polarization in 2D and 3D structures. Moreover, we demonstrated that different Rab35 effectors regulate podocalyxin trafficking in 2D and 3D environments; trafficking is mediated by OCRL in 2D monolayers and ACAP2 in 3D cysts. Our results give substantial insight into regulation of the transcytosis of this apical marker and highlight differences between trafficking mechanisms in 2D and 3D cell cultures.
Collapse
Affiliation(s)
- Paulina S Mrozowska
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
13
|
Basso M, Bonetto V. Extracellular Vesicles and a Novel Form of Communication in the Brain. Front Neurosci 2016; 10:127. [PMID: 27065789 PMCID: PMC4814526 DOI: 10.3389/fnins.2016.00127] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/14/2016] [Indexed: 01/08/2023] Open
Abstract
In numerous neurodegenerative diseases, the interplay between neurons and glia modulates the outcome and progression of pathology. One particularly intriguing mode of interaction between neurons, astrocytes, microglia, and oligodendrocytes is characterized by the release of extracellular vesicles that transport proteins, lipids, and nucleotides from one cell to another. Notably, several proteins that cause disease, including the prion protein and mutant SOD1, have been detected in glia-derived extracellular vesicles and observed to fuse with neurons and trigger pathology in vitro. Here we review the structural and functional characterization of such extracellular vesicles in neuron-glia interactions. Furthermore, we discuss possible mechanisms of extracellular vesicle biogenesis and release from activated glia and microglia, and their effects on neurons. Given that exosomes, the smallest type of extracellular vesicles, have been reported to recognize specific cellular populations and act as carriers of very specialized cargo, a thorough analysis of these vesicles may aid in their engineering in vitro and targeted delivery in vivo, opening opportunities for therapeutics.
Collapse
Affiliation(s)
- Manuela Basso
- Laboratory of Transcriptional Neurobiology, Centre for Integrative Biology, University of Trento Trento, Italy
| | - Valentina Bonetto
- Department of Molecular Biochemistry and Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri," Milano, Italy
| |
Collapse
|
14
|
Tang Y, Lin Y, Li C, Hu X, Liu Y, He M, Luo J, Sun G, Wang T, Li W, Guo M. MicroRNA-720 promotes in vitro cell migration by targeting Rab35 expression in cervical cancer cells. Cell Biosci 2015; 5:56. [PMID: 26413265 PMCID: PMC4583841 DOI: 10.1186/s13578-015-0047-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 09/22/2015] [Indexed: 12/18/2022] Open
Abstract
Background MicroRNA-720 (miR-720), a nonclassical miRNA, is involved in the initiation and progression of several tumors. In our previous studies, miR-720 was shown to be significantly upregulated in cervical cancer tissues compared with normal cervical tissues. However, the precise biological functions of miR-720, and its molecular mechanisms of action, are still unknown. Results Microarray expression profiles, luciferase reporter assays, and western blot assays were used to validate Rab35 as a target gene of miR-720 in HEK293T and HeLa cells. The regulation of Rab35 expression by miR-720 was assessed using qRT-PCR and western blot assays, and the effects of exogenous miR-720 and Rab35 on cell migration were evaluated in vitro using Transwell® assay, wound healing assay, and real-time analyses in HeLa cells. The influences of exogenous miR-720 on cell proliferation were evaluated in vitro by the MTT assay in HeLa cells. In addition, expression of E-cadherin and vimentin associated with epithelial-mesenchymal transition were also assessed using western blot analyses after transfection of miR-720 mimics and Rab35 expression vectors. The results showed that the small GTPase, Rab35, is a direct functional target of miR-720 in cervical cancer HeLa cells. By targeting Rab35, overexpression of miR-720 resulted in a decrease in E-cadherin expression and an increase in vimentin expression and finally led to promotion of HeLa cell migration. Furthermore, reintroduction of Rab35 3′-UTR(−) markedly reversed the induction of cell migration in miR-720-expressing HeLa cells. Conclusions The miR-720 promotes cell migration of HeLa cells by downregulating Rab35. The results show that miR-720 is a novel cell migration-associated gene in cervical cancer cells. Electronic supplementary material The online version of this article (doi:10.1186/s13578-015-0047-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yunlan Tang
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Yi Lin
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Chuang Li
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Xunwu Hu
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Yi Liu
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Mingyang He
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Jun Luo
- Department of Pathology, Zhongnan Hospital, Wuhan University, 430071 Wuhan, People's Republic of China
| | - Guihong Sun
- School of Basic Medical Sciences, Wuhan University, 430071 Wuhan, People's Republic of China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Wenxin Li
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Mingxiong Guo
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| |
Collapse
|
15
|
Activation-Inactivation Cycling of Rab35 and ARF6 Is Required for Phagocytosis of Zymosan in RAW264 Macrophages. J Immunol Res 2015; 2015:429439. [PMID: 26229970 PMCID: PMC4502309 DOI: 10.1155/2015/429439] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 05/25/2015] [Indexed: 12/26/2022] Open
Abstract
Phagocytosis of zymosan by phagocytes is a widely used model of microbial recognition by the innate immune system. Live-cell imaging showed that fluorescent protein-fused Rab35 accumulated in the membranes of phagocytic cups and then dissociated from the membranes of newly formed phagosomes. By our novel pull-down assay for Rab35 activity, we found that Rab35 is deactivated immediately after zymosan internalization into the cells. Phagosome formation was inhibited in cells expressing the GDP- or GTP-locked Rab35 mutant. Moreover, the simultaneous expression of ACAP2-a Rab35 effector protein-with GTP-locked Rab35 or the expression of plasma membrane-targeted ACAP2 showed a marked inhibitory effect on phagocytosis through ARF6 inactivation by the GAP activity of ACAP2. ARF6, a substrate for ACAP2, was also localized on the phagocytic cups and dissociated from the membranes of internalized phagosomes. In support of the microscopic observations, ARF6-GTP pull-down experiments showed that ARF6 is transiently activated during phagosome formation. Furthermore, the expression of GDP- or GTP-locked ARF6 mutants also suppresses the uptake of zymosan. These data suggest that the activation-inactivation cycles of Rab35 and ARF6 are required for the uptake of zymosan and that ACAP2 is an important component that links Rab35/ARF6 signaling during phagocytosis of zymosan.
Collapse
|