1
|
Luo Y, Pezacki AT, Matier CD, Wang WX. A novel route of intercellular copper transport and detoxification in oyster hemocytes. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135003. [PMID: 38917627 DOI: 10.1016/j.jhazmat.2024.135003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/03/2024] [Accepted: 06/20/2024] [Indexed: 06/27/2024]
Abstract
Bivalve hemocytes are oyster immune cells composed of several cellular subtypes with different functions. Hemocytes accumulate high concentrations of copper (Cu) and exert critical roles in metal sequestration and detoxification in oysters, however the specific biochemical mechanisms that govern this have yet to be fully uncovered. Herein, we demonstrate that Cu(I) is predominately sequestered in lysosomes via the Cu transporter ATP7A in hemocytes to reduce the toxic effects of intracellular Cu(I). We also found that Cu(I) is translocated along tunneling nanotubes (TNTs) relocating from high Cu(I) cells to low Cu(I) cells, effectively reducing the burden caused by overloaded Cu(I), and that ATP7A facilitates the efflux of intracellular Cu(I) in both TNTs and hemocyte subtypes. We identify that elevated glutathione (GSH) contents and heat-shock protein (Hsp) levels, as well as the activation of the cell cycle were critical in maintaining the cellular homeostasis and function of hemocytes exposed to Cu. Cu exposure also increased the expression of membrane proteins (MYOF, RalA, RalBP1, and cadherins) and lipid transporter activity which can induce TNT formation, and activated the lysosomal signaling pathway, promoting intercellular lysosomal trafficking dependent on increased hydrolase activity and ATP-dependent activity. This study explores the intracellular and intercellular transport and detoxification of Cu in oyster hemocytes, which may help in understanding the potential toxicity and fate of metals in marine animals.
Collapse
Affiliation(s)
- Yali Luo
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| | - Aidan T Pezacki
- Departments of Chemistry and Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Carson D Matier
- Departments of Chemistry and Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Wen-Xiong Wang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China.
| |
Collapse
|
2
|
Jin H, Qin S, He J, Xiao J, Li Q, Mao Y, Zhao L. Systematic pan-cancer analysis identifies RALA as a tumor targeting immune therapeutic and prognostic marker. Front Immunol 2022; 13:1046044. [PMID: 36466919 PMCID: PMC9713825 DOI: 10.3389/fimmu.2022.1046044] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/02/2022] [Indexed: 10/07/2023] Open
Abstract
INTRODUCTION RALA is a member of the small GTPase Ras superfamily and has been shown to play a role in promoting cell proliferation and migration in most tumors, and increase the resistance of anticancer drugs such as imatinib and cisplatin. Although many literatures have studied the cancer-promoting mechanism of RALA, there is a lack of relevant pan-cancer analysis. METHODS This study systematically analyzed the differential expression and mutation of RALA in pan-cancer, including different tissues and cancer cell lines, and studied the prognosis and immune infiltration associated with RALA in various cancers. Next, based on the genes co-expressed with RALA in pan-cancer, we selected 241 genes with high correlation for enrichment analysis. In terms of pan-cancer, we also analyzed the protein-protein interaction pathway of RALA and the application of small molecule drug Guanosine-5'-Diphosphate. We screened hepatocellular cancer (HCC) to further study RALA. RESULTS The results indicated that RALA was highly expressed in most cancers. RALA was significantly correlated with the infiltration of B cells and macrophages, as well as the expression of immune checkpoint molecules such as CD274, CTLA4, HAVCR2 and LAG3, suggesting that RALA can be used as a kind of new pan-cancer immune marker. The main functions of 241 genes are mitosis and protein localization to nucleosome, which are related to cell cycle. For HCC, the results displayed that RALA was positively correlated with common intracellular signaling pathways such as angiogenesis and apoptosis. DISCUSSION In summary, RALA was closely related to the clinical prognosis and immune infiltration of various tumors, and RALA was expected to become a broad-spectrum molecular immune therapeutic target and prognostic marker for pan-cancer.
Collapse
Affiliation(s)
- Haoer Jin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Sha Qin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jiang He
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juxiong Xiao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qingling Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yitao Mao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luqing Zhao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Richardson DS, Spehar JM, Han DT, Chakravarthy PA, Sizemore ST. The RAL Enigma: Distinct Roles of RALA and RALB in Cancer. Cells 2022; 11:cells11101645. [PMID: 35626682 PMCID: PMC9139244 DOI: 10.3390/cells11101645] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 11/16/2022] Open
Abstract
RALA and RALB are highly homologous small G proteins belonging to the RAS superfamily. Like other small GTPases, the RALs are molecular switches that can be toggled between inactive GDP-bound and active GTP-bound states to regulate diverse and critical cellular functions such as vesicle trafficking, filopodia formation, mitochondrial fission, and cytokinesis. The RAL paralogs are activated and inactivated by a shared set of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs) and utilize similar sets of downstream effectors. In addition to their important roles in normal cell biology, the RALs are known to be critical mediators of cancer cell survival, invasion, migration, and metastasis. However, despite their substantial similarities, the RALs often display striking functional disparities in cancer. RALA and RALB can have redundant, unique, or even antagonistic functions depending on cancer type. The molecular basis for these discrepancies remains an important unanswered question in the field of cancer biology. In this review we examine the functions of the RAL paralogs in normal cellular physiology and cancer biology with special consideration provided to situations where the roles of RALA and RALB are non-redundant.
Collapse
|
4
|
Chagraoui A, Di Giovanni G, De Deurwaerdère P. Neurobiological and Pharmacological Perspectives of D3 Receptors in Parkinson’s Disease. Biomolecules 2022; 12:biom12020243. [PMID: 35204744 PMCID: PMC8961531 DOI: 10.3390/biom12020243] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/19/2022] [Accepted: 01/26/2022] [Indexed: 12/02/2022] Open
Abstract
The discovery of the D3 receptor (D3R) subtypes of dopamine (DA) has generated an understandable increase in interest in the field of neurological diseases, especially Parkinson’s disease (PD). Indeed, although DA replacement therapy with l-DOPA has provided an effective treatment for patients with PD, it is responsible for invalidating abnormal involuntary movements, known as L-DOPA-induced dyskinesia, which constitutes a serious limitation of the use of this therapy. Of particular interest is the finding that chronic l-DOPA treatment can trigger the expression of D1R–D3R heteromeric interactions in the dorsal striatum. The D3R is expressed in various tissues of the central nervous system, including the striatum. Compelling research has focused on striatal D3Rs in the context of PD and motor side effects, including dyskinesia, occurring with DA replacement therapy. Therefore, this review will briefly describe the basal ganglia (BG) and the DA transmission within these brain regions, before going into more detail with regard to the role of D3Rs in PD and their participation in the current treatments. Numerous studies have also highlighted specific interactions between D1Rs and D3Rs that could promote dyskinesia. Finally, this review will also address the possibility that D3Rs located outside of the BG may mediate some of the effects of DA replacement therapy.
Collapse
Affiliation(s)
- Abdeslam Chagraoui
- Différenciation et Communication Neuroendocrine, Endocrine et Germinale Laboratory, Institute for Research and Innovation in Biomedicine of Normandy (IRIB), University of Rouen, INSERM 1239, 76000 Rouen, France
- Department of Medical Biochemistry, Rouen University Hospital, 76000 Rouen, France
- Correspondence: ; Tel.: +33-2-35-14-83-69
| | - Giuseppe Di Giovanni
- Laboratory of Neurophysiology, Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, 2080 Msida, Malta;
- Neuroscience Division, School of Biosciences, Cardiff University, Cardiff CF10 3AT, UK
| | - Philippe De Deurwaerdère
- Unité Mixte de Recherche (UMR) 5287, Centre National de la Recherche Scientifique (CNRS), CEDEX, 33000 Bordeaux, France;
| |
Collapse
|
5
|
Abstract
Ras is the most mutated oncoprotein in cancer. Among the three oncogenic effectors of Ras - Raf, PI3 Kinase and RalGEF>Ral - signalling through RalGEF>Ral (Ras-like) is by far the least well understood. A variety of signals and binding partners have been defined for Ral, yet we know little of how Ral functions in vivo. This review focuses on previous research in Drosophila that defined a function for Ral in apoptosis and established indirect relationships among Ral, the CNH-domain MAP4 Kinase misshapen, and the JNK MAP kinase basket. Most of the described signalling components are not essential in C. elegans, facilitating subsequent analysis using developmental patterning of the C. elegans vulval precursor cells (VPCs). The functions of two paralogous CNH-domain MAP4 Kinases were defined relative to Ras>Raf, Notch and Ras>RalGEF>Ral signalling in VPCs. MIG-15, the nematode ortholog of misshapen, antagonizes both the Ral-dependent and Ras>Raf-dependent developmental outcomes. In contrast, paralogous GCK-2, the C. elegans ortholog of Drosophila happyhour, propagates the 2°-promoting signal of Ral. Manipulations via CRISPR of Ral signalling through GCK-2 coupled with genetic epistasis delineated a Ras>RalGEF>Ral>Exo84>GCK-2>MAP3KMLK-1> p38PMK-1 cascade. Thus, genetic analysis using invertebrate experimental organisms defined a cascade from Ras to p38 MAP kinase.
Collapse
Affiliation(s)
| | - David J. Reiner
- Texas A&M University, Houston, TX, USA,CONTACT David J. Reiner Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, Houston, TX
| |
Collapse
|
6
|
Kano T, Tsumagari R, Nakashima A, Kikkawa U, Ueda S, Yamanoue M, Takei N, Shirai Y. RalA, PLD and mTORC1 Are Required for Kinase-Independent Pathways in DGKβ-Induced Neurite Outgrowth. Biomolecules 2021; 11:1814. [PMID: 34944458 PMCID: PMC8699322 DOI: 10.3390/biom11121814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Diacylglycerol kinase β (DGKβ) is an enzyme that converts diacylglycerol to phosphatidic acid and is mainly expressed in the cerebral cortex, hippocampus and striatum. We previously reported that DGKβ induces neurite outgrowth and spinogenesis, contributing to higher brain functions, including emotion and memory. To elucidate the mechanisms involved in neuronal development by DGKβ, we investigated the importance of DGKβ activity in the induction of neurite outgrowth using human neuroblastoma SH-SY5Y cells. Interestingly, both wild-type DGKβ and the kinase-negative (KN) mutant partially induced neurite outgrowth, and these functions shared a common pathway via the activation of mammalian target of rapamycin complex 1 (mTORC1). In addition, we found that DGKβ interacted with the small GTPase RalA and that siRNA against RalA and phospholipase D (PLD) inhibitor treatments abolished DGKβKN-induced neurite outgrowth. These results indicate that binding of RalA and activation of PLD and mTORC1 are involved in DGKβKN-induced neurite outgrowth. Taken together with our previous reports, mTORC1 is a key molecule in both kinase-dependent and kinase-independent pathways of DGKβ-mediated neurite outgrowth, which is important for higher brain functions.
Collapse
Affiliation(s)
- Takuya Kano
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (T.K.); (R.T.); (S.U.); (M.Y.)
| | - Ryosuke Tsumagari
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (T.K.); (R.T.); (S.U.); (M.Y.)
| | - Akio Nakashima
- Division of Signal Functions, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan; (A.N.); (U.K.)
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Ushio Kikkawa
- Division of Signal Functions, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan; (A.N.); (U.K.)
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Shuji Ueda
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (T.K.); (R.T.); (S.U.); (M.Y.)
| | - Minoru Yamanoue
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (T.K.); (R.T.); (S.U.); (M.Y.)
| | - Nobuyuki Takei
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan;
| | - Yasuhito Shirai
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (T.K.); (R.T.); (S.U.); (M.Y.)
| |
Collapse
|
7
|
Wang X, Gou L, Gao Y, Huang Y, Kuai R, Li Y, Wang Y, Chen Y, Li J, Cheng C, Feng Z, Wu X, Yao R. RalA exerts an inhibitory effect on IL-1β/IL-18 secretion by blocking NLRP3 inflammasome activation in levornidazole-treated human THP-1 macrophages. Int Immunopharmacol 2020; 88:106898. [PMID: 32866784 DOI: 10.1016/j.intimp.2020.106898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 12/17/2022]
Abstract
The NLRP3 inflammasome is an important mediator of inflammatory responses and its regulation is an active area of research. RalA is a Ras-like GTPase, which play pivotal roles in the biology of cells. So far, there have been very few studies on RalA regulating inflammatory responses. Bioinformatics analysis predicted that RalA might participate in the regulatory network of NLRP3 inflammasome, which has been confirmed in THP-1 macrophages. After virtual screening of compounds, it was found that levonidazole selected from our virtual small molecule compound library has the potential to bind to RalA. Of note, the interaction of RalA/levornidazole was verified by Surface Plasmon Resonance-Biacore T200, LC/MS analysis and Western blotting analysis. Molecular dynamics simulations revealed that the conformational changes of RalA might be regulated by levornidazole. Additionally, IL-1β/IL-18 secretion from ATP + LPS stimulated THP-1-derived macrophages was RalA-dependently suppressed by levornidazole, suggesting that RalA might have an inhibitory effect on NLRP3 inflammasome activation. The results of co-immunoprecipitation and RalA depletion experiments showed that levornidazole could induce RalA to block the assembly of NLRP3/ASC/pro-caspase-1 complex, thereby reducing the levels of cleaved-caspase-1 and IL-1β/IL-18 secretion. Our study has suggested an anti-inflammatory function of RalA and identified its targeting chemical compound. Overall, this study clarifies a novel pharmacological mechanism by which RalA/levornidazole inhibits NLRP3 inflammasome activation and IL-1β/IL-18 secretion.
Collapse
Affiliation(s)
- Xingqi Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu, China.
| | - Lingshan Gou
- Center for Genetic Medicine, Xuzhou Maternity and Child Health Care Hospital, Xuzhou 221009, Jiangsu, China
| | - Yuzhi Gao
- Department of Cell Biology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221009, Jiangsu, China
| | - Yuqing Huang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu, China
| | - Rui Kuai
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu, China
| | - Yu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu, China
| | - Yujing Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu, China
| | - Yanhong Chen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu, China
| | - Jun Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu, China
| | - Chao Cheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu, China
| | - Zhaojun Feng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu, China
| | - Xuefeng Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210093, Jiangsu, China.
| | - Ruiqin Yao
- Department of Cell Biology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221009, Jiangsu, China.
| |
Collapse
|
8
|
Pleiotropic Roles of Calmodulin in the Regulation of KRas and Rac1 GTPases: Functional Diversity in Health and Disease. Int J Mol Sci 2020; 21:ijms21103680. [PMID: 32456244 PMCID: PMC7279331 DOI: 10.3390/ijms21103680] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022] Open
Abstract
Calmodulin is a ubiquitous signalling protein that controls many biological processes due to its capacity to interact and/or regulate a large number of cellular proteins and pathways, mostly in a Ca2+-dependent manner. This complex interactome of calmodulin can have pleiotropic molecular consequences, which over the years has made it often difficult to clearly define the contribution of calmodulin in the signal output of specific pathways and overall biological response. Most relevant for this review, the ability of calmodulin to influence the spatiotemporal signalling of several small GTPases, in particular KRas and Rac1, can modulate fundamental biological outcomes such as proliferation and migration. First, direct interaction of calmodulin with these GTPases can alter their subcellular localization and activation state, induce post-translational modifications as well as their ability to interact with effectors. Second, through interaction with a set of calmodulin binding proteins (CaMBPs), calmodulin can control the capacity of several guanine nucleotide exchange factors (GEFs) to promote the switch of inactive KRas and Rac1 to an active conformation. Moreover, Rac1 is also an effector of KRas and both proteins are interconnected as highlighted by the requirement for Rac1 activation in KRas-driven tumourigenesis. In this review, we attempt to summarize the multiple layers how calmodulin can regulate KRas and Rac1 GTPases in a variety of cellular events, with biological consequences and potential for therapeutic opportunities in disease settings, such as cancer.
Collapse
|
9
|
Shafiq A, Campbell LJ, Owen D, Mott HR. NMR resonance assignments for the active and inactive conformations of the small G protein RalA. BIOMOLECULAR NMR ASSIGNMENTS 2020; 14:87-91. [PMID: 31916136 PMCID: PMC7069931 DOI: 10.1007/s12104-019-09925-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/26/2019] [Indexed: 06/10/2023]
Abstract
The Ral proteins (RalA and RalB) are small G proteins of the Ras family that have been implicated in exocytosis, endocytosis, transcriptional regulation and mitochondrial fission, as well as having a role in tumourigenesis. RalA and RalB are activated downstream of the master regulator, Ras, which causes the nucleotide exchange of GDP for GTP. Here we report the 1H, 15 N and 13C resonance assignments of RalA in its active form bound to the GTP analogue GMPPNP. We also report the backbone assignments of RalA in its inactive, GDP-bound form. The assignments give insight into the switch regions, which change conformation upon nucleotide exchange. These switch regions are invisible in the spectra of the active, GMPPNP bound form but the residues proximal to the switches can be monitored. RalA is also an important drug target due to its over activation in some cancers and these assignments will be extremely useful for NMR-based screening approaches.
Collapse
Affiliation(s)
- Arooj Shafiq
- Department of Biochemistry, 80, Tennis Court Road, Cambridge, CB2 1GA, UK
- Barrett Hodgson University, Korangi Creek, Salim Habib Campus, NC-24, Deh Dih, Korangi Creek, Karachi, 74900, Sindh, Pakistan
| | - Louise J Campbell
- Department of Biochemistry, 80, Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Darerca Owen
- Department of Biochemistry, 80, Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Helen R Mott
- Department of Biochemistry, 80, Tennis Court Road, Cambridge, CB2 1GA, UK.
| |
Collapse
|
10
|
Abstract
In this issue of Cell Stem Cell, Johansson et al. (2019) find evolutionarily conserved regulation of Wnt signaling through Ral GTPases. These GTPases promote internalization of Wnt receptor complexes and play a critical role in intestinal stem cell function in flies and mice.
Collapse
Affiliation(s)
- Helen Tauc
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Heinrich Jasper
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
11
|
Maurin J, Morel A, Hassen-Khodja C, Vives V, Jurdic P, Machuca-Gayet I, Blangy A. Combined strategy of siRNA and osteoclast actin cytoskeleton automated imaging to identify novel regulators of bone resorption shows a non-mitotic function for anillin. Eur J Cell Biol 2018; 97:568-579. [DOI: 10.1016/j.ejcb.2018.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 09/17/2018] [Accepted: 10/17/2018] [Indexed: 11/30/2022] Open
|
12
|
Abstract
Our understanding of fundamental biological processes within platelets is continually evolving. A critical feature of platelet biology relates to the intricate uptake, packaging and release of bioactive cargo from storage vesicles, essential in mediating a range of classical (haemostasis/thrombosis) and non-classical (regeneration/inflammation/metastasis) roles platelets assume. Pivotal to the molecular control of these vesicle trafficking events are the small GTPases of the Ras superfamily, which function as spatially distinct, molecular switches controlling essential cellular processes. Herein, we specifically focus on members of the Rab, Arf and Ras subfamilies, which comprise over 130 members and platelet proteomic datasets suggest that more than half of these are expressed in human platelets. We provide an update of current literature relating to trafficking roles for these GTPases in platelets, particularly regarding endocytic and exocytic events, but also vesicle biogenesis and provide speculative argument for roles that other related GTPases and regulatory proteins may adopt in platelets. Advances in our understanding of small GTPase function in the anucleate platelet has been hampered by the lack of specific molecular tools, but it is anticipated that this will be greatly accelerated in the years ahead and will be crucial to the identification of novel therapeutic targets controlling different platelet processes.
Collapse
Affiliation(s)
- Tony G Walsh
- a From the School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building , University of Bristol , Bristol , UK
| | - Yong Li
- a From the School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building , University of Bristol , Bristol , UK
| | - Andreas Wersäll
- a From the School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building , University of Bristol , Bristol , UK
| | - Alastair W Poole
- a From the School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building , University of Bristol , Bristol , UK
| |
Collapse
|
13
|
Oo HZ, Seiler R, Black PC, Daugaard M. Post-translational modifications in bladder cancer: Expanding the tumor target repertoire. Urol Oncol 2018; 38:858-866. [PMID: 30342880 DOI: 10.1016/j.urolonc.2018.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 07/09/2018] [Accepted: 09/03/2018] [Indexed: 12/17/2022]
Abstract
Over the past decade, genomic and transcriptomic analyses have uncovered promising tumor antigens including immunotherapeutic targets in bladder cancer (BCa). Conventional tumor antigens are proteins expressed on the plasma membrane of tumor cells such as EGFR, FGFR3, and ERBB2 in BCa, which can be targeted by antibodies or similar epitope-specific binding reagents. The cellular proteome consists of ∼100,000 proteins but the expression of these proteins is rarely unique to tumor cells. Many tumor-associated proteins are post-translationally modified with phosphorylation, glycosylation, ubiquitination, or SUMOylation moieties. Although these modifications expand the complexity, they potentially offer novel targeting opportunities across tumor sub-populations. Experimental targeting of cancer-specific post-translational modifications (PTMs) has shown encouraging results in pre-clinical models of BCa, which could potentially overcome issues with inherent intra-tumor heterogeneity due to simultaneous expression on different proteins. Here, we review current knowledge on post-translational modifications in BCa and highlight recent efforts in experimental targeting strategies.
Collapse
Affiliation(s)
- Htoo Zarni Oo
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada; Vancouver Prostate Centre, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| | - Roland Seiler
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada; Vancouver Prostate Centre, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; Department of Urology, University of Bern, Bern, Switzerland
| | - Peter C Black
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada; Vancouver Prostate Centre, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| | - Mads Daugaard
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada; Vancouver Prostate Centre, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| |
Collapse
|
14
|
Moghadam AR, Patrad E, Tafsiri E, Peng W, Fangman B, Pluard TJ, Accurso A, Salacz M, Shah K, Ricke B, Bi D, Kimura K, Graves L, Najad MK, Dolatkhah R, Sanaat Z, Yazdi M, Tavakolinia N, Mazani M, Amani M, Ghavami S, Gartell R, Reilly C, Naima Z, Esfandyari T, Farassati F. Ral signaling pathway in health and cancer. Cancer Med 2017; 6:2998-3013. [PMID: 29047224 PMCID: PMC5727330 DOI: 10.1002/cam4.1105] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 04/10/2017] [Accepted: 04/14/2017] [Indexed: 12/12/2022] Open
Abstract
The Ral (Ras-Like) signaling pathway plays an important role in the biology of cells. A plethora of effects is regulated by this signaling pathway and its prooncogenic effectors. Our team has demonstrated the overactivation of the RalA signaling pathway in a number of human malignancies including cancers of the liver, ovary, lung, brain, and malignant peripheral nerve sheath tumors. Additionally, we have shown that the activation of RalA in cancer stem cells is higher in comparison with differentiated cancer cells. In this article, we review the role of Ral signaling in health and disease with a focus on the role of this multifunctional protein in the generation of therapies for cancer. An improved understanding of this pathway can lead to development of a novel class of anticancer therapies that functions on the basis of intervention with RalA or its downstream effectors.
Collapse
Affiliation(s)
- Adel Rezaei Moghadam
- Department of Human Anatomy and Cell ScienceUniversity of ManitobaWinnipegCanada
| | - Elham Patrad
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Elham Tafsiri
- Department of Pediatrics, Columbia Presbyterian Medical CenterNew YorkNew York
| | - Warner Peng
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Benjamin Fangman
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Timothy J Pluard
- Saint Luke's HospitalUniversity of Missouri at Kansas CityKansas CityMissouri
| | - Anthony Accurso
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Michael Salacz
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Kushal Shah
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Brandon Ricke
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Danse Bi
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Kyle Kimura
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Leland Graves
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Marzieh Khajoie Najad
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Roya Dolatkhah
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Zohreh Sanaat
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Mina Yazdi
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Naeimeh Tavakolinia
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Mohammad Mazani
- Pasteur Institute of IranTehranIran
- Ardabil University of Medical Sciences, BiochemistryArdabilIran
| | - Mojtaba Amani
- Pasteur Institute of IranTehranIran
- Ardabil University of Medical Sciences, BiochemistryArdabilIran
| | - Saeid Ghavami
- Department of Human Anatomy and Cell ScienceUniversity of ManitobaWinnipegCanada
| | - Robyn Gartell
- Department of Pediatrics, Columbia Presbyterian Medical CenterNew YorkNew York
| | - Colleen Reilly
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Zaid Naima
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Tuba Esfandyari
- Department of Medicine, Molecular Medicine LaboratoryThe University of Kansas Medical SchoolKansas CityKansas
| | - Faris Farassati
- Research Service (151)Kansas City Veteran Affairs Medical Center & Midwest Biomedical Research Foundation4801 E Linwood BlvdKansas CityMissouri64128‐2226
| |
Collapse
|
15
|
Christodoulou EG, Yang H, Lademann F, Pilarsky C, Beyer A, Schroeder M. Detection of COPB2 as a KRAS synthetic lethal partner through integration of functional genomics screens. Oncotarget 2017; 8:34283-34297. [PMID: 28415695 PMCID: PMC5470967 DOI: 10.18632/oncotarget.16079] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 03/01/2017] [Indexed: 12/30/2022] Open
Abstract
Mutated KRAS plays an important role in many cancers. Although targeting KRAS directly is difficult, indirect inactivation via synthetic lethal partners (SLPs) is promising. Yet to date, there are no SLPs from high-throughput RNAi screening, which are supported by multiple screens. Here, we address this problem by aggregating and ranking data over three independent high-throughput screens. We integrate rankings by minimizing the displacement and by considering established methods such as RIGER and RSA.Our meta analysis reveals COPB2 as a potential SLP of KRAS with good support from all three screens. COPB2 is a coatomer subunit and its knock down has already been linked to disabled autophagy and reduced tumor growth. We confirm COPB2 as SLP in knock down experiments on pancreas and colorectal cancer cell lines.Overall, consistent integration of high throughput data can generate candidate synthetic lethal partners, which individual screens do not uncover. Concretely, we reveal and confirm that COPB2 is a synthetic lethal partner of KRAS and hence a promising cancer target. Ligands inhibiting COPB2 may, therefore, be promising new cancer drugs.
Collapse
Affiliation(s)
- Eleni G. Christodoulou
- Biotechnology Center, TU Dresden, Dresden, Germany
- Department of Medical Oncology, National Cancer Center of Singapore, Singapore
| | - Hai Yang
- Chirurgische Klinik, Translational Research Center, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Christian Pilarsky
- Chirurgische Klinik, Translational Research Center, Universitätsklinikum Erlangen, Erlangen, Germany
- Medizinische Fakultät Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Andreas Beyer
- Biotechnology Center, TU Dresden, Dresden, Germany
- Cellular Networks and Systems Biology, University of Cologne, Cologne, Germany
| | | |
Collapse
|
16
|
Zhang X, Kim KM. Multifactorial Regulation of G Protein-Coupled Receptor Endocytosis. Biomol Ther (Seoul) 2017; 25:26-43. [PMID: 28035080 PMCID: PMC5207461 DOI: 10.4062/biomolther.2016.186] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/21/2016] [Accepted: 11/30/2016] [Indexed: 12/26/2022] Open
Abstract
Endocytosis is a process by which cells absorb extracellular materials via the inward budding of vesicles formed from the plasma membrane. Receptor-mediated endocytosis is a highly selective process where receptors with specific binding sites for extracellular molecules internalize via vesicles. G protein-coupled receptors (GPCRs) are the largest single family of plasma-membrane receptors with more than 1000 family members. But the molecular mechanisms involved in the regulation of GPCRs are believed to be highly conserved. For example, receptor phosphorylation in collaboration with β-arrestins plays major roles in desensitization and endocytosis of most GPCRs. Nevertheless, a number of subsequent studies showed that GPCR regulation, such as that by endocytosis, occurs through various pathways with a multitude of cellular components and processes. This review focused on i) functional interactions between homologous and heterologous pathways, ii) methodologies applied for determining receptor endocytosis, iii) experimental tools to determine specific endocytic routes, iv) roles of small guanosine triphosphate-binding proteins in GPCR endocytosis, and v) role of post-translational modification of the receptors in endocytosis.
Collapse
Affiliation(s)
- Xiaohan Zhang
- Pharmacology Laboratory, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kyeong-Man Kim
- Pharmacology Laboratory, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
17
|
Zheng M, Zhang X, Sun N, Min C, Zhang X, Kim KM. RalA employs GRK2 and β-arrestins for the filamin A-mediated regulation of trafficking and signaling of dopamine D2 and D3 receptor. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2072-83. [PMID: 27188791 DOI: 10.1016/j.bbamcr.2016.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/10/2016] [Accepted: 05/13/2016] [Indexed: 12/13/2022]
Abstract
Filamin A (FLNA) is known to act as platform for the signaling and intracellular trafficking of various GPCRs including dopamine D2 and D3 receptors (D2R, D3R). To understand molecular mechanisms involved in the FLNA-mediated regulation of D2R and D3R, comparative studies were conducted on the signaling and intracellular trafficking of the D2R and D3R in FLNA-knockdown cells, with a specific focus on the roles of the proteins that interact with FLNA and the D2R and D3R. Lowering the level of cellular FLNA caused an elevation in RalA activity and resulted in selective interference with the normal intracellular trafficking and signaling of the D2R and D3R, through GRK2 and β-arrestins, respectively. Knockdown of FLNA or coexpression of active RalA interfered with the recycling of the internalized D2R and resulted in the development of receptor tolerance. Active RalA was found to interact with GRK2 to sequester it from D2R. Knockdown of FLNA or coexpression of active RalA prevented D3R from coupling with G protein. The selective involvement of GRK2- and β-arrestins in the RalA-mediated cellular processes of the D2R and D3R was achieved via their different modes of interactions with the receptor and their distinct functional roles in receptor regulation. Our results show that FLNA is a multi-functional protein that acts as a platform on which D2R and D3R can interact with various proteins, through which selective regulation of these receptors occurs in combination with GRK2 and β-arrestins.
Collapse
Affiliation(s)
- Mei Zheng
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Xiaohan Zhang
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - NingNing Sun
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Chengchun Min
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Xiaowei Zhang
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju 500-757, Republic of Korea.
| |
Collapse
|
18
|
Tracy K, Velentzas PD, Baehrecke EH. Ral GTPase and the exocyst regulate autophagy in a tissue-specific manner. EMBO Rep 2015; 17:110-21. [PMID: 26598552 DOI: 10.15252/embr.201541283] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 10/22/2015] [Indexed: 11/09/2022] Open
Abstract
Autophagy traffics cellular components to the lysosome for degradation. Ral GTPase and the exocyst have been implicated in the regulation of stress-induced autophagy, but it is unclear whether they are global regulators of this process. Here, we investigate Ral function in different cellular contexts in Drosophila and find that it is required for autophagy during developmentally regulated cell death in salivary glands, but does not affect starvation-induced autophagy in the fat body. Furthermore, knockdown of exocyst subunits has a similar effect, preventing autophagy in dying cells but not in cells of starved animals. Notch activity is elevated in dying salivary glands, this change in Notch signaling is influenced by Ral, and decreased Notch function influences autophagy. These data indicate that Ral and the exocyst regulate autophagy in a context-dependent manner, and that in dying salivary glands, Ral mediates autophagy, at least in part, by regulation of Notch.
Collapse
Affiliation(s)
- Kirsten Tracy
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Panagiotis D Velentzas
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
19
|
Zheng M, Zhang X, Guo S, Zhang X, Min C, Cheon SH, Oak MH, Kim YR, Kim KM. Agonist-induced changes in RalA activities allows the prediction of the endocytosis of G protein-coupled receptors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:77-90. [PMID: 26477566 DOI: 10.1016/j.bbamcr.2015.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 10/10/2015] [Accepted: 10/13/2015] [Indexed: 12/31/2022]
Abstract
GTP binding proteins are classified into two families: heterotrimeric large G proteins which are composed of three subunits, and one subunit of small G proteins. Roles of small G proteins in the intracellular trafficking of G protein-coupled receptors (GPCRs) were studied. Among various small G proteins tested, GTP-bound form (G23V) of RalA inhibited the internalization of dopamine D2 receptor independently of the previously reported downstream effectors of RalA, such as Ral-binding protein 1 and PLD. With high affinity for GRK2, active RalA inhibited the GPCR endocytosis by sequestering the GRK2 from receptors. When it was tested for several GPCRs including an endogenous GPCR, lysophosphatidic acid receptor 1, agonist-induced conversion of GTP-bound to GDP-bound RalA, which presumably releases the sequestered GRK2, was observed selectively with the GPCRs which have tendency to undergo endocytosis. Conversion of RalA from active to inactive state occurred by translocation of RGL, a guanine nucleotide exchange factor, from the plasma membrane to cytosol as a complex with Gβγ. These results suggest that agonist-induced Gβγ-mediated conversion of RalA from the GTP-bound form to the GDP-bound form could be a mechanism to facilitate agonist-induced internalization of GPCRs.
Collapse
Affiliation(s)
- Mei Zheng
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Xiaohan Zhang
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Shuohan Guo
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Xiaowei Zhang
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Chengchun Min
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Seung Hoon Cheon
- Department of Medicinal Chemistry, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Min-Ho Oak
- College of Pharmacy, Mokpo National University, Muan-gun, Jeollanamdo 534-729, Republic of Korea
| | - Young Ran Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea.
| |
Collapse
|
20
|
Papini D, Langemeyer L, Abad MA, Kerr A, Samejima I, Eyers PA, Jeyaprakash AA, Higgins JMG, Barr FA, Earnshaw WC. TD-60 links RalA GTPase function to the CPC in mitosis. Nat Commun 2015; 6:7678. [PMID: 26158537 PMCID: PMC4510650 DOI: 10.1038/ncomms8678] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 05/29/2015] [Indexed: 12/26/2022] Open
Abstract
TD-60 (also known as RCC2) is a highly conserved protein that structurally resembles the Ran guanine exchange factor (GEF) RCC1, but has not previously been shown to have GEF activity. TD-60 has a typical chromosomal passenger complex (CPC) distribution in mitotic cells, but associates with integrin complexes and is involved in cell motility during interphase. Here we show that TD-60 exhibits GEF activity, in vitro and in cells, for the small GTPase RalA. TD-60 or RalA depletion causes spindle abnormalities in prometaphase associated with abnormal centromeric accumulation of CPC components. TD-60 and RalA apparently work together to contribute to the regulation of kinetochore-microtubule interactions in early mitosis. Importantly, several mitotic phenotypes caused by TD-60 depletion are reverted by the expression of a GTP-locked mutant, RalA (Q72L). The demonstration that a small GTPase participates in the regulation of the CPC reveals a level of mitotic regulation not suspected in previous studies.
Collapse
Affiliation(s)
- Diana Papini
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
- Institute for Cell and Molecular Biosciences (ICaMB), Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Lars Langemeyer
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Maria A. Abad
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Alastair Kerr
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Itaru Samejima
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Patrick A. Eyers
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, UK
| | - A. Arockia Jeyaprakash
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Jonathan M. G. Higgins
- Institute for Cell and Molecular Biosciences (ICaMB), Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Francis A. Barr
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - William C. Earnshaw
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| |
Collapse
|
21
|
Sisakhtnezhad S, Khosravi L. Emerging physiological and pathological implications of tunneling nanotubes formation between cells. Eur J Cell Biol 2015; 94:429-43. [PMID: 26164368 DOI: 10.1016/j.ejcb.2015.06.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 06/20/2015] [Accepted: 06/23/2015] [Indexed: 12/21/2022] Open
Abstract
Cell-to-cell communication is a critical requirement to coordinate behaviors of the cells in a community and thereby achieve tissue homeostasis and conservation of the multicellular organisms. Tunneling nanotubes (TNTs), as a cell-to-cell communication over long distance, allow for bi- or uni-directional transfer of cellular components between cells. Identification of inducing agents and the cell and molecular mechanism underling the formation of TNTs and their structural and functional features may lead to finding new important roles for these intercellular bridges in vivo and in vitro. During the last decade, research has shown TNTs have different structural and functional properties, varying between and within cell systems. In this review, we will focus on TNTs and their cell and molecular mechanism of formation. Moreover, the latest findings into their functional roles in physiological and pathological processes, such as signal transduction, micro and nano-particles delivery, immune responses, embryogenesis, cellular reprogramming, apoptosis, cancer, and neurodegenerative diseases initiation and progression and pathogens transfer, will be discussed.
Collapse
Affiliation(s)
| | - Leila Khosravi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| |
Collapse
|
22
|
Armenti ST, Chan E, Nance J. Polarized exocyst-mediated vesicle fusion directs intracellular lumenogenesis within the C. elegans excretory cell. Dev Biol 2014; 394:110-21. [PMID: 25102190 DOI: 10.1016/j.ydbio.2014.07.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 07/08/2014] [Accepted: 07/26/2014] [Indexed: 12/18/2022]
Abstract
Lumenogenesis of small seamless tubes occurs through intracellular membrane growth and directed vesicle fusion events. Within the Caenorhabditis elegans excretory cell, which forms seamless intracellular tubes (canals) that mediate osmoregulation, lumens grow in length and diameter when vesicles fuse with the expanding lumenal surface. Here, we show that lumenal vesicle fusion depends on the small GTPase RAL-1, which localizes to vesicles and acts through the exocyst vesicle-tethering complex. Loss of either the exocyst or RAL-1 prevents excretory canal lumen extension. Within the excretory canal and other polarized cells, the exocyst co-localizes with the PAR polarity proteins PAR-3, PAR-6 and PKC-3. Using early embryonic cells to determine the functional relationships between the exocyst and PAR proteins, we show that RAL-1 recruits the exocyst to the membrane, while PAR proteins concentrate membrane-localized exocyst proteins to a polarized domain. These findings reveal that RAL-1 and the exocyst direct the polarized vesicle fusion events required for intracellular lumenogenesis of the excretory cell, suggesting mechanistic similarities in the formation of topologically distinct multicellular and intracellular lumens.
Collapse
Affiliation(s)
- Stephen T Armenti
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA; Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Emily Chan
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA; Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Jeremy Nance
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA; Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
23
|
Personnic N, Lakisic G, Gouin E, Rousseau A, Gautreau A, Cossart P, Bierne H. A role for Ral GTPase-activating protein subunit β in mitotic regulation. FEBS J 2014; 281:2977-89. [PMID: 24814574 DOI: 10.1111/febs.12836] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 04/30/2014] [Accepted: 05/02/2014] [Indexed: 01/05/2023]
Abstract
Ral proteins are small GTPases that play critical roles in normal physiology and in oncogenesis. There is little information on the GTPase-activating proteins (GAPs) that downregulate their activity. Here, we provide evidence that the noncatalytic β subunit of RalGAPα1/2 β complexes is involved in mitotic control. RalGAPβ localizes to the Golgi and nucleus during interphase, and relocalizes to the mitotic spindle and cytokinetic intercellular bridge during mitosis. Depletion of RalGAPβ causes chromosome misalignment and decreases the amount of mitotic cyclin B1, disturbing the metaphase-to-anaphase transition. Overexpression of RalGAPβ interferes with cell division, leading to binucleation and multinucleation, and cell death. We propose that RalGAPβ plays an essential role in the sequential progression of mitosis by controlling the spatial and temporal activation of Ral GTPases in the spindle assembly checkpoint (SAC) and cytokinesis. Deregulation of RalGAPβ might cause genomic instability, leading to human carcinogenesis.
Collapse
Affiliation(s)
- Nicolas Personnic
- Institut Pasteur, Unité des interactions Bactéries cellules, Paris, France; Inserm, U604, Paris, France; INRA, USC2020, Paris, France
| | | | | | | | | | | | | |
Collapse
|
24
|
Sim SE, Lee HR, Kim JI, Choi SL, Bakes J, Jang DJ, Lee K, Han K, Kim E, Kaang BK. Elevated RalA activity in the hippocampus of PI3Kγ knock-out mice lacking NMDAR-dependent long-term depression. BMB Rep 2013; 46:103-6. [PMID: 23433113 PMCID: PMC4133848 DOI: 10.5483/bmbrep.2013.46.2.143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Phosphoinositide 3-kinases (PI3Ks) play key roles in synaptic plasticity and cognitive functions in the brain. We recently found that genetic deletion of PI3Kγ, the only known member of class IB PI3Ks, results in impaired N-methyl-D-aspartate receptor-dependent long-term depression (NMDAR-LTD) in the hippocampus. The activity of RalA, a small GTP-binding protein, increases following NMDAR-LTD inducing stimuli, and this increase in RalA activity is essential for inducing NMDAR-LTD. We found that RalA activity increased significantly in PI3Kγ knockout mice. Furthermore, NMDAR-LTD-inducing stimuli did not increase RalA activity in PI3Kγ knockout mice. These results suggest that constitutively increased RalA activity occludes further increases in RalA activity during induction of LTD, causing impaired NMDAR-LTD. We propose that PI3Kγ regulates the activity of RalA, which is one of the molecular mechanisms inducing NMDAR dependent LTD.
Collapse
Affiliation(s)
- Su-Eon Sim
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 151-747, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Pathak R, Dermardirossian C. GEF-H1: orchestrating the interplay between cytoskeleton and vesicle trafficking. Small GTPases 2013; 4:174-9. [PMID: 23648943 DOI: 10.4161/sgtp.24616] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Vesicle trafficking is crucial for delivery of membrane compartments as well as signaling molecules to specific sites on the plasma membrane for regulation of diverse processes such as cell division, migration, polarity establishment and secretion. Rho GTPases are well-studied signaling molecules that regulate actin cytoskeleton in response to variety of extracellular stimuli. Increasing amounts of evidence suggest that Rho proteins play a critical role in vesicle trafficking in both the exocytic and endocytic pathways; however, the molecular mechanism underlying the process remains largely unclear. We recently defined a mechanism of action for RhoA in membrane trafficking pathways through regulation of the octameric complex exocyst in a manuscript published in Developmental Cell. We have shown that microtubule-associated RhoA-activating factor GEF-H1 is involved in endocytic and excocytic vesicle trafficking. GEF-H1 activates RhoA in response to RalA GTPase, which in turn regulates the localization and the assembly of exocyst components and exocytosis. Our work defines a mechanism for RhoA activation in response to RalA signaling and during vesicle trafficking. These results provide a framework for understanding how RhoA/GEF-H1 regulates the coordination of actin and microtubule cytoskeleton modulation and vesicle trafficking during migration and cell division.
Collapse
Affiliation(s)
- Ritu Pathak
- Departments of Immunology and Microbial Science; The Scripps Research Institute; La Jolla, CA USA
| | - Celine Dermardirossian
- Departments of Immunology and Microbial Science; The Scripps Research Institute; La Jolla, CA USA
| |
Collapse
|
26
|
Tazat K, Harsat M, Goldshmid-Shagal A, Ehrlich M, Henis YI. Dual effects of Ral-activated pathways on p27 localization and TGF-β signaling. Mol Biol Cell 2013; 24:1812-24. [PMID: 23576547 PMCID: PMC3667732 DOI: 10.1091/mbc.e13-01-0007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Constitutive activation or overactivation of Ras signaling pathways contributes to epithelial tumorigenesis in several ways, one of which is cytoplasmic mislocalization of the cyclin-dependent kinase inhibitor p27(Kip1) (p27). We previously showed that such an effect can be mediated by activation of the Ral-GEF pathway by oncogenic N-Ras. However, the mechanism(s) leading to p27 cytoplasmic accumulation downstream of activated Ral remained unknown. Here, we report a dual regulation of p27 cellular localization by Ral downstream pathways, based on opposing effects via the Ral effectors RalBP1 and phospholipase D1 (PLD1). Because RalA and RalB are equally effective in mislocalizing both murine and human p27, we focus on RalA and murine p27, which lacks the Thr-157 phosphorylation site of human p27. In experiments based on specific RalA and p27 mutants, complemented with short hairpin RNA-mediated knockdown of Ral downstream signaling components, we show that activation of RalBP1 induces cytoplasmic accumulation of p27 and that this event requires p27 Ser-10 phosphorylation by protein kinase B/Akt. Of note, activation of PLD1 counteracts this effect in a Ser-10-independent manner. The physiological relevance of the modulation of p27 localization by Ral is demonstrated by the ability of Ral-mediated activation of the RalBP1 pathway to abrogate transforming growth factor-β-mediated growth arrest in epithelial cells.
Collapse
Affiliation(s)
- Keren Tazat
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | | | |
Collapse
|
27
|
Abstract
The process of phagocytosis in multicellular organisms is required for homeostasis, clearance of foreign particles, and establishment of long-term immunity, yet the molecular determinants of uptake are not well characterized. Cdc42, a Rho guanosine triphosphatase, is thought to orchestrate critical actin remodeling events needed for internalization. In this paper, we show that Cdc42 controls exocytic events during phagosome formation. Cdc42 inactivation led to a selective defect in large particle phagocytosis as well as a general decrease in the rate of membrane flow to the cell surface. Supporting the connection between Cdc42 and exocytic function, we found that the overproduction of a regulator of exocytosis, Rab11, rescued the large particle uptake defect in the absence of Cdc42. Additionally, we demonstrated a temporal interaction between Cdc42 and the exocyst complex during large particle uptake. Furthermore, disruption of exocyst function through Exo70 depletion led to a defect in large particle internalization, thereby establishing a functional role for the exocyst complex during phagocytosis.
Collapse
Affiliation(s)
- Sina Mohammadi
- Howard Hughes Medical Institute, Tufts University School of Medicine, Boston, MA 02111, USA
| | | |
Collapse
|
28
|
Leto D, Uhm M, Williams A, Chen XW, Saltiel AR. Negative regulation of the RalGAP complex by 14-3-3. J Biol Chem 2013; 288:9272-83. [PMID: 23386617 DOI: 10.1074/jbc.m112.426106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
RGC1 and RGC2 comprise a functional RalGAP complex (RGC) that suppresses RalA activity. The PI3-kinase/Akt signaling pathway activates RalA through phosphorylation-mediated inhibition of the RGC. Here we identify a novel phosphorylation-dependent interaction between 14-3-3 and the RGC. 14-3-3 binds to the complex through an Akt-phosphorylated residue, threonine 715, on RGC2. Interaction with 14-3-3 does not alter in vitro activity of the GTPase-activating protein complex. However, blocking the interaction between 14-3-3 and RGC2 in cells increases suppression of RalA activity by the RGC, suggesting that 14-3-3 inhibits the complex through a non-catalytic mechanism. Together, these data show that 14-3-3 negatively regulates the RGC downstream of the PI3-kinase/Akt signaling pathway.
Collapse
Affiliation(s)
- Dara Leto
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | |
Collapse
|
29
|
Brymora A, Duggin IG, Berven LA, van Dam EM, Roufogalis BD, Robinson PJ. Identification and characterisation of the RalA-ERp57 interaction: evidence for GDI activity of ERp57. PLoS One 2012; 7:e50879. [PMID: 23226417 PMCID: PMC3511393 DOI: 10.1371/journal.pone.0050879] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 10/25/2012] [Indexed: 01/03/2023] Open
Abstract
RalA is a membrane-associated small GTPase that regulates vesicle trafficking. Here we identify a specific interaction between RalA and ERp57, an oxidoreductase and signalling protein. ERp57 bound specifically to the GDP-bound form of RalA, but not the GTP-bound form, and inhibited the dissociation of GDP from RalA in vitro. These activities were inhibited by reducing agents, but no disulphide bonds were detected between RalA and ERp57. Mutation of all four of ERp57’s active site cysteine residues blocked sensitivity to reducing agents, suggesting that redox-dependent conformational changes in ERp57 affect binding to RalA. Mutations in the switch II region of the GTPase domain of RalA specifically reduced or abolished binding to ERp57, but did not block GTP-specific binding to known RalA effectors, the exocyst and RalBP1. Oxidative treatment of A431 cells with H2O2 inhibited cellular RalA activity, and the effect was exacerbated by expression of recombinant ERp57. The oxidative treatment significantly increased the amount of RalA localised to the cytosol. These findings suggest that ERp57 regulates RalA signalling by acting as a redox-sensitive guanine-nucleotide dissociation inhibitor (RalGDI).
Collapse
Affiliation(s)
- Adam Brymora
- Cell Signalling Unit, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Iain G. Duggin
- Faculty of Pharmacy, The University of Sydney, Sydney, Australia
| | - Leise A. Berven
- Cell Signalling Unit, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Ellen M. van Dam
- Cell Signalling Unit, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | | | - Phillip J. Robinson
- Cell Signalling Unit, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
- * E-mail:
| |
Collapse
|
30
|
Rajasekar KV, Campbell LJ, Nietlispach D, Owen D, Mott HR. 1H, 13C and 15N resonance assignments of the GTPase-activating (GAP) and Ral binding domains (GBD) of RLIP76 (RalBP1). BIOMOLECULAR NMR ASSIGNMENTS 2012; 6:119-122. [PMID: 21915608 DOI: 10.1007/s12104-011-9337-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 08/25/2011] [Indexed: 05/31/2023]
Abstract
RLIP76 (also known as RalBP1) is an effector for Ral small G proteins. RLIP76 is a multifunctional, multi-domain protein that includes a GTPase activating domain for the Rho family (RhoGAP domain) and a GTPase binding domain (GBD) for the Ral small G proteins. The juxtaposition of these two domains (GAP and GBD) may be a strategy employed to co-ordinate regulation of Rho family and Ral-controlled signalling pathways at a crossover node. Here we present the (1)H, (15)N and (13)C NMR backbone and sidechain resonance assignments of the GAP and GBD di-domain (31 kDa).
Collapse
Affiliation(s)
- Karthik V Rajasekar
- Department of Biochemistry, University of Cambridge, 80, Tennis Court Road, Cambridge CB2 1GA, UK
| | | | | | | | | |
Collapse
|
31
|
Lim YS, Tang BL. Intercellular organelle trafficking by membranous nanotube connections: a possible new role in cellular rejuvenation? ACTA ACUST UNITED AC 2012; 19:39-44. [DOI: 10.3109/15419061.2012.712574] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
32
|
Cho B, Fischer JA. Ral inhibits ligand-independent Notch signaling in Drosophila. Small GTPases 2012; 3:186-91. [PMID: 22750761 DOI: 10.4161/sgtp.19802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We discovered recently that the Drosophila Ral GTPase regulates Notch signaling and thereby affects cell patterning in the eye. Although Ral functions in the ligand signaling cells, Ral does not stimulate ligand signaling directly. Rather, in cells that express both Notch receptor and ligand, Ral activity promotes a cell to become the signaler by inhibiting Notch receptor activation in that cell. Moreover, Ral inhibits a particular pathway of Notch activation-receptor activation that occurs independent of ligand binding. In this Commentary, we discuss the phenomenon of ligand-independent Notch receptor activation and how this event might be regulated by Ral.
Collapse
Affiliation(s)
- Bomsoo Cho
- Section of Molecular Cell and Developmental Biology, Institute for Cell and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
| | | |
Collapse
|
33
|
Hazelett CC, Yeaman C. Sec5 and Exo84 mediate distinct aspects of RalA-dependent cell polarization. PLoS One 2012; 7:e39602. [PMID: 22761837 PMCID: PMC3382198 DOI: 10.1371/journal.pone.0039602] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 05/25/2012] [Indexed: 01/06/2023] Open
Abstract
Metastasis is a complex process during which several gross cellular changes occur. Cells must dissociate from the tumor mass and gain the ability to degrade extracellular matrix and migrate in order to ultimately attach and form a satellite tumor. Regulation of the actin cytoskeleton is an indispensible aspect of cell migration, and many different factors have been implicated in this process. We identified interactions between RalA and its effectors in the Exocyst complex as directly necessary for migration and invasion of prostate cancer tumor cells. Blocking RalA-Exocyst binding caused significant morphological changes and defects in single and coordinated cell migration.
Collapse
Affiliation(s)
- C Clayton Hazelett
- Department of Anatomy and Cell Biology Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | | |
Collapse
|
34
|
Mollberg NM, Steinert G, Aigner M, Hamm A, Lin FJ, Elbers H, Reissfelder C, Weitz J, Buchler MW, Koch M. Overexpression of RalBP1 in colorectal cancer is an independent predictor of poor survival and early tumor relapse. Cancer Biol Ther 2012; 13:694-700. [PMID: 22549157 DOI: 10.4161/cbt.20087] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The non-ABC transport protein RalBP1 has been shown to be overexpressed in various cancer cell lines and implicated in the process of metastasis formation, but its expression in tissue samples and prognostic significance has not been shown. In this study matched tumor-mucosa tissue samples from 78 CRC patients were investigated. The RalBP1 mRNA and protein levels were quantified by real-time quantitative PCR (qPCR) and ELISA. RalBP1 was found to be overexpressed in tumor at the mRNA level both overall (p = 0.027), and for stages I (p = 0.024), II (p = 0.038) and IV (p = 0.004). At the protein level, RalBP1 was only significantly overexpressed in stage IV patients (p = 0.018). Expression of RalBP1 mRNA and protein were inversely correlated (r = 0.4173; p = 0.0004). Multivariate Cox regression analysis including sex, age, stage, grade, and nodal status as covariates showed that overexpression of RalBP1 protein, but not mRNA, was an independent predictor of both decreased disease free survival (p = 0.016, RR = 6.892) and overall survival (p = 0.039, RR = 5.986). These results suggest that RalBP1 protein is an independent predictor of poor survival and early relapse for CRC patients. Owing to its multifunctional intermediary role in cell survival, chemotherapeutic resistance, and metastasis formation, RalBP1 represents a promising novel therapeutic target.
Collapse
Affiliation(s)
- Nathan M Mollberg
- Department of Surgery, University of Illinois at Mount Sinai Hospital, Chicago, IL USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abounit S, Zurzolo C. Wiring through tunneling nanotubes--from electrical signals to organelle transfer. J Cell Sci 2012; 125:1089-98. [PMID: 22399801 DOI: 10.1242/jcs.083279] [Citation(s) in RCA: 254] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tunneling nanotubes (TNTs) represent a subset of F-actin-based transient tubular connections that allow direct communication between distant cells. Recent studies have provided new insights into the existence of TNTs in vivo, and this novel mechanism of intercellular communication is implicated in various essential processes, such as development, immunity, tissue regeneration and transmission of electrical signals. TNTs are versatile structures known to facilitate the transfer of various cargos, such as organelles, plasma membrane components, pathogens and Ca(2+). Recently, a new function of TNTs in the long-range transfer of electrical signals that involves gap junctions has been suggested. This indicates that different types of TNTs might exist, and supports the notion that TNTs might not be just passive open conduits but rather are regulated by gating mechanisms. Furthermore, TNTs have been found in different cell lines and are characterized by their diversity in terms of morphology. Here we discuss these novel findings in the context of the two models that have been proposed for TNT formation, and focus on putative proteins that could represent TNT specific markers. We also shed some light on the molecular mechanisms used by TNTs to transfer cargos, as well as chemical and electrical signals.
Collapse
Affiliation(s)
- Saïda Abounit
- Institut Pasteur, Unité de Trafic Membranaire et Pathogénèse, 25 rue du Docteur Roux, 75724 Paris, Cedex 15, France
| | | |
Collapse
|
36
|
Hazelett CC, Sheff D, Yeaman C. RalA and RalB differentially regulate development of epithelial tight junctions. Mol Biol Cell 2011; 22:4787-800. [PMID: 22013078 PMCID: PMC3237622 DOI: 10.1091/mbc.e11-07-0657] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Tight junctions (TJs) are structures indispensable to epithelial cells and are responsible for regulation of paracellular diffusion and maintenance of cellular polarity. Although many interactions between TJ constituents have been identified, questions remain concerning how specific functions of TJs are established and regulated. Here we investigated the roles of Ral GTPases and their common effector exocyst complex in the formation of nascent TJs. Unexpectedly, RNA interference-mediated suppression of RalA or RalB caused opposing changes in TJ development. RalA reduction increased paracellular permeability and decreased incorporation of components into TJs, whereas RalB reduction decreased paracellular permeability and increased incorporation of components into TJs. Activities of both Ral GTPases were mediated through the exocyst. Finally, we show that TJ-mediated separation of apical-basal membrane domains is established prior to equilibration of barrier function and that it is unaffected by Ral knockdown or specific composition of TJs.
Collapse
Affiliation(s)
- C Clayton Hazelett
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
37
|
Jeon H, Zheng LT, Lee S, Lee WH, Park N, Park JY, Heo WD, Lee MS, Suk K. Comparative analysis of the role of small G proteins in cell migration and cell death: cytoprotective and promigratory effects of RalA. Exp Cell Res 2011; 317:2007-18. [PMID: 21645515 DOI: 10.1016/j.yexcr.2011.05.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2011] [Revised: 05/18/2011] [Accepted: 05/19/2011] [Indexed: 12/30/2022]
Abstract
Small G protein superfamily consists of more than 150 members, and is classified into six families: the Ras, Rho, Rab, Arf, Ran, and RGK families. They regulate a wide variety of cell functions such as cell proliferation/differentiation, cytoskeletal reorganization, vesicle trafficking, nucleocytoplasmic transport and microtubule organization. The small G proteins have also been shown to regulate cell death/survival and cell shape. In this study, we compared the role of representative members of the six families of small G proteins in cell migration and cell death/survival, two cellular phenotypes that are associated with inflammation, tumorigenesis, and metastasis. Our results show that small G proteins of the six families differentially regulate cell death and cell cycle distribution. In particular, our results indicate that Rho family of small G proteins is antiapoptotic. Ras, Rho, and Ran families promoted cell migration. There was no significant correlation between the cell death- and cell migration-regulating activities of the small G proteins. Nevertheless, RalA was not only cytoprotective against multiple chemotherapeutic drugs, but also promigratory inducing stress fiber formation, which was accompanied by the activation of Akt and Erk pathways. Our study provides a framework for further systematic investigation of small G proteins in the perspectives of cell death/survival and motility in inflammation and cancer.
Collapse
Affiliation(s)
- Hyejin Jeon
- Department of Pharmacology, Brain Science and Engineering Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Cho B, Fischer JA. Ral GTPase promotes asymmetric Notch activation in the Drosophila eye in response to Frizzled/PCP signaling by repressing ligand-independent receptor activation. Development 2011; 138:1349-59. [PMID: 21350007 DOI: 10.1242/dev.056002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ral is a small Ras-like GTPase that regulates membrane trafficking and signaling. Here, we show that in response to planar cell polarity (PCP) signals, Ral modulates asymmetric Notch signaling in the Drosophila eye. Specification of the initially equivalent R3/R4 photoreceptor precursor cells in each developing ommatidium occurs in response to a gradient of Frizzled (Fz) signaling. The cell with the most Fz signal (R3) activates the Notch receptor in the adjacent cell (R4) via the ligand Delta, resulting in R3/R4 cell determination and their asymmetric positions within the ommatidium. Two mechanisms have been proposed for ensuring that the cell with the most Fz activation sends the Delta signal: Fz-dependent transcriptional upregulation in R3 of genes that promote Delta signaling, and direct blockage of Notch receptor activation in R3 by localization of an activated Fz/Disheveled protein complex to the side of the plasma membrane adjacent to R4. Here, we discover a distinct mechanism for biasing the direction of Notch signaling that depends on Ral. Using genetic experiments in vivo, we show that, in direct response to Fz signaling, Ral transcription is upregulated in R3, and Ral represses ligand-independent activation of Notch in R3. Thus, prevention of ligand-independent Notch activation is not simply a constitutive process, but is a target for regulation by Ral during cell fate specification and pattern formation.
Collapse
Affiliation(s)
- Bomsoo Cho
- Section of Molecular Cell and Developmental Biology, Institute for Cell and Molecular Biology, The University of Texas at Austin, 1 University Station A4800, Austin, TX 78712, USA
| | | |
Collapse
|
39
|
Shirai Y, Morioka S, Sakuma M, Yoshino KI, Otsuji C, Sakai N, Kashiwagi K, Chida K, Shirakawa R, Horiuchi H, Nishigori C, Ueyama T, Saito N. Direct binding of RalA to PKCη and its crucial role in morphological change during keratinocyte differentiation. Mol Biol Cell 2011; 22:1340-52. [PMID: 21346190 PMCID: PMC3078077 DOI: 10.1091/mbc.e10-09-0754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
A small G protein, RalA, was identified as a binding partner of PKCη. The binding led to activation of RalA and actin depolymerization associated with keratinocyte differentiation. These results provide new insight into the molecular mechanism of cytoskeletal regulation that leads to drastic change of cell shape. During differentiation, keratinocytes undergo a dramatic shape change from small and round to large and flat, in addition to production of proteins necessary for the formation of epidermis. It has been shown that protein kinase C (PKC) η is crucial for keratinocyte differentiation. However, its role in this process has yet to be fully elucidated. Here, we show that catalytic activity is not necessary for enlarged and flattened morphology of human keratinocytes induced by overexpression of PKCη, although it is important for gene expression of the marker proteins. In addition, we identify the small G protein RalA as a binding partner of PKCη, which binds to the C1 domain, an indispensable region for the morphological change. The binding led activation of RalA and actin depolymerization associated with keratinocyte differentiation. siRNA techniques proved that RalA is involved in not only the keratinocyte differentiation induced by PKCη overexpression but also normal keratinocyte differentiation induced by calcium and cholesterol sulfate. These results provide a new insight into the molecular mechanism of cytoskeletal regulation leading to drastic change of cell shape.
Collapse
Affiliation(s)
- Yasuhito Shirai
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Motile processes are critical for several physiological and pathological situations such as embryonic development, tumour dissemination and metastasis. Migrating cells, or developing neurons, need to establish front–rear polarity consisting of actin-driven extension of the leading edge and traffic of components that are essential for membrane extension and cell adhesion at the front. Previously, several studies have suggested that the exocyst complex is critical for the establishment and maintenance of cell polarity. This octameric complex controls the docking and insertion of exocytic vesicles to growing areas of the plasma membrane. The aim of the present review is to detail recent advances concerning the molecular and structural organization of the exocyst complex that help to elucidate its role in cell polarity. We will also review the function of the exocyst complex and some of its key interacting partners [including the small GTP-binding protein Ral, aPKCs (atypical protein kinase Cs) and proteins involved in actin assembly] in the formation of plasma extensions at the leading edge, growth cone formation during axonal extension and generation of cell movement.
Collapse
|
41
|
Abstract
Salmonella entry into host cells involves rearrangements of actin and mobilization of membranes. Here we discuss new findings showing that Salmonella recruits the exocyst complex, which plays a role in vesicle secretion, to the site of invasion to promote its entry.
Collapse
Affiliation(s)
- Virginie Braun
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada, M5G1X8
| | | |
Collapse
|
42
|
Wang H, Owens C, Chandra N, Conaway MR, Brautigan DL, Theodorescu D. Phosphorylation of RalB is important for bladder cancer cell growth and metastasis. Cancer Res 2010; 70:8760-9. [PMID: 20940393 DOI: 10.1158/0008-5472.can-10-0952] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RalA and RalB are monomeric G proteins that are 83% identical in amino acid sequence but have paralogue-specific effects on cell proliferation, metastasis, and apoptosis. Using in vitro kinase assays and phosphosite-specific antibodies, here we show phosphorylation of RalB by protein kinase C (PKC) and RalA by protein kinase A. We used mass spectrometry and site-directed mutagenesis to identify S198 as the primary PKC phosphorylation site in RalB. Phorbol ester [phorbol 12-myristate 13-acetate (PMA)] treatment of human bladder carcinoma cells induced S198 phosphorylation of stably expressed FLAG-RalB as well as endogenous RalB. PMA treatment caused RalB translocation from the plasma membrane to perinuclear regions in a S198 phosphorylation-dependent manner. Using RNA interference depletion of RalB followed by rescue with wild-type RalB or RalB(S198A) as well as overexpression of wild-type RalB or RalB(S198A) with and without PMA stimulation, we show that phosphorylation of RalB at S198 is necessary for actin cytoskeletal organization, anchorage-independent growth, cell migration, and experimental lung metastasis of T24 or UMUC3 human bladder cancer cells. In addition, UMUC3 cells transfected with a constitutively active RalB(G23V) exhibited enhanced subcutaneous tumor growth, whereas those transfected with phospho-deficient RalB(G23V-S198A) were indistinguishable from control cells. Our data show that RalA and RalB are phosphorylated by different kinases, and RalB phosphorylation is necessary for in vitro cellular functions and in vivo tumor growth and metastasis.
Collapse
Affiliation(s)
- Hong Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | | | | | | | | | |
Collapse
|
43
|
Corrotte M, Nyguyen APT, Harlay ML, Vitale N, Bader MF, Grant NJ. Ral isoforms are implicated in Fc gamma R-mediated phagocytosis: activation of phospholipase D by RalA. THE JOURNAL OF IMMUNOLOGY 2010; 185:2942-50. [PMID: 20679536 DOI: 10.4049/jimmunol.0903138] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Phagocytosis is an essential element of the immune response permitting the elimination of pathogens, cellular debris, apoptotic cells, and tumor cells. Recently, both phospholipase D (PLD) isoforms, PLD1 and PLD2, were shown to be necessary for efficient FcgammaR-mediated phagocytosis. In this study, we investigated the role of a potential PLD regulator, the Ral GTPases RalA and RalB, in murine RAW 264.7 macrophages. Both Ral isoforms are expressed in macrophages and are transiently activated following FcgammaR stimulation. When Ral expression levels were varied using Ral mutants or interference RNA, phagocytosis assays revealed that Ral isoforms have antagonistic effects; RalA is a positive modulator, whereas RalB plays a negative role. We then focused on RalA and its possible relationship with PLD. The increase in PLD activity that occurs when phagocytosis is stimulated was inhibited in cells with reduced RalA protein, but it was unaffected by reduced levels of RalB. Furthermore, in macrophages transfected with dsRed-RalA and GFP-PLD1 or GFP-PLD2, RalA colocalized with PLD1 and PLD2 at the phagocytic cup during phagosome formation. Additional results obtained from immunoprecipitation of PLD from macrophages transfected with myc-RalA and hemagglutinin-tagged PLD1 or PLD2 indicated an enhanced interaction of RalA with both PLD isoforms during phagocytic stimulation. The increase in RalA and PLD1 interaction was transient and correlated with the time course of RalA activation. These findings reveal a novel pathway involving RalA and PLD in the regulation of FcgammaR-mediated phagocytosis.
Collapse
Affiliation(s)
- Matthias Corrotte
- Département Neurotransmission et Sécrétion Neuroendocrine, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Unité Propre de Recherche 3212, Strasbourg, France
| | | | | | | | | | | |
Collapse
|
44
|
Fenwick RB, Prasannan S, Campbell LJ, Nietlispach D, Evetts KA, Camonis J, Mott HR, Owen D. Solution structure and dynamics of the small GTPase RalB in its active conformation: significance for effector protein binding. Biochemistry 2009; 48:2192-206. [PMID: 19166349 DOI: 10.1021/bi802129d] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The small G proteins RalA/B have a crucial function in the regulatory network that couples extracellular signals with appropriate cellular responses. RalA/B are an important component of the Ras signaling pathway and, in addition to their role in membrane trafficking, are implicated in the initiation and maintenance of tumorigenic transformation of human cells. RalA and RalB share 85% sequence identity and collaborate in supporting cancer cell proliferation but have markedly different effects. RalA is important in mediating proliferation, while depletion of RalB results in transformed cells undergoing apoptosis. Crystal structures of RalA in the free form and in complex with its effectors, Sec5 and Exo84, have been solved. Here we have determined the solution structure of free RalB bound to the GTP analogue GMPPNP to an RMSD of 0.6 A. We show that, while the overall architecture of RalB is very similar to the crystal structure of RalA, differences exist in the switch regions, which are sensitive to the bound nucleotide. Backbone 15N dynamics suggest that there are four regions of disorder in RalB: the P-loop, switch I, switch II, and the loop comprising residues 116-121, which has a single residue insertion compared to RalA. 31P NMR data and the structure of RalB.GMPPNP show that the switch regions predominantly adopt state 1 (Ras nomenclature) in the unbound form, which in Ras is not competent to bind effectors. In contrast, 31P NMR analysis of RalB.GTP reveals that conformations corresponding to states 1 and 2 are both sampled in solution and that addition of an effector protein only partially stabilizes state 2.
Collapse
|
45
|
Ceriani M, Amigoni L, Scandiuzzi C, Berruti G, Martegani E. The PH-PxxP domain of RalGPS2 promotes PC12 cells differentiation acting as a dominant negative for RalA GTPase activation. Neurosci Res 2009; 66:290-8. [PMID: 20025911 DOI: 10.1016/j.neures.2009.11.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 11/11/2009] [Accepted: 11/30/2009] [Indexed: 10/20/2022]
Abstract
RalGPS2 is a guanine nucleotide exchange factor for RalA GTPase characterized by a C-terminal Pleckstrin Homology (PH) domain; this GEF is endogenously expressed in PC12 cells and in rat brain but its role in PC12 cells and in cell differentiation is actually unknown. Here we have shown that transient expression of RalGPS2-PH-PxxP domain in PC12 and PC12-TrkA cells induces high level of neurite outgrowth; this differentiation is comparable with that of PC12 cells treated with RalGPS2 siRNA. Stable PC12 cell lines expressing the PH-PxxP domain of RalGPS2 have been generated; in these cell lines the PH-PxxP domain acts as a dominant negative for RalA activation, promotes cells differentiation and re-directs NGF signals towards MAPKs. Furthermore it has been also demonstrated that the PH-PxxP domain of RalGPS2 induces microspikes formation a typical feature of cells in which the Cdc42 GTPase is constitutively activated.
Collapse
Affiliation(s)
- Michela Ceriani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, Milan, Italy
| | | | | | | | | |
Collapse
|
46
|
Hase K, Kimura S, Takatsu H, Ohmae M, Kawano S, Kitamura H, Ito M, Watarai H, Hazelett CC, Yeaman C, Ohno H. M-Sec promotes membrane nanotube formation by interacting with Ral and the exocyst complex. Nat Cell Biol 2009; 11:1427-32. [PMID: 19935652 DOI: 10.1038/ncb1990] [Citation(s) in RCA: 275] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 11/04/2009] [Indexed: 12/14/2022]
Abstract
Cell-cell communication is essential for the development and homeostasis of multicellular organisms. Recently, a new type of cell-cell communication was discovered that is based on the formation of thin membranous nanotubes between remote cells. These long membrane tethers, termed tunneling nanotubes (TNTs), form an intercellular conduit and have been shown to enable the transport of various cellular components and signals. However, the molecular basis for TNT formation remains to be elucidated. Here we report that a mammalian protein, M-Sec, induces de novo formation of numerous membrane protrusions extending from the plasma membrane, some of which tether onto adjacent cells and subsequently form TNT-like structures. Depletion of M-Sec by RNA interference (RNAi) greatly reduced endogenous TNT formation as well as intercellular propagation of a calcium flux in a macrophage cell line. Furthermore, blockage of the interaction of M-Sec with Ral and the exocyst complex, which serves as a downstream effector of Ral, attenuated the formation of membrane nanotubes. Our results reveal that M-Sec functions as a key regulator of membrane nanotube formation through interaction with the Ral-exocyst pathway.
Collapse
Affiliation(s)
- Koji Hase
- Laboratory for Epithelial Immunobiology, Kanagawa 230-0045, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
The GTPase RalA regulates different steps of the secretory process in pancreatic beta-cells. PLoS One 2009; 4:e7770. [PMID: 19890390 PMCID: PMC2766836 DOI: 10.1371/journal.pone.0007770] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Accepted: 10/11/2009] [Indexed: 12/25/2022] Open
Abstract
Background RalA and RalB are multifuntional GTPases involved in a variety of cellular processes including proliferation, oncogenic transformation and membrane trafficking. Here we investigated the mechanisms leading to activation of Ral proteins in pancreatic β-cells and analyzed the impact on different steps of the insulin-secretory process. Methodology/Principal Findings We found that RalA is the predominant isoform expressed in pancreatic islets and insulin-secreting cell lines. Silencing of this GTPase in INS-1E cells by RNA interference led to a decrease in secretagogue-induced insulin release. Real-time measurements by fluorescence resonance energy transfer revealed that RalA activation in response to secretagogues occurs within 3–5 min and reaches a plateau after 10–15 min. The activation of the GTPase is triggered by increases in intracellular Ca2+ and cAMP and is prevented by the L-type voltage-gated Ca2+ channel blocker Nifedipine and by the protein kinase A inhibitor H89. Defective insulin release in cells lacking RalA is associated with a decrease in the secretory granules docked at the plasma membrane detected by Total Internal Reflection Fluorescence microscopy and with a strong impairment in Phospholipase D1 activation in response to secretagogues. RalA was found to be activated by RalGDS and to be severely hampered upon silencing of this GDP/GTP exchange factor. Accordingly, INS-1E cells lacking RalGDS displayed a reduction in hormone secretion induced by secretagogues and in the number of insulin-containing granules docked at the plasma membrane. Conclusions/Significance Taken together, our data indicate that RalA activation elicited by the exchange factor RalGDS in response to a rise in intracellular Ca2+ and cAMP controls hormone release from pancreatic β-cell by coordinating the execution of different events in the secretory pathway.
Collapse
|
48
|
Han K, Kim MH, Seeburg D, Seo J, Verpelli C, Han S, Chung HS, Ko J, Lee HW, Kim K, Heo WD, Meyer T, Kim H, Sala C, Choi SY, Sheng M, Kim E. Regulated RalBP1 binding to RalA and PSD-95 controls AMPA receptor endocytosis and LTD. PLoS Biol 2009; 7:e1000187. [PMID: 19823667 PMCID: PMC2730530 DOI: 10.1371/journal.pbio.1000187] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Accepted: 07/30/2009] [Indexed: 11/18/2022] Open
Abstract
Long-term depression (LTD) is a long-lasting activity-dependent decrease in synaptic strength. NMDA receptor (NMDAR)-dependent LTD, an extensively studied form of LTD, involves the endocytosis of AMPA receptors (AMPARs) via protein dephosphorylation, but the underlying mechanism has remained unclear. We show here that a regulated interaction of the endocytic adaptor RalBP1 with two synaptic proteins, the small GTPase RalA and the postsynaptic scaffolding protein PSD-95, controls NMDAR-dependent AMPAR endocytosis during LTD. NMDAR activation stimulates RalA, which binds and translocates widespread RalBP1 to synapses. In addition, NMDAR activation dephosphorylates RalBP1, promoting the interaction of RalBP1 with PSD-95. These two regulated interactions are required for NMDAR-dependent AMPAR endocytosis and LTD and are sufficient to induce AMPAR endocytosis in the absence of NMDAR activation. RalA in the basal state, however, maintains surface AMPARs. We propose that NMDAR activation brings RalBP1 close to PSD-95 to promote the interaction of RalBP1-associated endocytic proteins with PSD-95-associated AMPARs. This suggests that scaffolding proteins at specialized cellular junctions can switch their function from maintenance to endocytosis of interacting membrane proteins in a regulated manner.
Collapse
Affiliation(s)
- Kihoon Han
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Myoung-Hwan Kim
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Daniel Seeburg
- The Picower Institute for Learning and Memory, RIKEN-MIT Neuroscience Research Center
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Jinsoo Seo
- Department of Physiology and Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| | - Chiara Verpelli
- CNR Institute of Neuroscience and Department of Neurological Sciences, University of Milan, Milan, Italy
| | - Seungnam Han
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Hye Sun Chung
- Department of Anatomy and Division of Brain Korea 21 Biomedical Science, College of Medicine, Korea University, Seoul, Korea
| | - Jaewon Ko
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Hyun Woo Lee
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Karam Kim
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Won Do Heo
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Tobias Meyer
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
| | - Hyun Kim
- Department of Anatomy and Division of Brain Korea 21 Biomedical Science, College of Medicine, Korea University, Seoul, Korea
| | - Carlo Sala
- CNR Institute of Neuroscience and Department of Neurological Sciences, University of Milan, Milan, Italy
| | - Se-Young Choi
- Department of Physiology and Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| | - Morgan Sheng
- The Picower Institute for Learning and Memory, RIKEN-MIT Neuroscience Research Center
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Eunjoon Kim
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
- * E-mail:
| |
Collapse
|
49
|
Kardassis D, Murphy C, Fotsis T, Moustakas A, Stournaras C. Control of transforming growth factor β signal transduction by small GTPases. FEBS J 2009; 276:2947-65. [DOI: 10.1111/j.1742-4658.2009.07031.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
50
|
Abstract
Ras leads an important signaling pathway that is deregulated in neurofibromatosis type 1 and malignant peripheral nerve sheath tumor (MPNST). In this study, we show that overactivation of Ras and many of its downstream effectors occurred in only a fraction of MPNST cell lines. RalA, however, was overactivated in all MPNST cells and tumor samples compared to nontransformed Schwann cells. Silencing Ral or inhibiting it with a dominant-negative Ral (Ral S28N) caused a significant reduction in proliferation, invasiveness, and in vivo tumorigenicity of MPNST cells. Silencing Ral also reduced the expression of epithelial mesenchymal transition markers. Expression of the NF1-GTPase-related domain (NF1-GRD) diminished the levels of Ral activation, implicating a role for neurofibromin in regulating RalA activation. NF1-GRD treatment caused a significant decrease in proliferation, invasiveness, and cell cycle progression, but cell death increased. We propose Ral overactivation as a novel cell signaling abnormality in MPNST that leads to important biological outcomes with translational ramifications.
Collapse
|