1
|
Fakieh RA, Reiner DJ. RAP-2 and CNH-MAP4 Kinase MIG-15 confer resistance in bystander epithelium to cell-fate transformation by excess Ras or Notch activity. Proc Natl Acad Sci U S A 2025; 122:e2414321121. [PMID: 39739816 PMCID: PMC11725784 DOI: 10.1073/pnas.2414321121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/30/2024] [Indexed: 01/02/2025] Open
Abstract
Induction of cell fates by growth factors impacts many facets of developmental biology and disease. LIN-3/EGF induces the equipotent vulval precursor cells (VPCs) in Caenorhabditis elegans to assume the 3˚-3˚-2˚-1˚-2˚-3˚ pattern of cell fates. 1˚ and 2˚ cells become specialized epithelia and undergo stereotyped series of cell divisions to form the vulva. Conversely, 3˚ cells are relatively quiescent and nonspecialized; they divide once and fuse with the surrounding epithelium. 3˚ cells have thus been characterized as passive, uninduced, or ground state. Based on our previous studies, we hypothesized that a 3˚-promoting program would confer resistance to cell fate-transformation by inappropriately activated 1˚ and 2˚ fate-promoting LET-60/Ras and LIN-12/Notch, respectively. Deficient MIG-15/CNH-MAP4 Kinase meets the expectations of genetic interactions for a 3˚-promoting protein. Moreover, endogenous MIG-15 is required for expression of a fluorescent biomarker of 3˚ cell fate, is expressed in VPCs, and functions cell autonomously in VPCs. The Ras family small GTPase RAP-2, orthologs of which activate orthologs of MIG-15 in other systems, emulates these functions of MIG-15. However, gain of RAP-2 function has no effect on patterning, suggesting its activity is constitutive in VPCs. The 3˚ biomarker is expressed independently of the AC, raising questions about the cellular origin of 3˚-promoting activity. Activated LET-60/Ras and LIN-12/Notch repress expression of the 3˚ biomarker, suggesting that the 3˚-promoting program is both antagonized by as well as antagonizes 1˚- and 2˚- promoting programs. This study provides insight into developmental properties of cells historically considered to be nonresponding to growth factor signals.
Collapse
Affiliation(s)
- Razan A. Fakieh
- Department of Translational Medical Sciences, School of Medicine, Texas A&M Health Science Center, Texas A&M University, Houston, TX77030
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Imam Abdulrahman bin Faisal University, Dammam34212, Kingdom of Saudi Arabia
| | - David J. Reiner
- Department of Translational Medical Sciences, School of Medicine, Texas A&M Health Science Center, Texas A&M University, Houston, TX77030
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Texas A&M University, Houston, TX77030
| |
Collapse
|
2
|
Joachim J, Maselli D, Petsolari E, Aman J, Swiatlowska P, Killock D, Chaudhry H, Zarban AA, Sarker M, Fraser P, Cleary SJ, Amison R, Cuthbert I, Yang Y, Meier M, Fraternali F, Brain SD, Shah AM, Ivetic A. TNIK: A redox sensor in endothelial cell permeability. SCIENCE ADVANCES 2024; 10:eadk6583. [PMID: 39705357 DOI: 10.1126/sciadv.adk6583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/18/2024] [Indexed: 12/22/2024]
Abstract
Dysregulation of endothelial barrier integrity can lead to vascular leak and potentially fatal oedema. TNF-α controls endothelial permeability during inflammation and requires the actin organizing Ezrin-Radixin-Moesin (ERM) proteins. We identified TRAF2 and NCK-interacting kinase (TNIK) as a kinase directly phosphorylating and activating ERM, specifically at the plasma membrane of primary human endothelial cells. TNIK mediates TNF-α-dependent cellular stiffness and paracellular gap formation in vitro and is essential in driving inflammatory oedema formation in vivo. Unlike its homologs, TNIK activity is negatively and reversibly regulated by H2O2-mediated oxidation of C202 within the kinase domain. TNIK oxidation results in intermolecular disulfide bond formation and loss of kinase activity. Pharmacologic inhibition of endogenous reactive oxygen species production in endothelial cells elevated TNIK-dependent ERM phosphorylation, endothelial cell contraction, and cell rounding. Together, we highlight an interplay between TNIK, ERM phosphorylation, and redox signalling in regulating TNF-induced endothelial cell permeability.
Collapse
Affiliation(s)
- Justin Joachim
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Davide Maselli
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Emmanouela Petsolari
- Randall Centre for Cell and Molecular Biology, King's College London, London SE1 1UL, UK
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam University Medical Center, location VUMC, Amsterdam, The Netherlands
| | - Pamela Swiatlowska
- Myocardial Function, National Heart and Lung Institute, Imperial College London, ICTEM, Hammersmith Hospital, London, UK
| | - David Killock
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Hiba Chaudhry
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Ali A Zarban
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
- Department of Pharmacological Sciences, Faculty of Pharmacy, Jazan University, Saudi Arabia
| | - Mosharraf Sarker
- Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK
| | - Paul Fraser
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Simon J Cleary
- Institute of Pharmaceutical Science, King's College London, Floor 5, Southwark Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Richard Amison
- School of Cancer and Pharmaceutical Sciences, Pulmonary Pharmacology Unit, King's College London, London, UK
| | - Isabelle Cuthbert
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Yue Yang
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Magda Meier
- School of Genetics and Genomic Medicine, University College London Institute of Child Health, London, UK
| | - Franca Fraternali
- Randall Centre for Cell and Molecular Biology, King's College London, London SE1 1UL, UK
- Division of Biosciences, Structural and Molecular Biology Department, University College London, Darwin (SMB) Building, Gower Street, London WC1E 6BT, UK
- Department of Structural and Molecular Biology, Division of Biosciences and Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK Department of Biological Sciences, Birkbeck, University of London, London WC1E 7HX, United Kingdom
| | - Susan D Brain
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Ajay M Shah
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Aleksandar Ivetic
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| |
Collapse
|
3
|
Felipe-López A, Hansmeier N, Hensel M. Destruction of the brush border by Salmonella enterica sv. Typhimurium subverts resorption by polarized epithelial cells. Front Microbiol 2024; 15:1329798. [PMID: 38894970 PMCID: PMC11183102 DOI: 10.3389/fmicb.2024.1329798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 05/06/2024] [Indexed: 06/21/2024] Open
Abstract
Salmonella enterica serovar Typhimurium is an invasive, facultative intracellular gastrointestinal pathogen that destroys the brush border of polarized epithelial cells (PEC). The brush border is critical for the functions of PEC because it resorbs nutrients from the intestinal lumen and builds a physical barrier to infecting pathogens. The manipuation of PEC during infection by Salmonella was investigated by live-cell imaging and ultrastructural analysed of the brush border. We demonstrate that the destruction of the brush border by Salmonella significantly reduces the resorption surface of PEC along with the abrogation of endocytosis at the apical side of PEC. Both these changes in the physiology of PEC were associated with the translocation of type III secretion system effector protein SopE. Additionally, the F-actin polymerization rate at the apical side of PEC was highly altered by SopE, indicating that reduced endocytosis observed in infected PEC is related to the manipulation of F-actin polymerization mediated by SopE and, to a lesser extent, by effectors SopE2 or SipA. We further observed that in the absence of SopE, Salmonella effaced microvilli and induced reticular F-actin by bacterial accumulation during prolonged infection periods. In contrast to strains translocating SopE, strains lacking SopE did not alter resorption by PEC. Finally, we observed that after engulfment of Salmonella, ezrin was lost from the apical side of PEC and found later in early endosomes containing Salmonella. Our observations suggest that the destruction of the brush border by Salmonella may contribute to the pathogenesis of diarrhea.
Collapse
Affiliation(s)
| | | | - Michael Hensel
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany
- CellNanOs—Center of Cellular Nanoanalytics Osnabrück, Universität Osnabrück, Osnabrück, Germany
| |
Collapse
|
4
|
Bradford STJ, Wu H, Kirita Y, Chen C, Malvin NP, Yoshimura Y, Muto Y, Humphreys BD. TNIK depletion induces inflammation and apoptosis in injured renal proximal tubule epithelial cells. Am J Physiol Renal Physiol 2024; 326:F827-F838. [PMID: 38482555 PMCID: PMC11386974 DOI: 10.1152/ajprenal.00262.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 04/12/2024] Open
Abstract
In the aftermath of acute kidney injury (AKI), surviving proximal tubule epithelia repopulate injured tubules to promote repair. However, a portion of cells fail to repair [termed failed-repair proximal tubule cells (FR-PTCs)] and exert ongoing proinflammatory and profibrotic effects. To better understand the molecular drivers of the FR-PTC state, we reanalyzed a mouse ischemia-reperfusion injury single-nucleus RNA-sequencing (snRNA-seq) atlas to identify Traf2 and Nck interacting kinase (Tnik) to be exclusively expressed in FR-PTCs but not in healthy or acutely injured proximal tubules after AKI (2 and 6 wk) in mice. We confirmed expression of Tnik protein in injured mouse and human tissues by immunofluorescence. Then, to determine the functional role of Tnik in FR-PTCs, we depleted TNIK with siRNA in two human renal proximal tubule epithelial cell lines (primary and immortalized hRPTECs) and analyzed each by bulk RNA-sequencing. Pathway analysis revealed significant upregulation of inflammatory signaling pathways, whereas pathways associated with differentiated proximal tubules such as organic acid transport were significantly downregulated. TNIK gene knockdown drove reduced cell viability and increased apoptosis, including differentially expressed poly(ADP-ribose) polymerase (PARP) family members, cleaved PARP-1 fragments, and increased annexin V binding to phosphatidylserine. Together, these results indicate that Tnik upregulation in FR-PTCs acts in a compensatory fashion to suppress inflammation and promote proximal tubule epithelial cell survival after injury. Modulating TNIK activity may represent a prorepair therapeutic strategy after AKI.NEW & NOTEWORTHY The molecular drivers of successful and failed repair in the proximal tubule after acute kidney injury (AKI) are incompletely understood. We identified Traf2 and Nck interacting kinase (Tnik) to be exclusively expressed in failed-repair proximal tubule cells after AKI. We tested the effect of siTNIK depletion in two proximal tubule cell lines followed by bulk RNA-sequencing analysis. Our results indicate that TNIK acts to suppress inflammatory signaling and apoptosis in injured renal proximal tubule epithelial cells to promote cell survival.
Collapse
Affiliation(s)
- Shayna T J Bradford
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Haojia Wu
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Yuhei Kirita
- Department of Nephrology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Changfeng Chen
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Nicole P Malvin
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Yasuhiro Yoshimura
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Yoshiharu Muto
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| |
Collapse
|
5
|
Chau KM, Dominic A, Davis EL, Kotla S, Berrios ET, Fahim A, Arunesh A, Li S, Zhao D, Chen K, Davis AR, Nguyen MTH, Wang Y, Evans SE, Wang G, Cooke JP, Abe JI, Huston DP, Le NT. TNIK regulation of interferon signaling and endothelial cell response to virus infection. Front Cardiovasc Med 2024; 10:1213428. [PMID: 38264262 PMCID: PMC10803426 DOI: 10.3389/fcvm.2023.1213428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 11/27/2023] [Indexed: 01/25/2024] Open
Abstract
Background Traf2 and Nck-interacting kinase (TNIK) is known for its regulatory role in various processes within cancer cells. However, its role within endothelial cells (ECs) has remained relatively unexplored. Methods Leveraging RNA-seq data and Ingenuity Pathway Analysis (IPA), we probed the potential impact of TNIK depletion on ECs. Results Examination of RNA-seq data uncovered more than 450 Differentially Expressed Genes (DEGs) in TNIK-depleted ECs, displaying a fold change exceeding 2 with a false discovery rate (FDR) below 0.05. IPA analysis unveiled that TNIK depletion leads to the inhibition of the interferon (IFN) pathway [-log (p-value) >11], downregulation of IFN-related genes, and inhibition of Hypercytokinemia/Hyperchemokinemia [-log (p-value) >8]. The validation process encompassed qRT-PCR to evaluate mRNA expression of crucial IFN-related genes, immunoblotting to gauge STAT1 and STAT2 protein levels, and ELISA for the quantification of IFN and cytokine secretion in siTNIK-depleted ECs. These assessments consistently revealed substantial reductions upon TNIK depletion. When transducing HUVECs with replication incompetent E1-E4 deleted adenovirus expressing green fluorescent protein (Ad-GFP), it was demonstrated that TNIK depletion did not affect the uptake of Ad-GFP. Nonetheless, TNIK depletion induced cytopathic effects (CPE) in ECs transduced with wild-type human adenovirus serotype 5 (Ad-WT). Summary Our findings suggest that TNIK plays a crucial role in regulating the EC response to virus infections through modulation of the IFN pathway.
Collapse
Affiliation(s)
- Khanh M. Chau
- Department of Cardiovascular Sciences, Center for Cardiovascular Sciences, Academic Institute, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, United States
| | - Abishai Dominic
- Department of Cardiovascular Sciences, Center for Cardiovascular Sciences, Academic Institute, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, United States
- Department of Molecular and Cellular Medicine, College of Medicine Texas A&M University, College Station, TX, United States
| | - Eleanor L. Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, College Station, TX, United States
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Estefani Turcios Berrios
- Department of Cardiovascular Sciences, Center for Cardiovascular Sciences, Academic Institute, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, United States
| | - Arsany Fahim
- Center for Cell and Gene Therapy, Baylor College of Medicine, College Station, TX, United States
| | - Ashwin Arunesh
- Center for Cell and Gene Therapy, Baylor College of Medicine, College Station, TX, United States
| | - Shengyu Li
- Department of Cardiovascular Sciences, Center for Cardiovascular Sciences, Academic Institute, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, United States
| | - Dongyu Zhao
- Department of Molecular and Cellular Medicine, College of Medicine Texas A&M University, College Station, TX, United States
| | - Kaifu Chen
- Department of Cardiovascular Sciences, Center for Cardiovascular Sciences, Academic Institute, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, United States
| | - Alan R. Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, College Station, TX, United States
- Department of Cellular and Molecular Biology, Baylor College of Medicine, Houston, TX, United States
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Minh T. H. Nguyen
- Department of Cardiovascular Sciences, Center for Cardiovascular Sciences, Academic Institute, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, United States
| | - Yongxing Wang
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Scott E. Evans
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Center for Cardiovascular Sciences, Academic Institute, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, United States
| | - John P. Cooke
- Department of Cardiovascular Sciences, Center for Cardiovascular Sciences, Academic Institute, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, United States
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - David P. Huston
- Department of Microbial Pathogenesis and Immunology, College of Medicine Texas A&M University, College Station, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Center for Cardiovascular Sciences, Academic Institute, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, United States
| |
Collapse
|
6
|
Demouchy F, Nicolle O, Michaux G, Pacquelet A. PAR-4/LKB1 prevents intestinal hyperplasia by restricting endoderm specification in Caenorhabditis elegans embryos. Development 2024; 151:dev202205. [PMID: 38078543 DOI: 10.1242/dev.202205] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024]
Abstract
The kinase PAR-4/LKB1 is a major regulator of intestinal homeostasis, which prevents polyposis in humans. Moreover, its ectopic activation is sufficient to induce polarization and formation of microvilli-like structures in intestinal cell lines. Here, we use Caenorhabditis elegans to examine the role of PAR-4 during intestinal development in vivo. We show that it is not required to establish enterocyte polarity and plays only a minor role in brush border formation. By contrast, par-4 mutants display severe deformations of the intestinal lumen as well as supernumerary intestinal cells, thereby revealing a previously unappreciated function of PAR-4 in preventing intestinal hyperplasia. The presence of supernumerary enterocytes in par-4 mutants is not due to excessive cell proliferation, but rather to the abnormal expression of the intestinal cell fate factors end-1 and elt-2 outside the E lineage. Notably, par-4 mutants also display reduced expression of end-1 and elt-2 inside the E lineage. Our work thereby unveils an essential and dual role of PAR-4, which both restricts intestinal specification to the E lineage and ensures its robust differentiation.
Collapse
Affiliation(s)
- Flora Demouchy
- University of Rennes, CNRS, IGDR (Institut de Génétique et de Développement de Rennes), UMR 6290, F-35000 Rennes, France
| | - Ophélie Nicolle
- University of Rennes, CNRS, IGDR (Institut de Génétique et de Développement de Rennes), UMR 6290, F-35000 Rennes, France
| | - Grégoire Michaux
- University of Rennes, CNRS, IGDR (Institut de Génétique et de Développement de Rennes), UMR 6290, F-35000 Rennes, France
| | - Anne Pacquelet
- University of Rennes, CNRS, IGDR (Institut de Génétique et de Développement de Rennes), UMR 6290, F-35000 Rennes, France
| |
Collapse
|
7
|
Zhang N, Zhang H, Khan LA, Jafari G, Eun Y, Membreno E, Gobel V. The biosynthetic-secretory pathway, supplemented by recycling routes, determines epithelial membrane polarity. SCIENCE ADVANCES 2023; 9:eade4620. [PMID: 37379377 PMCID: PMC10306302 DOI: 10.1126/sciadv.ade4620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 05/24/2023] [Indexed: 06/30/2023]
Abstract
In prevailing epithelial polarity models, membrane-based polarity cues (e.g., the partitioning-defective PARs) position apicobasal cellular membrane domains. Intracellular vesicular trafficking expands these domains by sorting polarized cargo toward them. How the polarity cues themselves are polarized in epithelia and how sorting confers long-range apicobasal directionality to vesicles is still unclear. Here, a systems-based approach using two-tiered C. elegans genomics-genetics screens identifies trafficking molecules that are not implicated in apical sorting yet polarize apical membrane and PAR complex components. Live tracking of polarized membrane biogenesis indicates that the biosynthetic-secretory pathway, linked to recycling routes, is asymmetrically oriented toward the apical domain during this domain's biosynthesis, and that this directionality is regulated upstream of PARs and independent of polarized target membrane domains. This alternative mode of membrane polarization could offer solutions to open questions in current models of epithelial polarity and polarized trafficking.
Collapse
Affiliation(s)
- Nan Zhang
- Mucosal Immunology and Biology Research Center, Developmental Biology and Genetics Core, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA, USA
- Key Laboratory of Zoonosis Research by the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Hongjie Zhang
- Mucosal Immunology and Biology Research Center, Developmental Biology and Genetics Core, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA, USA
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Liakot A. Khan
- Mucosal Immunology and Biology Research Center, Developmental Biology and Genetics Core, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA, USA
| | - Gholamali Jafari
- Mucosal Immunology and Biology Research Center, Developmental Biology and Genetics Core, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA, USA
| | - Yong Eun
- Mucosal Immunology and Biology Research Center, Developmental Biology and Genetics Core, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA, USA
- Department of Medicine, NYC Health & Hospitals/Harlem, Columbia University, New York, NY, USA
| | - Edward Membreno
- Mucosal Immunology and Biology Research Center, Developmental Biology and Genetics Core, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA, USA
| | - Verena Gobel
- Mucosal Immunology and Biology Research Center, Developmental Biology and Genetics Core, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Hemha P, Chomphoo S, Polsan Y, Goto K, Watanabe M, Kondo H, Hipkaeo W. Discrete localization of phospholipase Cβ3 and diacylglycerol kinase ι along the renal proximal tubules of normal rat kidney and gentamicin-induced changes in their expression. Histochem Cell Biol 2023; 159:293-307. [PMID: 36478081 DOI: 10.1007/s00418-022-02166-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
Many signaling enzymes have multiple isozymes that are localized discretely at varying molecular levels in different compartments of cells where they play specific roles. In this study, among the various isozymes of phospholipase C (PLC) and diacylglycerol kinase (DGK), which work sequentially in the phosphoinositide cycle, both PLCβ3 and DGKι were found in renal brush-border microvilli, but found to replace each other along the proximal tubules: PLCβ3 in the proximal straight tubules (PST) of the outer stripe of the outer medulla (OSOM) and the medullary ray (MR), and DGKι in the proximal convoluted tubules (PCT) in the cortex and partially in the PST of the MR. Following daily injection of gentamicin for 1 week, the expression of PLCβ3 and DGKι was transiently enhanced, as demonstrated by western blot, and the increases were found to most likely occur in their original sites, that is, in the brush borders of the PST for PLCβ3 and in the PCT for DGKι. These findings showing differences in expression along the tubules suggest that the exertion of reabsorption and secretion through various ion channels and transporters in the microvillus membranes and the maintenance of microvillus turnover are regulated by a PLC-mediated signal with the balance shifted toward relative augmentation of the DAG function in the PST, and by a DGK-mediated signal with the balance shifted to relative augmentation of the phosphatidic acid function in the PCT. Our results also suggest the possibility that these isozymes are potential diagnostic signs for the early detection of acute kidney injury caused by gentamicin.
Collapse
Affiliation(s)
- Premrudee Hemha
- Electron Microscopy Laboratory, Division of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Surang Chomphoo
- Electron Microscopy Laboratory, Division of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Yada Polsan
- Electron Microscopy Laboratory, Division of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Kaoru Goto
- Department of Anatomy, School of Medicine, Yamagata University, Yamagata, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hisatake Kondo
- Electron Microscopy Laboratory, Division of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Wiphawi Hipkaeo
- Electron Microscopy Laboratory, Division of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
9
|
LIM CHANGHYUN, NUNES EVERSONA, CURRIER BRADS, MCLEOD JONATHANC, THOMAS AARONCQ, PHILLIPS STUARTM. An Evidence-Based Narrative Review of Mechanisms of Resistance Exercise-Induced Human Skeletal Muscle Hypertrophy. Med Sci Sports Exerc 2022; 54:1546-1559. [PMID: 35389932 PMCID: PMC9390238 DOI: 10.1249/mss.0000000000002929] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skeletal muscle plays a critical role in physical function and metabolic health. Muscle is a highly adaptable tissue that responds to resistance exercise (RE; loading) by hypertrophying, or during muscle disuse, RE mitigates muscle loss. Resistance exercise training (RET)-induced skeletal muscle hypertrophy is a product of external (e.g., RE programming, diet, some supplements) and internal variables (e.g., mechanotransduction, ribosomes, gene expression, satellite cells activity). RE is undeniably the most potent nonpharmacological external variable to stimulate the activation/suppression of internal variables linked to muscular hypertrophy or countering disuse-induced muscle loss. Here, we posit that despite considerable research on the impact of external variables on RET and hypertrophy, internal variables (i.e., inherent skeletal muscle biology) are dominant in regulating the extent of hypertrophy in response to external stimuli. Thus, identifying the key internal skeletal muscle-derived variables that mediate the translation of external RE variables will be pivotal to determining the most effective strategies for skeletal muscle hypertrophy in healthy persons. Such work will aid in enhancing function in clinical populations, slowing functional decline, and promoting physical mobility. We provide up-to-date, evidence-based perspectives of the mechanisms regulating RET-induced skeletal muscle hypertrophy.
Collapse
Affiliation(s)
- CHANGHYUN LIM
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - EVERSON A. NUNES
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
- Department of Physiological Science, Federal University of Santa Catarina, Florianópolis, Santa-Catarina, BRAZIL
| | - BRAD S. CURRIER
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - JONATHAN C. MCLEOD
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - AARON C. Q. THOMAS
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - STUART M. PHILLIPS
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| |
Collapse
|
10
|
Dubois F, Bazille C, Levallet J, Maille E, Brosseau S, Madelaine J, Bergot E, Zalcman G, Levallet G. Molecular Alterations in Malignant Pleural Mesothelioma: A Hope for Effective Treatment by Targeting YAP. Target Oncol 2022; 17:407-431. [PMID: 35906513 PMCID: PMC9345804 DOI: 10.1007/s11523-022-00900-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2022] [Indexed: 01/11/2023]
Abstract
Malignant pleural mesothelioma is a rare and aggressive neoplasm, which has primarily been attributed to the exposure to asbestos fibers (83% of cases); yet, despite a ban of using asbestos in many countries, the incidence of malignant pleural mesothelioma failed to decline worldwide. While little progress has been made in malignant pleural mesothelioma diagnosis, bevacizumab at first, then followed by double immunotherapy (nivolumab plus ipilumumab), were all shown to improve survival in large phase III randomized trials. The morphological analysis of the histological subtyping remains the primary indicator for therapeutic decision making at an advanced disease stage, while a platinum-based chemotherapy regimen combined with pemetrexed, either with or without bevacizumab, is still the main treatment option. Consequently, malignant pleural mesothelioma still represents a significant health concern owing to poor median survival (12-18 months). Given this context, both diagnosis and therapy improvements require better knowledge of the molecular mechanisms underlying malignant pleural mesothelioma's carcinogenesis and progression. Hence, the Hippo pathway in malignant pleural mesothelioma initiation and progression has recently received increasing attention, as the aberrant expression of its core components may be closely related to patient prognosis. The purpose of this review was to provide a critical analysis of our current knowledge on these topics, the main focus being on the available evidence concerning the role of each Hippo pathway's member as a promising biomarker, enabling detection of the disease at earlier stages and thus improving prognosis.
Collapse
Affiliation(s)
- Fatéméh Dubois
- Normandie University, UNICAEN, CNRS, ISTCT Unit, Avenue H. Becquerel, 14074, Caen, France
- Department of Pathology, CHU de Caen, Caen, France
- Federative Structure of Cyto-Molecular Oncogenetics (SF-MOCAE), CHU de Caen, Caen, France
| | - Céline Bazille
- Normandie University, UNICAEN, CNRS, ISTCT Unit, Avenue H. Becquerel, 14074, Caen, France
- Department of Pathology, CHU de Caen, Caen, France
| | - Jérôme Levallet
- Normandie University, UNICAEN, CNRS, ISTCT Unit, Avenue H. Becquerel, 14074, Caen, France
| | - Elodie Maille
- Normandie University, UNICAEN, CNRS, ISTCT Unit, Avenue H. Becquerel, 14074, Caen, France
| | - Solenn Brosseau
- Department of Thoracic Oncology and CIC1425, Hospital Bichat-Claude Bernard, Assistance Publique Hôpitaux de Paris, Université Paris-Diderot, Paris, France
- U830 INSERM "Genetics and Biology of Cancers, A.R.T Group", Curie Institute, Paris, France
| | - Jeannick Madelaine
- Department of Pulmonology and Thoracic Oncology, CHU de Caen, Caen, France
| | - Emmanuel Bergot
- Normandie University, UNICAEN, CNRS, ISTCT Unit, Avenue H. Becquerel, 14074, Caen, France
- Department of Pulmonology and Thoracic Oncology, CHU de Caen, Caen, France
| | - Gérard Zalcman
- Department of Thoracic Oncology and CIC1425, Hospital Bichat-Claude Bernard, Assistance Publique Hôpitaux de Paris, Université Paris-Diderot, Paris, France
- U830 INSERM "Genetics and Biology of Cancers, A.R.T Group", Curie Institute, Paris, France
| | - Guénaëlle Levallet
- Normandie University, UNICAEN, CNRS, ISTCT Unit, Avenue H. Becquerel, 14074, Caen, France.
- Department of Pathology, CHU de Caen, Caen, France.
- Federative Structure of Cyto-Molecular Oncogenetics (SF-MOCAE), CHU de Caen, Caen, France.
| |
Collapse
|
11
|
Comprehensive Analysis of RAPGEF2 for Predicting Prognosis and Immunotherapy Response in Patients with Hepatocellular Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:6560154. [PMID: 35518785 PMCID: PMC9064514 DOI: 10.1155/2022/6560154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 03/30/2022] [Indexed: 11/17/2022]
Abstract
Background Hepatocellular carcinoma (HCC) is the sixth most common tumor worldwide. Additionally, deletion of RAPGEF2 plays a critical role in CNV and related to tumor immune microenvironment, whereas the prognostic potential of RAPGEF2 in HCC patient needs to be explored. Methods We looked for prognostic potential genes in HCC using a variety of R programs. Then, using the LASSO Cox regression, we thoroughly evaluated and integrated the RAPGEF2-related genes from TCGA database. Meanwhile, utilizing TCGA and ICGA databases, the link between RAPGEF2 and immunotherapy response in HCC was studied. In vivo, the effect of RAPGEF2 on tumor development and the capacity of natural killer (NK) cells to recruit were confirmed. To ascertain the connection between RAPGEF2-related genes and the prognosis of HCC, a prognostic model was created and validated. Result We demonstrated RAPGEF2 has a differential expression, and patients with deletion of RAPGEF2 gene get shorter survival in HCC. Additionally, the tissues without RAPGEF2 have a weaker ability to recruit the NK cells and response to immunotherapy. After that, we scoured the database for eight RAPGEF2-related genes linked with a better prognosis in HCC patients. Additionally, silencing RAPGEF2 accelerated tumor development in the HCC mouse model and decreased CD56+ NK cell recruitment in HCC tissues. TCGA database was used to classify patients into low- and high-risk categories based on the expression of related genes. Patients in the low-risk group had a significantly greater overall survival than those in the high-risk group (P < 0.001). Meanwhile, the low-risk group demonstrated connections with the NK cell and immunotherapy response. Finally, the prognostic nomogram showed a high sensitivity and specificity for predicting the survival of HCC patients at 1, 2, and 3 years. Conclusion The prognostic model based on RAPGEF2 and RAPGEF2-related genes showed an excellent predictive performance in terms of prognosis and immunotherapy response in HCC, therefore establishing a unique prognostic model for clinical assessment of HCC patients.
Collapse
|
12
|
Duncan ED, Han KJ, Trout MA, Prekeris R. Ubiquitylation by Rab40b/Cul5 regulates Rap2 localization and activity during cell migration. J Cell Biol 2022; 221:213068. [PMID: 35293963 PMCID: PMC8931537 DOI: 10.1083/jcb.202107114] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/08/2021] [Accepted: 02/01/2022] [Indexed: 02/07/2023] Open
Abstract
Cell migration is a complex process that involves coordinated changes in membrane transport and actin cytoskeleton dynamics. Ras-like small monomeric GTPases, such as Rap2, play a key role in regulating actin cytoskeleton dynamics and cell adhesions. However, how Rap2 function, localization, and activation are regulated during cell migration is not fully understood. We previously identified the small GTPase Rab40b as a regulator of breast cancer cell migration. Rab40b contains a suppressor of cytokine signaling (SOCS) box, which facilitates binding to Cullin5, a known E3 ubiquitin ligase component responsible for protein ubiquitylation. In this study, we show that the Rab40b/Cullin5 complex ubiquitylates Rap2. Importantly, we demonstrate that ubiquitylation regulates Rap2 activation as well as recycling of Rap2 from the endolysosomal compartment to the lamellipodia of migrating breast cancer cells. Based on these data, we propose that Rab40b/Cullin5 ubiquitylates and regulates Rap2-dependent actin dynamics at the leading edge, a process that is required for breast cancer cell migration and invasion.
Collapse
Affiliation(s)
- Emily D Duncan
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Ke-Jun Han
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Margaret A Trout
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
13
|
Bugda Gwilt K, Thiagarajah JR. Membrane Lipids in Epithelial Polarity: Sorting out the PIPs. Front Cell Dev Biol 2022; 10:893960. [PMID: 35712665 PMCID: PMC9197455 DOI: 10.3389/fcell.2022.893960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
The development of cell polarity in epithelia, is critical for tissue morphogenesis and vectorial transport between the environment and the underlying tissue. Epithelial polarity is defined by the development of distinct plasma membrane domains: the apical membrane interfacing with the exterior lumen compartment, and the basolateral membrane directly contacting the underlying tissue. The de novo generation of polarity is a tightly regulated process, both spatially and temporally, involving changes in the distribution of plasma membrane lipids, localization of apical and basolateral membrane proteins, and vesicular trafficking. Historically, the process of epithelial polarity has been primarily described in relation to the localization and function of protein 'polarity complexes.' However, a critical and foundational role is emerging for plasma membrane lipids, and in particular phosphoinositide species. Here, we broadly review the evidence for a primary role for membrane lipids in the generation of epithelial polarity and highlight key areas requiring further research. We discuss the complex interchange that exists between lipid species and briefly examine how major membrane lipid constituents are generated and intersect with vesicular trafficking to be preferentially localized to different membrane domains with a focus on some of the key protein-enzyme complexes involved in these processes.
Collapse
Affiliation(s)
- Katlynn Bugda Gwilt
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Jay R Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
14
|
Honerlagen H, Reyer H, Oster M, Ponsuksili S, Trakooljul N, Kuhla B, Reinsch N, Wimmers K. Identification of Genomic Regions Influencing N-Metabolism and N-Excretion in Lactating Holstein- Friesians. Front Genet 2021; 12:699550. [PMID: 34335696 PMCID: PMC8318802 DOI: 10.3389/fgene.2021.699550] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/18/2021] [Indexed: 12/03/2022] Open
Abstract
Excreted nitrogen (N) of dairy cows contribute to environmental eutrophication. The main N-excretory metabolite of dairy cows is urea, which is synthesized as a result of N-metabolization in the liver and is excreted via milk and urine. Genetic variation in milk urea (MU) has been postulated but the complex physiology behind the trait as well as the tremendous diversity of processes regulating the N-metabolism impede the consistent determination of causal regions in the bovine genome. In order to map the genetic determinants affecting N-excretion, MU and eight other N-excretory metabolites in milk and urine were assessed in a genome-wide association study. Therefore phenotypes of 371 Holstein- Friesians were obtained in a trial on a dairy farm under near commercial conditions. Genotype data comprised SNP information of the Bovine 50K MD Genome chip (45,613 SNPs). Significantly associated genomic regions for MU concentration revealed GJA1 (BTA 9), RXFP1, and FRY1 (both BTA 12) as putative candidates. For milk urea yield (MUY) a promising QTL on BTA 17 including SH3D19 emerged, whereas RCAN2, CLIC5, ENPP4, and ENPP5 (BTA 23) are suggested to influence urinary urea concentration. Minor N-fractions in milk (MN) may be regulated by ELF2 and SLC7A11 (BTA 17), whilst ITPR2 and MYBPC1 (BTA 5), STIM2 (BTA 6), SGCD (BTA 7), SLC6A2 (BTA 18), TMCC2 and MFSD4A (BTA 16) are suggested to have an impact on various non-urea-N (NUN) fractions excreted via urine. Our results highlight genomic regions and candidate genes for N-excretory metabolites and provide a deeper insight into the predisposed component to regulate the N-metabolism in dairy cows.
Collapse
Affiliation(s)
- Hanne Honerlagen
- Genomics Unit, Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Henry Reyer
- Genomics Unit, Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Michael Oster
- Genomics Unit, Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Siriluck Ponsuksili
- Genomics Unit, Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Nares Trakooljul
- Genomics Unit, Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Björn Kuhla
- Metabolism Efficiency Unit, Institute of Nutritional Physiology "Oskar Kellner," Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Norbert Reinsch
- Livestock Genetics and Breeding Unit, Institute of Genetics and Biometry, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Klaus Wimmers
- Genomics Unit, Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany.,Faculty of Agricultural and Environmental Sciences, University of Rostock, Rostock, Germany
| |
Collapse
|
15
|
Zaman R, Lombardo A, Sauvanet C, Viswanatha R, Awad V, Bonomo LER, McDermitt D, Bretscher A. Effector-mediated ERM activation locally inhibits RhoA activity to shape the apical cell domain. J Cell Biol 2021; 220:211973. [PMID: 33836044 PMCID: PMC8185690 DOI: 10.1083/jcb.202007146] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/07/2021] [Accepted: 03/09/2021] [Indexed: 12/21/2022] Open
Abstract
Activated ezrin-radixin-moesin (ERM) proteins link the plasma membrane to the actin cytoskeleton to generate apical structures, including microvilli. Among many kinases implicated in ERM activation are the homologues LOK and SLK. CRISPR/Cas9 was used to knock out all ERM proteins or LOK/SLK in human cells. LOK/SLK knockout eliminates all ERM-activating phosphorylation. The apical domains of cells lacking LOK/SLK or ERMs are strikingly similar and selectively altered, with loss of microvilli and with junctional actin replaced by ectopic myosin-II–containing apical contractile structures. Constitutively active ezrin can reverse the phenotypes of either ERM or LOK/SLK knockouts, indicating that a central function of LOK/SLK is to activate ERMs. Both knockout lines have elevated active RhoA with concomitant enhanced myosin light chain phosphorylation, revealing that active ERMs are negative regulators of RhoA. As RhoA-GTP activates LOK/SLK to activate ERM proteins, the ability of active ERMs to negatively regulate RhoA-GTP represents a novel local feedback loop necessary for the proper apical morphology of epithelial cells.
Collapse
Affiliation(s)
- Riasat Zaman
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Andrew Lombardo
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Cécile Sauvanet
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Raghuvir Viswanatha
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Valerie Awad
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Locke Ezra-Ros Bonomo
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - David McDermitt
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Anthony Bretscher
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| |
Collapse
|
16
|
Song Y, Ma X, Zhang M, Wang M, Wang G, Ye Y, Xia W. Ezrin Mediates Invasion and Metastasis in Tumorigenesis: A Review. Front Cell Dev Biol 2020; 8:588801. [PMID: 33240887 PMCID: PMC7683424 DOI: 10.3389/fcell.2020.588801] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/21/2020] [Indexed: 12/13/2022] Open
Abstract
Ezrin, as encoded by the EZR gene, is a member of the Ezrin/Radixin/Moesin (ERM) family. The ERM family includes three highly related actin filament binding proteins, Ezrin, Radixin, and Moesin. These three members share similar structural properties containing an N-terminal domain named FERM, a central helical linker region, and a C-terminal domain that mediates the interaction with F-actin. Ezrin protein is highly regulated through the conformational change between a closed, inactivate form and an open, active form. As a membrane-cytoskeleton linker protein, Ezrin facilitates numerous signal transductions in tumorigenesis and mediates diverse essential functions through interactions with a variety of growth factor receptors and adhesion molecules. Emerging evidence has demonstrated that Ezrin is an oncogene protein, as high levels of Ezrin are associated with metastatic behavior in various types of cancer. The diverse functions attributed to Ezrin and the understanding of how Ezrin drives the deadly process of metastasis are complex and often controversial. Here by reviewing recent findings across a wide spectrum of cancer types we will highlight the structures, protein interactions and oncogenic roles of Ezrin as well as the emerging therapeutic agents targeting Ezrin. This review provides a comprehensive framework to guide future studies of Ezrin and other ERM proteins in basic and clinical studies.
Collapse
Affiliation(s)
- Yanan Song
- Central Laboratory, The Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaokun Ma
- Department of Nuclear Medicine, The Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Miao Zhang
- Central Laboratory, The Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Menghan Wang
- Department of Nuclear Medicine, The Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guoyu Wang
- Department of Nuclear Medicine, The Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Ye
- Central Laboratory, The Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Xia
- Department of Nuclear Medicine, The Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
17
|
Krahn MP. Phospholipids of the Plasma Membrane - Regulators or Consequence of Cell Polarity? Front Cell Dev Biol 2020; 8:277. [PMID: 32411703 PMCID: PMC7198698 DOI: 10.3389/fcell.2020.00277] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/31/2020] [Indexed: 12/15/2022] Open
Abstract
Cell polarity is a key feature of many eukaryotic cells, including neurons, epithelia, endothelia and asymmetrically dividing stem cells. Apart from the specific localization of proteins to distinct domains of the plasma membrane, most of these cells exhibit an asymmetric distribution of phospholipids within the plasma membrane too. Notably, research over the last years has revealed that many known conserved regulators of apical-basal polarity in epithelial cells are capable of binding to phospholipids, which in turn regulate the localization and to some extent the function of these proteins. Conversely, phospholipid-modifying enzymes are recruited and controlled by polarity regulators, demonstrating an elaborated balance between asymmetrically localized proteins and phospholipids, which are enriched in certain (micro)domains of the plasma membrane. In this review, we will focus on our current understanding of apical-basal polarity and the implication of phospholipids within the plasma membrane during the cell polarization of epithelia and migrating cells.
Collapse
Affiliation(s)
- Michael P. Krahn
- Department of Medical Cell Biology, Medical Clinic D, University Hospital of Münster, Münster, Germany
| |
Collapse
|
18
|
Luan D, Zhang Y, Yuan L, Chu Z, Ma L, Xu Y, Zhao S. MST4 modulates the neuro-inflammatory response by regulating IκBα signaling pathway and affects the early outcome of experimental ischemic stroke in mice. Brain Res Bull 2020; 154:43-50. [DOI: 10.1016/j.brainresbull.2019.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/16/2019] [Accepted: 10/26/2019] [Indexed: 10/25/2022]
|
19
|
Abstract
The Hippo pathway was initially discovered in Drosophila melanogaster as a key regulator of tissue growth. It is an evolutionarily conserved signaling cascade regulating numerous biological processes, including cell growth and fate decision, organ size control, and regeneration. The core of the Hippo pathway in mammals consists of a kinase cascade, MST1/2 and LATS1/2, as well as downstream effectors, transcriptional coactivators YAP and TAZ. These core components of the Hippo pathway control transcriptional programs involved in cell proliferation, survival, mobility, stemness, and differentiation. The Hippo pathway is tightly regulated by both intrinsic and extrinsic signals, such as mechanical force, cell-cell contact, polarity, energy status, stress, and many diffusible hormonal factors, the majority of which act through G protein-coupled receptors. Here, we review the current understanding of molecular mechanisms by which signals regulate the Hippo pathway with an emphasis on mechanotransduction and the effects of this pathway on basic biology and human diseases.
Collapse
Affiliation(s)
- Shenghong Ma
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA; , , ,
| | - Zhipeng Meng
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA; , , ,
| | - Rui Chen
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA; , , ,
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA; , , ,
| |
Collapse
|
20
|
Zhang J, Wei Y, Min J, Wang Y, Yin L, Cao G, Shen H. Knockdown of RAP2A gene expression suppresses cisplatin resistance in gastric cancer cells. Oncol Lett 2019; 19:350-358. [PMID: 31897147 PMCID: PMC6923840 DOI: 10.3892/ol.2019.11086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 07/09/2019] [Indexed: 01/05/2023] Open
Abstract
Cisplatin (DDP) resistance is closely associated with the failure of chemotherapy to manage various different types of human cancer. The GTPase protein Ras-related protein Rap-2a (RAP2A) regulates cancer cell proliferation, migration and invasion; however, little is currently known regarding its role in cancer cell resistance to chemotherapy. The present study investigated the potential roles of the RAP2A gene in gastric cancer cell resistance to DDP treatment. The DDP half maximal inhibitory concentration (IC50) values for the proliferation inhibition of MGC803 and MGC803/DDP gastric cancer cells were determined by treating the cells with a DDP concentration gradient and measuring their survival rates using the Cell Counting Kit-8 (CCK-8) assay; cell viability was also assessed using the CCK-8 assay. Cell migration and invasion were assessed using Transwell Matrigel assays, and apoptosis and DNA damage were evaluated using flow cytometry and Hoechst staining. RAP2A expression was knocked down by siRNA transfection, and RAP2A protein levels were examined using western blotting. The DDP IC50 values for DDP-resistant MGC803/DDP cells were greater than those for MGC803 cells. Furthermore, MGC803/DDP cells exhibited increased levels of viability, migration and invasion, and decreased levels of apoptosis and DNA damage during DDP treatment. Knockdown of RAP2A expression significantly promoted MGC803/DDP cell apoptosis and DNA damage, and decreased the viability and invasion capabilities of these cells following treatment with DDP. The results of the present study revealed that RAP2A expression promotes DDP resistance in gastric cancer cells by increasing their viability, migration and invasion capacities, and by suppressing apoptosis and DNA damage.
Collapse
Affiliation(s)
- Jinyu Zhang
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Yunhai Wei
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Jie Min
- Intensive Care Unit, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Yan Wang
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Lei Yin
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Guoliang Cao
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Hua Shen
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| |
Collapse
|
21
|
Faust JJ, Millis BA, Tyska MJ. Profilin-Mediated Actin Allocation Regulates the Growth of Epithelial Microvilli. Curr Biol 2019; 29:3457-3465.e3. [PMID: 31607529 DOI: 10.1016/j.cub.2019.08.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/17/2019] [Accepted: 08/20/2019] [Indexed: 01/18/2023]
Abstract
Transporting epithelial cells, like those that line the intestinal tract, are specialized for solute processing and uptake. One defining feature is the brush border, an array of microvilli that serves to amplify apical membrane surface area and increase functional capacity. During differentiation, upon exit from stem-cell-containing crypts, enterocytes build thousands of microvilli, each supported by a parallel bundle of actin filaments several microns in length. Given the high concentration of actin residing in mature brush borders, we sought to determine whether enterocytes were resource (i.e., actin monomer) limited in assembling this domain. To examine this possibility, we inhibited Arp2/3, the ubiquitous branched actin nucleator, to increase G-actin availability during brush border assembly. In native intestinal tissues, Arp2/3 inhibition led to increased microvilli length on the surface of crypt, but not villus, enterocytes. In a cell culture model of brush border assembly, Arp2/3 inhibition accelerated the growth and increased the length of microvilli; it also led to a redistribution of F-actin from cortical lateral networks into the brush border. Effects on brush border growth were rescued by treatment with the G-actin sequestering drug, latrunculin A. G-actin binding protein, profilin-1, colocalized in the terminal web with G-actin, and knockdown of this factor compromised brush border growth in a concentration-dependent manner. Finally, the acceleration in brush border assembly induced by Arp2/3 inhibition was abrogated by profilin-1 knockdown. Thus, brush border assembly is limited by G-actin availability, and profilin-1 directs unallocated actin monomers into microvillar core bundles during enterocyte differentiation.
Collapse
Affiliation(s)
- James J Faust
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Bryan A Millis
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Biomedical Engineering, Vanderbilt University School of Engineering, Nashville, TN 37232, USA; Cell Imaging Shared Resource, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Biophotonics Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
22
|
Dasgupta I, McCollum D. Control of cellular responses to mechanical cues through YAP/TAZ regulation. J Biol Chem 2019; 294:17693-17706. [PMID: 31594864 DOI: 10.1074/jbc.rev119.007963] [Citation(s) in RCA: 201] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To perceive their three-dimensional environment, cells and tissues must be able to sense and interpret various physical forces like shear, tensile, and compression stress. These forces can be generated both internally and externally in response to physical properties, like substrate stiffness, cell contractility, and forces generated by adjacent cells. Mechanical cues have important roles in cell fate decisions regarding proliferation, survival, and differentiation as well as the processes of tissue regeneration and wound repair. Aberrant remodeling of the extracellular space and/or defects in properly responding to mechanical cues likely contributes to various disease states, such as fibrosis, muscle diseases, and cancer. Mechanotransduction involves the sensing and translation of mechanical forces into biochemical signals, like activation of specific genes and signaling cascades that enable cells to adapt to their physical environment. The signaling pathways involved in mechanical signaling are highly complex, but numerous studies have highlighted a central role for the Hippo pathway and other signaling networks in regulating the YAP and TAZ (YAP/TAZ) proteins to mediate the effects of mechanical stimuli on cellular behavior. How mechanical cues control YAP/TAZ has been poorly understood. However, rapid progress in the last few years is beginning to reveal a surprisingly diverse set of pathways for controlling YAP/TAZ. In this review, we will focus on how mechanical perturbations are sensed through changes in the actin cytoskeleton and mechanosensors at focal adhesions, adherens junctions, and the nuclear envelope to regulate YAP/TAZ.
Collapse
Affiliation(s)
- Ishani Dasgupta
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Dannel McCollum
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
23
|
Abstract
In this issue of Molecular Cell, Han et al. (2018) show that phosphatidic acid inhibits the action of the LATS kinases, key mediators of Hippo pathway signaling, which leads to an increase in the nuclear activity of the transcriptional regulator YAP.
Collapse
Affiliation(s)
- Eleni Stampouloglou
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
24
|
Abstract
Mechanical cues can regulate cell proliferation and differentiation through the Hippo-YAP signaling network. Reporting in Nature, Meng et al. (2018) show that the Ras-related GTPase RAP2 connects extracellular matrix stiffness to Hippo pathway regulation, adding to our understanding of how mechanical cues are converted into changes in YAP activity.
Collapse
|
25
|
Polarized Organization of the Cytoskeleton: Regulation by Cell Polarity Proteins. J Mol Biol 2018; 430:3565-3584. [DOI: 10.1016/j.jmb.2018.06.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/09/2018] [Accepted: 06/13/2018] [Indexed: 01/02/2023]
|
26
|
Chen M, Zhang H, Shi Z, Li Y, Zhang X, Gao Z, Zhou L, Ma J, Xu Q, Guan J, Cheng Y, Jiao S, Zhou Z. The MST4-MOB4 complex disrupts the MST1-MOB1 complex in the Hippo-YAP pathway and plays a pro-oncogenic role in pancreatic cancer. J Biol Chem 2018; 293:14455-14469. [PMID: 30072378 DOI: 10.1074/jbc.ra118.003279] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/19/2018] [Indexed: 01/07/2023] Open
Abstract
The mammalian STE20-like protein kinase 1 (MST1)-MOB kinase activator 1 (MOB1) complex has been shown to suppress the oncogenic activity of Yes-associated protein (YAP) in the mammalian Hippo pathway, which is involved in the development of multiple tumors, including pancreatic cancer (PC). However, it remains unclear whether other MST-MOB complexes are also involved in regulating Hippo-YAP signaling and have potential roles in PC. Here, we report that mammalian STE20-like kinase 4 (MST4), a distantly related ortholog of the MST1 kinase, forms a complex with MOB4 in a phosphorylation-dependent manner. We found that the overall structure of the MST4-MOB4 complex resembles that of the MST1-MOB1 complex, even though the two complexes exhibited opposite biological functions in PC. In contrast to the tumor-suppressor effect of the MST1-MOB1 complex, the MST4-MOB4 complex promoted growth and migration of PANC-1 cells. Moreover, expression levels of MST4 and MOB4 were elevated in PC and were positively correlated with each other, whereas MST1 expression was down-regulated. Because of divergent evolution of key interface residues, MST4 and MOB4 could disrupt assembly of the MST1-MOB1 complex through alternative pairing and thereby increased YAP activity. Collectively, these findings identify the MST4-MOB4 complex as a noncanonical regulator of the Hippo-YAP pathway with an oncogenic role in PC. Our findings highlight that although MST-MOB complexes display some structural conservation, they functionally diverged during their evolution.
Collapse
Affiliation(s)
- Min Chen
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031
| | - Hui Zhang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031
| | - Zhubing Shi
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031
| | - Yehua Li
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031
| | - Xiaoman Zhang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031
| | - Ziyang Gao
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031
| | - Li Zhou
- the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, and
| | - Jian Ma
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031
| | - Qi Xu
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031
| | - Jingmin Guan
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031
| | - Yunfeng Cheng
- the Department of Hematology and Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shi Jiao
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031,
| | - Zhaocai Zhou
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, .,the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, and
| |
Collapse
|
27
|
Meng Z, Qiu Y, Lin KC, Kumar A, Placone JK, Fang C, Wang KC, Lu S, Pan M, Hong AW, Moroishi T, Luo M, Plouffe SW, Diao Y, Ye Z, Park HW, Wang X, Yu FX, Chien S, Wang CY, Ren B, Engler AJ, Guan KL. RAP2 mediates mechanoresponses of the Hippo pathway. Nature 2018; 560:655-660. [PMID: 30135582 PMCID: PMC6128698 DOI: 10.1038/s41586-018-0444-0] [Citation(s) in RCA: 260] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/12/2018] [Indexed: 02/05/2023]
Abstract
Mammalian cells are surrounded by neighbouring cells and extracellular matrix (ECM), which provide cells with structural support and mechanical cues that influence diverse biological processes1. The Hippo pathway effectors YAP (also known as YAP1) and TAZ (also known as WWTR1) are regulated by mechanical cues and mediate cellular responses to ECM stiffness2,3. Here we identified the Ras-related GTPase RAP2 as a key intracellular signal transducer that relays ECM rigidity signals to control mechanosensitive cellular activities through YAP and TAZ. RAP2 is activated by low ECM stiffness, and deletion of RAP2 blocks the regulation of YAP and TAZ by stiffness signals and promotes aberrant cell growth. Mechanistically, matrix stiffness acts through phospholipase Cγ1 (PLCγ1) to influence levels of phosphatidylinositol 4,5-bisphosphate and phosphatidic acid, which activates RAP2 through PDZGEF1 and PDZGEF2 (also known as RAPGEF2 and RAPGEF6). At low stiffness, active RAP2 binds to and stimulates MAP4K4, MAP4K6, MAP4K7 and ARHGAP29, resulting in activation of LATS1 and LATS2 and inhibition of YAP and TAZ. RAP2, YAP and TAZ have pivotal roles in mechanoregulated transcription, as deletion of YAP and TAZ abolishes the ECM stiffness-responsive transcriptome. Our findings show that RAP2 is a molecular switch in mechanotransduction, thereby defining a mechanosignalling pathway from ECM stiffness to the nucleus.
Collapse
Affiliation(s)
- Zhipeng Meng
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Yunjiang Qiu
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
- Bioinformatics and Systems Biology Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Kimberly C Lin
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Aditya Kumar
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Jesse K Placone
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Cao Fang
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Kuei-Chun Wang
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Institute of Engineering in Medicine, University of California San Diego, La Jolla, CA, USA
| | - Shicong Lu
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Margaret Pan
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Audrey W Hong
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Toshiro Moroishi
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Enzymology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Min Luo
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Steven W Plouffe
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Yarui Diao
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Zhen Ye
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Hyun Woo Park
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, South Korea
| | - Xiaoqiong Wang
- Robert J. Tomisch Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Fa-Xing Yu
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shu Chien
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Institute of Engineering in Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cun-Yu Wang
- Division of Oral Biology and Medicine, School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Bing Ren
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Adam J Engler
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
28
|
Chen X, Shibata AC, Hendi A, Kurashina M, Fortes E, Weilinger NL, MacVicar BA, Murakoshi H, Mizumoto K. Rap2 and TNIK control Plexin-dependent tiled synaptic innervation in C. elegans. eLife 2018; 7:38801. [PMID: 30063210 PMCID: PMC6067881 DOI: 10.7554/elife.38801] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/11/2018] [Indexed: 12/22/2022] Open
Abstract
During development, neurons form synapses with their fate-determined targets. While we begin to elucidate the mechanisms by which extracellular ligand-receptor interactions enhance synapse specificity by inhibiting synaptogenesis, our knowledge about their intracellular mechanisms remains limited. Here we show that Rap2 GTPase (rap-2) and its effector, TNIK (mig-15), act genetically downstream of Plexin (plx-1) to restrict presynaptic assembly and to form tiled synaptic innervation in C. elegans. Both constitutively GTP- and GDP-forms of rap-2 mutants exhibit synaptic tiling defects as plx-1 mutants, suggesting that cycling of the RAP-2 nucleotide state is critical for synapse inhibition. Consistently, PLX-1 suppresses local RAP-2 activity. Excessive ectopic synapse formation in mig-15 mutants causes a severe synaptic tiling defect. Conversely, overexpression of mig-15 strongly inhibited synapse formation, suggesting that mig-15 is a negative regulator of synapse formation. These results reveal that subcellular regulation of small GTPase activity by Plexin shapes proper synapse patterning in vivo. Genes do more than just direct the color of our hair or eyes. They produce proteins that are involved in almost every process in the body. In humans, the majority of active genes can be found in the brain, where they help it to develop and work properly – effectively controlling how we move and behave. The brain’s functional units, the nerve cells or neurons, communicate with each other by releasing messenger molecules in the gap between them, the synapse. These molecules are then picked up from specific receptor proteins of the receiving neuron. In the nervous system, neurons only form synapses with the cells they need to connect with, even though they are surrounded by many more cells. This implies that they use specific mechanisms to stop neurons from forming synapses with incorrect target cells. This is important, because if too many synapses were present or if synapses formed with incorrect target cells, it would compromise the information flow in the nervous system. This would ultimately lead to various neurological conditions, including Autism Spectrum Disorder. In 2013, researchers found that in the roundworm Caenorhabditis elegans, a receptor protein called Plexin, is located at the surface of the neurons and can inhibit the formation of nearby synapses. Now, Chen et al. – including one author involved in the previous research – wanted to find out what genes Plexin manipulates when it stops synapses from growing. Knowing what each of those genes does can help us understand how neurons can inhibit synapses. The results revealed that Plexin appears to regulate two genes, Rap2 and TNIK. Plexin reduced the activity of Rap2 in the neuron that released the messenger, which hindered the formation of synapses. The gene TNIK and its protein on the other hand, have the ability to modify other proteins and could so inhibit the growth of synapses. When TNIK was experimentally removed, the number of synapses increased, but when its activity was increased, the number of synapses was strongly reduced. These findings could help scientists understand how mutations in Rap2 or TNIK can lead to various neurological conditions. A next step will be to test if these genes also affect the formation of synapses in other species such as mice, which have a more complex nervous system that is structurally and functionally more similar to that of humans.
Collapse
Affiliation(s)
- Xi Chen
- Department of Zoology, The University of British Columbia, Vancouver, Canada
| | - Akihiro Ce Shibata
- Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Japan
| | - Ardalan Hendi
- Department of Zoology, The University of British Columbia, Vancouver, Canada
| | - Mizuki Kurashina
- Department of Zoology, The University of British Columbia, Vancouver, Canada
| | - Ethan Fortes
- Department of Zoology, The University of British Columbia, Vancouver, Canada
| | | | - Brian A MacVicar
- Department of Psychiatry, The University of British Columbia, Vancouver, Canada
| | - Hideji Murakoshi
- Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Japan
| | - Kota Mizumoto
- Department of Zoology, The University of British Columbia, Vancouver, Canada
| |
Collapse
|
29
|
Wang JC, Lee JYJ, Dang-Lawson M, Pritchard C, Gold MR. The Rap2c GTPase facilitates B cell receptor-induced reorientation of the microtubule-organizing center. Small GTPases 2018; 11:402-412. [PMID: 29457987 DOI: 10.1080/21541248.2018.1441626] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
When B lymphocytes encounter antigen-bearing surfaces, B-cell receptor (BCR) signaling initiates remodeling of the F-actin network and reorientation of the microtubule-organizing center (MTOC) towards the antigen contact site. We have previously shown that the Rap1 GTPase, an evolutionarily conserved regulator of cell polarity, is essential for these processes and that Rap1-regulated actin remodeling is required for MTOC polarization. The role of Rap2 proteins in establishing cell polarity is not well understood. We now show that depleting Rap2c, the only Rap2 isoform expressed in the A20 B-cell line, impairs BCR-induced MTOC reorientation as well as the actin remodeling that supports MTOC polarization. Thus Rap1 and Rap2 proteins may have similar but non-redundant functions in coupling the BCR to MTOC polarization.
Collapse
Affiliation(s)
- Jia C Wang
- Department of Microbiology & Immunology and the Life Sciences Institute, University of British Columbia , Vancouver, BC, Canada
| | - Jeff Y-J Lee
- Department of Microbiology & Immunology and the Life Sciences Institute, University of British Columbia , Vancouver, BC, Canada
| | - May Dang-Lawson
- Department of Microbiology & Immunology and the Life Sciences Institute, University of British Columbia , Vancouver, BC, Canada
| | - Caitlin Pritchard
- Department of Microbiology & Immunology and the Life Sciences Institute, University of British Columbia , Vancouver, BC, Canada
| | - Michael R Gold
- Department of Microbiology & Immunology and the Life Sciences Institute, University of British Columbia , Vancouver, BC, Canada
| |
Collapse
|
30
|
Ito Y, Honda A, Igarashi M. Glycoprotein M6a as a signaling transducer in neuronal lipid rafts. Neurosci Res 2018; 128:19-24. [DOI: 10.1016/j.neures.2017.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023]
|
31
|
Schneeberger K, Roth S, Nieuwenhuis EES, Middendorp S. Intestinal epithelial cell polarity defects in disease: lessons from microvillus inclusion disease. Dis Model Mech 2018; 11:11/2/dmm031088. [PMID: 29590640 PMCID: PMC5894939 DOI: 10.1242/dmm.031088] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The intestinal epithelium is a highly organized tissue. The establishment of epithelial cell polarity, with distinct apical and basolateral plasma membrane domains, is pivotal for both barrier formation and for the uptake and vectorial transport of nutrients. The establishment of cell polarity requires a specialized subcellular machinery to transport and recycle proteins to their appropriate location. In order to understand and treat polarity-associated diseases, it is necessary to understand epithelial cell-specific trafficking mechanisms. In this Review, we focus on cell polarity in the adult mammalian intestine. We discuss how intestinal epithelial polarity is established and maintained, and how disturbances in the trafficking machinery can lead to a polarity-associated disorder, microvillus inclusion disease (MVID). Furthermore, we discuss the recent developments in studying MVID, including the creation of genetically manipulated cell lines, mouse models and intestinal organoids, and their uses in basic and applied research. Summary: Microvillus inclusion disease serves as a useful model to enhance our understanding of the intestinal trafficking and polarity machinery in health and disease.
Collapse
Affiliation(s)
- Kerstin Schneeberger
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands
| | - Sabrina Roth
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands
| | - Edward E S Nieuwenhuis
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands
| | - Sabine Middendorp
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands .,Regenerative Medicine Center Utrecht, University Medical Centre (UMC) Utrecht, 3584 CT, Utrecht, The Netherlands
| |
Collapse
|
32
|
Tonucci FM, Ferretti A, Almada E, Cribb P, Vena R, Hidalgo F, Favre C, Tyska MJ, Kaverina I, Larocca MC. Microtubules regulate brush border formation. J Cell Physiol 2018; 233:1468-1480. [PMID: 28548701 PMCID: PMC5673559 DOI: 10.1002/jcp.26033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/25/2017] [Indexed: 01/01/2023]
Abstract
Most epithelial cells contain apical membrane structures associated to bundles of actin filaments, which constitute the brush border. Whereas microtubule participation in the maintenance of the brush border identity has been characterized, their contribution to de novo microvilli organization remained elusive. Hereby, using a cell model of individual enterocyte polarization, we found that nocodazole induced microtubule depolymerization prevented the de novo brush border formation. Microtubule participation in brush border actin organization was confirmed in polarized kidney tubule MDCK cells. We also found that centrosome, but not Golgi derived microtubules, were essential for the initial stages of brush border development. During this process, microtubule plus ends acquired an early asymmetric orientation toward the apical membrane, which clearly differs from their predominant basal orientation in mature epithelia. In addition, overexpression of the microtubule plus ends associated protein CLIP170, which regulate actin nucleation in different cell contexts, facilitated brush border formation. In combination, the present results support the participation of centrosomal microtubule plus ends in the activation of the polarized actin organization associated to brush border formation, unveiling a novel mechanism of microtubule regulation of epithelial polarity.
Collapse
Affiliation(s)
- Facundo M. Tonucci
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Anabela Ferretti
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Evangelina Almada
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Pamela Cribb
- Instituto de Biología Molecular y Celular de Rosario, Consejo de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Rodrigo Vena
- Instituto de Biología Molecular y Celular de Rosario, Consejo de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Florencia Hidalgo
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Cristián Favre
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Matt J. Tyska
- Department of Cell and Developmental Biology, Epithelial Biology Center, Vanderbilt-Ingram Cancer Center and the Nashville VA Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Irina Kaverina
- Department of Cell and Developmental Biology, Epithelial Biology Center, Vanderbilt-Ingram Cancer Center and the Nashville VA Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Maria C. Larocca
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| |
Collapse
|
33
|
Abstract
Our laboratory has studied Ras and Ras-like proteins since the discovery of the Ras oncogene 35 years ago. In this review, I will give an account of what we have done in these 35 years and indicate the main papers that have guided our research. Our efforts started with the early analysis of mutant Ras in human tumors followed by deciphering of the role of Ras in signal transduction pathways. In an attempt to interfere in Ras signaling we turned to Rap proteins. These proteins are the closest relatives of Ras and were initially identified as Ras antagonists. However, our studies revealed that the Rap signaling network primarily is involved in spatiotemporal control of cell adhesion, in part through regulation of the actin cytoskeleton. More recently we returned to Ras, trying to interfere in Ras signaling by combinatorial drug testing using the organoid technology.
Collapse
Affiliation(s)
- Johannes L Bos
- Molecular Cancer Research, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
34
|
Liu X, Lu J, Liu Z, Zhao J, Sun H, Wu N, Liu H, Liu W, Hu Z, Meng G, Shen L, Miller AW, Su B, Li X, Kang Z. Intestinal Epithelial Cell-Derived LKB1 Suppresses Colitogenic Microbiota. THE JOURNAL OF IMMUNOLOGY 2018; 200:1889-1900. [PMID: 29352002 DOI: 10.4049/jimmunol.1700547] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 12/20/2017] [Indexed: 12/14/2022]
Abstract
Dysregulation of the immune barrier function of the intestinal epithelium can often result in dysbiosis. In this study we report a novel role of intestinal epithelial cell (IEC)-derived liver kinase B1 (LKB1) in suppressing colitogenic microbiota. IEC-specific deletion of LKB1 (LKB1ΔIEC) resulted in an increased susceptibility to dextran sodium sulfate (DSS)-induced colitis and a definitive shift in the composition of the microbial population in the mouse intestine. Importantly, transfer of the microbiota from LKB1ΔIEC mice was sufficient to confer increased susceptibility to DSS-induced colitis in wild-type recipient mice. Collectively, the data indicate that LKB1 deficiency in intestinal epithelial cells nurtures the outgrowth of colitogenic bacteria in the commensal community. In addition, LKB1 deficiency in the intestinal epithelium reduced the production of IL-18 and antimicrobial peptides in the colon. Administration of exogenous IL-18 restored the expression of antimicrobial peptides, corrected the outgrowth of several bacterial genera, and rescued the LKB1ΔIEC mice from increased sensitivity to DSS challenge. Taken together, our study reveals an important function of LKB1 in IECs for suppressing colitogenic microbiota by IL-18 expression.
Collapse
Affiliation(s)
- Xia'nan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jinfeng Lu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhenshan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Junjie Zhao
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195
| | - Hongxiang Sun
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ningbo Wu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hongzhi Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weiwei Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhuqin Hu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guangxun Meng
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lei Shen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Aaron W Miller
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195.,Department of Urology, Cleveland Clinic, Cleveland, OH 44195; and
| | - Bing Su
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoxia Li
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| | - Zizhen Kang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; .,Department of Immunology, Cleveland Clinic, Cleveland, OH 44195.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| |
Collapse
|
35
|
Apodaca G. Role of Polarity Proteins in the Generation and Organization of Apical Surface Protrusions. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a027813. [PMID: 28264821 DOI: 10.1101/cshperspect.a027813] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protruding from the apical surfaces of epithelial cells are specialized structures, including cilia, microplicae, microvilli, and stereocilia. These contribute to epithelial function by cushioning the apical surface, by amplifying its surface area to facilitate nutrient absorption, and by promoting sensory transduction and barrier function. Despite these important roles, and the diseases that result when their formation is perturbed, there remain significant gaps in our understanding of the biogenesis of apical protrusions, or the pathways that promote their organization and orientation once at the apical surface. Here, I review some general aspects of these apical structures, and then discuss our current understanding of their formation and organization with respect to proteins that specify apicobasolateral polarity and planar cell polarity.
Collapse
Affiliation(s)
- Gerard Apodaca
- Department of Medicine Renal-Electrolyte Division and the Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
36
|
Engevik AC, Goldenring JR. Trafficking Ion Transporters to the Apical Membrane of Polarized Intestinal Enterocytes. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a027979. [PMID: 28264818 DOI: 10.1101/cshperspect.a027979] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Epithelial cells lining the gastrointestinal tract require distinct apical and basolateral domains to function properly. Trafficking and insertion of enzymes and transporters into the apical brush border of intestinal epithelial cells is essential for effective digestion and absorption of nutrients. Specific critical ion transporters are delivered to the apical brush border to facilitate fluid and electrolyte uptake. Maintenance of these apical transporters requires both targeted delivery and regulated membrane recycling. Examination of altered apical trafficking in patients with Microvillus Inclusion disease caused by inactivating mutations in MYO5B has led to insights into the regulation of apical trafficking by elements of the apical recycling system. Modeling of MYO5B loss in cell culture and animal models has led to recognition of Rab11a and Rab8a as critical regulators of apical brush border function. All of these studies show the importance of apical membrane trafficking dynamics in maintenance of polarized epithelial cell function.
Collapse
Affiliation(s)
- Amy Christine Engevik
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232.,Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232.,Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232.,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee 37232.,Nashville VA Medical Center, Nashville, Tennessee 37232
| |
Collapse
|
37
|
Abstract
The specificity of knowledge that Gene Ontology (GO) annotations currently can represent is still restricted by the legacy format of the GO annotation file, a format intentionally designed for simplicity to keep the barriers to entry low and thus encourage initial adoption. Historically, the information that could be captured in a GO annotation was simply the role or location of a gene product, although genetically interacting or binding partners could be specified. While there was no mechanism within the original GO annotation format for capturing additional information about the context of a GO term, such as the target gene of an activity or the location of a molecular function, the long-term vision for the GO Consortium was to provide greater expressivity in its annotations to capture physiologically relevant information.Thus, as a step forwards, the GO Consortium has introduced a new field into the annotation format, annotation extensions, which can be used to capture valuable contextual detail. This provides experimentally verified links between gene products and other physiological information that is crucial for accurate analysis of pathway and network data. This chapter will provide a simple overview of annotation extensions, illustrated with examples of their usage, and explain why they are useful for scientists and bioinformaticians alike.
Collapse
|
38
|
Yin K, Yin W, Wang Y, Zhou L, Liu Y, Yang G, Wang J, Lu J. MiR-206 suppresses epithelial mesenchymal transition by targeting TGF-β signaling in estrogen receptor positive breast cancer cells. Oncotarget 2017; 7:24537-48. [PMID: 27014911 PMCID: PMC5029720 DOI: 10.18632/oncotarget.8233] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 03/04/2016] [Indexed: 01/13/2023] Open
Abstract
Background: Previous reports have shown a mutual negative feedback loop between microRNA (miR)-206 and estrogen receptor (ER) expression. Furthermore, decreased miR-206 expression in breast cancer (BC) is associated with the advanced clinical stage and lymph node metastasis. However, its role and the mechanism underlying the migration and invasion of ER positive BC remain unclear. Results: In this study, miR-206 was stably transfected into ER positive cell lines MCF-7 and T47D to investigate the effect of miR-206. The results showed that miR-206 overexpression markedly impaired the migration and invasive abilities of these cells, followed by suppression of the epithelial mesenchymal transition (EMT). Mechanistic analyses showed that miR-206 inhibited the autocrine production of transforming growth factor (TGF)-β as well as the downstream expression of neuropilin-1 (NRP1) and SMAD2, responsible for the decreased migration, invasion, and EMT in these cells. Conclusions: Our data demonstrate that miR-206 inhibits TGF-β transcription and autocrine production, as well as downstream target genes of EMT. Restoring miR-206 expression may provide an effective therapeutic strategy for ER positive BC.
Collapse
Affiliation(s)
- Kai Yin
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenjin Yin
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yaohui Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liheng Zhou
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Gong Yang
- Breast Cancer Center, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jianhua Wang
- Breast Cancer Center, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jinsong Lu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
39
|
Larhammar M, Huntwork-Rodriguez S, Rudhard Y, Sengupta-Ghosh A, Lewcock JW. The Ste20 Family Kinases MAP4K4, MINK1, and TNIK Converge to Regulate Stress-Induced JNK Signaling in Neurons. J Neurosci 2017; 37:11074-11084. [PMID: 28993483 PMCID: PMC6596808 DOI: 10.1523/jneurosci.0905-17.2017] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 09/13/2017] [Accepted: 10/02/2017] [Indexed: 11/21/2022] Open
Abstract
The c-Jun-N-terminal kinase (JNK) signaling pathway regulates nervous system development, axon regeneration, and neuronal degeneration after acute injury or in chronic neurodegenerative disease. Dual leucine zipper kinase (DLK) is required for stress-induced JNK signaling in neurons, yet the factors that initiate DLK/JNK pathway activity remain poorly defined. In the present study, we identify the Ste20 kinases MAP4K4, misshapen-like kinase 1 (MINK1 or MAP4K6) and TNIK Traf2- and Nck-interacting kinase (TNIK or MAP4K7), as upstream regulators of DLK/JNK signaling in neurons. Using a trophic factor withdrawal-based model of neurodegeneration in both male and female embryonic mouse dorsal root ganglion neurons, we show that MAP4K4, MINK1, and TNIK act redundantly to regulate DLK activation and downstream JNK-dependent phosphorylation of c-Jun in response to stress. Targeting MAP4K4, MINK1, and TNIK, but not any of these kinases individually, is sufficient to protect neurons potently from degeneration. Pharmacological inhibition of MAP4Ks blocks stabilization and phosphorylation of DLK within axons and subsequent retrograde translocation of the JNK signaling complex to the nucleus. These results position MAP4Ks as important regulators of the DLK/JNK signaling pathway.SIGNIFICANCE STATEMENT Neuronal degeneration occurs in disparate circumstances: during development to refine neuronal connections, after injury to clear damaged neurons, or pathologically during disease. The dual leucine zipper kinase (DLK)/c-Jun-N-terminal kinase (JNK) pathway represents a conserved regulator of neuronal injury signaling that drives both neurodegeneration and axon regeneration, yet little is known about the factors that initiate DLK activity. Here, we uncover a novel role for a subfamily of MAP4 kinases consisting of MAP4K4, Traf2- and Nck-interacting kinase (TNIK or MAP4K7), and misshapen-like kinase 1 (MINK1 or MAP4K6) in regulating DLK/JNK signaling in neurons. Inhibition of these MAP4Ks blocks stress-induced retrograde JNK signaling and protects from neurodegeneration, suggesting that these kinases may represent attractive therapeutic targets.
Collapse
Affiliation(s)
- Martin Larhammar
- Department of Neuroscience, Genentech, Inc., San Francisco, California 94080
- Denali Therapeutics Inc., South San Francisco, California 94080
| | - Sarah Huntwork-Rodriguez
- Department of Neuroscience, Genentech, Inc., San Francisco, California 94080
- Denali Therapeutics Inc., South San Francisco, California 94080
| | - York Rudhard
- In Vitro Pharmacology, Evotec AG, Manfred Eigen Campus, 22419 Hamburg, Germany, and
| | | | - Joseph W Lewcock
- Department of Neuroscience, Genentech, Inc., San Francisco, California 94080,
- Denali Therapeutics Inc., South San Francisco, California 94080
| |
Collapse
|
40
|
Differential regulation of spermatogenic process by Lkb1 isoforms in mouse testis. Cell Death Dis 2017; 8:e3121. [PMID: 29022902 PMCID: PMC5682689 DOI: 10.1038/cddis.2017.527] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 07/28/2017] [Accepted: 09/04/2017] [Indexed: 01/27/2023]
Abstract
Liver serine/threonine kinase B1 (LKB1) is a tumor suppressor associated with the pathogenesis of Peutz-Jeghers syndrome. Affected males are at increased risk of developing Sertoli cell tumors and display defective spermatogenesis. Male mice lacking the short isoform (Lkb1S) of Lkb1 were sterile and exhibited abnormal spermiogenesis. In addition to the short isoform, the long isoform of Lkb1 (Lkb1L) is also expressed in testis; however, the requirement of the long isoform for fertility and the functional difference between the isoforms remain unknown. Herein, different from the spermiation failure reported in Lkb1S knockout mice, conditional deletion (cKO) of both isoforms of Lkb1 in germ cells resulted in male sterility stemming from defects in acrosome formation, as well as nuclear elongation and condensation during spermatid differentiation. Additionally, cKO mice showed a progressive germ cell loss that was never reported in mice with Lkb1S deletion. Further experiments revealed that the defect resulted from the failure of spermatogonial stem/progenitor cells (SPCs) maintenance. Although increased mTORC1 activity in postnatal cKO testes was consistent with a tendency toward germline stem cell differentiation, in vivo inhibition of the pathway by rapamycin treatment failed to rescue the phenotype. Concurrently, we detected a significant reduction of mitochondrial activity in Lkb1deficient SPCs. The results suggest that the regulation of LKB1 on SPCs' maintenance is associated with mitochondrial functions but not through the mTOR signaling pathway. In summary, our study supports different roles of Lkb1 isoforms in spermatogenesis with Lkb1L directing SPCs maintenance, and Lkb1L and Lkb1S coordinately regulating spermatid differentiation.
Collapse
|
41
|
Yano T, Kanoh H, Tamura A, Tsukita S. Apical cytoskeletons and junctional complexes as a combined system in epithelial cell sheets. Ann N Y Acad Sci 2017; 1405:32-43. [DOI: 10.1111/nyas.13432] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/12/2017] [Accepted: 06/14/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Tomoki Yano
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine; Osaka University; Osaka Japan
| | - Hatsuho Kanoh
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine; Osaka University; Osaka Japan
- Graduate School of Biostudies; Kyoto University; Kyoto Japan
| | - Atsushi Tamura
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine; Osaka University; Osaka Japan
| | - Sachiko Tsukita
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine; Osaka University; Osaka Japan
| |
Collapse
|
42
|
Ma LL, Shen L, Tong GH, Tang N, Luo Y, Guo LL, Hu CT, Huang YX, Huang G, Jing FY, Liu C, Li ZY, Zhou N, Yan QW, Lei Y, Zhu SJ, Cheng ZQ, Cao GW, Deng YJ, Ding YQ. Prohibitin, relocated to the front ends, can control the migration directionality of colorectal cancer cells. Oncotarget 2017; 8:76340-76356. [PMID: 29100316 PMCID: PMC5652710 DOI: 10.18632/oncotarget.19394] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/20/2017] [Indexed: 01/05/2023] Open
Abstract
Directional migration is a cost-effective movement allowing invasion and metastatic spread of cancer cells. Although migration related to cytoskeletal assembly and microenvironmental chemotaxis has been elucidated, little is known about interaction between extracellular and intracellular molecules for controlling the migrational directionality. A polarized expression of prohibitin (PHB) in the front ends of CRC cells favors metastasis and is correlated with poor prognosis for 545 CRC patients. A high level of vascular endothelial growth factor (VEGF) in the interstitial tissue of CRC patients is associated with metastasis. VEGF bound to its receptor, neuropilin-1, can stimulate the activation of cell division cycle 42, which recruits intra-mitochondrial PHB to the front end of a CRC cell. This intracellular relocation of PHB results in the polymerization and reorganization of filament actin extending to the front end of the cell. As a result, the migration directionality of CRC cells is targeted towards VEGF. Together, these findings identify PHB as a key modulator of directional migration of CRC cells and a target for metastasis.
Collapse
Affiliation(s)
- Li-Li Ma
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China.,Department of Cardiothoracic Surgery, Taishan City People's Hospital, Taishan 529200, China
| | - Lan Shen
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Gui-Hui Tong
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Na Tang
- Department of Pathology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen 518020, China
| | - Yang Luo
- Department of Urinary Surgery, Nanfang Hospital and the Fifth Affiliated Hospital of Southern Medical University, Guangzhou 510900, China
| | - Li-Li Guo
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Chun-Ting Hu
- Pathology Center, Shanghai General Hospital Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ying-Xin Huang
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Guan Huang
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China.,Department of Pathology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Fang-Yan Jing
- Department of Anorectal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chao Liu
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Zhuo-Yi Li
- Department of Cardiothoracic Surgery, Taishan City People's Hospital, Taishan 529200, China
| | - Na Zhou
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Qian-Wen Yan
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Yan Lei
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Shi-Jie Zhu
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Zhi-Qiang Cheng
- Department of Pathology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen 518020, China
| | - Guang-Wen Cao
- Department of Epidemiology, Second Military Medical University, Shanghai 200433, China
| | - Yong-Jian Deng
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Yan-Qing Ding
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| |
Collapse
|
43
|
Oh YS, Heo K, Kim EK, Jang JH, Bae SS, Park JB, Kim YH, Song M, Kim SR, Ryu SH, Kim IH, Suh PG. Dynamic relocalization of NHERF1 mediates chemotactic migration of ovarian cancer cells toward lysophosphatidic acid stimulation. Exp Mol Med 2017; 49:e351. [PMID: 28684865 PMCID: PMC5565956 DOI: 10.1038/emm.2017.88] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/28/2016] [Accepted: 01/09/2017] [Indexed: 01/05/2023] Open
Abstract
NHERF1/EBP50 (Na+/H+ exchanger regulating
factor 1; Ezrin-binding phosphoprotein of 50 kDa) organizes stable
protein complexes beneath the apical membrane of polar epithelial cells. By
contrast, in cancer cells without any fixed polarity, NHERF1 often localizes in
the cytoplasm. The regulation of cytoplasmic NHERF1 and its role in cancer
progression remain unclear. In this study, we found that, upon lysophosphatidic
acid (LPA) stimulation, cytoplasmic NHERF1 rapidly translocated to the plasma
membrane, and subsequently to cortical protrusion structures, of ovarian cancer
cells. This movement depended on direct binding of NHERF1 to C-terminally
phosphorylated ERM proteins (cpERMs). Moreover, NHERF1 depletion downregulated
cpERMs and further impaired cpERM-dependent remodeling of the cell cortex,
suggesting reciprocal regulation between these proteins. The LPA-induced protein
complex was highly enriched in migratory pseudopodia, whose formation was
impaired by overexpression of NHERF1 truncation mutants. Consistent with this,
NHERF1 depletion in various types of cancer cells abolished chemotactic cell
migration toward a LPA gradient. Taken together, our findings suggest that the
high dynamics of cytosolic NHERF1 provide cancer cells with a means of
controlling chemotactic migration. This capacity is likely to be essential for
ovarian cancer progression in tumor microenvironments containing LPA.
Collapse
Affiliation(s)
- Yong-Seok Oh
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Kyun Heo
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Eung-Kyun Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Jin-Hyeok Jang
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Sun Sik Bae
- MRC for Ischemic Tissue Regeneration, Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Jong Bae Park
- Research Institute, National Cancer Center, Goyang, Republic of Korea.,Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Yun Hee Kim
- Research Institute, National Cancer Center, Goyang, Republic of Korea.,Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Minseok Song
- Synaptic Circuit Plasticity Laboratory, Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Sang Ryong Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Institute of Life Science and Biotechnology, Kyungpook National University, Daegu, Republic of Korea
| | - Sung Ho Ryu
- Division of Molecular and Life Science, Department of Life Science, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - In-Hoo Kim
- Research Institute, National Cancer Center, Goyang, Republic of Korea.,Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Pann-Ghill Suh
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| |
Collapse
|
44
|
Shah B, Püschel AW. Regulation of Rap GTPases in mammalian neurons. Biol Chem 2017; 397:1055-69. [PMID: 27186679 DOI: 10.1515/hsz-2016-0165] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/06/2016] [Indexed: 12/15/2022]
Abstract
Small GTPases are central regulators of many cellular processes. The highly conserved Rap GTPases perform essential functions in the mammalian nervous system during development and in mature neurons. During neocortical development, Rap1 is required to regulate cadherin- and integrin-mediated adhesion. In the adult nervous system Rap1 and Rap2 regulate the maturation and plasticity of dendritic spine and synapses. Although genetic studies have revealed important roles of Rap GTPases in neurons, their regulation by guanine nucleotide exchange factors (GEFs) that activate them and GTPase activating proteins (GAPs) that inactivate them by stimulating their intrinsic GTPase activity is just beginning to be explored in vivo. Here we review how GEFs and GAPs regulate Rap GTPases in the nervous system with a focus on their in vivo function.
Collapse
|
45
|
Extracellular Signals Induce Glycoprotein M6a Clustering of Lipid Rafts and Associated Signaling Molecules. J Neurosci 2017; 37:4046-4064. [PMID: 28275160 DOI: 10.1523/jneurosci.3319-16.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/25/2017] [Accepted: 02/18/2017] [Indexed: 01/08/2023] Open
Abstract
Lipid raft domains, where sphingolipids and cholesterol are enriched, concentrate signaling molecules. To examine how signaling protein complexes are clustered in rafts, we focused on the functions of glycoprotein M6a (GPM6a), which is expressed at a high concentration in developing mouse neurons. Using imaging of lipid rafts, we found that GPM6a congregated in rafts in a GPM6a palmitoylation-dependent manner, thereby contributing to lipid raft clustering. In addition, we found that signaling proteins downstream of GPM6a, such as Rufy3, Rap2, and Tiam2/STEF, accumulated in lipid rafts in a GPM6a-dependent manner and were essential for laminin-dependent polarity during neurite formation in neuronal development. In utero RNAi targeting of GPM6a resulted in abnormally polarized neurons with multiple neurites. These results demonstrate that GPM6a induces the clustering of lipid rafts, which supports the raft aggregation of its associated downstream molecules for acceleration of neuronal polarity determination. Therefore, GPM6a acts as a signal transducer that responds to extracellular signals.SIGNIFICANCE STATEMENT Lipid raft domains, where sphingolipids and cholesterol are enriched, concentrate signaling molecules. We focused on glycoprotein M6a (GPM6a), which is expressed at a high concentration in developing neurons. Using imaging of lipid rafts, we found that GPM6a congregated in rafts in a palmitoylation-dependent manner, thereby contributing to lipid raft clustering. In addition, we found that signaling proteins downstream of GPM6a accumulated in lipid rafts in a GPM6a-dependent manner and were essential for laminin-dependent polarity during neurite formation. In utero RNAi targeting of GPM6a resulted in abnormally polarized neurons with multiple neurites. These results demonstrate that GPM6a induces the clustering of lipid rafts, which supports the raft aggregation of its associated downstream molecules for acceleration of polarity determination. Therefore, GPM6a acts as a signal transducer that responds to extracellular signals.
Collapse
|
46
|
Mardakheh FK, Self A, Marshall CJ. RHO binding to FAM65A regulates Golgi reorientation during cell migration. J Cell Sci 2016; 129:4466-4479. [PMID: 27807006 PMCID: PMC5201024 DOI: 10.1242/jcs.198614] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 10/24/2016] [Indexed: 12/31/2022] Open
Abstract
Directional cell migration involves reorientation of the secretory machinery. However, the molecular mechanisms that control this reorientation are not well characterised. Here, we identify a new Rho effector protein, named FAM65A, which binds to active RHOA, RHOB and RHOC. FAM65A links RHO proteins to Golgi-localising cerebral cavernous malformation-3 protein (CCM3; also known as PDCD10) and its interacting proteins mammalian STE20-like protein kinases 3 and 4 (MST3 and MST4; also known as STK24 and STK26, respectively). Binding of active RHO proteins to FAM65A does not affect the kinase activity of MSTs but results in their relocation from the Golgi in a CCM3-dependent manner. This relocation is crucial for reorientation of the Golgi towards the leading edge and subsequent directional cell migration. Our results reveal a previously unidentified pathway downstream of RHO that regulates the polarity of migrating cells through Golgi reorientation in a FAM65A-, CCM3- and MST3- and MST4-dependent manner.
Collapse
Affiliation(s)
- Faraz K Mardakheh
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK
| | - Annette Self
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK
| | - Christopher J Marshall
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK
| |
Collapse
|
47
|
Baldanzi G, Bettio V, Malacarne V, Graziani A. Diacylglycerol Kinases: Shaping Diacylglycerol and Phosphatidic Acid Gradients to Control Cell Polarity. Front Cell Dev Biol 2016; 4:140. [PMID: 27965956 PMCID: PMC5126041 DOI: 10.3389/fcell.2016.00140] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 11/14/2016] [Indexed: 12/24/2022] Open
Abstract
Diacylglycerol kinases (DGKs) terminate diacylglycerol (DAG) signaling and promote phosphatidic acid (PA) production. Isoform specific regulation of DGKs activity and localization allows DGKs to shape the DAG and PA gradients. The capacity of DGKs to constrain the areas of DAG signaling is exemplified by their role in defining the contact interface between T cells and antigen presenting cells: the immune synapse. Upon T cell receptor engagement, both DGK α and ζ metabolize DAG at the immune synapse thus constraining DAG signaling. Interestingly, their activity and localization are not fully redundant because DGKζ activity metabolizes the bulk of DAG in the cell, whereas DGKα limits the DAG signaling area localizing specifically at the periphery of the immune synapse. When DGKs terminate DAG signaling, the local PA production defines a new signaling domain, where PA recruits and activates a second wave of effector proteins. The best-characterized example is the role of DGKs in protrusion elongation and cell migration. Indeed, upon growth factor stimulation, several DGK isoforms, such as α, ζ, and γ, are recruited and activated at the plasma membrane. Here, local PA production controls cell migration by finely modulating cytoskeletal remodeling and integrin recycling. Interestingly, DGK-produced PA also controls the localization and activity of key players in cell polarity such as aPKC, Par3, and integrin β1. Thus, T cell polarization and directional migration may be just two instances of the general contribution of DGKs to the definition of cell polarity by local specification of membrane identity signaling.
Collapse
Affiliation(s)
- Gianluca Baldanzi
- Department of Translational Medicine, University of Piemonte OrientaleNovara, Italy; Institute for Research and Cure of Autoimmune DiseasesNovara, Italy
| | - Valentina Bettio
- Department of Translational Medicine, University of Piemonte OrientaleNovara, Italy; Institute for Research and Cure of Autoimmune DiseasesNovara, Italy
| | - Valeria Malacarne
- Department of Translational Medicine, University of Piemonte OrientaleNovara, Italy; Division of Experimental Oncology, School of Medicine, University Vita e Salute San RaffaeleMilan, Italy
| | - Andrea Graziani
- Department of Translational Medicine, University of Piemonte OrientaleNovara, Italy; Division of Experimental Oncology, School of Medicine, University Vita e Salute San RaffaeleMilan, Italy
| |
Collapse
|
48
|
Coch RA, Leube RE. Intermediate Filaments and Polarization in the Intestinal Epithelium. Cells 2016; 5:E32. [PMID: 27429003 PMCID: PMC5040974 DOI: 10.3390/cells5030032] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 01/02/2023] Open
Abstract
The cytoplasmic intermediate filament cytoskeleton provides a tissue-specific three-dimensional scaffolding with unique context-dependent organizational features. This is particularly apparent in the intestinal epithelium, in which the intermediate filament network is localized below the apical terminal web region and is anchored to the apical junction complex. This arrangement is conserved from the nematode Caenorhabditis elegans to humans. The review summarizes compositional, morphological and functional features of the polarized intermediate filament cytoskeleton in intestinal cells of nematodes and mammals. We emphasize the cross talk of intermediate filaments with the actin- and tubulin-based cytoskeleton. Possible links of the intermediate filament system to the distribution of apical membrane proteins and the cell polarity complex are highlighted. Finally, we discuss how these properties relate to the establishment and maintenance of polarity in the intestine.
Collapse
Affiliation(s)
- Richard A Coch
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, Aachen 52074, Germany.
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, Aachen 52074, Germany.
| |
Collapse
|
49
|
aPKC regulates apical localization of Lgl to restrict elongation of microridges in developing zebrafish epidermis. Nat Commun 2016; 7:11643. [PMID: 27249668 PMCID: PMC4895443 DOI: 10.1038/ncomms11643] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 04/15/2016] [Indexed: 12/05/2022] Open
Abstract
Epithelial cells exhibit apical membrane protrusions, which confer specific functions to epithelial tissues. Microridges are short actin protrusions that are laterally long and form a maze-like pattern in the apical domain. They are widely found on vertebrate squamous epithelia including epidermis and have functions in mucous retention, membrane storage and abrasion resistance. It is largely unknown how the formation of these laterally long actin projections is regulated. Here, we show that antagonistic interactions between aPKC and Lgl–regulators of apical and basolateral domain identity, respectively,–control the length of microridges in the zebrafish periderm, the outermost layer of the epidermis. aPKC regulates the levels of Lgl and the active form of non-muscle myosinII at the apical cortex to prevent actin polymerization-dependent precocious fusion and elongation of microridges. Our data unravels the functional significance of exclusion of Lgl from the apical domain in epithelial cells. Squamous epithelia present actin-rich microridges on the apical surface, but the mechanism of their formation is not known. Here the authors show that, in zebrafish epidermis, the exclusion of the basolateral regulator Lgl from the apical domain by atypical protein kinase C prevents precocious elongation and fusion of microridges.
Collapse
|
50
|
Khanal I, Elbediwy A, Diaz de la Loza MDC, Fletcher GC, Thompson BJ. Shot and Patronin polarise microtubules to direct membrane traffic and biogenesis of microvilli in epithelia. J Cell Sci 2016; 129:2651-9. [PMID: 27231092 PMCID: PMC4958304 DOI: 10.1242/jcs.189076] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/19/2016] [Indexed: 01/08/2023] Open
Abstract
In epithelial tissues, polarisation of microtubules and actin microvilli occurs along the apical-basal axis of each cell, yet how these cytoskeletal polarisation events are coordinated remains unclear. Here, we examine the hierarchy of events during cytoskeletal polarisation in Drosophila melanogaster epithelia. Core apical-basal polarity determinants polarise the spectrin cytoskeleton to recruit the microtubule-binding proteins Patronin (CAMSAP1, CAMSAP2 and CAMSAP3 in humans) and Shortstop [Shot; MACF1 and BPAG1 (also known as DST) in humans] to the apical membrane domain. Patronin and Shot then act to polarise microtubules along the apical-basal axis to enable apical transport of Rab11 endosomes by the Nuf-Dynein microtubule motor complex. Finally, Rab11 endosomes are transferred to the MyoV (also known as Didum in Drosophila) actin motor to deliver the key microvillar determinant Cadherin 99C to the apical membrane to organise the biogenesis of actin microvilli.
Collapse
Affiliation(s)
- Ichha Khanal
- The Francis Crick Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Ahmed Elbediwy
- The Francis Crick Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | | | | | - Barry J Thompson
- The Francis Crick Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| |
Collapse
|