1
|
Tong CS, Su M, Sun H, Chua XL, Xiong D, Guo S, Raj R, Ong NWP, Lee AG, Miao Y, Wu M. Collective dynamics of actin and microtubule and its crosstalk mediated by FHDC1. Front Cell Dev Biol 2024; 11:1261117. [PMID: 38567385 PMCID: PMC10985548 DOI: 10.3389/fcell.2023.1261117] [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: 07/18/2023] [Accepted: 12/19/2023] [Indexed: 04/04/2024] Open
Abstract
The coordination between actin and microtubule network is crucial, yet this remains a challenging problem to dissect and our understanding of the underlying mechanisms remains limited. In this study, we used travelling waves in the cell cortex to characterize the collective dynamics of cytoskeletal networks. Our findings show that Cdc42 and F-BAR-dependent actin waves in mast cells are mainly driven by formin-mediated actin polymerization, with the microtubule-binding formin FH2 domain-containing protein 1 (FHDC1) as an early regulator. Knocking down FHDC1 inhibits actin wave formation, and this inhibition require FHDC1's interaction with both microtubule and actin. The phase of microtubule depolymerization coincides with the nucleation of actin waves and microtubule stabilization inhibit actin waves, leading us to propose that microtubule shrinking and the concurrent release of FHDC1 locally regulate actin nucleation. Lastly, we show that FHDC1 is crucial for multiple cellular processes such as cell division and migration. Our data provided molecular insights into the nucleation mechanisms of actin waves and uncover an antagonistic interplay between microtubule and actin polymerization in their collective dynamics.
Collapse
Affiliation(s)
- Chee San Tong
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biological Sciences, Centre for Bioimaging Sciences, Singapore, Singapore
| | - Maohan Su
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biological Sciences, Centre for Bioimaging Sciences, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - He Sun
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Xiang Le Chua
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biological Sciences, Centre for Bioimaging Sciences, Singapore, Singapore
| | - Ding Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Su Guo
- Department of Biological Sciences, Centre for Bioimaging Sciences, Singapore, Singapore
| | - Ravin Raj
- Special Programme in Science, National University of Singapore, Singapore, Singapore
| | - Nicole Wen Pei Ong
- Special Programme in Science, National University of Singapore, Singapore, Singapore
| | - Ann Gie Lee
- Special Programme in Science, National University of Singapore, Singapore, Singapore
| | - Yansong Miao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Min Wu
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biological Sciences, Centre for Bioimaging Sciences, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
2
|
Aglago EK, Kim A, Lin Y, Qu C, Evangelou M, Ren Y, Morrison J, Albanes D, Arndt V, Barry EL, Baurley JW, Berndt SI, Bien SA, Bishop DT, Bouras E, Brenner H, Buchanan DD, Budiarto A, Carreras-Torres R, Casey G, Cenggoro TW, Chan AT, Chang-Claude J, Chen X, Conti DV, Devall M, Diez-Obrero V, Dimou N, Drew D, Figueiredo JC, Gallinger S, Giles GG, Gruber SB, Gsur A, Gunter MJ, Hampel H, Harlid S, Hidaka A, Harrison TA, Hoffmeister M, Huyghe JR, Jenkins MA, Jordahl K, Joshi AD, Kawaguchi ES, Keku TO, Kundaje A, Larsson SC, Marchand LL, Lewinger JP, Li L, Lynch BM, Mahesworo B, Mandic M, Obón-Santacana M, Moreno V, Murphy N, Nan H, Nassir R, Newcomb PA, Ogino S, Ose J, Pai RK, Palmer JR, Papadimitriou N, Pardamean B, Peoples AR, Platz EA, Potter JD, Prentice RL, Rennert G, Ruiz-Narvaez E, Sakoda LC, Scacheri PC, Schmit SL, Schoen RE, Shcherbina A, Slattery ML, Stern MC, Su YR, Tangen CM, Thibodeau SN, Thomas DC, Tian Y, Ulrich CM, van Duijnhoven FJB, Van Guelpen B, Visvanathan K, Vodicka P, Wang J, White E, Wolk A, Woods MO, Wu AH, Zemlianskaia N, Hsu L, Gauderman WJ, Peters U, Tsilidis KK, Campbell PT. A Genetic Locus within the FMN1/GREM1 Gene Region Interacts with Body Mass Index in Colorectal Cancer Risk. Cancer Res 2023; 83:2572-2583. [PMID: 37249599 PMCID: PMC10391330 DOI: 10.1158/0008-5472.can-22-3713] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/25/2023] [Accepted: 05/24/2023] [Indexed: 05/31/2023]
Abstract
Colorectal cancer risk can be impacted by genetic, environmental, and lifestyle factors, including diet and obesity. Gene-environment interactions (G × E) can provide biological insights into the effects of obesity on colorectal cancer risk. Here, we assessed potential genome-wide G × E interactions between body mass index (BMI) and common SNPs for colorectal cancer risk using data from 36,415 colorectal cancer cases and 48,451 controls from three international colorectal cancer consortia (CCFR, CORECT, and GECCO). The G × E tests included the conventional logistic regression using multiplicative terms (one degree of freedom, 1DF test), the two-step EDGE method, and the joint 3DF test, each of which is powerful for detecting G × E interactions under specific conditions. BMI was associated with higher colorectal cancer risk. The two-step approach revealed a statistically significant G×BMI interaction located within the Formin 1/Gremlin 1 (FMN1/GREM1) gene region (rs58349661). This SNP was also identified by the 3DF test, with a suggestive statistical significance in the 1DF test. Among participants with the CC genotype of rs58349661, overweight and obesity categories were associated with higher colorectal cancer risk, whereas null associations were observed across BMI categories in those with the TT genotype. Using data from three large international consortia, this study discovered a locus in the FMN1/GREM1 gene region that interacts with BMI on the association with colorectal cancer risk. Further studies should examine the potential mechanisms through which this locus modifies the etiologic link between obesity and colorectal cancer. SIGNIFICANCE This gene-environment interaction analysis revealed a genetic locus in FMN1/GREM1 that interacts with body mass index in colorectal cancer risk, suggesting potential implications for precision prevention strategies.
Collapse
Affiliation(s)
- Elom K. Aglago
- Department of Epidemiology and Biostatistics, Imperial College London, School of Public Health, London, United Kingdom
| | - Andre Kim
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Yi Lin
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Conghui Qu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Marina Evangelou
- Department of Epidemiology and Biostatistics, Imperial College London, School of Public Health, London, United Kingdom
| | - Yu Ren
- Department of Epidemiology and Biostatistics, Imperial College London, School of Public Health, London, United Kingdom
| | - John Morrison
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Volker Arndt
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elizabeth L. Barry
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - James W. Baurley
- Bioinformatics and Data Science Research Center, Bina Nusantara University, Jakarta, Indonesia
- BioRealm LLC, Walnut, California
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephanie A. Bien
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - D. Timothy Bishop
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Emmanouil Bouras
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel D. Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, Victoria, Australia
- Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Arif Budiarto
- Bioinformatics and Data Science Research Center, Bina Nusantara University, Jakarta, Indonesia
- Computer Science Department, School of Computer Science, Bina Nusantara University, Jakarta, Indonesia
| | - Robert Carreras-Torres
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), Salt, Girona, Spain
| | - Graham Casey
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia
| | - Tjeng Wawan Cenggoro
- Bioinformatics and Data Science Research Center, Bina Nusantara University, Jakarta, Indonesia
| | - Andrew T. Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- University Medical Centre Hamburg-Eppendorf, University Cancer Centre Hamburg (UCCH), Hamburg, Germany
| | - Xuechen Chen
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - David V. Conti
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Matthew Devall
- Department of Family Medicine, University of Virginia, Charlottesville, Virginia
| | - Virginia Diez-Obrero
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Unit of Biomarkers and Susceptibility (UBS), Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, Spain
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Niki Dimou
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - David Drew
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jane C. Figueiredo
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Steven Gallinger
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Graham G. Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Stephen B. Gruber
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte California
| | - Andrea Gsur
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Marc J. Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Heather Hampel
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte California
| | - Sophia Harlid
- Department of Radiation Sciences, Oncology Unit, Umeå University, Umeå, Sweden
| | - Akihisa Hidaka
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Tabitha A. Harrison
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jeroen R. Huyghe
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mark A. Jenkins
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Kristina Jordahl
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Amit D. Joshi
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Eric S. Kawaguchi
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Temitope O. Keku
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, North Carolina
| | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, California
- Department of Computer Science, Stanford University, Stanford, California
| | - Susanna C. Larsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Juan Pablo Lewinger
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Li Li
- Department of Family Medicine, University of Virginia, Charlottesville, Virginia
| | - Brigid M. Lynch
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
- Physical Activity Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Bharuno Mahesworo
- Bioinformatics and Data Science Research Center, Bina Nusantara University, Jakarta, Indonesia
| | - Marko Mandic
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Mireia Obón-Santacana
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Unit of Biomarkers and Susceptibility (UBS), Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Victor Moreno
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Unit of Biomarkers and Susceptibility (UBS), Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Neil Murphy
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Hongmei Nan
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indianapolis, Indiana
- IU Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Rami Nassir
- Department of Pathology, School of Medicine, Umm Al-Qura'a University, Mecca, Saudi Arabia
| | - Polly A. Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington
| | - Shuji Ogino
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jennifer Ose
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Rish K. Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Julie R. Palmer
- Department of Medicine, Boston University School of Medicine, Slone Epidemiology Center, Boston University, Boston, Massachusetts
| | - Nikos Papadimitriou
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Bens Pardamean
- Bioinformatics and Data Science Research Center, Bina Nusantara University, Jakarta, Indonesia
| | - Anita R. Peoples
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - John D. Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington
- Research Centre for Hauora and Health, Massey University, Wellington, New Zealand
| | - Ross L. Prentice
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Gad Rennert
- Department of Community Medicine and Epidemiology, Lady Davis Carmel Medical Center, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Clalit National Cancer Control Center, Haifa, Israel
| | - Edward Ruiz-Narvaez
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Lori C. Sakoda
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Peter C. Scacheri
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | | | - Robert E. Schoen
- Department of Medicine and Epidemiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Anna Shcherbina
- Department of Genetics, Stanford University, Stanford, California
- Department of Computer Science, Stanford University, Stanford, California
| | - Martha L. Slattery
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Mariana C. Stern
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Yu-Ru Su
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Catherine M. Tangen
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Stephen N. Thibodeau
- Division of Laboratory Genetics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Duncan C. Thomas
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Yu Tian
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- School of Public Health, Capital Medical University, Beijing, China
| | - Cornelia M. Ulrich
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Franzel JB van Duijnhoven
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, the Netherlands
| | - Bethany Van Guelpen
- Department of Radiation Sciences, Oncology Unit, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Pavel Vodicka
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Jun Wang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Emily White
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington
| | - Alicja Wolk
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Michael O. Woods
- Memorial University of Newfoundland, Discipline of Genetics, St. John's, Canada
| | - Anna H. Wu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Natalia Zemlianskaia
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - W. James Gauderman
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington
| | - Konstantinos K. Tsilidis
- Department of Epidemiology and Biostatistics, Imperial College London, School of Public Health, London, United Kingdom
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Peter T. Campbell
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
3
|
Romero MD, Carabeo RA. Distinct roles of the Chlamydia trachomatis effectors TarP and TmeA in the regulation of formin and Arp2/3 during entry. J Cell Sci 2022; 135:jcs260185. [PMID: 36093837 PMCID: PMC9659389 DOI: 10.1242/jcs.260185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/05/2022] [Indexed: 12/13/2022] Open
Abstract
The obligate intracellular pathogen Chlamydia trachomatis manipulates the host actin cytoskeleton to assemble actin-rich structures that drive pathogen entry. The recent discovery of TmeA, which, like TarP, is an invasion-associated type III effector implicated in actin remodeling, raised questions regarding the nature of their functional interaction. Quantitative live-cell imaging of actin remodeling at invasion sites revealed differences in recruitment and turnover kinetics associated with the TarP and TmeA pathways, with the former accounting for most of the robust actin dynamics at invasion sites. TarP-mediated recruitment of actin nucleators, i.e. formins and the Arp2/3 complex, was crucial for rapid actin kinetics, generating a collaborative positive feedback loop that enhanced their respective actin-nucleating activities within invasion sites. In contrast, the formin Fmn1 was not recruited to invasion sites and did not collaborate with Arp2/3 within the context of TmeA-associated actin recruitment. Although the TarP-Fmn1-Arp2/3 signaling axis is responsible for the majority of actin dynamics, its inhibition had similar effects as the deletion of TmeA on invasion efficiency, consistent with the proposed model that TarP and TmeA act on different stages of the same invasion pathway.
Collapse
Affiliation(s)
- Matthew D. Romero
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA
| | - Rey A. Carabeo
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA
| |
Collapse
|
4
|
Ahangar P, Cowin AJ. Reforming the Barrier: The Role of Formins in Wound Repair. Cells 2022; 11:cells11182779. [PMID: 36139355 PMCID: PMC9496773 DOI: 10.3390/cells11182779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 12/04/2022] Open
Abstract
The restoration of an intact epidermal barrier after wound injury is the culmination of a highly complex and exquisitely regulated physiological process involving multiple cells and tissues, overlapping dynamic events and protein synthesis and regulation. Central to this process is the cytoskeleton, a system of intracellular proteins that are instrumental in regulating important processes involved in wound repair including chemotaxis, cytokinesis, proliferation, migration, and phagocytosis. One highly conserved family of cytoskeletal proteins that are emerging as major regulators of actin and microtubule nucleation, polymerization, and stabilization are the formins. The formin family includes 15 different proteins categorized into seven subfamilies based on three formin homology domains (FH1, FH2, and FH3). The formins themselves are regulated in different ways including autoinhibition, activation, and localization by a range of proteins, including Rho GTPases. Herein, we describe the roles and effects of the formin family of cytoskeletal proteins on the fundamental process of wound healing and highlight recent advances relating to their important functions, mechanisms, and regulation at the molecular and cellular levels.
Collapse
|
5
|
Tóth K, Földi I, Mihály J. A Comparative Study of the Role of Formins in Drosophila Embryonic Dorsal Closure. Cells 2022; 11:cells11091539. [PMID: 35563844 PMCID: PMC9102720 DOI: 10.3390/cells11091539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 12/10/2022] Open
Abstract
Dorsal closure is a late embryogenesis process required to seal the epidermal hole on the dorsal side of the Drosophila embryo. This process involves the coordination of several forces generated in the epidermal cell layer and in the amnioserosa cells, covering the hole. Ultimately, these forces arise due to cytoskeletal rearrangements that induce changes in cell shape and result in tissue movement. While a number of cytoskeleton regulatory proteins have already been linked to dorsal closure, here we expand this list by demonstrating that four of the six Drosophila formin type actin assembly factors are needed to bring about the proper fusion of the epithelia. An analysis of the morphological and dynamic properties of dorsal closure in formin mutants revealed a differential contribution for each formin, although we found evidence for functional redundancies as well. Therefore, we propose that the four formins promote the formation of several, and only partly identical, actin structures each with a specific role in the mechanics of dorsal closure.
Collapse
Affiliation(s)
- Krisztina Tóth
- Biological Research Centre, Institute of Genetics, Temesvári krt. 62, H-6726 Szeged, Hungary; (K.T.); (I.F.)
- Doctoral School of Multidisciplinary Medical Science, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary
| | - István Földi
- Biological Research Centre, Institute of Genetics, Temesvári krt. 62, H-6726 Szeged, Hungary; (K.T.); (I.F.)
| | - József Mihály
- Biological Research Centre, Institute of Genetics, Temesvári krt. 62, H-6726 Szeged, Hungary; (K.T.); (I.F.)
- Department of Genetics, University of Szeged, H-6726 Szeged, Hungary
- Correspondence:
| |
Collapse
|
6
|
Molecular Dissection of DAAM Function during Axon Growth in Drosophila Embryonic Neurons. Cells 2022; 11:cells11091487. [PMID: 35563792 PMCID: PMC9102401 DOI: 10.3390/cells11091487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
Axonal growth is mediated by coordinated changes of the actin and microtubule (MT) cytoskeleton. Ample evidence suggests that members of the formin protein family are involved in the coordination of these cytoskeletal rearrangements, but the molecular mechanisms of the formin-dependent actin–microtubule crosstalk remains largely elusive. Of the six Drosophila formins, DAAM was shown to play a pivotal role during axonal growth in all stages of nervous system development, while FRL was implicated in axonal development in the adult brain. Here, we aimed to investigate the potentially redundant function of these two formins, and we attempted to clarify which molecular activities are important for axonal growth. We used a combination of genetic analyses, cellular assays and biochemical approaches to demonstrate that the actin-processing activity of DAAM is indispensable for axonal growth in every developmental condition. In addition, we identified a novel MT-binding motif within the FH2 domain of DAAM, which is required for proper growth and guidance of the mushroom body axons, while being dispensable during embryonic axon development. Together, these data suggest that DAAM is the predominant formin during axonal growth in Drosophila, and highlight the contribution of multiple formin-mediated mechanisms in cytoskeleton coordination during axonal growth.
Collapse
|
7
|
Sallem A, Denizot AL, Ziyyat A, L'Hostis A, Favier S, Burlet P, Lapierre JM, Dimby SF, Patrat C, Sifer C, Vicaut E, Steffann J, Vaiman D, Romana SP, Wolf JP. A fertilin-derived peptide improves in vitro maturation and ploidy of human oocytes. F&S SCIENCE 2022; 3:21-28. [PMID: 35559993 DOI: 10.1016/j.xfss.2021.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/19/2021] [Accepted: 10/29/2021] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To analyze the effect of a cyclic fertilin-derived peptide (cFEE) on in vitro maturation of human oocytes. DESIGN Randomized study. SETTING Fertility center in an academic hospital. PATIENT(S) Not applicable. INTERVENTION(S) Human immature germinal vesicle-stage oocytes (n = 1,629) donated for research according to French bioethics laws were randomly allocated to groups treated with 1 or 100 μM of cFEE or to a control group. They were incubated at 37 °C in 6% CO2 and 5% O2, and their maturation was assessed using time-lapse microscopy over 24 hours. In vitro maturated metaphase II oocytes were analyzed for chromosomal content using microarray comparative genomic hybridization, and their transcriptomes were analyzed using Affymetrix Clariom D microarrays. MAIN OUTCOME MEASURE(S) The percentage of oocytes undergoing maturation in vitro was observed. Aneuploidy and euploidy were assessed for all chromosomes, and differential gene expression was analyzed in oocytes treated with cFEE compared with the control to obtain insights into its mechanism of action. RESULT(S) cFEE significantly increased the percentage of oocytes that matured in vitro and improved euploidy in meiosis II oocytes by the up-regulation of FMN1 and FLNA genes, both of which encode proteins involved in spindle structure. CONCLUSION(S) cFEE improves human oocyte maturation in vitro and reduces aneuploidy. It may prove useful for treating oocytes before fertilization in assisted reproductive technology and for in vitro maturation in fertility preservation programs to improve oocyte quality and the chances for infertile couples to conceive.
Collapse
Affiliation(s)
- Amira Sallem
- Team "From Gametes to Birth," Département Développement, Reproduction, Cancer, Institut Cochin, Institut de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université de Paris, 22 rue Mechain, Paris, France; Laboratoire d'Histologie-Embryologie et Cytogénétique, Faculté de Médecine de Monastir, Tunisie
| | - Anne-Lyse Denizot
- Team "From Gametes to Birth," Département Développement, Reproduction, Cancer, Institut Cochin, Institut de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université de Paris, 22 rue Mechain, Paris, France; Service d'Histologie-Embryologie-Biologie de la Reproduction, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Ahmed Ziyyat
- Team "From Gametes to Birth," Département Développement, Reproduction, Cancer, Institut Cochin, Institut de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université de Paris, 22 rue Mechain, Paris, France; Service d'Histologie-Embryologie-Biologie de la Reproduction, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Audrey L'Hostis
- Team "From Gametes to Birth," Département Développement, Reproduction, Cancer, Institut Cochin, Institut de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université de Paris, 22 rue Mechain, Paris, France
| | - Sophie Favier
- Team "From Gametes to Birth," Département Développement, Reproduction, Cancer, Institut Cochin, Institut de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université de Paris, 22 rue Mechain, Paris, France
| | - Philippe Burlet
- Service de Génétique Moléculaire, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Michel Lapierre
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Solohaja Faniaha Dimby
- Unité de Recherche Clinique, ACTION Study Group, Hôpital Fernand Widal, Assistance Publique-Hôpitaux de Paris, Paris, France; Statistique, Analyse et Modélisation Multidisciplinaire-EA 4543, Université Paris 1 Panthéon Sorbonne, Paris, France
| | - Catherine Patrat
- Team "From Gametes to Birth," Département Développement, Reproduction, Cancer, Institut Cochin, Institut de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université de Paris, 22 rue Mechain, Paris, France; Service d'Histologie-Embryologie-Biologie de la Reproduction, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Christophe Sifer
- Service d'Histologie-Embryologie-Cytogénétique-Centre d'Etude et de Conservation des Œufs et du Sperme humains, Centre Hospitalo-Universitaire Jean Verdier, Assistance Publique-Hôpitaux de Paris, Bondy, France
| | - Eric Vicaut
- Unité de Recherche Clinique, ACTION Study Group, Hôpital Fernand Widal, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Julie Steffann
- Service de Génétique Moléculaire, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; Institut Imagine, Université de Paris, Laboratoire des Maladies Génétiques Mitochondriales. Inserm 1163, Paris, France
| | - Daniel Vaiman
- Team "From Gametes to Birth," Département Développement, Reproduction, Cancer, Institut Cochin, Institut de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université de Paris, 22 rue Mechain, Paris, France
| | - Serge Pierrick Romana
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; Institut Imagine, Université de Paris, Laboratoire d'Embryologie et de Génétique des Malformations Congénitales, Institut de la Santé et de la Recherche Médicale 1163, Paris, France
| | - Jean-Philippe Wolf
- Team "From Gametes to Birth," Département Développement, Reproduction, Cancer, Institut Cochin, Institut de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université de Paris, 22 rue Mechain, Paris, France; Service d'Histologie-Embryologie-Biologie de la Reproduction, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France.
| |
Collapse
|
8
|
Adaptive mechanoproperties mediated by the formin FMN1 characterize glioblastoma fitness for invasion. Dev Cell 2021; 56:2841-2855.e8. [PMID: 34559979 DOI: 10.1016/j.devcel.2021.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/23/2021] [Accepted: 09/03/2021] [Indexed: 11/22/2022]
Abstract
Glioblastoma are heterogeneous tumors composed of highly invasive and highly proliferative clones. Heterogeneity in invasiveness could emerge from discrete biophysical properties linked to specific molecular expression. We identified clones of patient-derived glioma propagating cells that were either highly proliferative or highly invasive and compared their cellular architecture, migratory, and biophysical properties. We discovered that invasiveness was linked to cellular fitness. The most invasive cells were stiffer, developed higher mechanical forces on the substrate, and moved stochastically. The mechano-chemical-induced expression of the formin FMN1 conferred invasive strength that was confirmed in patient samples. Moreover, FMN1 expression was also linked to motility in other cancer and normal cell lines, and its ectopic expression increased fitness parameters. Mechanistically, FMN1 acts from the microtubule lattice and promotes a robust mechanical cohesion, leading to highly invasive motility.
Collapse
|
9
|
Tangeman JA, Luz-Madrigal A, Sreeskandarajan S, Grajales-Esquivel E, Liu L, Liang C, Tsonis PA, Del Rio-Tsonis K. Transcriptome Profiling of Embryonic Retinal Pigment Epithelium Reprogramming. Genes (Basel) 2021; 12:genes12060840. [PMID: 34072522 PMCID: PMC8226911 DOI: 10.3390/genes12060840] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/05/2021] [Accepted: 05/22/2021] [Indexed: 12/27/2022] Open
Abstract
The plasticity of human retinal pigment epithelium (RPE) has been observed during proliferative vitreoretinopathy, a defective repair process during which injured RPE gives rise to fibrosis. In contrast, following injury, the RPE of the embryonic chicken can be reprogrammed to regenerate neural retina in a fibroblast growth factor 2 (FGF2)-dependent manner. To better explore the mechanisms underlying embryonic RPE reprogramming, we used laser capture microdissection to isolate RNA from (1) intact RPE, (2) transiently reprogrammed RPE (t-rRPE) 6 h post-retinectomy, and (3) reprogrammed RPE (rRPE) 6 h post-retinectomy with FGF2 treatment. Using RNA-seq, we observed the acute repression of genes related to cell cycle progression in the injured t-rRPE, as well as up-regulation of genes associated with injury. In contrast, the rRPE was strongly enriched for mitogen-activated protein kinase (MAPK)-responsive genes and retina development factors, confirming that FGF2 and the downstream MAPK cascade are the main drivers of embryonic RPE reprogramming. Clustering and pathway enrichment analysis was used to create an integrated network of the core processes associated with RPE reprogramming, including key terms pertaining to injury response, migration, actin dynamics, and cell cycle progression. Finally, we employed gene set enrichment analysis to suggest a previously uncovered role for epithelial-mesenchymal transition (EMT) machinery in the initiation of embryonic chick RPE reprogramming. The EMT program is accompanied by extensive, coordinated regulation of extracellular matrix (ECM) associated factors, and these observations together suggest an early role for ECM and EMT-like dynamics during reprogramming. Our study provides for the first time an in-depth transcriptomic analysis of embryonic RPE reprogramming and will prove useful in guiding future efforts to understand proliferative disorders of the RPE and to promote retinal regeneration.
Collapse
Affiliation(s)
- Jared A. Tangeman
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
| | - Agustín Luz-Madrigal
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sutharzan Sreeskandarajan
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Erika Grajales-Esquivel
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
| | - Lin Liu
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
| | - Chun Liang
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
- Department of Computer Science and Software Engineering, Miami University, Oxford, OH 45056, USA
| | - Panagiotis A. Tsonis
- Department of Biology, University of Dayton and Center for Tissue Regeneration and Engineering at the University of Dayton (TREND), Dayton, OH 45469, USA;
| | - Katia Del Rio-Tsonis
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
- Correspondence: ; Tel.: +513-529-3128; Fax: +513-529-6900
| |
Collapse
|
10
|
Martier R, Sogorb-Gonzalez M, Stricker-Shaver J, Hübener-Schmid J, Keskin S, Klima J, Toonen LJ, Juhas S, Juhasova J, Ellederova Z, Motlik J, Haas E, van Deventer S, Konstantinova P, Nguyen HP, Evers MM. Development of an AAV-Based MicroRNA Gene Therapy to Treat Machado-Joseph Disease. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:343-358. [PMID: 31828177 PMCID: PMC6889651 DOI: 10.1016/j.omtm.2019.10.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/22/2019] [Indexed: 01/06/2023]
Abstract
Spinocerebellar ataxia type 3 (SCA3), or Machado-Joseph disease (MJD), is a progressive neurodegenerative disorder caused by a CAG expansion in the ATXN3 gene. The expanded CAG repeat is translated into a prolonged polyglutamine repeat in the ataxin-3 protein and accumulates within inclusions, acquiring toxic properties, which results in degeneration of the cerebellum and brain stem. In the current study, a non-allele-specific ATXN3 silencing approach was investigated using artificial microRNAs engineered to target various regions of the ATXN3 gene (miATXN3). The miATXN3 candidates were screened in vitro based on their silencing efficacy on a luciferase (Luc) reporter co-expressing ATXN3. The three best miATXN3 candidates were further tested for target engagement and potential off-target activity in induced pluripotent stem cells (iPSCs) differentiated into frontal brain-like neurons and in a SCA3 knockin mouse model. Besides a strong reduction of ATXN3 mRNA and protein, small RNA sequencing revealed efficient guide strand processing without passenger strands being produced. We used different methods to predict alteration of off-target genes upon AAV5-miATXN3 treatment and found no evidence for unwanted effects. Furthermore, we demonstrated in a large animal model, the minipig, that intrathecal delivery of AAV5 can transduce the main areas affected in SCA3 patients. These results proved a strong basis to move forward to investigate distribution, efficacy, and safety of AAV5-miATXN3 in large animals.
Collapse
Affiliation(s)
- Raygene Martier
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands.,Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marina Sogorb-Gonzalez
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands.,Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Janice Stricker-Shaver
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | | | - Sonay Keskin
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands
| | - Jiri Klima
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Lodewijk J Toonen
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands
| | - Stefan Juhas
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Jana Juhasova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Zdenka Ellederova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Jan Motlik
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Eva Haas
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Sander van Deventer
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands.,Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Pavlina Konstantinova
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands
| | - Huu Phuc Nguyen
- Department of Human Genetics, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Melvin M Evers
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands
| |
Collapse
|
11
|
Regulation of Blood-Testis Barrier (BTB) Dynamics, Role of Actin-, and Microtubule-Based Cytoskeletons. Methods Mol Biol 2019; 1748:229-243. [PMID: 29453575 DOI: 10.1007/978-1-4939-7698-0_16] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The blood-testis barrier (BTB) is an important ultrastructure in the testis that supports meiosis and postmeiotic spermatid development since a delay in the establishment of a functional Sertoli cell barrier during postnatal development in rats or mice by 17-20 day postpartum (dpp) would lead to a delay of the first wave of meiosis. Furthermore, irreversible disruption of the BTB by toxicants also induces infertility in rodents. Herein, we summarize recent findings that BTB dynamics (i.e., disassembly, reassembly, and stabilization) are supported by the concerted efforts of the actin- and microtubule (MT)-based cytoskeletons. We focus on the role of two actin nucleation protein complexes, namely, the Arp2/3 (actin-related protein 2/3) complex and formin 1 (or the formin 1/spire 1 complex) known to induce actin nucleation, respectively, by conferring plasticity to actin cytoskeleton. We also focus on the MT plus (+)-end tracking protein (+TIP) EB1 (end-binding protein 1) which is known to confer MT stabilization. Furthermore, we discuss in particular how the interactions of these proteins modulate BTB dynamics during spermatogenesis. These findings also yield a novel hypothetical concept regarding the molecular mechanism that modulates BTB function.
Collapse
|
12
|
Courtemanche N. Mechanisms of formin-mediated actin assembly and dynamics. Biophys Rev 2018; 10:1553-1569. [PMID: 30392063 DOI: 10.1007/s12551-018-0468-6] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/18/2018] [Indexed: 12/14/2022] Open
Abstract
Cellular viability requires tight regulation of actin cytoskeletal dynamics. Distinct families of nucleation-promoting factors enable the rapid assembly of filament nuclei that elongate and are incorporated into diverse and specialized actin-based structures. In addition to promoting filament nucleation, the formin family of proteins directs the elongation of unbranched actin filaments. Processive association of formins with growing filament ends is achieved through continuous barbed end binding of the highly conserved, dimeric formin homology (FH) 2 domain. In cooperation with the FH1 domain and C-terminal tail region, FH2 dimers mediate actin subunit addition at speeds that can dramatically exceed the rate of spontaneous assembly. Here, I review recent biophysical, structural, and computational studies that have provided insight into the mechanisms of formin-mediated actin assembly and dynamics.
Collapse
Affiliation(s)
- Naomi Courtemanche
- Department of Genetics, Cell and Developmental Biology, University of Minnesota, 420 Washington Ave SE, 6-130 MCB, Minneapolis, MN, 55455, USA.
| |
Collapse
|
13
|
Higashi T, Stephenson RE, Miller AL. Comprehensive analysis of formin localization in Xenopus epithelial cells. Mol Biol Cell 2018; 30:82-95. [PMID: 30379611 PMCID: PMC6337911 DOI: 10.1091/mbc.e18-02-0133] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Reorganization of the actin cytoskeleton is crucial for cellular processes, including cytokinesis and cell–cell junction remodeling. Formins are conserved processive actin-polymerizing machines that regulate actin dynamics by nucleating, elongating, and bundling linear actin filaments. Because the formin family is large, with at least 15 members in vertebrates, there have not been any comprehensive studies examining formin localization and function within a common cell type. Here, we characterized the localization of all 15 formins in epithelial cells of Xenopus laevis gastrula-stage embryos. Dia1 and Dia2 localized to tight junctions, while Fhod1 and Fhod3 localized to adherens junctions. Only Dia3 strongly localized at the cytokinetic contractile ring. The Diaphanous inhibitory domain–dimerization domain (DID-DD) region of Dia1 was sufficient for Dia1 localization, and overexpression of a Dia1 DID-DD fragment competitively removed Dia1 and Dia2 from cell–cell junctions. In Dia1 DID-DD–overexpressing cells, Dia1 and Dia2 were mislocalized to the contractile ring, and cells exhibited increased cytokinesis failure. This work provides a comprehensive analysis of the localization of all 15 vertebrate formins in epithelial cells and suggests that misregulated formin localization results in epithelial cytokinesis failure.
Collapse
Affiliation(s)
- Tomohito Higashi
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Rachel E Stephenson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Ann L Miller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
14
|
Fernández-Barrera J, Alonso MA. Coordination of microtubule acetylation and the actin cytoskeleton by formins. Cell Mol Life Sci 2018; 75:3181-3191. [PMID: 29947928 PMCID: PMC11105221 DOI: 10.1007/s00018-018-2855-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/22/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022]
Abstract
The acetylation of the lysine 40 residue of α-tubulin was described more than 30 years ago and has been the subject of intense research ever since. Although the exact function of this covalent modification of tubulin in the cell remains unknown, it has been established that tubulin acetylation confers resilience to mechanical stress on the microtubules. Formins have a dual role in the fate of the actin and tubulin cytoskeletons. On the one hand, they catalyze the formation of actin filaments, and on the other, they bind microtubules, act on their stability, and regulate their acetylation and alignment with actin fibers. Recent evidence indicates that formins coordinate the actin cytoskeleton and tubulin acetylation by modulating the levels of free globular actin (G-actin). G-actin, in turn, controls the activity of the myocardin-related transcription factor-serum response factor transcriptional complex that regulates the expression of the α-tubulin acetyltransferase 1 (α-TAT1) gene, which encodes the main enzyme responsible for tubulin acetylation. The effect of formins on tubulin acetylation is the combined result of their ability to activate α-TAT1 gene transcription and of their capacity to regulate microtubule stabilization. The contribution of these two mechanisms in different formins is discussed, particularly with respect to INF2, a formin that is mutated in hereditary human renal and neurodegenerative disorders.
Collapse
Affiliation(s)
- Jaime Fernández-Barrera
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma, Madrid, Spain
| | - Miguel A Alonso
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma, Madrid, Spain.
| |
Collapse
|
15
|
Fernández-Barrera J, Bernabé-Rubio M, Casares-Arias J, Rangel L, Fernández-Martín L, Correas I, Alonso MA. The actin-MRTF-SRF transcriptional circuit controls tubulin acetylation via α-TAT1 gene expression. J Cell Biol 2018; 217:929-944. [PMID: 29321169 PMCID: PMC5839776 DOI: 10.1083/jcb.201702157] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/28/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023] Open
Abstract
The role of formins in microtubules is not well understood. In this study, we have investigated the mechanism by which INF2, a formin mutated in degenerative renal and neurological hereditary disorders, controls microtubule acetylation. We found that silencing of INF2 in epithelial RPE-1 cells produced a dramatic drop in tubulin acetylation, increased the G-actin/F-actin ratio, and impaired myocardin-related transcription factor (MRTF)/serum response factor (SRF)-dependent transcription, which is known to be repressed by increased levels of G-actin. The effect on tubulin acetylation was caused by the almost complete absence of α-tubulin acetyltransferase 1 (α-TAT1) messenger RNA (mRNA). Activation of the MRTF-SRF transcriptional complex restored α-TAT1 mRNA levels and tubulin acetylation. Several functional MRTF-SRF-responsive elements were consistently identified in the α-TAT1 gene. The effect of INF2 silencing on microtubule acetylation was also observed in epithelial ECV304 cells, but not in Jurkat T cells. Therefore, the actin-MRTF-SRF circuit controls α-TAT1 transcription. INF2 regulates the circuit, and hence microtubule acetylation, in cell types where it has a prominent role in actin polymerization.
Collapse
Affiliation(s)
- Jaime Fernández-Barrera
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel Bernabé-Rubio
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Javier Casares-Arias
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Rangel
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain.,Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Fernández-Martín
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Isabel Correas
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain.,Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel A Alonso
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
16
|
Buzanskas ME, Grossi DDA, Ventura RV, Schenkel FS, Chud TCS, Stafuzza NB, Rola LD, Meirelles SLC, Mokry FB, Mudadu MDA, Higa RH, da Silva MVGB, de Alencar MM, Regitano LCDA, Munari DP. Candidate genes for male and female reproductive traits in Canchim beef cattle. J Anim Sci Biotechnol 2017; 8:67. [PMID: 28852499 PMCID: PMC5569548 DOI: 10.1186/s40104-017-0199-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 07/13/2017] [Indexed: 01/20/2023] Open
Abstract
Background Beef cattle breeding programs in Brazil have placed greater emphasis on the genomic study of reproductive traits of males and females due to their economic importance. In this study, genome-wide associations were assessed for scrotal circumference at 210 d of age, scrotal circumference at 420 d of age, age at first calving, and age at second calving, in Canchim beef cattle. Data quality control was conducted resulting in 672,778 SNPs and 392 animals. Results Associated SNPs were observed for scrotal circumference at 420 d of age (435 SNPs), followed by scrotal circumference at 210 d of age (12 SNPs), age at first calving (six SNPs), and age at second calving (four SNPs). We investigated whether significant SNPs were within genic or surrounding regions. Biological processes of genes were associated with immune system, multicellular organismal process, response to stimulus, apoptotic process, cellular component organization or biogenesis, biological adhesion, and reproduction. Conclusions Few associations were observed for scrotal circumference at 210 d of age, age at first calving, and age at second calving, reinforcing their polygenic inheritance and the complexity of understanding the genetic architecture of reproductive traits. Finding many associations for scrotal circumference at 420 d of age in various regions of the Canchim genome also reveals the difficulty of targeting specific candidate genes that could act on fertility; nonetheless, the high linkage disequilibrium between loci herein estimated could aid to overcome this issue. Therefore, all relevant information about genomic regions influencing reproductive traits may contribute to target candidate genes for further investigation of causal mutations and aid in future genomic studies in Canchim cattle to improve the breeding program. Electronic supplementary material The online version of this article (doi:10.1186/s40104-017-0199-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marcos Eli Buzanskas
- Departamento de Zootecnia, Universidade Federal da Paraíba (UFPB), Areia, Paraíba 58397-000 Brazil
| | | | | | - Flavio Schramm Schenkel
- Department of Animal and Poultry Science, University of Guelph, Centre for Genetic Improvement of Livestock (CGIL), Guelph, ON N1G 2W1 Canada
| | - Tatiane Cristina Seleguim Chud
- Departamento de Ciências Exatas, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista (Unesp), Jaboticabal, São Paulo 14884-900 Brazil
| | - Nedenia Bonvino Stafuzza
- Departamento de Ciências Exatas, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista (Unesp), Jaboticabal, São Paulo 14884-900 Brazil
| | - Luciana Diniz Rola
- Departamento de Zootecnia, Núcleo de Pesquisa e Conservação de Cervídeos, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista (Unesp), Jaboticabal, São Paulo 14884-900 Brazil
| | | | - Fabiana Barichello Mokry
- Department of Genetics and Evolution, Federal University of São Carlos (UFSCar), São Carlos, São Paulo 13565-905 Brazil
| | | | | | | | | | | | - Danísio Prado Munari
- Departamento de Ciências Exatas, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista (Unesp), Jaboticabal, São Paulo 14884-900 Brazil
| |
Collapse
|
17
|
De Conto F, Fazzi A, Razin SV, Arcangeletti MC, Medici MC, Belletti S, Chezzi C, Calderaro A. Mammalian Diaphanous-related formin-1 restricts early phases of influenza A/NWS/33 virus (H1N1) infection in LLC-MK2 cells by affecting cytoskeleton dynamics. Mol Cell Biochem 2017; 437:185-201. [PMID: 28744815 DOI: 10.1007/s11010-017-3107-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 07/01/2017] [Indexed: 12/15/2022]
Abstract
Viruses depend on cellular machinery to efficiently replicate. The host cytoskeleton is one of the first cellular systems hijacked by viruses in order to ensure their intracellular transport and promote the development of infection. Our previous results demonstrated that stable microfilaments and microtubules interfered with human influenza A/NWS/33 virus (H1N1) infection in semi-permissive LLC-MK2 cells. Although formins play a key role in cytoskeletal remodelling, few studies addressed a possible role of these proteins in development of viral infection. Here, we have demonstrated that mammalian Diaphanous-related formin-1 (mDia1) is involved in the control of cytoskeleton dynamics during human influenza A virus infection. First, by employing cytoskeleton-perturbing drugs, we evidenced a cross-talk occurring between microtubules and microfilaments that also has implications on the intracellular localization of mDia1. In influenza A/NWS/33 virus-infected LLC-MK2 cells, mDia1 showed a highly dynamic intracellular localization and partially co-localized with actin and tubulin. A depletion of mDia1 by RNA-mediated RNA interference was found to improve the outcome of influenza A/NWS/33 virus infection and to increase the dynamics of microfilament and microtubule networks in LLC-MK2 cells. Consistent with these findings, observations made in epithelial respiratory cells from paediatric patients with acute respiratory disease assessed that the expression of mDia1 is stimulated by influenza A virus but not by respiratory syncytial virus. Taken together, the obtained results suggest that mDia1 restricts the initiation of influenza A/NWS/33 virus infection in LLC-MK2 cells by counteracting cytoskeletal dynamics.
Collapse
Affiliation(s)
- Flora De Conto
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | - Alessandra Fazzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Sergey V Razin
- Institute of Gene Biology, Russian Academy of Sciences and Lomonosow Moscow State University, Moscow, Russia
| | | | | | - Silvana Belletti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Carlo Chezzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Adriana Calderaro
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
18
|
Szikora S, Földi I, Tóth K, Migh E, Vig A, Bugyi B, Maléth J, Hegyi P, Kaltenecker P, Sanchez-Soriano N, Mihály J. The formin DAAM is required for coordination of the actin and microtubule cytoskeleton in axonal growth cones. J Cell Sci 2017; 130:2506-2519. [PMID: 28606990 DOI: 10.1242/jcs.203455] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/05/2017] [Indexed: 01/10/2023] Open
Abstract
Directed axonal growth depends on correct coordination of the actin and microtubule cytoskeleton in the growth cone. However, despite the relatively large number of proteins implicated in actin-microtubule crosstalk, the mechanisms whereby actin polymerization is coupled to microtubule stabilization and advancement in the peripheral growth cone remained largely unclear. Here, we identified the formin Dishevelled-associated activator of morphogenesis (DAAM) as a novel factor playing a role in concerted regulation of actin and microtubule remodeling in Drosophilamelanogaster primary neurons. In vitro, DAAM binds to F-actin as well as to microtubules and has the ability to crosslink the two filament systems. Accordingly, DAAM associates with the neuronal cytoskeleton, and a significant fraction of DAAM accumulates at places where the actin filaments overlap with that of microtubules. Loss of DAAM affects growth cone and microtubule morphology, and several aspects of microtubule dynamics; and biochemical and cellular assays revealed a microtubule stabilization activity and binding to the microtubule tip protein EB1. Together, these data suggest that, besides operating as an actin assembly factor, DAAM is involved in linking actin remodeling in filopodia to microtubule stabilization during axonal growth.
Collapse
Affiliation(s)
- Szilárd Szikora
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - István Földi
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Krisztina Tóth
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Ede Migh
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Andrea Vig
- University of Pécs, Medical School, Department of Biophysics, Szigeti str. 12, Pécs H-7624, Hungary
| | - Beáta Bugyi
- University of Pécs, Medical School, Department of Biophysics, Szigeti str. 12, Pécs H-7624, Hungary.,Szentágothai Research Center, Ifjúság str. 34, Pécs H-7624, Hungary
| | - József Maléth
- MTA-SZTE Translational Gastroenterology Research Group, First Department of Internal Medicine, Szeged H-6720, Hungary
| | - Péter Hegyi
- MTA-SZTE Translational Gastroenterology Research Group, First Department of Internal Medicine, Szeged H-6720, Hungary.,Institute for Translational Medicine, Department of Pathophysiology, University of Pécs, Pécs H-7624, Hungary
| | - Péter Kaltenecker
- Institute for Translational Medicine, Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool L69 3BX, UK
| | - Natalia Sanchez-Soriano
- Institute for Translational Medicine, Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool L69 3BX, UK
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| |
Collapse
|
19
|
Shevchenko G. Participation of proteins binding both actin filaments and microtubules in higher plant cell growth. CYTOL GENET+ 2015. [DOI: 10.3103/s009545271504009x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
Li N, Mruk DD, Tang EI, Wong CK, Lee WM, Silvestrini B, Cheng CY. Formins: Actin nucleators that regulate cytoskeletal dynamics during spermatogenesis. SPERMATOGENESIS 2015; 5:e1066476. [PMID: 26413414 DOI: 10.1080/21565562.2015.1066476] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/18/2015] [Accepted: 06/18/2015] [Indexed: 12/21/2022]
Abstract
Formins are a growing class of actin nucleation proteins that promote the polymerization of actin microfilaments, forming long stretches of actin microfilaments to confer actin filament bundling in mammalian cells. As such, microfilament bundles can be formed in specific cellular domains, in particular in motile mammalian cells, such as filopodia. Since ectoplasmic specialization (ES), a testis-specific adherens junction (AJ), at the Sertoli cell-cell and Sertoli-spermatid interface is constituted by arrays of actin microfilament bundles, it is likely that formins are playing a significant physiological role on the homeostasis of ES during the epithelial cycle of spermatogenesis. In this Commentary, we provide a timely discussion on formin 1 which was recently shown to be a crucial regulator of actin microfilaments at the ES in the rat testis (Li N et al. Endocrinology, 2015, in press; DOI: 10.1210/en.2015-1161, PMID:25901598). We also highlight research that is needed to unravel the functional significance of formins in spermatogenesis.
Collapse
Affiliation(s)
- Nan Li
- The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; Population Council ; New York, NY USA
| | - Dolores D Mruk
- The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; Population Council ; New York, NY USA
| | - Elizabeth I Tang
- The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; Population Council ; New York, NY USA
| | - Chris Kc Wong
- Department of Biology; Hong Kong Baptist University ; Hong Kong, China
| | - Will M Lee
- School of Biological Sciences; University of Hong Kong ; Hong Kong, China
| | | | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; Population Council ; New York, NY USA
| |
Collapse
|
21
|
SMIFH2 has effects on Formins and p53 that perturb the cell cytoskeleton. Sci Rep 2015; 5:9802. [PMID: 25925024 PMCID: PMC5386218 DOI: 10.1038/srep09802] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 03/19/2015] [Indexed: 01/08/2023] Open
Abstract
Formin proteins are key regulators of the cytoskeleton involved in developmental and homeostatic programs, and human disease. For these reasons, small molecules interfering with Formins' activity have gained increasing attention. Among them, small molecule inhibitor of Formin Homology 2 domains (SMIFH2) is often used as a pharmacological Formin blocker. Although SMIFH2 inhibits actin polymerization by Formins and affects the actin cytoskeleton, its cellular mechanism of action and target specificity remain unclear. Here we show that SMIFH2 induces remodelling of actin filaments, microtubules and the Golgi complex as a result of its effects on Formins and p53. We found that SMIFH2 triggers alternated depolymerization-repolymerization cycles of actin and tubulin, increases cell migration, causes scattering of the Golgi complex, and also cytotoxicity at high dose. Moreover, SMIFH2 reduces expression and activity of p53 through a post-transcriptional, proteasome-independent mechanism that influences remodelling of the cytoskeleton. As the action of SMIFH2 may go beyond Formin inhibition, only short-term and low-dose SMIFH2 treatments minimize confounding effects induced by loss of p53 and cytotoxicity.
Collapse
|
22
|
Roth-Johnson EA, Vizcarra CL, Bois JS, Quinlan ME. Interaction between microtubules and the Drosophila formin Cappuccino and its effect on actin assembly. J Biol Chem 2013; 289:4395-404. [PMID: 24362037 DOI: 10.1074/jbc.m113.499921] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Formin family actin nucleators are potential coordinators of the actin and microtubule cytoskeletons, as they can both nucleate actin filaments and bind microtubules in vitro. To gain a more detailed mechanistic understanding of formin-microtubule interactions and formin-mediated actin-microtubule cross-talk, we studied microtubule binding by Cappuccino (Capu), a formin involved in regulating actin and microtubule organization during Drosophila oogenesis. We found that two distinct domains within Capu, FH2 and tail, work together to promote high-affinity microtubule binding. The tail domain appears to bind microtubules through nonspecific charge-based interactions. In contrast, distinct residues within the FH2 domain are important for microtubule binding. We also report the first visualization of a formin polymerizing actin filaments in the presence of microtubules. Interestingly, microtubules are potent inhibitors of the actin nucleation activity of Capu but appear to have little effect on Capu once it is bound to the barbed end of an elongating filament. Because Capu does not simultaneously bind microtubules and assemble actin filaments in vitro, its actin assembly and microtubule binding activities likely require spatial and/or temporal regulation within the Drosophila oocyte.
Collapse
|
23
|
Thurston SF, Kulacz WA, Shaikh S, Lee JM, Copeland JW. The ability to induce microtubule acetylation is a general feature of formin proteins. PLoS One 2012; 7:e48041. [PMID: 23110170 PMCID: PMC3480493 DOI: 10.1371/journal.pone.0048041] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 09/20/2012] [Indexed: 01/23/2023] Open
Abstract
Cytoplasmic microtubules exist as distinct dynamic and stable populations within the cell. Stable microtubules direct and maintain cell polarity and it is thought that their stabilization is dependent on coordinative organization between the microtubule network and the actin cytoskeleton. A growing body of work suggests that some members of the formin family of actin remodeling proteins also regulate microtubule organization and stability. For example, we showed previously that expression of the novel formin INF1 is sufficient to induce microtubule stabilization and tubulin acetylation, but not tubulin detyrosination. An important issue with respect to the relationship between formins and microtubules is the determination of which formin domains mediate microtubule stabilization. INF1 has a distinct microtubule-binding domain at its C-terminus and the endogenous INF1 protein is associated with the microtubule network. Surprisingly, the INF1 microtubule-binding domain is not essential for INF1-induced microtubule acetylation. We show here that expression of the isolated FH1 + FH2 functional unit of INF1 is sufficient to induce microtubule acetylation independent of the INF1 microtubule-binding domain. It is not yet clear whether or not microtubule stabilization is a general property of all mammalian formins; therefore we expressed constitutively active derivatives of thirteen of the fifteen mammalian formin proteins in HeLa and NIH3T3 cells and measured their effects on stress fiber formation, MT organization and MT acetylation. We found that expression of the FH1 + FH2 unit of the majority of mammalian formins is sufficient to induce microtubule acetylation. Our results suggest that the regulation of microtubule acetylation is likely a general formin activity and that the FH2 should be thought of as a dual-function domain capable of regulating both actin and microtubule networks.
Collapse
Affiliation(s)
- Susan F. Thurston
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Wojciech A. Kulacz
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Sahir Shaikh
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Jonathan M. Lee
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - John W. Copeland
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- * E-mail:
| |
Collapse
|
24
|
Santos-Ledo A, Jenny A, Marlow FL. Comparative gene expression analysis of the fmnl family of formins during zebrafish development and implications for tissue specific functions. Gene Expr Patterns 2012; 13:30-7. [PMID: 23072729 DOI: 10.1016/j.gep.2012.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 08/14/2012] [Accepted: 09/06/2012] [Indexed: 10/27/2022]
Abstract
Fmlns belong to the Formin family, catalysts of linear actin polymerization with mostly unknown roles in vivo. In cell culture Fmnls are involved in cell migration and adhesion and the formation of different types of protrusions including filopodia and blebs, suggesting important roles during development. Moreover, Fmnls can act downstream of Rac and Cdc42, mediators of cytoskeletal changes as targets of important pathways required for shaping tissues. The zebrafish genome encodes five Fmnls. Here we report their tissue specific expression patterns during early development and pharyngula stages. The fmnls show overlapping and distinct expression patterns, which suggest that they could regulate similar processes during development, but may also have independent functions. In particular, we find a strong maternal contribution of all fmnls, but distinct expression patterns in the developing brain eye, ear, heart and vascular system.
Collapse
Affiliation(s)
- Adrián Santos-Ledo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA
| | | | | |
Collapse
|
25
|
Wang J, Xue X, Ren H. New insights into the role of plant formins: regulating the organization of the actin and microtubule cytoskeleton. PROTOPLASMA 2012; 249 Suppl 2:S101-7. [PMID: 22215231 DOI: 10.1007/s00709-011-0368-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 12/13/2011] [Indexed: 05/03/2023]
Abstract
Formins are well-known as important regulators participating in the organization of the actin cytoskeleton in organisms. For many years in the past, research on plant formins is more difficult than that in other eukaryotic formins and is limited to class I formins. Nevertheless, positive progress has been made in plant formin research recently, especially the investigations on class II formins. New functions of plant formins are identified gradually, such as regulating cell division and affecting diffuse cell expansion. More significantly, plant formins are also verified to interact with microtubules in vivo and in vitro. They may probably function as linking proteins between microtubules and microfilaments to participate in various cellular processes.
Collapse
Affiliation(s)
- Jiaojiao Wang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education and College of Life Science, Beijing Normal University, Beijing 100875, China
| | | | | |
Collapse
|
26
|
Schevzov G, Curthoys NM, Gunning PW, Fath T. Functional diversity of actin cytoskeleton in neurons and its regulation by tropomyosin. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 298:33-94. [PMID: 22878104 DOI: 10.1016/b978-0-12-394309-5.00002-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurons comprise functionally, molecularly, and spatially distinct subcellular compartments which include the soma, dendrites, axon, branches, dendritic spines, and growth cones. In this chapter, we detail the remarkable ability of the neuronal cytoskeleton to exquisitely regulate all these cytoplasmic distinct partitions, with particular emphasis on the microfilament system and its plethora of associated proteins. Importance will be given to the family of actin-associated proteins, tropomyosin, in defining distinct actin filament populations. The ability of tropomyosin isoforms to regulate the access of actin-binding proteins to the filaments is believed to define the structural diversity and dynamics of actin filaments and ultimately be responsible for the functional outcome of these filaments.
Collapse
Affiliation(s)
- Galina Schevzov
- Oncology Research Unit, Department of Pharmacology, School of Medical Sciences, University of New South Wales, Kensington, Australia
| | | | | | | |
Collapse
|
27
|
Gaillard J, Ramabhadran V, Neumanne E, Gurel P, Blanchoin L, Vantard M, Higgs HN. Differential interactions of the formins INF2, mDia1, and mDia2 with microtubules. Mol Biol Cell 2011; 22:4575-87. [PMID: 21998204 PMCID: PMC3226476 DOI: 10.1091/mbc.e11-07-0616] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Three mammalian formins, although binding microtubules with high affinity, differ dramatically in their microtubule-binding mechanisms. In addition, the ability of one formin (mDia2) to bind actin is strongly inhibited by microtubules, whereas the ability of another formin (INF2) to bind microtubules is strongly inhibited by actin monomers. A number of cellular processes use both microtubules and actin filaments, but the molecular machinery linking these two cytoskeletal elements remains to be elucidated in detail. Formins are actin-binding proteins that have multiple effects on actin dynamics, and one formin, mDia2, has been shown to bind and stabilize microtubules through its formin homology 2 (FH2) domain. Here we show that three formins, INF2, mDia1, and mDia2, display important differences in their interactions with microtubules and actin. Constructs containing FH1, FH2, and C-terminal domains of all three formins bind microtubules with high affinity (Kd < 100 nM). However, only mDia2 binds microtubules at 1:1 stoichiometry, with INF2 and mDia1 showing saturating binding at approximately 1:3 (formin dimer:tubulin dimer). INF2-FH1FH2C is a potent microtubule-bundling protein, an effect that results in a large reduction in catastrophe rate. In contrast, neither mDia1 nor mDia2 is a potent microtubule bundler. The C-termini of mDia2 and INF2 have different functions in microtubule interaction, with mDia2's C-terminus required for high-affinity binding and INF2's C-terminus required for bundling. mDia2's C-terminus directly binds microtubules with submicromolar affinity. These formins also differ in their abilities to bind actin and microtubules simultaneously. Microtubules strongly inhibit actin polymerization by mDia2, whereas they moderately inhibit mDia1 and have no effect on INF2. Conversely, actin monomers inhibit microtubule binding/bundling by INF2 but do not affect mDia1 or mDia2. These differences in interactions with microtubules and actin suggest differential function in cellular processes requiring both cytoskeletal elements.
Collapse
Affiliation(s)
- Jeremie Gaillard
- CEA, iRTSV, Laboratoire Physiologie Cellulaire & Végétale, CNRS, UMR5168, Université Joseph Fourier-Grenoble I, F-38054 Grenoble, France
| | | | | | | | | | | | | |
Collapse
|
28
|
Simon-Areces J, Dopazo A, Dettenhofer M, Rodriguez-Tebar A, Garcia-Segura LM, Arevalo MA. Formin1 mediates the induction of dendritogenesis and synaptogenesis by neurogenin3 in mouse hippocampal neurons. PLoS One 2011; 6:e21825. [PMID: 21818269 PMCID: PMC3139584 DOI: 10.1371/journal.pone.0021825] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 06/13/2011] [Indexed: 11/21/2022] Open
Abstract
Neurogenin3, a proneural transcription factor controlled by Notch receptor, has been recently shown to regulate dendritogenesis and synaptogenesis in mouse hippocampal neurons. However, little is known about the molecular mechanisms involved in these actions of Ngn3. We have used a microarray analysis to identify Ngn3 regulated genes related with cytoskeleton dynamics. One of such genes is Fmn1, whose protein, Formin1, is associated with actin and microtubule cytoskeleton. Overexpression of the Fmn1 isoform-Ib in cultured mouse hippocampal neurons induced an increase in the number of primary dendrites and in the number of glutamatergic synaptic inputs at 4 days in vitro. The same changes were provoked by overexpression of Ngn3. In addition downregulation of Fmn1 by the use of Fmn1-siRNAs impaired such morphological and synaptic changes induced by Ngn3 overexpression in neurons. These results reveal a previously unknown involvement of Formin1 in dendritogenesis and synaptogenesis and indicate that this protein is a key component of the Ngn3 signaling pathway that controls neuronal differentiation.
Collapse
Affiliation(s)
- Julia Simon-Areces
- Laboratory of Neuroactive Steroids, Instituto Cajal, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid, Spain
| | - Ana Dopazo
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Markus Dettenhofer
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Alfredo Rodriguez-Tebar
- Centro Andaluz de Biología Molecular y Medicina Regenerativa/Consejo Superior de Investigaciones Cientificas (CABIMER/CSIC), Seville, Spain
| | - Luis Miguel Garcia-Segura
- Laboratory of Neuroactive Steroids, Instituto Cajal, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid, Spain
| | - Maria-Angeles Arevalo
- Laboratory of Neuroactive Steroids, Instituto Cajal, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid, Spain
- * E-mail:
| |
Collapse
|
29
|
Zhang Z, Zhang Y, Tan H, Wang Y, Li G, Liang W, Yuan Z, Hu J, Ren H, Zhang D. RICE MORPHOLOGY DETERMINANT encodes the type II formin FH5 and regulates rice morphogenesis. THE PLANT CELL 2011; 23:681-700. [PMID: 21307283 PMCID: PMC3077795 DOI: 10.1105/tpc.110.081349] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 01/01/2011] [Accepted: 01/18/2011] [Indexed: 05/19/2023]
Abstract
Multicellular organisms contain a large number of formins; however, their physiological roles in plants remain poorly understood. Here, we reveal that formin homology 5 (FH5), a type II formin mutated in rice morphology determinant (rmd), plays a crucial role in determining rice (Oryza sativa) morphology. FH5/RMD encodes a formin-like protein consisting of an N-terminal phosphatase tensin (PTEN)-like domain, an FH1 domain, and an FH2 domain. The rmd mutants display a bending growth pattern in seedlings, are stunted as adult plants, and have aberrant inflorescence (panicle) and seed shape. Cytological analysis showed that rmd mutants have severe cell elongation defects and abnormal microtubule and microfilament arrays. FH5/RMD is ubiquitously expressed in rice tissues, and its protein localization to the chloroplast surface is mediated by the PTEN domain. Biochemical assays demonstrated that recombinant FH5 protein can nucleate actin polymerization from monomeric G-actin or actin/profilin complexes, cap the barbed end of actin filaments, and bundle actin filaments in vitro. Moreover, FH5 can directly bind to and bundle microtubules through its FH2 domain in vitro. Our findings suggest that the rice formin protein FH5 plays a critical role in determining plant morphology by regulating actin dynamics and proper spatial organization of microtubules and microfilaments.
Collapse
Affiliation(s)
- Zheng Zhang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yi Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology of the Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, China
| | - Hexin Tan
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ying Wang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Gang Li
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wanqi Liang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zheng Yuan
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianping Hu
- Michigan State University–Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, Michigan 48824
- Department of Plant Biology, Michigan State University, East Lansing, Michigan 48824
| | - Haiyun Ren
- Key Laboratory of Cell Proliferation and Regulation Biology of the Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, China
| | - Dabing Zhang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Bio-X Center, Key Laboratory of Genetics and Development and Neuropsychiatric Diseases, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
- Address correspondence to
| |
Collapse
|
30
|
Kwon S, Shin H, Lim HJ. Dynamic interaction of formin proteins and cytoskeleton in mouse oocytes during meiotic maturation. Mol Hum Reprod 2010; 17:317-27. [DOI: 10.1093/molehr/gaq088] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
31
|
Li Y, Shen Y, Cai C, Zhong C, Zhu L, Yuan M, Ren H. The type II Arabidopsis formin14 interacts with microtubules and microfilaments to regulate cell division. THE PLANT CELL 2010; 22:2710-26. [PMID: 20709814 PMCID: PMC2947165 DOI: 10.1105/tpc.110.075507] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 07/17/2010] [Accepted: 07/28/2010] [Indexed: 05/18/2023]
Abstract
Formins have long been known to regulate microfilaments but have also recently been shown to associate with microtubules. In this study, Arabidopsis thaliana FORMIN14 (AFH14), a type II formin, was found to regulate both microtubule and microfilament arrays. AFH14 expressed in BY-2 cells was shown to decorate preprophase bands, spindles, and phragmoplasts and to induce coalignment of microtubules with microfilaments. These effects perturbed the process of cell division. Localization of AFH14 to microtubule-based structures was confirmed in Arabidopsis suspension cells. Knockdown of AFH14 in mitotic cells altered interactions between microtubules and microfilaments, resulting in the formation of an abnormal mitotic apparatus. In Arabidopsis afh14 T-DNA insertion mutants, microtubule arrays displayed abnormalities during the meiosis-associated process of microspore formation, which corresponded to altered phenotypes during tetrad formation. In vitro biochemical experiments showed that AFH14 bound directly to either microtubules or microfilaments and that the FH2 domain was essential for cytoskeleton binding and bundling. However, in the presence of both microtubules and microfilaments, AFH14 promoted interactions between microtubules and microfilaments. These results demonstrate that AFH14 is a unique plant formin that functions as a linking protein between microtubules and microfilaments and thus plays important roles in the process of plant cell division.
Collapse
Affiliation(s)
- Yanhua Li
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Yuan Shen
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Chao Cai
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Chenchun Zhong
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Lei Zhu
- State Key Laboratory of Plant Physiology and Biochemistry, China Agricultural University, Beijing 100094, People's Republic of China
| | - Ming Yuan
- State Key Laboratory of Plant Physiology and Biochemistry, China Agricultural University, Beijing 100094, People's Republic of China
| | - Haiyun Ren
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, People's Republic of China
| |
Collapse
|
32
|
Fitzgerald LM, McDonnell SK, Carlson EE, Langeberg W, McIntosh LM, Deutsch K, Ostrander EA, Schaid DJ, Stanford JL. Genome-wide linkage analyses of hereditary prostate cancer families with colon cancer provide further evidence for a susceptibility locus on 15q11-q14. Eur J Hum Genet 2010; 18:1141-7. [PMID: 20407467 DOI: 10.1038/ejhg.2010.49] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The search for susceptibility loci in hereditary prostate cancer (HPC) is challenging because of locus and disease heterogeneity. One approach to reduce disease heterogeneity is to stratify families on the basis of the occurrence of multiple cancer types. This method may increase the power for detecting susceptibility loci, including those with pleiotropic effects. We have completed a genome-wide SNP linkage analysis of 96 HPC families, each of which has one or more first-degree relatives with colon cancer (CCa), and further analyzed the subset of families with two or more CCa cases (n = 27). When only a prostate cancer (PCa) phenotype was considered to be affected, we observed suggestive evidence for linkage (LOD ≥1.86) at 15q14, 18q21 and 19q13 in all families, and at 1p32 and 15q11-q14 in families with two or more CCa cases. When both the PCa and CCa phenotypes were considered affected, suggestive evidence for linkage was observed at 11q25, 15q14 and 18q21 in all families, and at 1q31, 11q14 and 15q11-14 in families with two or more CCa cases. The strongest linkage signal was identified at 15q14 when both PCa and CCa phenotypes were considered to be affected in families with two or more CCa cases (recessive HLOD = 3.88). These results provide further support for the presence of HPC susceptibility loci on chromosomes 11q14, 15q11-q14 and 19q13 and highlight loci at 1q31, 11q, 15q11-14 and 18q21 as having possible pleiotropic effects. This study shows the benefit of using a comprehensive family cancer history to create more genetically homogenous subsets of HPC families for linkage analyses.
Collapse
Affiliation(s)
- Liesel M Fitzgerald
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Schönichen A, Geyer M. Fifteen formins for an actin filament: a molecular view on the regulation of human formins. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1803:152-63. [PMID: 20102729 DOI: 10.1016/j.bbamcr.2010.01.014] [Citation(s) in RCA: 182] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2009] [Revised: 12/24/2009] [Accepted: 01/11/2010] [Indexed: 10/19/2022]
Abstract
The regulation of the actin cytoskeleton is a key process for the stability and motility of eukaryotic cells. Besides the Arp2/3 complex and its nucleation promoting factors, WH2 domain-containing proteins and a diverse family of formin proteins have recently been recognized as actin nucleators and potent polymerization factors of actin filaments. Formins are defined by the presence of a catalytic formin homology 2 (FH2) domain, yet, the modular domain architecture appears significantly different for the eight formin families identified in humans. A diverse picture of protein localization, interaction partners and cell specific regulation emerged, suggesting various functions of formins in the building and maintenance of actin filaments. This review focuses on the domain architecture of human formins, the regulation mechanisms of their activation and the diversity in formin cellular functions.
Collapse
Affiliation(s)
- André Schönichen
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Physikalische Biochemie, Otto-Hahn-Str. 11, 44227 Dortmund, Germany
| | | |
Collapse
|
34
|
Chesarone MA, DuPage AG, Goode BL. Unleashing formins to remodel the actin and microtubule cytoskeletons. Nat Rev Mol Cell Biol 2009; 11:62-74. [PMID: 19997130 DOI: 10.1038/nrm2816] [Citation(s) in RCA: 403] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Formins are highly conserved proteins that have essential roles in remodelling the actin and microtubule cytoskeletons to influence eukaryotic cell shape and behaviour. Recent work has identified numerous cellular factors that locally recruit, activate or inactivate formins to bridle and unleash their potent effects on actin nucleation and elongation. The effects of formins on microtubules have also begun to be described, which places formins in a prime position to coordinate actin and microtubule dynamics. The emerging complexity in the mechanisms governing formins mirrors the wide range of essential functions that they perform in cell motility, cell division and cell and tissue morphogenesis.
Collapse
Affiliation(s)
- Melissa A Chesarone
- Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, Massachusetts 02454, USA
| | | | | |
Collapse
|
35
|
Bartolini F, Gundersen GG. Formins and microtubules. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1803:164-73. [PMID: 19631698 DOI: 10.1016/j.bbamcr.2009.07.006] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 05/29/2009] [Accepted: 07/04/2009] [Indexed: 02/06/2023]
Abstract
Formins have recently been recognized as prominent regulators of the microtubule (MT) cytoskeleton where they modulate the dynamics of selected MTs in interphase and mitosis. The association of formins with the MT cytoskeleton and their action on MT dynamics are relatively unexplored areas, yet growing evidence supports a direct role in their regulation of MT stability independent of their activity on actin. Formins regulate MT stability alone or in combination with accessory MT binding proteins that have previously been implicated in the stabilization of MTs downstream of polarity cues. As actin and MT arrays are typically remodeled downstream of signaling pathways that orchestrate cell shape and division, formins are emerging as excellent candidates for coordinating the responses of the cytoskeletal in diverse regulated and homeostatic processes.
Collapse
Affiliation(s)
- F Bartolini
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | | |
Collapse
|
36
|
Zhou F, Leder P, Zuniga A, Dettenhofer M. Formin1 disruption confers oligodactylism and alters Bmp signaling. Hum Mol Genet 2009; 18:2472-82. [PMID: 19383632 DOI: 10.1093/hmg/ddp185] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Proper limb development requires concerted communication between cells within the developing limb bud. Several molecules have been identified which contribute to the formation of a circuitry loop consisting in large part of secreted proteins. The intracellular actin nucleator, Formin 1 (Fmn1), has previously been implicated in limb development, but questions remain after the identification of a Gremlin transcriptional enhancer within the 3' end of the Fmn 1 locus. To resolve this issue, a knockout mouse devoid of Fmn1 protein was created and characterized. The mice exhibit a reduction of digit number to four, a deformed posterior metatarsal, phalangeal soft tissue fusion as well as the absence of a fibula to 100% penetrance in the FVB genetic background. Importantly, this mutant allele does not genetically disrupt the characterized Gremlin enhancer, and indeed Gremlin RNA expression is upregulated at the 35 somite stage of development. Our data reveal increased Bone Morphogenetic Protein (Bmp) activity in mice which carry a disruption in Fmn1, as evidenced by upregulation of Msx1 and a decrease in Fgf4 within the apical ectodermal ridge. Additionally, these studies show enhanced activity downstream of the Bmp receptor in cells where Fmn1 is perturbed, suggesting a role for Fmn1 in repression of Bmp signaling.
Collapse
Affiliation(s)
- Fen Zhou
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
37
|
Formin 1-isoform IV deficient cells exhibit defects in cell spreading and focal adhesion formation. PLoS One 2008; 3:e2497. [PMID: 18560567 PMCID: PMC2423616 DOI: 10.1371/journal.pone.0002497] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Accepted: 05/22/2008] [Indexed: 12/23/2022] Open
Abstract
Background Regulation of the cytoskeleton is a central feature of cell migration. The formin family of proteins controls the rate of actin nucleation at its barbed end. Thus, formins are predicted to contribute to several important cell processes such as locomotion, membrane ruffling, vesicle endocytosis, and stress fiber formation and disassociation. Methodology/Principal Findings In this study we investigated the functional role of Formin1-isoform4 (Fmn1-IV) by using genetically null primary cells that displayed augmented protrusive behaviour during wound healing and delayed cell spreading. Cells deficient of Fmn1-IV also showed reduced efficiency of focal adhesion formation. Additionally, we generated an enhanced green fluorescence protein (EGFP)-fused Fmn1-IV knock-in mouse to monitor the endogenous subcellular localization of Fmn1-IV. Its localization was found within the cytoplasm and along microtubules, yet it was largely excluded from adherens junctions. Conclusions/Significance It was determined that Fmn1-IV, as an actin nucleator, contributes to protrusion of the cell's leading edge and focal adhesion formation, thus contributing to cell motility.
Collapse
|
38
|
Shtutman M, Chausovsky A, Prager-Khoutorsky M, Schiefermeier N, Boguslavsky S, Kam Z, Fuchs E, Geiger B, Borisy GG, Bershadsky AD. Signaling function of alpha-catenin in microtubule regulation. Cell Cycle 2008; 7:2377-83. [PMID: 18677116 DOI: 10.4161/cc.6362] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Centrosomes control microtubule dynamics in many cell types, and their removal from the cytoplasm leads to a shift from dynamic instability to treadmilling behavior and to a dramatic decrease of microtubule mass (Rodionov et al., 1999; PNAS 96:115). In cadherin-expressing cells, these effects can be reversed:non-centrosomal cytoplasts that form cadherin-mediated adherens junctions display dense arrays of microtubules (Chausovsky et al., 2000; Nature Cell Biol 2:797). In adherens junctions, cadherin's cytoplasmic domain binds p120 catenin and beta-catenin, which in turn binds alpha-catenin. To elucidate the roles of the cadherin-associated proteins in regulating microtubule dynamics, we prepared GFP-tagged, plasma membrane targeted or untargeted p120 catenin, alpha-catenin and beta-catenin and tested their ability to rescue the loss of microtubule mass caused by centrosomal removal in the poorly adhesive cell line CHO-K1. Only membrane targeting of alpha-catenin led to a significant increase in microtubule length and density in centrosome-free cytoplasts. Expression of non-membrane-targeted alpha-catenin produced only a slight effect, while both membrane-targeted and non-targeted p120 and beta-catenin were ineffective in this assay. Together, these findings suggest that alpha-catenin is able to regulate microtubule dynamics in a centrosome-independent manner.
Collapse
Affiliation(s)
- Michael Shtutman
- Cancer Center, Ordway Research Institute, Albany, New York, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Profilins are small proteins involved in actin dynamics. In accordance with this function, they are found in all eukaryotes and are structurally highly conserved. However, their precise role in regulating actin-related functions is just beginning to emerge. This article recapitulates the wealth of information on structure, expression and functions accumulated on profilins from many different organisms in the 30 years after their discovery as actin-binding proteins. Emphasis is given to their interaction with a plethora of many different ligands in the cytoplasm as well as in the nucleus, which is considered the basis for their various activities and the significance of the tissue-specific expression of profilin isoforms.
Collapse
Affiliation(s)
- B M Jockusch
- Cell Biology, Zoological Institute, Technical University of Braunschweig, 38092 Braunschweig, Germany.
| | | | | |
Collapse
|