1
|
Luan Y, Deng Z, Zhu Y, Dai L, Yang Y, Xia Z. Decoupling actin assembly from microtubule disassembly by TBC1D3C-mediated direct GEF-H1 activation. Life Sci Alliance 2025; 8:e202402585. [PMID: 39467635 PMCID: PMC11519374 DOI: 10.26508/lsa.202402585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 10/30/2024] Open
Abstract
Actin and microtubules are essential cytoskeletal components and coordinate their dynamics through multiple coupling and decoupling mechanisms. However, how actin and microtubule dynamics are decoupled remains incompletely understood. Here, we identified TBC1D3C as a new regulator that can decouple actin filament assembly from microtubule disassembly. We showed that TBC1D3C induces the release of GEF-H1 from microtubules into the cytosol without perturbing microtubule arrays, leading to RhoA activation and actin filament assembly. Mechanistically, we found that TBC1D3C directly binds to GEF-H1, disrupting its interaction with the Tctex-DIC-14-3-3 complex and thereby displacing GEF-H1 from microtubules independently of microtubule disassembly. Super-resolution microscopy and live-cell imaging further confirmed that TBC1D3C triggers GEF-H1 release and actin filament assembly while maintaining microtubule integrity. Therefore, our findings demonstrated that TBC1D3C functions as a direct GEF activator and a novel regulator in decoupling actin assembly from microtubule disassembly, providing new insights into cytoskeletal regulation.
Collapse
Affiliation(s)
- Yi Luan
- https://ror.org/056swr059 Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, China
- https://ror.org/056swr059 Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- https://ror.org/056swr059 NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhifeng Deng
- https://ror.org/056swr059 Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, China
- https://ror.org/056swr059 Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- https://ror.org/056swr059 NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yutong Zhu
- Research and Development Center, Beijing, China
| | - Lisi Dai
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Yang Yang
- https://ror.org/056swr059 Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zongping Xia
- https://ror.org/056swr059 Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, China
- https://ror.org/056swr059 Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- https://ror.org/056swr059 NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| |
Collapse
|
2
|
de Castro Sampaio SS, Ramalho MCC, de Souza CS, de Almeida Rodrigues B, de Mendonça GRS, Lazarini M. RHO subfamily of small GTPases in the development and function of hematopoietic cells. J Cell Physiol 2024:e31469. [PMID: 39434451 DOI: 10.1002/jcp.31469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/16/2024] [Accepted: 10/03/2024] [Indexed: 10/23/2024]
Abstract
RHOA, RHOB, and RHOC comprise a subfamily of RHO GTPase proteins famed for controlling cytoskeletal dynamics. RHO proteins operate downstream of multiple signals emerging from the microenvironment, leading to diverse cell responses, such as proliferation, adhesion, and migration. Therefore, RHO signaling has been centrally placed in the regulation of blood cells. Despite their high homology, unique roles of RHOA, RHOB, and RHOC have been described in hematopoietic cells. In this article, we overview the contribution of RHO proteins in the development and function of each blood cell lineage. Additionally, we highlight the aberrations of the RHO signaling pathways found in hematological malignancies, providing clues for the identification of new therapeutic targets.
Collapse
Affiliation(s)
| | | | - Caroline Santos de Souza
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Mariana Lazarini
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Tran QTH, Kondo N, Ueda H, Matsuo Y, Tsukaguchi H. Altered Endoplasmic Reticulum Integrity and Organelle Interactions in Living Cells Expressing INF2 Variants. Int J Mol Sci 2024; 25:9783. [PMID: 39337270 PMCID: PMC11431639 DOI: 10.3390/ijms25189783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
The cytoskeleton mediates fundamental cellular processes by organizing inter-organelle interactions. Pathogenic variants of inverted formin 2 (INF2) CAAX isoform, an actin assembly factor that is predominantly expressed in the endoplasmic reticulum (ER), are linked to focal segmental glomerulosclerosis (FSGS) and Charcot-Marie-Tooth (CMT) neuropathy. To investigate how pathogenic INF2 variants alter ER integrity, we used high-resolution live imaging of HeLa cells. Cells expressing wild-type (WT) INF2 showed a predominant tubular ER with perinuclear clustering. Cells expressing INF2 FSGS variants that cause mild and intermediate disease induced more sheet-like ER, a pattern similar to that seen for cells expressing WT-INF2 that were treated with actin and microtubule (MT) inhibitors. Dual CMT-FSGS INF2 variants led to more severe ER dysmorphism, with a diffuse, fragmented ER and coarse INF2 aggregates. Proper organization of both F-actin and MT was needed to modulate the tubule vs. sheet conformation balance, while MT arrays regulated spatial expansion of tubular ER in the cell periphery. Pathogenic INF2 variants also induced mitochondria fragmentation and dysregulated mitochondria distribution. Such mitochondrial abnormalities were more prominent for cells expressing CMT-FSGS compared to those with FSGS variants, indicating that the severity of the dysfunction is linked to the degree of cytoskeletal disorganization. Our observations suggest that pathogenic INF2 variants disrupt ER continuity by altering interactions between the ER and the cytoskeleton that in turn impairs inter-organelle communication, especially at ER-mitochondria contact sites. ER continuity defects may be a common disease mechanism involved in both peripheral neuropathy and glomerulopathy.
Collapse
Affiliation(s)
- Quynh Thuy Huong Tran
- Second Department of Internal Medicine, Division of Nephrology, Kansai Medical University, Hirakata 573-1010, Japan
| | - Naoyuki Kondo
- Department of Molecular Genetics, Institute of Biochemical Science, Kansai Medical University, Hirakata 573-1010, Japan
| | - Hiroko Ueda
- Second Department of Internal Medicine, Division of Nephrology, Kansai Medical University, Hirakata 573-1010, Japan
| | - Yoshiyuki Matsuo
- Central Research Center, Institute of Biomedical Science, Kansai Medical University, Hirakata 573-1010, Japan
| | - Hiroyasu Tsukaguchi
- Second Department of Internal Medicine, Division of Nephrology, Kansai Medical University, Hirakata 573-1010, Japan
- Clinical Genetics Center, Kansai Medical University Hospital, Hirakata 573-1191, Japan
| |
Collapse
|
4
|
Azizoglu ZB, Babayeva R, Haskologlu ZS, Acar MB, Ayaz-Guner S, Okus FZ, Alsavaf MB, Can S, Basaran KE, Canatan MF, Ozcan A, Erkmen H, Leblebici CB, Yilmaz E, Karakukcu M, Kose M, Canoz O, Özen A, Karakoc-Aydiner E, Ceylaner S, Gümüş G, Per H, Gumus H, Canatan H, Ozcan S, Dogu F, Ikinciogullari A, Unal E, Baris S, Eken A. DIAPH1-Deficiency is Associated with Major T, NK and ILC Defects in Humans. J Clin Immunol 2024; 44:175. [PMID: 39120629 PMCID: PMC11315734 DOI: 10.1007/s10875-024-01777-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024]
Abstract
Loss of function mutations in Diaphanous related formin 1 (DIAPH1) are associated with seizures, cortical blindness, and microcephaly syndrome (SCBMS) and are recently linked to combined immunodeficiency. However, the extent of defects in T and innate lymphoid cells (ILCs) remain unexplored. Herein, we characterized the primary T, natural killer (NK) and helper ILCs of six patients carrying two novel loss of function mutation in DIAPH1 and Jurkat cells after DIAPH1 knockdown. Mutations were identified by whole exome sequencing. T-cell immunophenotyping, proliferation, migration, cytokine signaling, survival, and NK cell cytotoxicity were studied via flow cytometry-based assays, confocal microscopy, and real-time qPCR. CD4+ T cell proteome was analyzed by mass spectrometry. p.R351* and p.R322*variants led to a significant reduction in the DIAPH1 mRNA and protein levels. DIAPH1-deficient T cells showed proliferation, activation, as well as TCR-mediated signaling defects. DIAPH1-deficient PBMCs also displayed impaired transwell migration, defective STAT5 phosphorylation in response to IL-2, IL-7 and IL-15. In vitro generation/expansion of Treg cells from naïve T cells was significantly reduced. shRNA-mediated silencing of DIAPH1 in Jurkat cells reduced DIAPH1 protein level and inhibited T cell proliferation and IL-2/STAT5 axis. Additionally, NK cells from patients had diminished cytotoxic activity, function and IL-2/STAT5 axis. Lastly, DIAPH1-deficient patients' peripheral blood contained dramatically reduced numbers of all helper ILC subsets. DIAPH1 deficiency results in major functional defects in T, NK cells and helper ILCs underlining the critical role of formin DIAPH1 in the biology of those cell subsets.
Collapse
Affiliation(s)
- Zehra Busra Azizoglu
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye
| | - Royala Babayeva
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Zehra Sule Haskologlu
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | | | - Serife Ayaz-Guner
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Türkiye
| | - Fatma Zehra Okus
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
| | | | - Salim Can
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Kemal Erdem Basaran
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey
| | | | - Alper Ozcan
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey
| | - Hasret Erkmen
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Can Berk Leblebici
- Department of Medical Genetics, Ankara University Faculty of Medicine, Ankara, Türkiye
| | - Ebru Yilmaz
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey
| | - Musa Karakukcu
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey
| | - Mehmet Kose
- Division of Pediatric Pulmonology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
| | - Ozlem Canoz
- Department of Pathology, Faculty of Medicine, Erciyes University, 38039, Kayseri, Türkiye
| | - Ahmet Özen
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Elif Karakoc-Aydiner
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Serdar Ceylaner
- Intergen, Genetic, Rare and Undiagnosed Diseases, Diagnosis and Research Center, Ankara, Türkiye
| | - Gülsüm Gümüş
- Division of Pediatric Radiology, Department of Radiology, Erciyes University Faculty of Medicine, Kayseri, Türkiye
| | - Huseyin Per
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
| | - Hakan Gumus
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
| | - Halit Canatan
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye
| | - Servet Ozcan
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye
- Department of Biology, Faculty of Science, Erciyes University, Kayseri, 38039, Türkiye
| | - Figen Dogu
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Aydan Ikinciogullari
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Ekrem Unal
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey.
- School of Health Sciences, Hasan Kalyoncu University, Gaziantep, Türkiye.
- Medical Point Hospital, Pediatric Hematology Oncology and BMT Unit, Gaziantep, Türkiye.
| | - Safa Baris
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Ahmet Eken
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye.
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye.
| |
Collapse
|
5
|
Kumari R, Ven K, Chastney M, Kokate SB, Peränen J, Aaron J, Kogan K, Almeida-Souza L, Kremneva E, Poincloux R, Chew TL, Gunning PW, Ivaska J, Lappalainen P. Focal adhesions contain three specialized actin nanoscale layers. Nat Commun 2024; 15:2547. [PMID: 38514695 PMCID: PMC10957975 DOI: 10.1038/s41467-024-46868-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024] Open
Abstract
Focal adhesions (FAs) connect inner workings of cell to the extracellular matrix to control cell adhesion, migration and mechanosensing. Previous studies demonstrated that FAs contain three vertical layers, which connect extracellular matrix to the cytoskeleton. By using super-resolution iPALM microscopy, we identify two additional nanoscale layers within FAs, specified by actin filaments bound to tropomyosin isoforms Tpm1.6 and Tpm3.2. The Tpm1.6-actin filaments, beneath the previously identified α-actinin cross-linked actin filaments, appear critical for adhesion maturation and controlled cell motility, whereas the adjacent Tpm3.2-actin filament layer beneath seems to facilitate adhesion disassembly. Mechanistically, Tpm3.2 stabilizes ACF-7/MACF1 and KANK-family proteins at adhesions, and hence targets microtubule plus-ends to FAs to catalyse their disassembly. Tpm3.2 depletion leads to disorganized microtubule network, abnormally stable FAs, and defects in tail retraction during migration. Thus, FAs are composed of distinct actin filament layers, and each may have specific roles in coupling adhesions to the cytoskeleton, or in controlling adhesion dynamics.
Collapse
Affiliation(s)
- Reena Kumari
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Katharina Ven
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Megan Chastney
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Shrikant B Kokate
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Johan Peränen
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Jesse Aaron
- Advanced Imaging Center, HHMI Janelia Research Campus, Ashburn, VA, 20147, USA
| | - Konstantin Kogan
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Leonardo Almeida-Souza
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Elena Kremneva
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Renaud Poincloux
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Teng-Leong Chew
- Advanced Imaging Center, HHMI Janelia Research Campus, Ashburn, VA, 20147, USA
| | - Peter W Gunning
- School of Biomedical Sciences, UNSW Sydney, Wallace Wurth Building, Sydney, NSW 2052, Australia
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
- Department of Life Technologies, University of Turku, FI-20520, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Foundation for the Finnish Cancer Institute, Tukholmankatu 8, FI-00014, Helsinki, Finland
| | - Pekka Lappalainen
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland.
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
6
|
Chen M, Xu L, Wu Y, Soba P, Hu C. The organization and function of the Golgi apparatus in dendrite development and neurological disorders. Genes Dis 2023; 10:2425-2442. [PMID: 37554209 PMCID: PMC10404969 DOI: 10.1016/j.gendis.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/13/2022] [Accepted: 11/05/2022] [Indexed: 12/24/2022] Open
Abstract
Dendrites are specialized neuronal compartments that sense, integrate and transfer information in the neural network. Their development is tightly controlled and abnormal dendrite morphogenesis is strongly linked to neurological disorders. While dendritic morphology ranges from relatively simple to extremely complex for a specified neuron, either requires a functional secretory pathway to continually replenish proteins and lipids to meet dendritic growth demands. The Golgi apparatus occupies the center of the secretory pathway and is regulating posttranslational modifications, sorting, transport, and signal transduction, as well as acting as a non-centrosomal microtubule organization center. The neuronal Golgi apparatus shares common features with Golgi in other eukaryotic cell types but also forms distinct structures known as Golgi outposts that specifically localize in dendrites. However, the organization and function of Golgi in dendrite development and its impact on neurological disorders is just emerging and so far lacks a systematic summary. We describe the organization of the Golgi apparatus in neurons, review the current understanding of Golgi function in dendritic morphogenesis, and discuss the current challenges and future directions.
Collapse
Affiliation(s)
- Meilan Chen
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Lu Xu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| | - Yi Wu
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Peter Soba
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Bonn 53115, Germany
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Chun Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| |
Collapse
|
7
|
Ueda H, Tran QTH, Tran LNT, Higasa K, Ikeda Y, Kondo N, Hashiyada M, Sato C, Sato Y, Ashida A, Nishio S, Iwata Y, Iida H, Matsuoka D, Hidaka Y, Fukui K, Itami S, Kawashita N, Sugimoto K, Nozu K, Hattori M, Tsukaguchi H. Characterization of cytoskeletal and structural effects of INF2 variants causing glomerulopathy and neuropathy. Sci Rep 2023; 13:12003. [PMID: 37491439 PMCID: PMC10368640 DOI: 10.1038/s41598-023-38588-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 07/11/2023] [Indexed: 07/27/2023] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is a common glomerular injury leading to end-stage renal disease. Monogenic FSGS is primarily ascribed to decreased podocyte integrity. Variants between residues 184 and 245 of INF2, an actin assembly factor, produce the monogenic FSGS phenotype. Meanwhile, variants between residues 57 and 184 cause a dual-faceted disease involving peripheral neurons and podocytes (Charcot-Marie-Tooth CMT/FSGS). To understand the molecular basis for INF2 disorders, we compared structural and cytoskeletal effects of INF2 variants classified into two subgroups: One (G73D, V108D) causes the CMT/FSGS phenotype, and the other (T161N, N202S) produces monogenic FSGS. Molecular dynamics analysis revealed that all INF2 variants show distinct flexibility compared to the wild-type INF2 and could affect stability of an intramolecular interaction between their N- and C-terminal segments. Immunocytochemistry of cells expressing INF2 variants showed fewer actin stress fibers, and disorganization of cytoplasmic microtubule arrays. Notably, CMT/FSGS variants caused more prominent changes in mitochondrial distribution and fragmentation than FSGS variants and these changes correlated with the severity of cytoskeletal disruption. Our results indicate that CMT/FSGS variants are associated with more severe global cellular defects caused by disrupted cytoskeleton-organelle interactions than are FSGS variants. Further study is needed to clarify tissue-specific pathways and/or cellular functions implicated in FSGS and CMT phenotypes.
Collapse
Affiliation(s)
- Hiroko Ueda
- Division of Nephrology, Second Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1191, Japan
| | - Quynh Thuy Huong Tran
- Division of Nephrology, Second Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1191, Japan
| | - Linh Nguyen Truc Tran
- Division of Nephrology, Second Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1191, Japan
| | - Koichiro Higasa
- Department of Genome Analysis, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Yoshiki Ikeda
- Department of Molecular Genetics, Kansai Medical University, Hirakata, Japan
| | - Naoyuki Kondo
- Department of Molecular Genetics, Kansai Medical University, Hirakata, Japan
| | - Masaki Hashiyada
- Department of Legal Medicine, Kansai Medical University, Hirakata, Japan
| | - Chika Sato
- Department of Gynecology and Obstetrics, Kansai Medical University, Hirakata, Japan
| | - Yoshinori Sato
- Division of Nephrology, Department of Medicine, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan
| | - Akira Ashida
- Department of Pediatrics, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Saori Nishio
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasunori Iwata
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Hiroyuki Iida
- Department of Internal Medicine, Toyama Prefectural Central Hospital, Toyama, Japan
- Toyama Transplantation Promotion Foundation, Toyama, Japan
| | - Daisuke Matsuoka
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoshihiko Hidaka
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kenji Fukui
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Suzu Itami
- Major in Science, Graduate School of Science and Engineering, Kindai University, Higashiosaka, Japan
| | - Norihito Kawashita
- Department of Energy and Materials, Faculty of Science and Engineering, Kindai University, Higashiosaka, Japan
| | - Keisuke Sugimoto
- Department of Pediatrics, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Motoshi Hattori
- Department of Pediatric Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Hiroyasu Tsukaguchi
- Division of Nephrology, Second Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1191, Japan.
| |
Collapse
|
8
|
Lin WH, Feathers RW, Cooper LM, Lewis-Tuffin LJ, Chen J, Sarkaria JN, Anastasiadis PZ. A Syx-RhoA-Dia1 signaling axis regulates cell cycle progression, DNA damage, and therapy resistance in glioblastoma. JCI Insight 2023; 8:e157491. [PMID: 37427593 PMCID: PMC10371349 DOI: 10.1172/jci.insight.157491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/25/2023] [Indexed: 07/11/2023] Open
Abstract
Glioblastomas (GBM) are aggressive tumors that lack effective treatments. Here, we show that the Rho family guanine nucleotide exchange factor Syx promotes GBM cell growth both in vitro and in orthotopic xenografts derived from patients with GBM. Growth defects upon Syx depletion are attributed to prolonged mitosis, increased DNA damage, G2/M cell cycle arrest, and cell apoptosis, mediated by altered mRNA and protein expression of various cell cycle regulators. These effects are phenocopied by depletion of the Rho downstream effector Dia1 and are due, at least in part, to increased phosphorylation, cytoplasmic retention, and reduced activity of the YAP/TAZ transcriptional coactivators. Furthermore, targeting Syx signaling cooperates with radiation treatment and temozolomide (TMZ) to decrease viability in GBM cells, irrespective of their inherent response to TMZ. The data indicate that a Syx-RhoA-Dia1-YAP/TAZ signaling axis regulates cell cycle progression, DNA damage, and therapy resistance in GBM and argue for its targeting for cancer treatment.
Collapse
Affiliation(s)
- Wan-Hsin Lin
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Ryan W. Feathers
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Lisa M. Cooper
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Jiaxiang Chen
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
9
|
Legátová A, Pelantová M, Rösel D, Brábek J, Škarková A. The emerging role of microtubules in invasion plasticity. Front Oncol 2023; 13:1118171. [PMID: 36860323 PMCID: PMC9969133 DOI: 10.3389/fonc.2023.1118171] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
The ability of cells to switch between different invasive modes during metastasis, also known as invasion plasticity, is an important characteristic of tumor cells that makes them able to resist treatment targeted to a particular invasion mode. Due to the rapid changes in cell morphology during the transition between mesenchymal and amoeboid invasion, it is evident that this process requires remodeling of the cytoskeleton. Although the role of the actin cytoskeleton in cell invasion and plasticity is already quite well described, the contribution of microtubules is not yet fully clarified. It is not easy to infer whether destabilization of microtubules leads to higher invasiveness or the opposite since the complex microtubular network acts differently in diverse invasive modes. While mesenchymal migration typically requires microtubules at the leading edge of migrating cells to stabilize protrusions and form adhesive structures, amoeboid invasion is possible even in the absence of long, stable microtubules, albeit there are also cases of amoeboid cells where microtubules contribute to effective migration. Moreover, complex crosstalk of microtubules with other cytoskeletal networks participates in invasion regulation. Altogether, microtubules play an important role in tumor cell plasticity and can be therefore targeted to affect not only cell proliferation but also invasive properties of migrating cells.
Collapse
Affiliation(s)
- Anna Legátová
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Markéta Pelantová
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Daniel Rösel
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Jan Brábek
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Aneta Škarková
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia,*Correspondence: Aneta Škarková,
| |
Collapse
|
10
|
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
|
11
|
Noh J, Isogai T, Chi J, Bhatt K, Danuser G. Granger-causal inference of the lamellipodial actin regulator hierarchy by live cell imaging without perturbation. Cell Syst 2022; 13:471-487.e8. [PMID: 35675823 DOI: 10.1016/j.cels.2022.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/08/2022] [Accepted: 05/10/2022] [Indexed: 11/03/2022]
Abstract
Many cell regulatory systems implicate nonlinearity and redundancy among components. The regulatory network governing lamellipodial and lamellar actin structures is prototypical of such a system, containing tens of actin-nucleating and -modulating molecules with functional overlap and feedback loops. Due to instantaneous and long-term compensation, phenotyping the system response to perturbation provides limited information on the roles the targeted component plays in the unperturbed system. Accordingly, how individual actin regulators contribute to lamellipodial dynamics remains ambiguous. Here, we present a perturbation-free reconstruction of cause-effect relations among actin regulators by applying Granger-causal inference to constitutive image fluctuations that indicate regulator recruitment as a proxy for activity. Our analysis identifies distinct zones of actin regulator activation and of causal effects on filament assembly and delineates actin-dependent and actin-independent regulator roles in controlling edge motion. We propose that edge motion is driven by assembly of two independently operating actin filament systems.
Collapse
Affiliation(s)
- Jungsik Noh
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tadamoto Isogai
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph Chi
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kushal Bhatt
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
12
|
Grespi F, Vianello C, Cagnin S, Giacomello M, De Mario A. The Interplay of Microtubules with Mitochondria–ER Contact Sites (MERCs) in Glioblastoma. Biomolecules 2022; 12:biom12040567. [PMID: 35454156 PMCID: PMC9030160 DOI: 10.3390/biom12040567] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Gliomas are heterogeneous neoplasms, classified into grade I to IV according to their malignancy and the presence of specific histological/molecular hallmarks. The higher grade of glioma is known as glioblastoma (GB). Although progress has been made in surgical and radiation treatments, its clinical outcome is still unfavorable. The invasive properties of GB cells and glioma aggressiveness are linked to the reshaping of the cytoskeleton. Recent works suggest that the different susceptibility of GB cells to antitumor immune response is also associated with the extent and function of mitochondria–ER contact sites (MERCs). The presence of MERCs alterations could also explain the mitochondrial defects observed in GB models, including abnormalities of energy metabolism and disruption of apoptotic and calcium signaling. Based on this evidence, the question arises as to whether a MERCs–cytoskeleton crosstalk exists, and whether GB progression is linked to an altered cytoskeleton–MERCs interaction. To address this possibility, in this review we performed a meta-analysis to compare grade I and grade IV GB patients. From this preliminary analysis, we found that GB samples (grade IV) are characterized by altered expression of cytoskeletal and MERCs related genes. Among them, the cytoskeleton-associated protein 4 (CKAP4 or CLIMP-63) appears particularly interesting as it encodes a MERCs protein controlling the ER anchoring to microtubules (MTs). Although further in-depth analyses remain necessary, this perspective review may provide new hints to better understand GB molecular etiopathogenesis, by suggesting that cytoskeletal and MERCs alterations cooperate to exacerbate the cellular phenotype of high-grade GB and that MERCs players can be exploited as novel biomarkers/targets to enhance the current therapy for GB.
Collapse
Affiliation(s)
- Francesca Grespi
- Department of Biology, University of Padua, Via Ugo Bassi 58b, 35100 Padua, Italy; (F.G.); (C.V.); (S.C.)
| | - Caterina Vianello
- Department of Biology, University of Padua, Via Ugo Bassi 58b, 35100 Padua, Italy; (F.G.); (C.V.); (S.C.)
| | - Stefano Cagnin
- Department of Biology, University of Padua, Via Ugo Bassi 58b, 35100 Padua, Italy; (F.G.); (C.V.); (S.C.)
- CRIBI Biotechnology Center, University of Padua, Via Ugo Bassi 58b, 35100 Padua, Italy
- CIR-Myo Myology Center, University of Padua, Via Ugo Bassi 58b, 35100 Padua, Italy
| | - Marta Giacomello
- Department of Biology, University of Padua, Via Ugo Bassi 58b, 35100 Padua, Italy; (F.G.); (C.V.); (S.C.)
- Department of Biomedical Sciences, University of Padua, Via Ugo Bassi 58b, 35100 Padua, Italy
- Correspondence: (M.G.); (A.D.M.)
| | - Agnese De Mario
- Department of Biomedical Sciences, University of Padua, Via Ugo Bassi 58b, 35100 Padua, Italy
- Correspondence: (M.G.); (A.D.M.)
| |
Collapse
|
13
|
Sun J, Zhong X, Fu X, Miller H, Lee P, Yu B, Liu C. The Actin Regulators Involved in the Function and Related Diseases of Lymphocytes. Front Immunol 2022; 13:799309. [PMID: 35371070 PMCID: PMC8965893 DOI: 10.3389/fimmu.2022.799309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/01/2022] [Indexed: 11/21/2022] Open
Abstract
Actin is an important cytoskeletal protein involved in signal transduction, cell structure and motility. Actin regulators include actin-monomer-binding proteins, Wiskott-Aldrich syndrome (WAS) family of proteins, nucleation proteins, actin filament polymerases and severing proteins. This group of proteins regulate the dynamic changes in actin assembly/disassembly, thus playing an important role in cell motility, intracellular transport, cell division and other basic cellular activities. Lymphocytes are important components of the human immune system, consisting of T-lymphocytes (T cells), B-lymphocytes (B cells) and natural killer cells (NK cells). Lymphocytes are indispensable for both innate and adaptive immunity and cannot function normally without various actin regulators. In this review, we first briefly introduce the structure and fundamental functions of a variety of well-known and newly discovered actin regulators, then we highlight the role of actin regulators in T cell, B cell and NK cell, and finally provide a landscape of various diseases associated with them. This review provides new directions in exploring actin regulators and promotes more precise and effective treatments for related diseases.
Collapse
Affiliation(s)
- Jianxuan Sun
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingyu Zhong
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Fu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- Cytek Biosciences, R&D Clinical Reagents, Fremont, CA, United States
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Bing Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Abstract
Almost 25 years have passed since a mutation of a formin gene, DIAPH1, was identified as being responsible for a human inherited disorder: a form of sensorineural hearing loss. Since then, our knowledge of the links between formins and disease has deepened considerably. Mutations of DIAPH1 and six other formin genes (DAAM2, DIAPH2, DIAPH3, FMN2, INF2 and FHOD3) have been identified as the genetic cause of a variety of inherited human disorders, including intellectual disability, renal disease, peripheral neuropathy, thrombocytopenia, primary ovarian insufficiency, hearing loss and cardiomyopathy. In addition, alterations in formin genes have been associated with a variety of pathological conditions, including developmental defects affecting the heart, nervous system and kidney, aging-related diseases, and cancer. This review summarizes the most recent discoveries about the involvement of formin alterations in monogenic disorders and other human pathological conditions, especially cancer, with which they have been associated. In vitro results and experiments in modified animal models are discussed. Finally, we outline the directions for future research in this field.
Collapse
Affiliation(s)
| | - Miguel A. Alonso
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| |
Collapse
|
15
|
Chia S, Leung T, Tan I. Cyclical phosphorylation of LRAP35a and CLASP2 by GSK3β and CK1δ regulates EB1-dependent MT dynamics in cell migration. Cell Rep 2021; 36:109687. [PMID: 34525355 DOI: 10.1016/j.celrep.2021.109687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 07/02/2021] [Accepted: 08/19/2021] [Indexed: 11/30/2022] Open
Abstract
Mammalian cell cytoskeletal reorganization for efficient directional movement requires tight coordination of actomyosin and microtubule networks. In this study, we show that LRAP35a potentiates microtubule stabilization by promoting CLASP2/EB1 interaction besides its complex formation with MRCK/MYO18A for retrograde actin flow. The alternate regulation of these two networks by LRAP35a is tightly regulated by a series of phosphorylation events that dictated its specificity. Sequential phosphorylation of LRAP35a by Protein Kinase A (PKA) and Glycogen Synthase Kinase-3β (GSK3β) initiates the association of LRAP35a with CLASP2, while subsequent binding and further phosphorylation by Casein Kinase 1δ (CK1δ) induce their dissociation, which facilitates LRAP35a/MRCK association in driving lamellar actomyosin flow. Importantly, microtubule dynamics is directly moderated by CK1δ activity on CLASP2 to regulate GSK3β phosphorylation of the SxIP motifs that blocks EB1 binding, an event countered by LRAP35a interaction and its competition for CK1δ activity. Overall this study reveals an essential role for LRAP35a in coordinating lamellar contractility and microtubule polarization in cell migration.
Collapse
Affiliation(s)
- Shumei Chia
- Institute of Molecular and Cell Biology, A(∗)STAR, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Genome Institute of Singapore, A(∗)STAR, 60 Biopolis Street, #02-01 Genome, Singapore 138672, Singapore; Department of Anatomy, Yong Loo Lin School of Medicine, MD10, 4 Medical Drive, Singapore 117594, Singapore.
| | - Thomas Leung
- Institute of Molecular and Cell Biology, A(∗)STAR, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Department of Anatomy, Yong Loo Lin School of Medicine, MD10, 4 Medical Drive, Singapore 117594, Singapore
| | - Ivan Tan
- Institute of Molecular and Cell Biology, A(∗)STAR, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Bioprocessing Technology Institute, A(∗)STAR, 20 Biopolis Way, #06-01, Centros, Singapore 138668, Singapore.
| |
Collapse
|
16
|
Kundishora AJ, Peters ST, Pinard A, Duran D, Panchagnula S, Barak T, Miyagishima DF, Dong W, Smith H, Ocken J, Dunbar A, Nelson-Williams C, Haider S, Walker RL, Li B, Zhao H, Thumkeo D, Marlier A, Duy PQ, Diab NS, Reeves BC, Robert SM, Sujijantarat N, Stratman AN, Chen YH, Zhao S, Roszko I, Lu Q, Zhang B, Mane S, Castaldi C, López-Giráldez F, Knight JR, Bamshad MJ, Nickerson DA, Geschwind DH, Chen SSL, Storm PB, Diluna ML, Matouk CC, Orbach DB, Alper SL, Smith ER, Lifton RP, Gunel M, Milewicz DM, Jin SC, Kahle KT. DIAPH1 Variants in Non-East Asian Patients With Sporadic Moyamoya Disease. JAMA Neurol 2021; 78:993-1003. [PMID: 34125151 PMCID: PMC8204259 DOI: 10.1001/jamaneurol.2021.1681] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/31/2021] [Indexed: 12/18/2022]
Abstract
Importance Moyamoya disease (MMD), a progressive vasculopathy leading to narrowing and ultimate occlusion of the intracranial internal carotid arteries, is a cause of childhood stroke. The cause of MMD is poorly understood, but genetic factors play a role. Several familial forms of MMD have been identified, but the cause of most cases remains elusive, especially among non-East Asian individuals. Objective To assess whether ultrarare de novo and rare, damaging transmitted variants with large effect sizes are associated with MMD risk. Design, Setting, and Participants A genetic association study was conducted using whole-exome sequencing case-parent MMD trios in a small discovery cohort collected over 3.5 years (2016-2019); data were analyzed in 2020. Medical records from US hospitals spanning a range of 1 month to 1.5 years were reviewed for phenotyping. Exomes from a larger validation cohort were analyzed to identify additional rare, large-effect variants in the top candidate gene. Participants included patients with MMD and, when available, their parents. All participants who met criteria and were presented with the option to join the study agreed to do so; none were excluded. Twenty-four probands (22 trios and 2 singletons) composed the discovery cohort, and 84 probands (29 trios and 55 singletons) composed the validation cohort. Main Outcomes and Measures Gene variants were identified and filtered using stringent criteria. Enrichment and case-control tests assessed gene-level variant burden. In silico modeling estimated the probability of variant association with protein structure. Integrative genomics assessed expression patterns of MMD risk genes derived from single-cell RNA sequencing data of human and mouse brain tissue. Results Of the 24 patients in the discovery cohort, 14 (58.3%) were men and 18 (75.0%) were of European ancestry. Three of 24 discovery cohort probands contained 2 do novo (1-tailed Poisson P = 1.1 × 10-6) and 1 rare, transmitted damaging variant (12.5% of cases) in DIAPH1 (mammalian diaphanous-1), a key regulator of actin remodeling in vascular cells and platelets. Four additional ultrarare damaging heterozygous DIAPH1 variants (3 unphased) were identified in 3 other patients in an 84-proband validation cohort (73.8% female, 77.4% European). All 6 patients were non-East Asian. Compound heterozygous variants were identified in ena/vasodilator-stimulated phosphoproteinlike protein EVL, a mammalian diaphanous-1 interactor that regulates actin polymerization. DIAPH1 and EVL mutant probands had severe, bilateral MMD associated with transfusion-dependent thrombocytopenia. DIAPH1 and other MMD risk genes are enriched in mural cells of midgestational human brain. The DIAPH1 coexpression network converges in vascular cell actin cytoskeleton regulatory pathways. Conclusions and Relevance These findings provide the largest collection to date of non-East Asian individuals with sporadic MMD harboring pathogenic variants in the same gene. The results suggest that DIAPH1 is a novel MMD risk gene and impaired vascular cell actin remodeling in MMD pathogenesis, with diagnostic and therapeutic ramifications.
Collapse
Affiliation(s)
- Adam J. Kundishora
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Samuel T. Peters
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson
| | - Amélie Pinard
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Daniel Duran
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson
| | | | - Tanyeri Barak
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, Connecticut
| | - Danielle F. Miyagishima
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, Connecticut
| | - Weilai Dong
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, New York
| | - Hannah Smith
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Jack Ocken
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Ashley Dunbar
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
| | | | - Shozeb Haider
- Department of Pharmaceutical and Biological Chemistry, University College London School of Pharmacy, London, United Kingdom
| | - Rebecca L. Walker
- Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles
| | - Boyang Li
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Dean Thumkeo
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan
| | - Arnaud Marlier
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
| | - Phan Q. Duy
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Nicholas S. Diab
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
| | - Benjamin C. Reeves
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | | | | | - Amber N. Stratman
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, Missouri
| | - Yi-Hsien Chen
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri
| | - Shujuan Zhao
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri
| | - Isabelle Roszko
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Qiongshi Lu
- Department of Biostatistics & Medical Informatics, University of Wisconsin, Madison
| | - Bo Zhang
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Shrikant Mane
- Yale Center for Genome Analysis, West Haven, Connecticut
| | | | | | | | | | | | - Daniel H. Geschwind
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles
| | - Shih-Shan Lang Chen
- Division of Neurosurgery, Children's Hospital of Philadelphia, Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Phillip B. Storm
- Division of Neurosurgery, Children's Hospital of Philadelphia, Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Michael L. Diluna
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
| | - Charles C. Matouk
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
| | - Darren B. Orbach
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Seth L. Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Edward R. Smith
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Richard P. Lifton
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, New York
| | - Murat Gunel
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
| | - Dianna M. Milewicz
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Sheng Chih Jin
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri
| | - Kristopher T. Kahle
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
17
|
DIAPH1 regulates chromosomal instability of cancer cells by controlling microtubule dynamics. Eur J Cell Biol 2021; 100:151156. [PMID: 33689956 DOI: 10.1016/j.ejcb.2021.151156] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
Chromosomal instability (CIN) is a hallmark of cancer, resulting from misalignment and missegregation of chromosomes during meta- and anaphase, due to non-precise regulation of spindle-MT dynamics. Diaphanous Related Formin 1 (DIAPH1) is an actin nucleator and also binds microtubule (MT) with high affinity. In this study, we analyzed the role of DIAPH1 in regulation of spindle MT-dynamics and CIN in HT29 and HCT-116 colorectal cancer (CRC) cells. Our data show that down-regulation of DIAPH1 in these cell lines decreased spindle-MT speed by 50 % and the fraction of cells with misaligned and missegregated chromosomes was significantly increased. Furthermore, in HCT-116 DIAPH1 depleted cells deviation of chromosome number was elevated and the number of cells with micronuclei and cytosolic DNA was increased in both DIAPH1-knock down cell lines. In line with these results, database analysis revealed a significant correlation with low DIAPH1 mRNA expression and aneuploidy. Thus, DIAPH1 is substantially involved in the control of CIN in CRC cells. Since in vitro, DIAPH1 directly increased MT-polymerization, we assume that DIAPH1 controls CIN by regulating spindle-MT dynamics.
Collapse
|
18
|
Finding MEMO-Emerging Evidence for MEMO1's Function in Development and Disease. Genes (Basel) 2020; 11:genes11111316. [PMID: 33172038 PMCID: PMC7694686 DOI: 10.3390/genes11111316] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 11/24/2022] Open
Abstract
Although conserved throughout animal kingdoms, the protein encoded by the gene Mediator of ERBB2 Driven Cell Motility 1 or MEMO1, has only recently come into focus. True to its namesake, MEMO1 first emerged from a proteomic screen of molecules bound to the ERBB2 receptor and was found to be necessary for efficient cell migration upon receptor activation. While initially placed within the context of breast cancer metastasis—a pathological state that has provided tremendous insight into MEMO1′s cellular roles—MEMO1′s function has since expanded to encompass additional cancer cell types, developmental processes during embryogenesis and homeostatic regulation of adult organ systems. Owing to MEMO1′s deep conservation, a variety of model organisms have been amenable to uncovering biological facets of this multipurpose protein; facets ranging from the cellular (e.g., receptor signaling, cytoskeletal regulation, redox flux) to the organismal (e.g., mineralization and mineral homeostasis, neuro/gliogenesis, vasculogenesis) level. Although these facets emerge at the intersection of numerous biological and human disease processes, how and if they are interconnected remains to be resolved. Here, we review our current understanding of this ‘enigmatic’ molecule, its role in development and disease and open questions emerging from these previous studies.
Collapse
|
19
|
Labat-de-Hoz L, Alonso MA. The formin INF2 in disease: progress from 10 years of research. Cell Mol Life Sci 2020; 77:4581-4600. [PMID: 32451589 PMCID: PMC11104792 DOI: 10.1007/s00018-020-03550-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 05/04/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023]
Abstract
Formins are a conserved family of proteins that primarily act to form linear polymers of actin. Despite their importance to the normal functioning of the cytoskeleton, for a long time, the only two formin genes known to be a genetic cause of human disorders were DIAPH1 and DIAPH3, whose mutation causes two distinct forms of hereditary deafness. In the last 10 years, however, the formin INF2 has emerged as an important target of mutations responsible for the appearance of focal segmental glomerulosclerosis, which are histological lesions associated with glomerulus degeneration that often leads to end-stage renal disease. In some rare cases, focal segmental glomerulosclerosis concurs with Charcot-Marie-Tooth disease, which is a degenerative neurological disorder affecting peripheral nerves. All known INF2 gene mutations causing disease map to the exons encoding the amino-terminal domain. In this review, we summarize the structure, biochemical features and functions of INF2, conduct a systematic and comprehensive analysis of the pathogenic INF2 mutations, including a detailed study exon-by-exon of patient cases and mutations, address the impact of the pathogenic mutations on the structure, regulation and known functions of INF2, draw a series of conclusions that could be useful for INF2-related disease diagnosis, and suggest lines of research for future work on the molecular mechanisms by which INF2 causes disease.
Collapse
Affiliation(s)
- Leticia Labat-de-Hoz
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel A Alonso
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
20
|
López-Gay JM, Nunley H, Spencer M, di Pietro F, Guirao B, Bosveld F, Markova O, Gaugue I, Pelletier S, Lubensky DK, Bellaïche Y. Apical stress fibers enable a scaling between cell mechanical response and area in epithelial tissue. Science 2020; 370:370/6514/eabb2169. [PMID: 33060329 DOI: 10.1126/science.abb2169] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022]
Abstract
Biological systems tailor their properties and behavior to their size throughout development and in numerous aspects of physiology. However, such size scaling remains poorly understood as it applies to cell mechanics and mechanosensing. By examining how the Drosophila pupal dorsal thorax epithelium responds to morphogenetic forces, we found that the number of apical stress fibers (aSFs) anchored to adherens junctions scales with cell apical area to limit larger cell elongation under mechanical stress. aSFs cluster Hippo pathway components, thereby scaling Hippo signaling and proliferation with area. This scaling is promoted by tricellular junctions mediating an increase in aSF nucleation rate and lifetime in larger cells. Development, homeostasis, and repair entail epithelial cell size changes driven by mechanical forces; our work highlights how, in turn, mechanosensitivity scales with cell size.
Collapse
Affiliation(s)
- Jesús M López-Gay
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - Hayden Nunley
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Meryl Spencer
- Department of Physics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Florencia di Pietro
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - Boris Guirao
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - Floris Bosveld
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - Olga Markova
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - Isabelle Gaugue
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - Stéphane Pelletier
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - David K Lubensky
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109, USA. .,Department of Physics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yohanns Bellaïche
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France. .,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| |
Collapse
|
21
|
Seetharaman S, Etienne-Manneville S. Cytoskeletal Crosstalk in Cell Migration. Trends Cell Biol 2020; 30:720-735. [DOI: 10.1016/j.tcb.2020.06.004] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 01/15/2023]
|
22
|
Pimm ML, Hotaling J, Henty-Ridilla JL. Profilin choreographs actin and microtubules in cells and cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 355:155-204. [PMID: 32859370 PMCID: PMC7461721 DOI: 10.1016/bs.ircmb.2020.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Actin and microtubules play essential roles in aberrant cell processes that define and converge in cancer including: signaling, morphology, motility, and division. Actin and microtubules do not directly interact, however shared regulators coordinate these polymers. While many of the individual proteins important for regulating and choreographing actin and microtubule behaviors have been identified, the way these molecules collaborate or fail in normal or disease contexts is not fully understood. Decades of research focus on Profilin as a signaling molecule, lipid-binding protein, and canonical regulator of actin assembly. Recent reports demonstrate that Profilin also regulates microtubule dynamics and polymerization. Thus, Profilin can coordinate both actin and microtubule polymer systems. Here we reconsider the biochemical and cellular roles for Profilin with a focus on the essential cytoskeletal-based cell processes that go awry in cancer. We also explore how the use of model organisms has helped to elucidate mechanisms that underlie the regulatory essence of Profilin in vivo and in the context of disease.
Collapse
Affiliation(s)
- Morgan L Pimm
- Department of Cell and Developmental Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Jessica Hotaling
- Department of Cell and Developmental Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Jessica L Henty-Ridilla
- Department of Cell and Developmental Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States; Department of Biochemistry and Molecular Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States.
| |
Collapse
|
23
|
Biber G, Ben-Shmuel A, Sabag B, Barda-Saad M. Actin regulators in cancer progression and metastases: From structure and function to cytoskeletal dynamics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:131-196. [PMID: 33066873 DOI: 10.1016/bs.ircmb.2020.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cytoskeleton is a central factor contributing to various hallmarks of cancer. In recent years, there has been increasing evidence demonstrating the involvement of actin regulatory proteins in malignancy, and their dysregulation was shown to predict poor clinical prognosis. Although enhanced cytoskeletal activity is often associated with cancer progression, the expression of several inducers of actin polymerization is remarkably reduced in certain malignancies, and it is not completely clear how these changes promote tumorigenesis and metastases. The complexities involved in cytoskeletal induction of cancer progression therefore pose considerable difficulties for therapeutic intervention; it is not always clear which cytoskeletal regulator should be targeted in order to impede cancer progression, and whether this targeting may inadvertently enhance alternative invasive pathways which can aggravate tumor growth. The entire constellation of cytoskeletal machineries in eukaryotic cells are numerous and complex; the system is comprised of and regulated by hundreds of proteins, which could not be covered in a single review. Therefore, we will focus here on the actin cytoskeleton, which encompasses the biological machinery behind most of the key cellular functions altered in cancer, with specific emphasis on actin nucleating factors and nucleation-promoting factors. Finally, we discuss current therapeutic strategies for cancer which aim to target the cytoskeleton.
Collapse
Affiliation(s)
- G Biber
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - A Ben-Shmuel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - B Sabag
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - M Barda-Saad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
24
|
Wu K, Wang H, Guan J, Lan L, Zhao C, Zhang M, Wang D, Wang Q. A novel variant in diaphanous homolog 1 (DIAPH1) as the cause of auditory neuropathy in a Chinese family. Int J Pediatr Otorhinolaryngol 2020; 133:109947. [PMID: 32087478 DOI: 10.1016/j.ijporl.2020.109947] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/11/2020] [Accepted: 02/11/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To determine the genetic cause of non-syndromic autosomal dominant deafness segregating in a Chinese Auditory neuropathy (AN) family. INTRODUCTION AN is a genetically related rare disease characterized by sensorineural hearing loss and retention of hair cell function. Diaphanous Homolog 1 (DIAPH1) is the causative gene of DFNA1. To date, no evidence has been detected to reveal the connection between gene DIAPH1 and AN. MATERIAL AND METHODS Audiological and imageological examinations, genome-wide linkage analysis, and whole exome sequencing (WES) were carried out on the family members. RESULTS In the 13-member branch of the family, 4 patients with preserved otoacoustic emission or cochlear microphonic and abnormal auditory brainstem responses were diagnosed with AN. Linkage analysis detected an interval with a LOD (log odds) score >4 on chr5:138.845-149.509 cM. Using WES we identified a novel frameshift variant c.3551_3552del (p.Glu1184AlafsTer11) in exon 26 of DIAPH1 located in the linkage region. The variant was co-segregated with hearing impairment phenotype in the family except 4 members below the average age of onset. We have found sufficient evidence conforming with the American College of Medical Genetics and Genomics Guideline to consider c.3551_3552del as the genetic cause of the family patients. CONCLUSION It is the first report to expand DIAPH1-related phenotypic spectrum to include AN. Our findings could facilitate the clinical diagnosis and genetic counselling for AN, especially for those with DIAPH1 variants.
Collapse
Affiliation(s)
- Kan Wu
- Department of Otolaryngology-Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Hongyang Wang
- Department of Otolaryngology-Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jing Guan
- Department of Otolaryngology-Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lan Lan
- Department of Otolaryngology-Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Cui Zhao
- Department of Otolaryngology-Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Mengqian Zhang
- Department of Otolaryngology-Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Dayong Wang
- Department of Otolaryngology-Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Qiuju Wang
- Department of Otolaryngology-Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
25
|
Caveolin-1 Modulates Mechanotransduction Responses to Substrate Stiffness through Actin-Dependent Control of YAP. Cell Rep 2019; 25:1622-1635.e6. [PMID: 30404014 PMCID: PMC6231326 DOI: 10.1016/j.celrep.2018.10.024] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/16/2018] [Accepted: 10/03/2018] [Indexed: 02/04/2023] Open
Abstract
The transcriptional regulator YAP orchestrates many cellular functions, including tissue homeostasis, organ growth control, and tumorigenesis. Mechanical stimuli are a key input to YAP activity, but the mechanisms controlling this regulation remain largely uncharacterized. We show that CAV1 positively modulates the YAP mechanoresponse to substrate stiffness through actin-cytoskeleton-dependent and Hippo-kinase-independent mechanisms. RHO activity is necessary, but not sufficient, for CAV1-dependent mechanoregulation of YAP activity. Systematic quantitative interactomic studies and image-based small interfering RNA (siRNA) screens provide evidence that this actin-dependent regulation is determined by YAP interaction with the 14-3-3 protein YWHAH. Constitutive YAP activation rescued phenotypes associated with CAV1 loss, including defective extracellular matrix (ECM) remodeling. CAV1-mediated control of YAP activity was validated in vivo in a model of pancreatitis-driven acinar-to-ductal metaplasia. We propose that this CAV1-YAP mechanotransduction system controls a significant share of cell programs linked to these two pivotal regulators, with potentially broad physiological and pathological implications.
Collapse
|
26
|
Seetharaman S, Etienne-Manneville S. Microtubules at focal adhesions – a double-edged sword. J Cell Sci 2019; 132:132/19/jcs232843. [DOI: 10.1242/jcs.232843] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
ABSTRACT
Cell adhesion to the extracellular matrix is essential for cellular processes, such as migration and invasion. In response to cues from the microenvironment, integrin-mediated adhesions alter cellular behaviour through cytoskeletal rearrangements. The tight association of the actin cytoskeleton with adhesive structures has been extensively studied, whereas the microtubule network in this context has gathered far less attention. In recent years, however, microtubules have emerged as key regulators of cell adhesion and migration through their participation in adhesion turnover and cellular signalling. In this Review, we focus on the interactions between microtubules and integrin-mediated adhesions, in particular, focal adhesions and podosomes. Starting with the association of microtubules with these adhesive structures, we describe the classical role of microtubules in vesicular trafficking, which is involved in the turnover of cell adhesions, before discussing how microtubules can also influence the actin–focal adhesion interplay through RhoGTPase signalling, thereby orchestrating a very crucial crosstalk between the cytoskeletal networks and adhesions.
Collapse
Affiliation(s)
- Shailaja Seetharaman
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France
- Université Paris Descartes, Center for Research and Interdisciplinarity, Sorbonne Paris Cité, 12 Rue de l'École de Médecine, 75006 Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France
| |
Collapse
|
27
|
Shakhov AS, Dugina VB, Alieva IB. Structural Features of Actin Cytoskeleton Required for Endotheliocyte Barrier Function. BIOCHEMISTRY (MOSCOW) 2019; 84:358-369. [PMID: 31228927 DOI: 10.1134/s0006297919040035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cytoplasmic actin structures are essential components of the eukaryotic cytoskeleton. According to the classic concepts, actin structures perform contractile and motor functions, ensuring the possibility of cell shape changes during cell spreading, polarization, and movement both in vitro and in vivo, from the early embryogenesis stages and throughout the life of a multicellular organism. Intracellular organization of actin structures, their biochemical composition, and dynamic properties play a key role in the realization of specific cellular and tissue functions and vary in different cell types. This paper is a review of recent studies on the organization and properties of actin structures in endotheliocytes, interaction of these structures with other cytoskeletal components and elements involved in cell adhesion, as well as their role in the functional activity of endothelial cells.
Collapse
Affiliation(s)
- A S Shakhov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
| | - V B Dugina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
| | - I B Alieva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.
| |
Collapse
|
28
|
Saleh A, Subramaniam G, Raychaudhuri S, Dhawan J. Cytoplasmic sequestration of the RhoA effector mDiaphanous1 by Prohibitin2 promotes muscle differentiation. Sci Rep 2019; 9:8302. [PMID: 31165762 PMCID: PMC6549159 DOI: 10.1038/s41598-019-44749-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/23/2019] [Indexed: 02/06/2023] Open
Abstract
Muscle differentiation is controlled by adhesion and growth factor-dependent signalling through common effectors that regulate muscle-specific transcriptional programs. Here we report that mDiaphanous1, an effector of adhesion-dependent RhoA-signalling, negatively regulates myogenesis at the level of Myogenin expression. In myotubes, over-expression of mDia1ΔN3, a RhoA-independent mutant, suppresses Myogenin promoter activity and expression. We investigated mDia1-interacting proteins that may counteract mDia1 to permit Myogenin expression and timely differentiation. Using yeast two-hybrid and mass-spectrometric analysis, we report that mDia1 has a stage-specific interactome, including Prohibitin2, MyoD, Akt2, and β-Catenin, along with a number of proteosomal and mitochondrial components. Of these interacting partners, Prohibitin2 colocalises with mDia1 in cytoplasmic punctae in myotubes. We mapped the interacting domains of mDia1 and Phb2, and used interacting (mDia1ΔN3/Phb2 FL or mDia1ΔN3/Phb2-Carboxy) and non-interacting pairs (mDia1H + P/Phb2 FL or mDia1ΔN3/Phb2-Amino) to dissect the functional consequences of this partnership on Myogenin promoter activity. Co-expression of full-length as well as mDia1-interacting domains of Prohibitin2 reverse the anti-myogenic effects of mDia1ΔN3, while non-interacting regions do not. Our results suggest that Prohibitin2 sequesters mDia1, dampens its anti-myogenic activity and fine-tunes RhoA-mDia1 signalling to promote differentiation. Overall, we report that mDia1 is multi-functional signalling effector whose anti-myogenic activity is modulated by a differentiation-dependent interactome. The data have been deposited to the ProteomeXchange with identifier PXD012257.
Collapse
Affiliation(s)
- Amena Saleh
- Institute for Stem Cell Science & Regenerative Medicine, Bangalore, Karnataka, 560065, India
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
- Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Gunasekaran Subramaniam
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Swasti Raychaudhuri
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
| | - Jyotsna Dhawan
- Institute for Stem Cell Science & Regenerative Medicine, Bangalore, Karnataka, 560065, India.
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India.
| |
Collapse
|
29
|
Plessner M, Knerr J, Grosse R. Centrosomal Actin Assembly Is Required for Proper Mitotic Spindle Formation and Chromosome Congression. iScience 2019; 15:274-281. [PMID: 31096079 PMCID: PMC6520610 DOI: 10.1016/j.isci.2019.04.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/29/2019] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
Cytoskeletal cross talk between actin filaments and microtubules is a common mechanism governing the assembly of cellular structures, i.e., during filopodia formation or cilia organization. However, potential actin-microtubule interactions during mammalian cell divisions are less well understood. At mitotic entry, centrosomes propagate the formation of the mitotic spindle, thereby aligning individual chromosomes to the metaphase plate, a process coined chromosome congression. Here, we identify actin filament assembly spatially defined at centrosomes contemporaneously with spindle microtubules forming during prometaphase. We show that pharmacological Arp2/3 complex inhibition as well as overexpression of the Arp2/3 complex inhibitory protein Arpin decreased spindle actin. As a consequence, mitotic spindle formation is impaired, which resulted in disorganized chromosome congression and ultimately mitotic defects in non-transformed cells. Thus centrosomal Arp2/3 complex activity plays a role in the maintenance of genomic integrity during mitosis.
Collapse
Affiliation(s)
- Matthias Plessner
- Institute of Pharmacology, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestr. 18, 79104 Freiburg, Germany.
| | - Julian Knerr
- Institute of Pharmacology, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany
| | - Robert Grosse
- Institute of Pharmacology, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestr. 18, 79104 Freiburg, Germany.
| |
Collapse
|
30
|
The Cytoskeleton-A Complex Interacting Meshwork. Cells 2019; 8:cells8040362. [PMID: 31003495 PMCID: PMC6523135 DOI: 10.3390/cells8040362] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022] Open
Abstract
The cytoskeleton of animal cells is one of the most complicated and functionally versatile structures, involved in processes such as endocytosis, cell division, intra-cellular transport, motility, force transmission, reaction to external forces, adhesion and preservation, and adaptation of cell shape. These functions are mediated by three classical cytoskeletal filament types, as follows: Actin, microtubules, and intermediate filaments. The named filaments form a network that is highly structured and dynamic, responding to external and internal cues with a quick reorganization that is orchestrated on the time scale of minutes and has to be tightly regulated. Especially in brain tumors, the cytoskeleton plays an important role in spreading and migration of tumor cells. As the cytoskeletal organization and regulation is complex and many-faceted, this review aims to summarize the findings about cytoskeletal filament types, including substructures formed by them, such as lamellipodia, stress fibers, and interactions between intermediate filaments, microtubules and actin. Additionally, crucial regulatory aspects of the cytoskeletal filaments and the formed substructures are discussed and integrated into the concepts of cell motility. Even though little is known about the impact of cytoskeletal alterations on the progress of glioma, a final point discussed will be the impact of established cytoskeletal alterations in the cellular behavior and invasion of glioma.
Collapse
|
31
|
Grimes KM, Prasad V, McNamara JW. Supporting the heart: Functions of the cardiomyocyte's non-sarcomeric cytoskeleton. J Mol Cell Cardiol 2019; 131:187-196. [PMID: 30978342 DOI: 10.1016/j.yjmcc.2019.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 02/06/2023]
Abstract
The non-contractile cytoskeleton in cardiomyocytes is comprised of cytoplasmic actin, microtubules, and intermediate filaments. In addition to providing mechanical support to these cells, these structures are important effectors of tension-sensing and signal transduction and also provide networks for the transport of proteins and organelles. The majority of our knowledge on the function and structure of these cytoskeletal networks comes from research on proliferative cell types. However, in recent years, researchers have begun to show that there are important cardiomyocyte-specific functions of the cytoskeleton. Here we will discuss the current state of cytoskeletal biology in cardiomyocytes, as well as research from other cell types, that together suggest there is a wealth of knowledge on cardiac health and disease waiting to be uncovered through exploration of the complex signaling networks of cardiomyocyte non-sarcomeric cytoskeletal proteins.
Collapse
Affiliation(s)
- Kelly M Grimes
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Vikram Prasad
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - James W McNamara
- Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
32
|
Ughy B, Schmidthoffer I, Szilak L. Heparan sulfate proteoglycan (HSPG) can take part in cell division: inside and outside. Cell Mol Life Sci 2019; 76:865-871. [PMID: 30465083 PMCID: PMC11105504 DOI: 10.1007/s00018-018-2964-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/30/2018] [Accepted: 11/02/2018] [Indexed: 11/26/2022]
Abstract
Prior to the cytokinesis, the cell-matrix interactions should be disrupted, and the mitotic cells round up. Prerequisite of mitosis, the centrosomes duplicate, spindle fibers are generated and move away from each other to opposite sides of the cells marking the cell poles. Later, an invagination in the plasma membrane is formed a few minutes after anaphase. This furrow ingression is driven by a contractile actomyosin ring, whose assembly is regulated by RhoA GTPase. At the completion of cytokinesis, the two daughter cells are still connected by a thin intercellular bridge, which is subjected to abscission, as the terminal step of cytokinesis. Here, it is overviewed, how syndecan-4, a transmembrane, heparan sulfate proteoglycan, can contribute to these processes in a phosphorylation-dependent manner.
Collapse
Affiliation(s)
- Bettina Ughy
- Institute of Plant Biology, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, 6726, Hungary
| | - Ildiko Schmidthoffer
- Antal Wittmann Crop, Animal and Food Sciences Multidisciplinary Doctoral School, Mosonmagyaróvár, 9200, Hungary
| | - Laszlo Szilak
- Szilak Laboratories Bioinformatics and Molecule-Design Ltd., Szeged, 6723, Hungary.
- Cereal Research Non-profit Ltd., Szeged, 6726, Hungary.
| |
Collapse
|
33
|
Rangel L, Bernabé-Rubio M, Fernández-Barrera J, Casares-Arias J, Millán J, Alonso MA, Correas I. Caveolin-1α regulates primary cilium length by controlling RhoA GTPase activity. Sci Rep 2019; 9:1116. [PMID: 30718762 PMCID: PMC6362014 DOI: 10.1038/s41598-018-38020-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/26/2018] [Indexed: 11/08/2022] Open
Abstract
The primary cilium is a single non-motile protrusion of the plasma membrane of most types of mammalian cell. The structure, length and function of the primary cilium must be tightly controlled because their dysfunction is associated with disease. Caveolin 1 (Cav1), which is best known as a component of membrane invaginations called caveolae, is also present in non-caveolar membrane domains whose function is beginning to be understood. We show that silencing of α and β Cav1 isoforms in different cell lines increases ciliary length regardless of the route of primary ciliogenesis. The sole expression of Cav1α, which is distributed at the apical membrane, restores normal cilium size in Cav1 KO MDCK cells. Cells KO for only Cav1α, which also show long cilia, have a disrupted actin cytoskeleton and reduced RhoA GTPase activity at the apical membrane, and a greater accumulation of Rab11 vesicles at the centrosome. Subsequent experiments showed that DIA1 and ROCK help regulate ciliary length. Since MDCK cells lack apical caveolae, our results imply that non-caveolar apical Cav1α is an important regulator of ciliary length, exerting its effect via RhoA and its effectors, ROCK and DIA1.
Collapse
Affiliation(s)
- 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
| | - 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
| | - 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
| | - 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
| | - Jaime Millán
- 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 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.
| | - 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.
| |
Collapse
|
34
|
Gupta SK, Li Y, Guo M. Anisotropic mechanics and dynamics of a living mammalian cytoplasm. SOFT MATTER 2019; 15:190-199. [PMID: 30488938 DOI: 10.1039/c8sm01708e] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
During physiological processes, cells can undergo morphological changes that can result in a significant redistribution of the cytoskeleton causing anisotropic behavior. Evidence of anisotropy in cells under mechanical stimuli exists; however, the role of cytoskeletal restructuring resulting from changes in cell shape in mechanical anisotropy and its effects remain unclear. In the present study, we examine the role of cell morphology in inducing anisotropy in both intracellular mechanics and dynamics. We change the aspect ratio of cells by confining the cell width and measuring the mechanical properties of the cytoplasm using optical tweezers in both the longitudinal and transverse directions to quantify the degree of mechanical anisotropy. These active microrheology measurements are then combined with intracellular movement to calculate the intracellular force spectrum using force spectrum microscopy (FSM), from which the degree of anisotropy in dynamics and force can be quantified. We find that unrestricted cells with aspect ratio (AR) ∼1 are isotropic; however, when cells break symmetry, they exhibit significant anisotropy in cytoplasmic mechanics and dynamics.
Collapse
Affiliation(s)
- Satish Kumar Gupta
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | | | |
Collapse
|
35
|
Abstract
Formin homology proteins (formins) are a highly conserved family of cytoskeletal remodeling proteins that are involved in a diverse array of cellular functions. Formins are best known for their ability to regulate actin dynamics, but the same functional domains also govern stability and organization of microtubules. It is thought that this dual activity allows them to coordinate the activity of these two major cytoskeletal networks and thereby influence cellular architecture. Golgi ribbon assembly is dependent upon cooperative interactions between actin filaments and cytoplasmic microtubules originating both at the Golgi itself and from the centrosome. Similarly, centrosome assembly, centriole duplication, and centrosome positioning are also reliant on a dialogue between both cytoskeletal networks. As presented in this chapter, a growing body of evidence suggests that multiple formin proteins play essential roles in these central cellular processes.
Collapse
Affiliation(s)
- John Copeland
- Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
36
|
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: 2.7] [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
|
37
|
Zuidscherwoude M, Green HLH, Thomas SG. Formin proteins in megakaryocytes and platelets: regulation of actin and microtubule dynamics. Platelets 2018; 30:23-30. [PMID: 29913076 PMCID: PMC6406210 DOI: 10.1080/09537104.2018.1481937] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The platelet and megakaryocyte cytoskeletons are essential for formation and function of these cells. A dynamic, properly organised tubulin and actin cytoskeleton is critical for the development of the megakaryocyte and the extension of proplatelets. Tubulin in particular plays a pivotal role in the extension of these proplatelets and the release of platelets from them. Tubulin is further required for the maintenance of platelet size, and actin is the driving force for shape change, spreading and platelet contraction during platelet activation. Whilst several key proteins which regulate these cytoskeletons have been described in detail, the formin family of proteins has received less attention. Formins are intriguing as, although they were initially believed to simply be a nucleator of actin polymerisation, increasing evidence shows they are important regulators of the crosstalk between the actin and microtubule cytoskeletons. In this review, we will introduce the formin proteins and consider the recent evidence that they play an important role in platelets and megakaryocytes in mediating both the actin and tubulin cytoskeletons.
Collapse
Affiliation(s)
- Malou Zuidscherwoude
- a Institute of Cardiovascular Sciences , University of Birmingham , Birmingham , UK.,b Centre of Membrane Proteins and Receptors (COMPARE) , University of Birmingham and University of Nottingham , Midlands , UK
| | - Hannah L H Green
- a Institute of Cardiovascular Sciences , University of Birmingham , Birmingham , UK
| | - Steven G Thomas
- a Institute of Cardiovascular Sciences , University of Birmingham , Birmingham , UK.,b Centre of Membrane Proteins and Receptors (COMPARE) , University of Birmingham and University of Nottingham , Midlands , UK
| |
Collapse
|
38
|
BIG2-ARF1-RhoA-mDia1 Signaling Regulates Dendritic Golgi Polarization in Hippocampal Neurons. Mol Neurobiol 2018; 55:7701-7716. [PMID: 29455446 DOI: 10.1007/s12035-018-0954-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 02/05/2018] [Indexed: 01/08/2023]
Abstract
Proper dendrite development is essential for establishing neural circuitry, and Rho GTPases play key regulatory roles in this process. From mouse brain lysates, we identified Brefeldin A-inhibited guanine exchange factor 2 (BIG2) as a novel Rho GTPase regulatory protein involved in dendrite growth and maintenance. BIG2 was highly expressed during early development, and knockdown of the ARFGEF2 gene encoding BIG2 significantly reduced total dendrite length and the number of branches. Expression of the constitutively active ADP-ribosylation factor 1 ARF1 Q71L rescued the defective dendrite morphogenesis of ARFGEF2-null neurons, indicating that BIG2 controls dendrite growth and maintenance by activating ARF1. Moreover, BIG2 co-localizes with the Golgi apparatus and is required for Golgi deployment into major dendrites in cultured hippocampal neurons. Simultaneous overexpression of BIG2 and ARF1 activated RhoA, and treatment with the RhoA activator lysophosphatidic acid in neurons lacking BIG2 or ARF1 increased the number of cells with dendritic Golgi, suggesting that BIG2 and ARF1 activate RhoA to promote dendritic Golgi polarization. mDia1 was identified as a downstream effector of BIG2-ARF1-RhoA axis, mediating Golgi polarization and dendritic morphogenesis. Furthermore, in utero electroporation of ARFGEF2 shRNA into the embryonic mouse brain confirmed an in vivo role of BIG2 for Golgi deployment into the apical dendrite. Taken together, our results suggest that BIG2-ARF1-RhoA-mDia1 signaling regulates dendritic Golgi polarization and dendrite growth and maintenance in hippocampal neurons.
Collapse
|
39
|
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: 4.3] [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
|
40
|
Ganaha A, Kaname T, Shinjou A, Chinen Y, Yanagi K, Higa T, Kondo S, Suzuki M. Progressive macrothrombocytopenia and hearing loss in a large family with DIAPH1
related disease. Am J Med Genet A 2017; 173:2826-2830. [DOI: 10.1002/ajmg.a.38411] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/27/2017] [Accepted: 07/29/2017] [Indexed: 01/17/2023]
Affiliation(s)
- Akira Ganaha
- Department of Otorhinolaryngology-Head and Neck Surgery; University of the Ryukyus; Okinawa Japan
| | - Tadashi Kaname
- Department of Genome Medicine; National Center for Child Health and Development; Tokyo Japan
| | - Ayano Shinjou
- Department of Otorhinolaryngology-Head and Neck Surgery; University of the Ryukyus; Okinawa Japan
| | - Yasutsugu Chinen
- Department of Pediatrics; Faculty of Medicine; University of the Ryukyus; Nishihara Japan
| | - Kumiko Yanagi
- Department of Genome Medicine; National Center for Child Health and Development; Tokyo Japan
| | - Teruyuki Higa
- Department of Otorhinolaryngology-Head and Neck Surgery; University of the Ryukyus; Okinawa Japan
| | - Shunsuke Kondo
- Department of Otorhinolaryngology-Head and Neck Surgery; University of the Ryukyus; Okinawa Japan
| | - Mikio Suzuki
- Department of Otorhinolaryngology-Head and Neck Surgery; University of the Ryukyus; Okinawa Japan
| |
Collapse
|
41
|
Distinct functions of diaphanous-related formins regulate HIV-1 uncoating and transport. Proc Natl Acad Sci U S A 2017; 114:E6932-E6941. [PMID: 28760985 DOI: 10.1073/pnas.1700247114] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Diaphanous (Dia)-related formins (DRFs) coordinate cytoskeletal remodeling by controlling actin nucleation and microtubule (MT) stabilization to facilitate processes such as cell polarization and migration; yet the full extent of their activities remains unknown. Here, we uncover two discrete roles and functions of DRFs during early human immunodeficiency virus type 1 (HIV-1) infection. Independent of their actin regulatory activities, Dia1 and Dia2 facilitated HIV-1-induced MT stabilization and the intracellular motility of virus particles. However, DRFs also bound in vitro assembled capsid-nucleocapsid complexes and promoted the disassembly of HIV-1 capsid (CA) shell. This process, also known as "uncoating," is among the most poorly understood stages in the viral lifecycle. Domain analysis and structure modeling revealed that regions of Dia2 that bound viral CA and mediated uncoating as well as early infection contained coiled-coil domains, and that these activities were genetically separable from effects on MT stabilization. Our findings reveal that HIV-1 exploits discrete functions of DRFs to coordinate critical steps in early infection and identifies Dia family members as regulators of the poorly understood process of HIV-1 uncoating.
Collapse
|
42
|
Integrin-Dependent Regulation of Small GTPases: Role in Cell Migration. J Indian Inst Sci 2017. [DOI: 10.1007/s41745-016-0010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
43
|
Remodeling of the Inner Hair Cell Microtubule Meshwork in a Mouse Model of Auditory Neuropathy AUNA1. eNeuro 2016; 3:eN-NWR-0295-16. [PMID: 28058271 PMCID: PMC5197407 DOI: 10.1523/eneuro.0295-16.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 11/26/2022] Open
Abstract
Auditory neuropathy 1 (AUNA1) is a form of human deafness resulting from a point mutation in the 5′ untranslated region of the Diaphanous homolog 3 (DIAPH3) gene. Notably, the DIAPH3 mutation leads to the overexpression of the DIAPH3 protein, a formin family member involved in cytoskeleton dynamics. Through study of diap3-overexpressing transgenic (Tg) mice, we examine in further detail the anatomical, functional, and molecular mechanisms underlying AUNA1. We identify diap3 as a component of the hair cells apical pole in wild-type mice. In the diap3-overexpressing Tg mice, which show a progressive threshold shift associated with a defect in inner hair cells (IHCs), the neurotransmitter release and potassium conductances are not affected. Strikingly, the overexpression of diap3 results in a selective and early-onset alteration of the IHC cuticular plate. Molecular dissection of the apical components revealed that the microtubule meshwork first undergoes aberrant targeting into the cuticular plate of Tg IHCs, followed by collapse of the stereociliary bundle, with eventual loss of the IHC capacity to transmit incoming auditory stimuli.
Collapse
|
44
|
Kumar B, Chandran B. KSHV Entry and Trafficking in Target Cells-Hijacking of Cell Signal Pathways, Actin and Membrane Dynamics. Viruses 2016; 8:v8110305. [PMID: 27854239 PMCID: PMC5127019 DOI: 10.3390/v8110305] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/24/2016] [Accepted: 11/03/2016] [Indexed: 01/27/2023] Open
Abstract
Kaposi's sarcoma associated herpesvirus (KSHV) is etiologically associated with human endothelial cell hyperplastic Kaposi's sarcoma and B-cell primary effusion lymphoma. KSHV infection of adherent endothelial and fibroblast cells are used as in vitro models for infection and KSHV enters these cells by host membrane bleb and actin mediated macropinocytosis or clathrin endocytosis pathways, respectively. Infection in endothelial and fibroblast cells is initiated by the interactions between multiple viral envelope glycoproteins and cell surface associated heparan sulfate (HS), integrins (α3β1, αVβ3 and αVβ5), and EphA2 receptor tyrosine kinase (EphA2R). This review summarizes the accumulated studies demonstrating that KSHV manipulates the host signal pathways to enter and traffic in the cytoplasm of the target cells, to deliver the viral genome into the nucleus, and initiate viral gene expression. KSHV interactions with the cell surface receptors is the key platform for the manipulations of host signal pathways which results in the simultaneous induction of FAK, Src, PI3-K, Rho-GTPase, ROS, Dia-2, PKC ζ, c-Cbl, CIB1, Crk, p130Cas and GEF-C3G signal and adaptor molecules that play critical roles in the modulation of membrane and actin dynamics, and in the various steps of the early stages of infection such as entry and trafficking towards the nucleus. The Endosomal Sorting Complexes Required for Transport (ESCRT) proteins are also recruited to assist in viral entry and trafficking. In addition, KSHV interactions with the cell surface receptors also induces the host transcription factors NF-κB, ERK1/2, and Nrf2 early during infection to initiate and modulate viral and host gene expression. Nuclear delivery of the viral dsDNA genome is immediately followed by the host innate responses such as the DNA damage response (DDR), inflammasome and interferon responses. Overall, these studies form the initial framework for further studies of simultaneous targeting of KSHV glycoproteins, host receptor, signal molecules and trafficking machinery that would lead into novel therapeutic methods to prevent KSHV infection of target cells and consequently the associated malignancies.
Collapse
Affiliation(s)
- Binod Kumar
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| | - Bala Chandran
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| |
Collapse
|
45
|
Wang X, Tang P, Guo F, Zhang M, Chen Y, Yan Y, Tian Z, Xu P, Zhang L, Zhang L, Zhang L. RhoA regulates Activin B-induced stress fiber formation and migration of bone marrow-derived mesenchymal stromal cell through distinct signaling. Biochim Biophys Acta Gen Subj 2016; 1861:3011-3018. [PMID: 27693126 DOI: 10.1016/j.bbagen.2016.09.027] [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: 04/12/2016] [Revised: 09/04/2016] [Accepted: 09/26/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND In our previous study, Activin B induced actin stress fiber formation and cell migration in Bone marrow-derived mesenchymal stem cells (BMSCs) in vitro. However, the underlying molecular mechanisms are not well studied. RhoA is recognized to play a critical role in the regulation of actomyosin cytoskeletal organization and cell migration. METHODS Pull-down assay was performed to investigate the activity of RhoA. The dominant-negative mutants of RhoA (RhoA(N19)) was used to determine whether RhoA has a role in Activin B-induced cytoskeleton organization and cell migration in BMSCs. Cytoskeleton organization was examined by fluorescence Rhodamine-phalloidin staining, and cell migration by transwell and cell scratching assay. Western blot was carried out to investigate downstream signaling cascade of RhoA. Inhibitor and siRNAs were used to detect the role of downstream signaling in stress fiber formation and/or cell migration. RESULTS RhoA was activated by Activin B in BMSCs. RhoA(N19) blocked Activin B-induced stress fiber formation and cell migration. ROCK inhibitor blocked Activin B-induced stress fiber formation but enhanced BMSCs migration. Activin B induced phosphorylation of LIMK2 and Cofilin, which was abolished by ROCK inhibition. Both of siRNA LIMK2 and siRNA Cofilin inhibited Activin B-induced stress fiber formation. CONCLUSIONS RhoA regulates Activin B-induced stress fiber formation and migration of BMSCs. A RhoA-ROCK-LIMK2-Cofilin signaling node exists and regulates actin stress fiber formation. RhoA regulates Activin B-induced cell migration independent of ROCK. GENERAL SIGNIFICANCE Better understanding of the molecular mechanisms of BMSCs migration will help optimize therapeutic strategy to target BMSCs at injured tissues.
Collapse
Affiliation(s)
- Xueer Wang
- Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province, Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Pei Tang
- Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province, Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Min Zhang
- Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province, Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yinghua Chen
- Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province, Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yuan Yan
- Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province, Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhihui Tian
- Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province, Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Pengcheng Xu
- Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province, Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lei Zhang
- Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province, Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lu Zhang
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Lin Zhang
- Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province, Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
46
|
Interphase adhesion geometry is transmitted to an internal regulator for spindle orientation via caveolin-1. Nat Commun 2016; 7:ncomms11858. [PMID: 27292265 PMCID: PMC4910015 DOI: 10.1038/ncomms11858] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 05/09/2016] [Indexed: 12/31/2022] Open
Abstract
Despite theoretical and physical studies implying that cell-extracellular matrix adhesion geometry governs the orientation of the cell division axis, the molecular mechanisms that translate interphase adhesion geometry to the mitotic spindle orientation remain elusive. Here, we show that the cellular edge retraction during mitotic cell rounding correlates with the spindle axis. At the onset of mitotic cell rounding, caveolin-1 is targeted to the retracting cortical region at the proximal end of retraction fibres, where ganglioside GM1-enriched membrane domains with clusters of caveola-like structures are formed in an integrin and RhoA-dependent manner. Furthermore, Gαi1–LGN–NuMA, a well-known regulatory complex of spindle orientation, is targeted to the caveolin-1-enriched cortical region to guide the spindle axis towards the cellular edge retraction. We propose that retraction-induced cortical heterogeneity of caveolin-1 during mitotic cell rounding sets the spindle orientation in the context of adhesion geometry. Studies imply that cell adhesion geometry during interphase dictates the orientation of the cell division axis. Here the authors show that accumulation of caveolin-1 to rapidly retracting regions during cell rounding sets the spindle orientation by recruiting Gαi1-LGN-NuMA to the cortex.
Collapse
|
47
|
Lawrence EJ, Boucher E, Mandato CA. Mitochondria-cytoskeleton associations in mammalian cytokinesis. Cell Div 2016; 11:3. [PMID: 27030796 PMCID: PMC4812650 DOI: 10.1186/s13008-016-0015-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 03/02/2016] [Indexed: 12/27/2022] Open
Abstract
Background The role of the cytoskeleton in regulating mitochondrial distribution in dividing mammalian cells is poorly understood. We previously demonstrated that mitochondria are transported to the cleavage furrow during cytokinesis in a microtubule-dependent manner. However, the exact subset of spindle microtubules and molecular machinery involved remains unknown. Methods We employed quantitative imaging techniques and structured illumination microscopy to analyse the spatial and temporal relationship of mitochondria with microtubules and actin of the contractile ring during cytokinesis in HeLa cells. Results Superresolution microscopy revealed that mitochondria were associated with astral microtubules of the mitotic spindle in cytokinetic cells. Dominant-negative mutants of KIF5B, the heavy chain of kinesin-1 motor, and of Miro-1 disrupted mitochondrial transport to the furrow. Live imaging revealed that mitochondrial enrichment at the cell equator occurred simultaneously with the appearance of the contractile ring in cytokinesis. Inhibiting RhoA activity and contractile ring assembly with C3 transferase, caused mitochondrial mislocalisation during division. Conclusions Taken together, the data suggest a model in which mitochondria are transported by a microtubule-mediated mechanism involving equatorial astral microtubules, Miro-1, and KIF5B to the nascent actomyosin contractile ring in cytokinesis. Electronic supplementary material The online version of this article (doi:10.1186/s13008-016-0015-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- E J Lawrence
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC Canada
| | - E Boucher
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC Canada
| | - C A Mandato
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC Canada
| |
Collapse
|
48
|
A gain-of-function variant in DIAPH1 causes dominant macrothrombocytopenia and hearing loss. Blood 2016; 127:2903-14. [PMID: 26912466 DOI: 10.1182/blood-2015-10-675629] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/16/2016] [Indexed: 12/25/2022] Open
Abstract
Macrothrombocytopenia (MTP) is a heterogeneous group of disorders characterized by enlarged and reduced numbers of circulating platelets, sometimes resulting in abnormal bleeding. In most MTP, this phenotype arises because of altered regulation of platelet formation from megakaryocytes (MKs). We report the identification of DIAPH1, which encodes the Rho-effector diaphanous-related formin 1 (DIAPH1), as a candidate gene for MTP using exome sequencing, ontological phenotyping, and similarity regression. We describe 2 unrelated pedigrees with MTP and sensorineural hearing loss that segregate with a DIAPH1 R1213* variant predicting partial truncation of the DIAPH1 diaphanous autoregulatory domain. The R1213* variant was linked to reduced proplatelet formation from cultured MKs, cell clustering, and abnormal cortical filamentous actin. Similarly, in platelets, there was increased filamentous actin and stable microtubules, indicating constitutive activation of DIAPH1. Overexpression of DIAPH1 R1213* in cells reproduced the cytoskeletal alterations found in platelets. Our description of a novel disorder of platelet formation and hearing loss extends the repertoire of DIAPH1-related disease and provides new insight into the autoregulation of DIAPH1 activity.
Collapse
|
49
|
Cammarata GM, Bearce EA, Lowery LA. Cytoskeletal social networking in the growth cone: How +TIPs mediate microtubule-actin cross-linking to drive axon outgrowth and guidance. Cytoskeleton (Hoboken) 2016; 73:461-76. [PMID: 26783725 DOI: 10.1002/cm.21272] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/08/2016] [Accepted: 01/08/2016] [Indexed: 11/08/2022]
Abstract
The growth cone is a unique structure capable of guiding axons to their proper destinations. Within the growth cone, extracellular guidance cues are interpreted and then transduced into physical changes in the actin filament (F-actin) and microtubule cytoskeletons, providing direction and movement. While both cytoskeletal networks individually possess important growth cone-specific functions, recent data over the past several years point towards a more cooperative role between the two systems. Facilitating this interaction between F-actin and microtubules, microtubule plus-end tracking proteins (+TIPs) have been shown to link the two cytoskeletons together. Evidence suggests that many +TIPs can couple microtubules to F-actin dynamics, supporting both microtubule advance and retraction in the growth cone periphery. In addition, growing in vitro and in vivo data support a secondary role for +TIPs in which they may participate as F-actin nucleators, thus directly influencing F-actin dynamics and organization. This review focuses on how +TIPs may link F-actin and microtubules together in the growth cone, and how these interactions may influence axon guidance. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | | | - Laura Anne Lowery
- Department of Biology, Boston College, Chestnut Hill, Massachusetts.
| |
Collapse
|
50
|
Kasuya K, Nagakawa Y, Hosokawa Y, Sahara Y, Takishita C, Nakajima T, Hijikata Y, Soya R, Katsumata K, Tsuchida A. RhoA activity increases due to hypermethylation of ARHGAP28 in a highly liver-metastatic colon cancer cell line. Biomed Rep 2016; 4:335-339. [PMID: 26998271 DOI: 10.3892/br.2016.582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 01/20/2016] [Indexed: 11/06/2022] Open
Abstract
Certain cell lines exhibit metastatic ability (highly metastatic cell lines) while their parent cell lines have no metastatic ability. Differences in methylation, which are not derived from differences in the gene sequence between cell lines, were extensively analyzed. Using an established highly metastatic cell line, KM12SM, and its parent cell line, KM12C, differences in the frequency of methylation were analyzed in the promoter regions of ~480,000 gene sites using Infinium HumanMethylation450. The promoter region of the Rho GTPase-activating protein 28 (ARHGAP28) gene was the most markedly methylated region in KM12SM compared with KM12C. ARHGAP28 is a GTPase-activating protein (GAP), and it converts activated RhoA to inactivated RhoA via GTPase. RhoA activity was compared between these two cell lines. The activated RhoA level was compared using western blot analysis and G-LISA. The activated RhoA level was higher in KM12SM compared to KM12C for western blot analysis and G-LISA analysis. RhoA is a protein involved in cytoskeleton formation and cell motility. RhoA, for which ARHGAP28 acts as a GAP, is possibly a factor involved in the metastatic ability of cancer.
Collapse
Affiliation(s)
- Kazuhiko Kasuya
- Department of Gastrointestinal Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Yuichi Nagakawa
- Department of Gastrointestinal Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Yuichi Hosokawa
- Department of Gastrointestinal Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Yatsuka Sahara
- Department of Gastrointestinal Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Chie Takishita
- Department of Gastrointestinal Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Tetsushi Nakajima
- Department of Gastrointestinal Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Yosuke Hijikata
- Department of Gastrointestinal Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Ryoko Soya
- Department of Gastrointestinal Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Kenji Katsumata
- Department of Gastrointestinal Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Akihiko Tsuchida
- Department of Gastrointestinal Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| |
Collapse
|