1
|
Maupérin M, Sun Y, Glandorf T, Oswald TA, Klatt N, Geil B, Mutero-Maeda A, Méan I, Jond L, Janshoff A, Yan J, Citi S. A feedback circuitry involving γ-actin, β-actin and nonmuscle myosin-2 A controls tight junction and apical cortex mechanics. Nat Commun 2025; 16:2514. [PMID: 40082413 PMCID: PMC11906862 DOI: 10.1038/s41467-025-57428-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/21/2025] [Indexed: 03/16/2025] Open
Abstract
Cytoplasmic β- and γ-actin isoforms, along with non-muscle myosin 2 isoforms, are tightly regulated in epithelial cells and compose the actomyosin cytoskeleton at the apical junctional complex. However, their specific role in regulating the mechanics of the membrane cortex and the organization of junctions, and which biomechanical circuitries modulate their expression remain poorly understood. Here, we show that γ-actin depletion in MDCK and other epithelial cells results in increased expression and junctional accumulation of β-actin and increased tight junction membrane tortuosity, both dependent on nonmuscle myosin-2A upregulation. The knock-out of γ-actin also decreases apical membrane stiffness and increases dynamic exchange of the cytoplasmic tight junction proteins like ZO-1 and cingulin, without affecting tight junction organization and barrier function. In summary, our findings uncover a biomechanical circuitry linking γ-actin to β-actin expression through nonmuscle myosin-2A and reveal γ-actin as a key regulator of tight junction and apical membrane cortex mechanics, and the dynamics of cytoskeleton-associated tight junction proteins in epithelial cells.
Collapse
Affiliation(s)
- Marine Maupérin
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Yuze Sun
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Thomas Glandorf
- Georg-August Universität, Institute for Physical Chemistry, Göttingen, Germany
| | - Tabea Anne Oswald
- Georg-August Universität, Institute for Organic and Biomolecular Chemistry, Göttingen, Germany
| | - Niklas Klatt
- Georg-August Universität, Institute for Physical Chemistry, Göttingen, Germany
| | - Burkhard Geil
- Georg-August Universität, Institute for Physical Chemistry, Göttingen, Germany
| | - Annick Mutero-Maeda
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Isabelle Méan
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Lionel Jond
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Andreas Janshoff
- Georg-August Universität, Institute for Physical Chemistry, Göttingen, Germany
| | - Jie Yan
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Sandra Citi
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
2
|
Ni K, Liu Y, DI P, Wang L, Huang H, Holsinger RMD, Kiang KMY, Jiao J. Chromobox protein homolog 7 suppresses the stem-like phenotype of glioblastoma cells by regulating the myosin heavy chain 9-NF-κB signaling pathway. Cell Death Discov 2025; 11:74. [PMID: 39988672 PMCID: PMC11847914 DOI: 10.1038/s41420-025-02362-7] [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: 10/28/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
Cancer stem cells (CSCs) are significant factors in the treatment resistance and recurrence of glioblastoma. Chromobox protein homolog 7 (CBX7) can inhibit the progression of various tumors, but its impact on the stem cell-like properties of glioblastoma cells remains unclear. Clinically, low levels of CBX7 are associated with poor prognosis and increased distant metastasis in glioblastoma patients, and this low expression is caused by methylation of the CBX7 promoter. Our current research indicates that CBX7 plays a key role in suppressing the stem-like phenotype of glioblastoma. In this study, through bioinformatics analysis, we found that CBX7 is the most significantly downregulated member of the CBX family in glioblastoma and is closely associated with the stem-like phenotype of glioblastoma cells. We show that CBX7 promotes the degradation of myosin heavy chain 9 (MYH9) protein through the ubiquitin-proteasome pathway via the polycomb repressive complex 1 (PRC1) and suppresses the stem-like phenotype of glioblastoma cells by inhibiting the nuclear factor kappa-B (NF-κB) signaling pathway. Furthermore, overexpression of MYH9 in glioblastoma cells reverses the inhibitory effects of CBX7 on migration, proliferation, invasion, and stemness of glioblastoma cells. In summary, CBX7 acts as a tumor suppressor by inhibiting the stem cell-like characteristics of glioblastoma. The CBX7-MYH9-NF-κB signaling axis may serve as a potential therapeutic target for glioblastoma.
Collapse
Affiliation(s)
- Kaixiang Ni
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 214023, Wuxi, Jiangsu, China
| | - Yuankun Liu
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 214023, Wuxi, Jiangsu, China
| | - Pinggang DI
- Department of Emergency, Wulian County People's Hospital, 262300, Rizhao, Shandong, China
| | - Lu Wang
- Department of Pathology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Nanjing Medical University, 214023, Wuxi, Jiangsu, China
| | - Hui Huang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 214023, Wuxi, Jiangsu, China
| | - R M Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2050, Australia.
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Karrie Mei-Yee Kiang
- Division of Neurosurgery, Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Jiantong Jiao
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, China.
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 214023, Wuxi, Jiangsu, China.
| |
Collapse
|
3
|
Vale-Silva R, de Paes de Faria J, Seixas AI, Brakebusch C, Franklin RJM, Relvas JB. RhoA regulates oligodendrocyte differentiation and myelination by orchestrating cortical and membrane tension. Glia 2025; 73:381-398. [PMID: 39495111 DOI: 10.1002/glia.24640] [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: 07/24/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Timely differentiation and myelin formation by oligodendrocytes are essential for the physiological functioning of the central nervous system (CNS). While the Rho GTPase RhoA has been hinted as a negative regulator of myelin sheath formation, the precise in vivo mechanisms have remained elusive. Here we show that RhoA controls the timing and progression of myelination by oligodendrocytes through a fine-tuned balance between cortical tension, membrane tension and cell shape. Using a conditional mouse model, we observe that Rhoa ablation results in the acceleration of myelination driven by hastened differentiation and facilitated through membrane expansion induced by changes in MLCII activity and in F-actin redistribution and turnover within the cell. These findings reveal RhoA as a central molecular integrator of alterations in actin cytoskeleton, actomyosin contractility and membrane tension underlying precise morphogenesis of oligodendrocytes and normal myelination of the CNS.
Collapse
Affiliation(s)
- Raquel Vale-Silva
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Joana de Paes de Faria
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Ana Isabel Seixas
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Cord Brakebusch
- Biotech Research and Innovation Centre (BRIC), Københavns Biocenter, Copenhagen, Denmark
| | | | - João B Relvas
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Department of Biomedicine, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|
4
|
Busselman BW, Ratnayake I, Terasaki MR, Thakkar VP, Ilyas A, Otterpohl KL, Zimmerman JL, Chandrasekar I. Actin cytoskeleton and associated myosin motors within the renal epithelium. Am J Physiol Renal Physiol 2024; 327:F553-F565. [PMID: 39052845 PMCID: PMC11483076 DOI: 10.1152/ajprenal.00078.2024] [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: 03/07/2024] [Revised: 07/09/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024] Open
Abstract
This review highlights the complexity of renal epithelial cell membrane architectures and organelles through careful review of ultrastructural and physiological studies published over the past several decades. We also showcase the vital roles played by the actin cytoskeleton and actin-associated myosin motor proteins in regulating cell type-specific physiological functions within the cells of the renal epithelium. The purpose of this review is to provide a fresh conceptual framework to explain the structure-function relationships that exist between the actin cytoskeleton, organelle structure, and cargo transport within the mammalian kidney. With recent advances in technologies to visualize the actin cytoskeleton and associated proteins within intact kidneys, it has become increasingly imperative to reimagine the functional roles of these proteins in situ to provide a rationale for their unique, cell type-specific functions that are necessary to establish and maintain complex physiological processes.
Collapse
Affiliation(s)
- Brook W Busselman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, United States
| | | | - Mark R Terasaki
- Department of Cell Biology, University of Connecticut, Farmington, Connecticut, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, United States
| | - Vedant P Thakkar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Arooba Ilyas
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, United States
| | - Karla L Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Jenna L Zimmerman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Department of Cell Biology, University of Connecticut, Farmington, Connecticut, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, United States
| |
Collapse
|
5
|
Jaganathan A, Toth J, Chen X, Basir R, Pieuchot L, Shen Y, Reinhart-King C, Shenoy VB. Mechano-metabolism of metastatic breast cancer cells in 2D and 3D microenvironments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591879. [PMID: 38746096 PMCID: PMC11092625 DOI: 10.1101/2024.04.30.591879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Cells regulate their shape and metabolic activity in response to the mechano-chemical properties of their microenvironment. To elucidate the impact of matrix stiffness and ligand density on the bioenergetics of mesenchymal cells, we developed a nonequilibrium, active chemo-mechanical model that accounts for the mechanical energy of the cell and matrix, chemical energy from ATP hydrolysis, interfacial energy, and mechano-sensitive regulation of stress fiber assembly through signaling. By integrating the kinetics and energetics of these processes, we define the cell "metabolic potential" that, when minimized, provides testable predictions of cell contractility, shape, and ATP consumption. Specifically, we show that the morphology of MDA-MB-231 breast cancer cells in 3D collagen changes from spherical to elongated to spherical with increasing matrix stiffness, which is consistent with experimental observations. On 2D hydrogels, our model predicts a hemispherical-to-spindle-to-disc shape transition with increasing gel stiffness. In both cases, we show that these shape transitions emerge from competition between the energy of ATP hydrolysis associated with increased contractility that drives cell elongation and the interfacial energy that favors a rounded shape. Furthermore, our model can predict how increased energy demand in stiffer microenvironments is met by AMPK activation, which is confirmed experimentally in both 2D and 3D microenvironments and found to correlate with the upregulation of mitochondrial potential, glucose uptake, and ATP levels, as well as provide estimates of changes in intracellular adenosine nucleotide concentrations with changing environmental stiffness. Overall, we present a framework for relating adherent cell energy levels and contractility through biochemical regulation of underlying physical processes. Statement of Significance Increasing evidence indicates that cellular metabolism is regulated by mechanical cues from the extracellular environment. Forces transmitted from the microenvironment activate mechanotransduction pathways in the cell, which trigger a cascade of biochemical events that impact cytoskeletal tension, cellular morphology and energy budget available to the cell. Using a nonequilibrium free energy-based theory, we can predict the ATP consumption, contractility, and shape of mesenchymal cancer cells, as well as how cells regulate energy levels dependent on the mechanosensitive metabolic regulator AMPK. The insights from our model can be used to understand the mechanosensitive regulation of metabolism during metastasis and tumor progression, during which cells experience dynamic changes in their microenvironment and metabolic state.
Collapse
|
6
|
Foglio E, D'Avorio E, Nieri R, Russo MA, Limana F. Epicardial EMT and cardiac repair: an update. Stem Cell Res Ther 2024; 15:219. [PMID: 39026298 PMCID: PMC11264588 DOI: 10.1186/s13287-024-03823-z] [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: 05/16/2024] [Accepted: 06/30/2024] [Indexed: 07/20/2024] Open
Abstract
Epicardial epithelial-to-mesenchymal transition (EMT) plays a pivotal role in both heart development and injury response and involves dynamic cellular changes that are essential for cardiogenesis and myocardial repair. Specifically, epicardial EMT is a crucial process in which epicardial cells lose polarity, migrate into the myocardium, and differentiate into various cardiac cell types during development and repair. Importantly, following EMT, the epicardium becomes a source of paracrine factors that support cardiac growth at the last stages of cardiogenesis and contribute to cardiac remodeling after injury. As such, EMT seems to represent a fundamental step in cardiac repair. Nevertheless, endogenous EMT alone is insufficient to stimulate adequate repair. Redirecting and amplifying epicardial EMT pathways offers promising avenues for the development of innovative therapeutic strategies and treatment approaches for heart disease. In this review, we present a synthesis of recent literature highlighting the significance of epicardial EMT reactivation in adult heart disease patients.
Collapse
Affiliation(s)
- Eleonora Foglio
- Technoscience, Parco Scientifico e Tecnologico Pontino, Latina, Italy
| | - Erica D'Avorio
- Dipartimento di Promozione delle Scienze Umane e della Qualità della Vita, San Raffaele University of Rome, Rome, Italy
| | - Riccardo Nieri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Federica Limana
- Dipartimento di Promozione delle Scienze Umane e della Qualità della Vita, San Raffaele University of Rome, Rome, Italy.
- Laboratorio di Patologia Cellulare e Molecolare, IRCCS San Raffaele Roma, Rome, Italy.
| |
Collapse
|
7
|
Lecarpentier Y, Tremblay B, Locher C, Schussler O, Vallée A, Locher C, Pho D. Lung Cancers: Parenchymal Biochemistry and Mechanics. Cells 2024; 13:427. [PMID: 38474391 PMCID: PMC10931145 DOI: 10.3390/cells13050427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/14/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Parenchyma of pulmonary cancers acquires contractile properties that resemble those of muscles but presents some particularities. These non-muscle contractile tissues could be stimulated either electrically or chemically (KCl). They present the Frank-Starling mechanism, the Hill hyperbolic tension-velocity relationship, and the tridimensional time-independent tension-velocity-length relationship. Relaxation could be obtained by the inhibition of crossbridge molecular motors or by a decrease in the intracellular calcium concentration. They differ from muscles in that their kinetics are ultraslow as evidenced by their low shortening velocity and myosin ATPase activity. Contractility is generated by non-muscle myosin type II A and II B. The activation of the β-catenin/WNT pathway is accompanied by the high level of the non-muscle myosin observed in lung cancers.
Collapse
Affiliation(s)
- Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l’Est Francilien, 77100 Meaux, France
| | - Bruno Tremblay
- Service de Chirurgie Thoracique et Vasculaire, Grand Hôpital de l’Est Francilien, 77100 Meaux, France;
| | - Christèle Locher
- Service de Pneumologie, Grand Hôpital de l’Est Francilien, 77100 Meaux, France
| | - Olivier Schussler
- Département de Chirurgie Thoracique, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
| | - Alexandre Vallée
- Department of Clinical Research and Innovation, Foch Hospital, 92150 Suresnes, France
| | - Christophe Locher
- Service d’Hépato-Gastro-Entérologie, Grand Hôpital de l’Est Francilien, 77100 Meaux, France
| | - David Pho
- Service d’Anatomie et Cytologie Pathologiques, Grand Hôpital de l’Est Francilien, 77600 Jossigny, France
| |
Collapse
|
8
|
Kim OV, Litvinov RI, Gagne AL, French DL, Brass LF, Weisel JW. Megakaryocyte-induced contraction of plasma clots: cellular mechanisms and structural mechanobiology. Blood 2024; 143:548-560. [PMID: 37944157 PMCID: PMC11033616 DOI: 10.1182/blood.2023021545] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/17/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
ABSTRACT Nonmuscle cell contractility is an essential feature underlying diverse cellular processes such as motility, morphogenesis, division and genome replication, intracellular transport, and secretion. Blood clot contraction is a well-studied process driven by contracting platelets. Megakaryocytes (MKs), which are the precursors to platelets, can be found in bone marrow and lungs. Although they express many of the same proteins and structures found in platelets, little is known about their ability to engage with extracellular proteins such as fibrin and contract. Here, we have measured the ability of MKs to compress plasma clots. Megakaryocytes derived from human induced pluripotent stem cells (iPSCs) were suspended in human platelet-free blood plasma and stimulated with thrombin. Using real-time macroscale optical tracking, confocal microscopy, and biomechanical measurements, we found that activated iPSC-derived MKs (iMKs) caused macroscopic volumetric clot shrinkage, as well as densification and stiffening of the fibrin network via fibrin-attached plasma membrane protrusions undergoing extension-retraction cycles that cause shortening and bending of fibrin fibers. Contraction induced by iMKs involved 2 kinetic phases with distinct rates and durations. It was suppressed by inhibitors of nonmuscle myosin IIA, actin polymerization, and integrin αIIbβ3-fibrin interactions, indicating that the molecular mechanisms of iMK contractility were similar or identical to those in activated platelets. Our findings provide new insights into MK biomechanics and suggest that iMKs can be used as a model system to study platelet contractility. Physiologically, the ability of MKs to contract plasma clots may play a role in the mechanical remodeling of intravascular blood clots and thrombi.
Collapse
Affiliation(s)
- Oleg V. Kim
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute, Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA
| | - Rustem I. Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Alyssa L. Gagne
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Deborah L. French
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Lawrence F. Brass
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - John W. Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
9
|
Casas-Mao D, Carrington G, Pujol MG, Peckham M. Effects of specific disease mutations in non-muscle myosin 2A on its structure and function. J Biol Chem 2024; 300:105514. [PMID: 38042490 PMCID: PMC10770755 DOI: 10.1016/j.jbc.2023.105514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/04/2023] Open
Abstract
Non-muscle myosin 2A (NM2A), a widely expressed class 2 myosin, is important for organizing actin filaments in cells. It cycles between a compact inactive 10S state in which its regulatory light chain (RLC) is dephosphorylated and a filamentous state in which the myosin heads interact with actin, and the RLC is phosphorylated. Over 170 missense mutations in MYH9, the gene that encodes the NM2A heavy chain, have been described. These cause MYH9 disease, an autosomal-dominant disorder that leads to bleeding disorders, kidney disease, cataracts, and deafness. Approximately two-thirds of these mutations occur in the coiled-coil tail. These mutations could destabilize the 10S state and/or disrupt filament formation or both. To test this, we determined the effects of six specific mutations using multiple approaches, including circular dichroism to detect changes in secondary structure, negative stain electron microscopy to analyze 10S and filament formation in vitro, and imaging of GFP-NM2A in fixed and live cells to determine filament assembly and dynamics. Two mutations in D1424 (D1424G and D1424N) and V1516M strongly decrease 10S stability and have limited effects on filament formation in vitro. In contrast, mutations in D1447 and E1841K, decrease 10S stability less strongly but increase filament lengths in vitro. The dynamic behavior of all mutants was altered in cells. Thus, the positions of mutated residues and their roles in filament formation and 10S stabilization are key to understanding their contributions to NM2A in disease.
Collapse
Affiliation(s)
- David Casas-Mao
- Astbury Centre for Structural Molecular Biology & School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Glenn Carrington
- Astbury Centre for Structural Molecular Biology & School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Marta Giralt Pujol
- Astbury Centre for Structural Molecular Biology & School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Michelle Peckham
- Astbury Centre for Structural Molecular Biology & School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
10
|
Zhang Q, Feng P, Zhu XH, Zhou SQ, Ye ML, Yang XJ, Gong S, Huang SY, Tan XR, He SW, Li YQ. DNAJA4 suppresses epithelial-mesenchymal transition and metastasis in nasopharyngeal carcinoma via PSMD2-mediated MYH9 degradation. Cell Death Dis 2023; 14:697. [PMID: 37875476 PMCID: PMC10598267 DOI: 10.1038/s41419-023-06225-w] [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: 02/19/2023] [Revised: 10/09/2023] [Accepted: 10/16/2023] [Indexed: 10/26/2023]
Abstract
Emerging evidence indicates that DNA methylation plays an important role in the initiation and progression of nasopharyngeal carcinoma (NPC). DNAJA4 is hypermethylated in NPC, while its role in regulating NPC progression remains unclear. Here, we revealed that the promoter of DNAJA4 was hypermethylated and its expression was downregulated in NPC tissues and cells. Overexpression of DNAJA4 significantly suppressed NPC cell migration, invasion, and EMT in vitro, and markedly inhibited the inguinal lymph node metastasis and lung metastatic colonization in vivo, while it did not affect NPC cell viability and proliferation capability. Mechanistically, DNAJA4 facilitated MYH9 protein degradation via the ubiquitin-proteasome pathway by recruiting PSMD2. Furthermore, the suppressive effects of DNAJA4 on NPC cell migration, invasion, and EMT were reversed by overexpression of MYH9 in NPC cells. Clinically, a low level of DNAJA4 indicated poor prognosis and an increased probability of distant metastasis in NPC patients. Collectively, DNAJA4 serves as a crucial driver for NPC invasion and metastasis, and the DNAJA4-PSMD2-MYH9 axis might contain potential targets for NPC treatments.
Collapse
Affiliation(s)
- Qun Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, PR China
| | - Ping Feng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Xun-Hua Zhu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Shi-Qing Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Ming-Liang Ye
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Xiao-Jing Yang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Sha Gong
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Sheng-Yan Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Xi-Rong Tan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Shi-Wei He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.
| | - Ying-Qing Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.
| |
Collapse
|
11
|
Zhang M, Yang L, Chen D, Heisterkamp N. Drug-tolerant persister B-cell precursor acute lymphoblastic leukemia cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.28.530540. [PMID: 36909619 PMCID: PMC10002708 DOI: 10.1101/2023.02.28.530540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Reduced responsiveness of precursor B-acute lymphoblastic leukemia (BCP-ALL) to chemotherapy can be inferred when leukemia cells persist after 28 days of initial treatment. Survival of these long-term persister (LTP) / minimal residual disease (MRD) cells is partly due to bone marrow stromal cells that protect them under conditions of chemotherapy stress. We used RNA-seq to analyse BCP-ALL cells that survived a long-term, 30-day vincristine chemotherapy treatment while in co-culture with bone marrow stromal cells. RNAs of as many as 10% of the protein-encoding genes were differentially expressed. There was substantial overlap with genes associated with MRD cell persistence reported in other studies. The top pathway regulated in the LTP cells was that involving p53, a master regulator of a spectrum of responses relevant to drug resistance and cytotoxic drug exposure including control of autophagy. We tested a select number of genes for contribution to BCP-ALL cell survival using Cas9/CRISPR in a 2-step selection, initially for overall effect on cell fitness, followed by 21 days of exposure to vincristine. Many genes involved in autophagy and lysosomal function were found to contribute to survival both at steady-state and during drug treatment. We also identified MYH9, NCSTN and KIAA2013 as specific genes contributing to fitness of BCP-ALL cells. CD44 was not essential for growth under steady state conditions but was needed for survival of vincristine treatment. Finally, although the drug transporter ABCC1/MRP1 is not overexpressed in BCP-ALL, a functional gene was needed for DTP cells to survive treatment with vincristine. This suggests that addition of possible ABCC1 inhibitors during induction therapy could provide benefit in eradication of minimal residual disease in patients treated with a chemotherapy regimen that includes vincristine.
Collapse
|
12
|
Mahé C, Lavigne R, Com E, Pineau C, Zlotkowska AM, Tsikis G, Mermillod P, Schoen J, Saint-Dizier M. The sperm-interacting proteome in the bovine isthmus and ampulla during the periovulatory period. J Anim Sci Biotechnol 2023; 14:30. [PMID: 36797800 PMCID: PMC9936689 DOI: 10.1186/s40104-022-00811-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/24/2022] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Spermatozoa interact with oviduct secretions before fertilization in vivo but the molecular players of this dialog and underlying dynamics remain largely unknown. Our objectives were to identify an exhaustive list of sperm-interacting proteins (SIPs) in the bovine oviduct fluid and to evaluate the impact of the oviduct anatomical region (isthmus vs. ampulla) and time relative to ovulation (pre-ovulatory vs. post-ovulatory) on SIPs number and abundance. METHODS Pools of oviduct fluid (OF) from the pre-ovulatory ampulla, pre-ovulatory isthmus, post-ovulatory ampulla, and post-ovulatory isthmus in the side of ovulation were collected from the slaughterhouse. Frozen-thawed bull sperm were incubated with OF or phosphate-buffered saline (control) for 60 min at 38.5 °C. After protein extraction and digestion, sperm and OF samples were analyzed by nanoLC-MS/MS and label-free protein quantification. RESULTS A quantitative comparison between proteins identified in sperm and OF samples (2333 and 2471 proteins, respectively) allowed for the identification of 245 SIPs. The highest number (187) were found in the pre-ovulatory isthmus, i.e., time and place of the sperm reservoir. In total, 41 SIPs (17%) were differentially abundant between stages in a given region or between regions at a given stage and 76 SIPs (31%) were identified in only one region × stage condition. Functional analysis of SIPs predicted roles in cell response to stress, regulation of cell motility, fertilization, and early embryo development. CONCLUSION This study provides a comprehensive list of SIPs in the bovine oviduct and evidences dynamic spatio-temporal changes in sperm-oviduct interactions around ovulation time. Moreover, these data provide protein candidates to improve sperm conservation and in vitro fertilization media.
Collapse
Affiliation(s)
- Coline Mahé
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380, Nouzilly, France.
| | - Régis Lavigne
- grid.410368.80000 0001 2191 9284Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement Et Travail) - UMR-S 1085, F-35000 Rennes, France ,grid.410368.80000 0001 2191 9284Univ Rennes, CNRS, Inserm, Biosit UAR 3480 US_S 018, Protim Core Facility, 35000 Rennes, France
| | - Emmanuelle Com
- grid.410368.80000 0001 2191 9284Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement Et Travail) - UMR-S 1085, F-35000 Rennes, France ,grid.410368.80000 0001 2191 9284Univ Rennes, CNRS, Inserm, Biosit UAR 3480 US_S 018, Protim Core Facility, 35000 Rennes, France
| | - Charles Pineau
- grid.410368.80000 0001 2191 9284Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement Et Travail) - UMR-S 1085, F-35000 Rennes, France ,grid.410368.80000 0001 2191 9284Univ Rennes, CNRS, Inserm, Biosit UAR 3480 US_S 018, Protim Core Facility, 35000 Rennes, France
| | - Aleksandra Maria Zlotkowska
- grid.418188.c0000 0000 9049 5051Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology, FBN, Dummerstorf, Germany ,grid.418779.40000 0001 0708 0355Present Address: Department of Reproduction Biology, Leibniz Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
| | - Guillaume Tsikis
- grid.464126.30000 0004 0385 4036CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France
| | - Pascal Mermillod
- grid.464126.30000 0004 0385 4036CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France
| | - Jennifer Schoen
- grid.418188.c0000 0000 9049 5051Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology, FBN, Dummerstorf, Germany ,grid.418779.40000 0001 0708 0355Present Address: Department of Reproduction Biology, Leibniz Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
| | - Marie Saint-Dizier
- grid.464126.30000 0004 0385 4036CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France
| |
Collapse
|
13
|
Bharda AV, Jung HS. Review on the structural understanding of the 10S myosin II in the era of Cryo-electron microscopy. Appl Microsc 2022; 52:9. [PMID: 36216992 PMCID: PMC9550946 DOI: 10.1186/s42649-022-00078-x] [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: 12/21/2021] [Accepted: 09/23/2022] [Indexed: 11/10/2022] Open
Abstract
The compact smooth muscle 10S myosin II is a type of a monomer with folded tail and the heads bending back to interact with each other. This inactivated form is associated with regulatory and enzymatic activities affecting myosin processivity with actin filaments as well as ATPase activity. Phosphorylation by RLC can however, shuttle myosin from the inhibited 10S state to an activated 6S state, dictating the equilibrium. Multiple studies contributed by TEM have provided insights in the structural understanding of the 10S form. However, it is only recently that the true potential of Cryo-EM in deciphering the intramolecular interactions of 10S myosin state has been realized. This has led to an influx of new revelations on the 10S inactivation, unfolding mechanism and association in various diseases. This study reviews the gradual development in the structural interpretation of 10S species from TEM to Cryo-EM era. Furthermore, we discuss the utility of Cryo-EM in future myosin 10S studies and its contribution to human health.
Collapse
Affiliation(s)
- Anahita Vispi Bharda
- grid.412010.60000 0001 0707 9039Division of Chemistry & Biochemistry, Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon 24341 Republic of Korea
| | - Hyun Suk Jung
- grid.412010.60000 0001 0707 9039Division of Chemistry & Biochemistry, Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon 24341 Republic of Korea
| |
Collapse
|
14
|
Shi X, Yu X, Wang J, Bian S, Li Q, Fu F, Zou X, Zhang L, Bast RC, Lu Z, Guo L, Chen Y, Zhou J. SIK2 promotes ovarian cancer cell motility and metastasis by phosphorylating MYLK. Mol Oncol 2022; 16:2558-2574. [PMID: 35278271 PMCID: PMC9251837 DOI: 10.1002/1878-0261.13208] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/11/2022] [Accepted: 03/10/2022] [Indexed: 11/10/2022] Open
Abstract
Salt‐inducible kinase 2 (SIK2; also known as serine/threonine‐protein kinase SIK2) is overexpressed in several cancers and has been implicated in cancer progression. However, the mechanisms by which SIK2 regulates cancer cell motility, migration and metastasis in ovarian cancer have not been fully discovered. Here, we identify that SIK2 promotes ovarian cancer cell motility, migration and metastasis in vitro and in vivo. Mechanistically, SIK2 regulated cancer cell motility and migration by myosin light chain kinase, smooth muscle (MYLK)‐meditated phosphorylation of myosin light chain 2 (MYL2). SIK2 directly phosphorylated MYLK at Ser343 and activated its downstream effector MYL2, promoting ovarian cancer cell motility and metastasis. In addition, we found that adipocytes induced SIK2 phosphorylation at Ser358 and MYLK phosphorylation at Ser343, enhancing ovarian cancer cell motility. Moreover, SIK2 protein expression was positively correlated with the expression of MYLK‐pS343 in ovarian cancer cell lines and tissues. The co‐expression of SIK2 and MYLK‐pS343 was associated with reduced median overall survival in human ovarian cancer samples. Taken together, SIK2 positively regulates ovarian cancer motility, migration and metastasis, suggesting that SIK2 is a potential candidate for ovarian cancer treatment.
Collapse
Affiliation(s)
- Xiu Shi
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
- Clinical Research Center of Obstetrics and Gynecology Jiangsu Key Laboratory of Clinical Immunology Soochow University Suzhou People’s Republic of China
- Jiangsu Institute of Clinical Immunology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Xuejiao Yu
- Department of Imaging Department The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Juan Wang
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Shimin Bian
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Qiutong Li
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Fengqing Fu
- Clinical Research Center of Obstetrics and Gynecology Jiangsu Key Laboratory of Clinical Immunology Soochow University Suzhou People’s Republic of China
- Jiangsu Institute of Clinical Immunology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Xinwei Zou
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Lin Zhang
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Robert C. Bast
- Department of Imaging Department The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Zhen Lu
- Department of Imaging Department The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Lingchuan Guo
- Department of Experimental Therapeutics University of Texas M.D. Anderson Cancer Center Houston Texas USA
| | - Youguo Chen
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
- Clinical Research Center of Obstetrics and Gynecology Jiangsu Key Laboratory of Clinical Immunology Soochow University Suzhou People’s Republic of China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
- Clinical Research Center of Obstetrics and Gynecology Jiangsu Key Laboratory of Clinical Immunology Soochow University Suzhou People’s Republic of China
| |
Collapse
|
15
|
Wang AY, Coelho NM, Arora PD, Wang Y, Eymael D, Ji C, Wang Q, Lee W, Xu J, Kapus A, Carneiro KMM, McCulloch CA. DDR1 associates with TRPV4 in cell-matrix adhesions to enable calcium-regulated myosin activity and collagen compaction. J Cell Physiol 2022; 237:2451-2468. [PMID: 35150133 DOI: 10.1002/jcp.30696] [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: 06/24/2021] [Revised: 01/10/2022] [Accepted: 01/27/2022] [Indexed: 11/10/2022]
Abstract
Tissue fibrosis manifests as excessive deposition of compacted, highly aligned collagen fibrils, which interfere with organ structure and function. Cells in collagen-rich lesions often exhibit marked overexpression of discoidin domain receptor 1 (DDR1), which is linked to increased collagen compaction through the association of DDR1 with the Ca2+ -dependent nonmuscle myosin IIA (NMIIA). We examined the functional relationship between DDR1 and the transient receptor potential vanilloid type 4 (TRPV4) channel, a Ca2+ -permeable ion channel that is implicated in collagen compaction. Fibroblasts expressing high levels of DDR1 were used to model cells in lesions with collagen compaction. In these cells, the expression of the β1 integrin was deleted to simplify studies of DDR1 function. Compared with DDR1 wild-type cells, high DDR1 expression was associated with increased Ca2+ influx through TRPV4, enrichment of TRPV4 in collagen adhesions, and enhanced contractile activity mediated by NMIIA. At cell adhesion sites to collagen, DDR1 associated with TRPV4, which enhanced DDR1-mediated collagen alignment and compaction. We conclude that DDR1 regulates Ca2+ influx through the TRPV4 channel to promote critical, DDR1-mediated processes that are important in lesions with collagen compaction and alignment.
Collapse
Affiliation(s)
- Andrew Y Wang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Nuno M Coelho
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Pamma D Arora
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Yongqiang Wang
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Denise Eymael
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Chenfan Ji
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Qin Wang
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Wilson Lee
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Jessica Xu
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Andras Kapus
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital and Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Karina M M Carneiro
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Christopher A McCulloch
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Meng L, Li J, Xu H, Wu D, Shan M, Chen Y, Xu J, Liu L, Chen Z, Li Y, Gong T, Liu D. A potential biomarker for clinical atherosclerosis: A novel insight derived from myosin heavy chain 10 promoting transformation of vascular smooth muscle cells. Clin Transl Med 2022; 12:e672. [PMID: 35075787 PMCID: PMC8787098 DOI: 10.1002/ctm2.672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/26/2021] [Accepted: 11/19/2021] [Indexed: 11/07/2022] Open
Affiliation(s)
- Ling‐bing Meng
- Department of Cardiology Beijing Hospital National Center of Gerontology Chinese Academy of Medical Sciences Institute of Geriatric Medicine Beijing China
- Graduate School Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Jian‐yi Li
- Department of Cardiology Beijing Hospital National Center of Gerontology Chinese Academy of Medical Sciences Institute of Geriatric Medicine Beijing China
- Graduate School Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Hong‐xuan Xu
- Department of Cardiology Beijing Hospital National Center of Gerontology Chinese Academy of Medical Sciences Institute of Geriatric Medicine Beijing China
| | - Di‐shan Wu
- Department of Cardiology Beijing Hospital National Center of Gerontology Chinese Academy of Medical Sciences Institute of Geriatric Medicine Beijing China
- Graduate School Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Meng‐jie Shan
- Graduate School Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Department of Plastic Surgery Peking Union Medical College Hospital Beijing China
| | - Yu‐hui Chen
- Department of Neurology Beijing Hospital National Center of Gerontology Institute of Geriatric Medicine Chinese Academy of Medical Sciences Beijing China
| | - Jia‐pei Xu
- Department of Cardiology Beijing Hospital National Center of Gerontology Chinese Academy of Medical Sciences Institute of Geriatric Medicine Beijing China
| | - Long‐teng Liu
- Department of Pathology Beijing Hospital National Center of Gerontology Institute of Geriatric Medicine Chinese Academy of Medical Sciences Beijing China
| | - Zuoguan Chen
- Department of Vascular Surgery Beijing Hospital National Center of Gerontology Institute of Geriatric Medicine Chinese Academy of Medical Sciences Beijing China
| | - Yong‐jun Li
- Department of Vascular Surgery Beijing Hospital National Center of Gerontology Institute of Geriatric Medicine Chinese Academy of Medical Sciences Beijing China
| | - Tao Gong
- Department of Neurology Beijing Hospital National Center of Gerontology Institute of Geriatric Medicine Chinese Academy of Medical Sciences Beijing China
| | - De‐ping Liu
- Department of Cardiology Beijing Hospital National Center of Gerontology Chinese Academy of Medical Sciences Institute of Geriatric Medicine Beijing China
- Graduate School Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| |
Collapse
|
17
|
Lv M, Luo L, Chen X. The landscape of prognostic and immunological role of myosin light chain 9 (MYL9) in human tumors. IMMUNITY INFLAMMATION AND DISEASE 2021; 10:241-254. [PMID: 34729929 PMCID: PMC8767521 DOI: 10.1002/iid3.557] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/09/2021] [Accepted: 10/20/2021] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Recent studies have shown that myosin light chain 9 (MYL9) plays a vital role in immune infiltration, tumor invasion, and metastasis; however, the prognostic and immunological role of MYL9 has not been reported. The purpose of this study was to explore the potential prognostic and immunological roles of MYL9 in human cancers by public datasets mainly including the cancer genome atlas (TCGA) and Gene expression omnibus. METHODS The expression pattern and prognostic value of MYL9 were analyzed across multiple public datasets in different cancer. The correlations between MYL9 expression and immune infiltration among multiple cancers were analyzed by using the TIMER2.0. The MYL9-related gene enrichment analysis was implemented by mainly using KEGG and GO datasets. RESULTS MYL9 was lowly expressed in most cancers, such as breast cancer, lung adenocarcinoma and squamous cell carcinoma, and stomach adenocarcinoma; but it was highly expressed in several cancers, such as cholangiocarcinoma, head and neck squamous cell carcinoma, and liver hepatocellular carcinoma. Furthermore, MYL9 expression was distinctively associated with prognosis in adrenocortical carcinoma, colon adenocarcinoma, brain glioma, lung cancer, ovarian cancer, gastric cancer, breast cancer, blood cancer, and prostate cancer patients. The expressions of MYL9 were significantly associated with the infiltration of cancer-associated fibroblasts, B cell, CD8+ T cell, CD4+ T cell, macrophage, neutrophil, dendritic cell in different tumors as well as immune markers. In addition, we found that the functional mechanisms of MYL9 involved muscle contraction and focal adhesion. CONCLUSION MYL9 can serve as a prognostic signature in pan-cancer and is associated with immune infiltration. This pan-cancer study is the first to show a relatively comprehensive understanding of the prognostic and immunological roles of MYL9 across different cancers.
Collapse
Affiliation(s)
- Minghe Lv
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lumeng Luo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xue Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Moruzzi M, Nestor-Bergmann A, Goddard GK, Tarannum N, Brennan K, Woolner S. Generation of anisotropic strain dysregulates wild-type cell division at the interface between host and oncogenic tissue. Curr Biol 2021; 31:3409-3418.e6. [PMID: 34111402 PMCID: PMC8360906 DOI: 10.1016/j.cub.2021.05.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 03/19/2021] [Accepted: 05/13/2021] [Indexed: 12/11/2022]
Abstract
Epithelial tissues are highly sensitive to anisotropies in mechanical force, with cells altering fundamental behaviors, such as cell adhesion, migration, and cell division.1-5 It is well known that, in the later stages of carcinoma (epithelial cancer), the presence of tumors alters the mechanical properties of a host tissue and that these changes contribute to disease progression.6-9 However, in the earliest stages of carcinoma, when a clonal cluster of oncogene-expressing cells first establishes in the epithelium, the extent to which mechanical changes alter cell behavior in the tissue as a whole remains unclear. This is despite knowledge that many common oncogenes, such as oncogenic Ras, alter cell stiffness and contractility.10-13 Here, we investigate how mechanical changes at the cellular level of an oncogenic cluster can translate into the generation of anisotropic strain across an epithelium, altering cell behavior in neighboring host tissue. We generated clusters of oncogene-expressing cells within otherwise normal in vivo epithelium, using Xenopus laevis embryos. We find that cells in kRasV12, but not cMYC, clusters have increased contractility, which introduces radial stress in the tissue and deforms surrounding host cells. The strain imposed by kRasV12 clusters leads to increased cell division and altered division orientation in neighboring host tissue, effects that can be rescued by reducing actomyosin contractility specifically in the kRasV12 cells. Our findings indicate that some oncogenes can alter the mechanical and proliferative properties of host tissue from the earliest stages of cancer development, changes that have the potential to contribute to tumorigenesis.
Collapse
Affiliation(s)
- Megan Moruzzi
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Alexander Nestor-Bergmann
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK; School of Mathematics, University of Manchester, Manchester M13 9PL, UK
| | - Georgina K Goddard
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Nawseen Tarannum
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Keith Brennan
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK
| | - Sarah Woolner
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
19
|
Garrido-Casado M, Asensio-Juárez G, Vicente-Manzanares M. Nonmuscle Myosin II Regulation Directs Its Multiple Roles in Cell Migration and Division. Annu Rev Cell Dev Biol 2021; 37:285-310. [PMID: 34314591 DOI: 10.1146/annurev-cellbio-042721-105528] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nonmuscle myosin II (NMII) is a multimeric protein complex that generates most mechanical force in eukaryotic cells. NMII function is controlled at three main levels. The first level includes events that trigger conformational changes that extend the complex to enable its assembly into filaments. The second level controls the ATPase activity of the complex and its binding to microfilaments in extended NMII filaments. The third level includes events that modulate the stability and contractility of the filaments. They all work in concert to finely control force generation inside cells. NMII is a common endpoint of mechanochemical signaling pathways that control cellular responses to physical and chemical extracellular cues. Specific phosphorylations modulate NMII activation in a context-dependent manner. A few kinases control these phosphorylations in a spatially, temporally, and lineage-restricted fashion, enabling functional adaptability to the cellular microenvironment. Here, we review mechanisms that control NMII activity in the context of cell migration and division. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Marina Garrido-Casado
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas, University of Salamanca, 37007 Salamanca, Spain;
| | - Gloria Asensio-Juárez
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas, University of Salamanca, 37007 Salamanca, Spain;
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas, University of Salamanca, 37007 Salamanca, Spain;
| |
Collapse
|
20
|
Lecarpentier Y, Claes V, Hébert JL, Schussler O, Vallée A. Mechanical and Thermodynamic Properties of Non-Muscle Contractile Tissues: The Myofibroblast and the Molecular Motor Non-Muscle Myosin Type IIA. Int J Mol Sci 2021; 22:7738. [PMID: 34299379 PMCID: PMC8306181 DOI: 10.3390/ijms22147738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 01/11/2023] Open
Abstract
Myofibroblasts are contractile cells found in multiple tissues. They are physiological cells as in the human placenta and can be obtained from bone marrow mesenchymal stem cells after differentiation by transforming growth factor-β (TGF-β). They are also found in the stroma of cancerous tissues and can be located in non-muscle contractile tissues. When stimulated by an electric current or after exposure to KCl, these tissues contract. They relax either by lowering the intracellular Ca2+ concentration (by means of isosorbide dinitrate or sildenafil) or by inhibiting actin-myosin interactions (by means of 2,3-butanedione monoxime or blebbistatin). Their shortening velocity and their developed tension are dramatically low compared to those of muscles. Like sarcomeric and smooth muscles, they obey Frank-Starling's law and exhibit the Hill hyperbolic tension-velocity relationship. The molecular motor of the myofibroblast is the non-muscle myosin type IIA (NMIIA). Its essential characteristic is the extreme slowness of its molecular kinetics. In contrast, NMIIA develops a unitary force similar to that of muscle myosins. From a thermodynamic point of view, non-muscle contractile tissues containing NMIIA operate extremely close to equilibrium in a linear stationary mode.
Collapse
Affiliation(s)
- Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l’Est Francilien, 77100 Meaux, France
| | - Victor Claes
- Department of Pharmaceutical Sciences, University of Antwerp, 2180 Wilrijk, Belgium;
| | - Jean-Louis Hébert
- Institut de Cardiologie, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France;
| | - Olivier Schussler
- Département de Chirurgie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, APHP, Paris-Descartes Université, 75014 Paris, France;
| | - Alexandre Vallée
- Department of Clinical Research and Innovation, Foch Hospital, 92150 Suresnes, France;
| |
Collapse
|
21
|
Wang S, Li S, Li Y, Jiang Q, Li X, Wang Y, Han JD, Liu Y, Chen YG. Non-muscle myosin heavy chain 9 maintains intestinal homeostasis by preventing epithelium necroptosis and colitis adenoma formation. Stem Cell Reports 2021; 16:1290-1301. [PMID: 33891868 PMCID: PMC8185465 DOI: 10.1016/j.stemcr.2021.03.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Non-muscle myosin IIA plays an important role in cell adhesion, cell migration, and tissue architecture. We previously showed that low activity of the heavy chain of non-muscle myosin II Myh9 is beneficial to LGR5+ intestinal stem cell maintenance. However, the function of Myh9 in adult mouse intestinal epithelium is largely unclear. In this study, we used the inducible Villin-creERT2 knockout approach to delete Myh9 in adult mouse intestinal epithelium and observed that homozygous deletion of Myh9 causes colitis-like morphologic changes in intestine, leads to a high sensitivity to dextran sulfate sodium and promotes colitis-related adenoma formation in the colon. Myh9 deletion disturbs cell junctions and impairs intestinal lumen barrier integrity, promoting the necroptosis of epithelial cells. Consistently, these changes can be partially rescued by Ripk3 knockout. Our results indicate that Myh9 is required for the maintenance of intestinal epithelium integrity and the prevention of cell necroptosis.
Collapse
Affiliation(s)
- Shan Wang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Siqi Li
- Max-Planck Center for Tissue Stem Cell Research and Regenerative Medicine, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Yehua Li
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Quanlong Jiang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology, Peking University, Beijing 100871, P.R. China
| | - Xintong Li
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yalong Wang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jing-Dong Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology, Peking University, Beijing 100871, P.R. China
| | - Yuan Liu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; Max-Planck Center for Tissue Stem Cell Research and Regenerative Medicine, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| |
Collapse
|
22
|
Engemann VI, Rink I, Kilb MF, Hungsberg M, Helmer D, Schmitz K. Cell-based actin polymerization assay to analyze chemokine inhibitors. J Pharmacol Toxicol Methods 2021; 109:107056. [PMID: 33819607 DOI: 10.1016/j.vascn.2021.107056] [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: 10/12/2020] [Revised: 03/24/2021] [Accepted: 03/27/2021] [Indexed: 11/29/2022]
Abstract
Chemokines play an important role in various diseases as signaling molecules for immune cells. Therefore, the inhibition of the chemokine-receptor interaction and the characterization of potential inhibitors are important steps in the development of new therapies. Here, we present a new cell-based assay for chemokine-receptor interaction, using chemokine-dependent actin polymerization as a readout. We used interleukin-8 (IL-8, CXCL8) as a model chemokine and measured the IL-8-dependent actin polymerization with Atto565-phalloidin by monitoring the fluorescence intensity in the cell layer after activation with IL-8. This assay needs no transfection, is easy to perform and requires only a few working steps. It can be used to confirm receptor activation and to characterize the effect of chemokine receptor antagonists. Experiments with the well-known CXCR1/2 inhibitor reparixin confirmed that the observed increase in fluorescence intensity is a result of chemokine receptor activation and can be inhibited in a dose-dependent manner. With optimized parameters, the difference between positive and negative control was highly significant and statistical Z´-factors of 0.4 were determined on average.
Collapse
Affiliation(s)
- Victoria I Engemann
- Technical University of Darmstadt, Clemens-Schöpf-Institute for Organic Chemistry and Biochemistry, 64287 Darmstadt, Germany
| | - Ina Rink
- Technical University of Darmstadt, Clemens-Schöpf-Institute for Organic Chemistry and Biochemistry, 64287 Darmstadt, Germany
| | - Michelle F Kilb
- Technical University of Darmstadt, Clemens-Schöpf-Institute for Organic Chemistry and Biochemistry, 64287 Darmstadt, Germany.
| | - Maximilian Hungsberg
- Technical University of Darmstadt, Clemens-Schöpf-Institute for Organic Chemistry and Biochemistry, 64287 Darmstadt, Germany.
| | - Dorothea Helmer
- Albert-Ludwigs-University of Freiburg, Department of Microsystems Engineering (IMTEK), 79110 Freiburg im Breisgau, Germany.
| | - Katja Schmitz
- Technical University of Darmstadt, Clemens-Schöpf-Institute for Organic Chemistry and Biochemistry, 64287 Darmstadt, Germany.
| |
Collapse
|
23
|
Megakaryocyte migration defects due to nonmuscle myosin IIA mutations underlie thrombocytopenia in MYH9-related disease. Blood 2021; 135:1887-1898. [PMID: 32315395 DOI: 10.1182/blood.2019003064] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/29/2020] [Indexed: 12/17/2022] Open
Abstract
Megakaryocytes (MKs), the precursor cells for platelets, migrate from the endosteal niche of the bone marrow (BM) toward the vasculature, extending proplatelets into sinusoids, where circulating blood progressively fragments them into platelets. Nonmuscle myosin IIA (NMIIA) heavy chain gene (MYH9) mutations cause macrothrombocytopenia characterized by fewer platelets with larger sizes leading to clotting disorders termed myosin-9-related disorders (MYH9-RDs). MYH9-RD patient MKs have proplatelets with thicker and fewer branches that produce fewer and larger proplatelets, which is phenocopied in mouse Myh9-RD models. Defective proplatelet formation is considered to be the principal mechanism underlying the macrothrombocytopenia phenotype. However, MYH9-RD patient MKs may have other defects, as NMII interactions with actin filaments regulate physiological processes such as chemotaxis, cell migration, and adhesion. How MYH9-RD mutations affect MK migration and adhesion in BM or NMIIA activity and assembly prior to proplatelet production remain unanswered. NMIIA is the only NMII isoform expressed in mature MKs, permitting exploration of these questions without complicating effects of other NMII isoforms. Using mouse models of MYH9-RD (NMIIAR702C+/-GFP+/-, NMIIAD1424N+/-, and NMIIAE1841K+/-) and in vitro assays, we investigated MK distribution in BM, chemotaxis toward stromal-derived factor 1, NMIIA activity, and bipolar filament assembly. Results indicate that different MYH9-RD mutations suppressed MK migration in the BM without compromising bipolar filament formation but led to divergent adhesion phenotypes and NMIIA contractile activities depending on the mutation. We conclude that MYH9-RD mutations impair MK chemotaxis by multiple mechanisms to disrupt migration toward the vasculature, impairing proplatelet release and causing macrothrombocytopenia.
Collapse
|
24
|
Nanodiamonds inhibit scratch-wound repair in lung epithelial cell monolayers by blocking cell migration and inhibiting cell proliferation. Toxicol Lett 2021; 341:83-93. [PMID: 33508333 DOI: 10.1016/j.toxlet.2021.01.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/11/2021] [Accepted: 01/20/2021] [Indexed: 12/29/2022]
Abstract
Proliferation and migration of lung epithelial cells following the injury to the epithelial lining of alveoli and airways in the lung are pivotal for remodeling and repair of the wound to restore normal lung function. In the present study, we examined the modulatory effect of carboxylated nanodiamonds (cNDs) on the cell division, migration, and adhesion of epithelial cells in the well-established in vitro model of wound repair and cell migration. Flow cytometry and confocal microscopy results indicated that both LA4 and A549 cells effectively internalized fluorescent carboxylated nanodiamonds (cFNDs) and the internalized nanodiamonds were essentially localized in the cytoplasmic region. Treatment with cNDs blocked the division and migration of cells to fill the scratch wound. Live cell imaging and time-lapse videography of the wound healing process indicated a significant inhibition of cell proliferation activity in cND-treated cells and blocked the wound repair process. Trans-well cell-migration assay results further support the inhibitory effect of cNDs on the cell migration process. Western blotting and immunofluorescence staining indicated that the crucial proteins involved in epithelial-mesenchymal transition (EMT) and cell migration i.e. β-catenin, Vimentin, NM-myosin, and Focal Adhesion Kinase (FAK) were downregulated after treatment with cNDs, while the expression of E-cadherin and Claudin-1, major cell adhesion markers remained unaltered. Taken together, our results indicate that the decline in cell proliferation activity, downregulation in the expression of various crucial protein like β-Catenin, NM-myosin, FAK, and Vimentin involved in the cell migration and unaltered expression of cell adhesion molecules E-cadherin and Claudin-1, may be the factors that contribute to the cND-mediated inhibition of EMT during the wound repair process in the monolayers of lung epithelial cells.
Collapse
|
25
|
Wang K, Okada H, Bi E. Comparative Analysis of the Roles of Non-muscle Myosin-IIs in Cytokinesis in Budding Yeast, Fission Yeast, and Mammalian Cells. Front Cell Dev Biol 2020; 8:593400. [PMID: 33330476 PMCID: PMC7710916 DOI: 10.3389/fcell.2020.593400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/30/2020] [Indexed: 12/31/2022] Open
Abstract
The contractile ring, which plays critical roles in cytokinesis in fungal and animal cells, has fascinated biologists for decades. However, the basic question of how the non-muscle myosin-II and actin filaments are assembled into a ring structure to drive cytokinesis remains poorly understood. It is even more mysterious why and how the budding yeast Saccharomyces cerevisiae, the fission yeast Schizosaccharomyces pombe, and humans construct the ring structure with one, two, and three myosin-II isoforms, respectively. Here, we provide a comparative analysis of the roles of the non-muscle myosin-IIs in cytokinesis in these three model systems, with the goal of defining the common and unique features and highlighting the major questions regarding this family of proteins.
Collapse
Affiliation(s)
- Kangji Wang
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hiroki Okada
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Erfei Bi
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
26
|
Yao Y, Feng Q, Shen J. Myosin light chain kinase regulates intestinal permeability of mucosal homeostasis in Crohn's disease. Expert Rev Clin Immunol 2020; 16:1127-1141. [PMID: 33183108 DOI: 10.1080/1744666x.2021.1850269] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Researchers have investigated the potential role of intestinal permeability in Crohn's disease pathogenesis. Intestinal permeability is usually mediated by cytoskeleton and intercellular junctions. The myosin light chain kinase (MLCK) is an enzyme that activates the myosin light chain to exert its function related to cytoskeleton contraction and tight junction regulation. The correlation between MLCK and Crohn's disease pathogenesis has been consistently proven. Areas covered: This study aims to expand the understanding of the regulation and function of MLCK in Crohn's disease. An extensive literature search in the MEDLINE database (via PubMed) has been performed up to Oct. 2020. The roles of MLCK in tight junction activation, intestinal permeability enhancement, and cell signal regulation are comprehensively discussed. Expert opinion: Targeting the MLCK-related pathways such as TNF-α in CD treatment has been put into clinical use. More accurate targeting such as MLCK and TNFR2 has been proposed to reduce side effects. MLCK may also have the potential to become biomarkers in fields like CD activity. With the application of cutting age research methods and tools, the MLCK research could be accelerated.
Collapse
Affiliation(s)
- Yiran Yao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center; Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University , Shanghai, China
| | - Qi Feng
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center; Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University , Shanghai, China
| |
Collapse
|
27
|
Sung DC, Ahmad M, Lerma Cervantes CB, Zhang Y, Adelstein RS, Ma X. Mutations in non-muscle myosin 2A disrupt the actomyosin cytoskeleton in Sertoli cells and cause male infertility. Dev Biol 2020; 470:49-61. [PMID: 33188738 DOI: 10.1016/j.ydbio.2020.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 10/23/2022]
Abstract
Mutations in non-muscle myosin 2A (NM2A) encompass a wide spectrum of anomalies collectively known as MYH9-Related Disease (MYH9-RD) in humans that can include macrothrombocytopenia, glomerulosclerosis, deafness, and cataracts. We previously created mouse models of the three mutations most frequently found in humans: R702C, D1424N, and E1841K. While homozygous R702C and D1424N mutations are embryonic lethal, we found homozygous mutant E1841K mice to be viable. However the homozygous male, but not female, mice were infertile. Here, we report that these mice have reduced testis size and defects in actin-associated junctions in Sertoli cells, resulting in inability to form the blood-testis barrier and premature germ cell loss. Moreover, compound double heterozygous (R702C/E1841K and D1424/E1841K) males show the same abnormalities in testes as E1841K homozygous males. Conditional ablation of either NM2A or NM2B alone in Sertoli cells has no effect on fertility and testis size, however deletion of both NM2A and NM2B in Sertoli cells results in infertility. Isolation of mutant E1841K Sertoli cells reveals decreased NM2A and F-actin colocalization and thicker NM2A filaments. Furthermore, AE1841K/AE1841K and double knockout Sertoli cells demonstrate microtubule disorganization and increased tubulin acetylation, suggesting defects in the microtubule cytoskeleton. Together, these results demonstrate that NM2A and 2B paralogs play redundant roles in Sertoli cells and are essential for testes development and normal fertility.
Collapse
Affiliation(s)
- Derek C Sung
- Laboratory of Molecular Cardiology, Cell and Developmental Biology Center, Bethesda, MD, 20892-1583, United States; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892-1583, United States
| | - Mohsin Ahmad
- Laboratory of Molecular Cardiology, Cell and Developmental Biology Center, Bethesda, MD, 20892-1583, United States; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892-1583, United States
| | - Connie B Lerma Cervantes
- Laboratory of Molecular Cardiology, Cell and Developmental Biology Center, Bethesda, MD, 20892-1583, United States; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892-1583, United States
| | - Yingfan Zhang
- Laboratory of Molecular Cardiology, Cell and Developmental Biology Center, Bethesda, MD, 20892-1583, United States; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892-1583, United States
| | - Robert S Adelstein
- Laboratory of Molecular Cardiology, Cell and Developmental Biology Center, Bethesda, MD, 20892-1583, United States; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892-1583, United States
| | - Xuefei Ma
- Laboratory of Molecular Cardiology, Cell and Developmental Biology Center, Bethesda, MD, 20892-1583, United States; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892-1583, United States.
| |
Collapse
|
28
|
Otterpohl KL, Busselman BW, Ratnayake I, Hart RG, Hart KR, Evans CM, Phillips CL, Beach JR, Ahrenkiel P, Molitoris BA, Surendran K, Chandrasekar I. Conditional Myh9 and Myh10 inactivation in adult mouse renal epithelium results in progressive kidney disease. JCI Insight 2020; 5:138530. [PMID: 33001861 PMCID: PMC7710296 DOI: 10.1172/jci.insight.138530] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/23/2020] [Indexed: 01/07/2023] Open
Abstract
Actin-associated nonmuscle myosin II (NM2) motor proteins play critical roles in a myriad of cellular functions, including endocytosis and organelle transport pathways. Cell type-specific expression and unique subcellular localization of the NM2 proteins, encoded by the Myh9 and Myh10 genes, in the mouse kidney tubules led us to hypothesize that these proteins have specialized functional roles within the renal epithelium. Inducible conditional knockout (cKO) of Myh9 and Myh10 in the renal tubules of adult mice resulted in progressive kidney disease. Prior to overt renal tubular injury, we observed intracellular accumulation of the glycosylphosphatidylinositol-anchored protein uromodulin (UMOD) and gradual loss of Na+ K+ 2Cl- cotransporter from the apical membrane of the thick ascending limb epithelia. The UMOD accumulation coincided with expansion of endoplasmic reticulum (ER) tubules and activation of ER stress and unfolded protein response pathways in Myh9&10-cKO kidneys. We conclude that NM2 proteins are required for localization and transport of UMOD and loss of function results in accumulation of UMOD and ER stress-mediated progressive renal tubulointerstitial disease. These observations establish cell type-specific role(s) for NM2 proteins in regulation of specialized renal epithelial transport pathways and reveal the possibility that human kidney disease associated with MYH9 mutations could be of renal epithelial origin.
Collapse
Affiliation(s)
- Karla L. Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Brook W. Busselman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, USA
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, USA
| | - Ishara Ratnayake
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, South Dakota, USA
| | - Ryan G. Hart
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Kimberly R. Hart
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, USA
| | - Claire M. Evans
- Histology and Imaging Core, Sanford Research, Sioux Falls, South Dakota, USA
| | - Carrie L. Phillips
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jordan R. Beach
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
| | - Phil Ahrenkiel
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, South Dakota, USA
| | - Bruce A. Molitoris
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kameswaran Surendran
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, USA
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, USA
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, USA
| |
Collapse
|
29
|
The Oncogenic PRL Protein Causes Acid Addiction of Cells by Stimulating Lysosomal Exocytosis. Dev Cell 2020; 55:387-397.e8. [DOI: 10.1016/j.devcel.2020.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/15/2020] [Accepted: 08/20/2020] [Indexed: 12/27/2022]
|
30
|
Edwards-Jorquera SS, Bosveld F, Bellaïche YA, Lennon-Duménil AM, Glavic Á. Trpml controls actomyosin contractility and couples migration to phagocytosis in fly macrophages. J Cell Biol 2020; 219:133603. [PMID: 31940424 PMCID: PMC7055000 DOI: 10.1083/jcb.201905228] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 11/13/2019] [Accepted: 12/07/2019] [Indexed: 12/29/2022] Open
Abstract
Phagocytes use their actomyosin cytoskeleton to migrate as well as to probe their environment by phagocytosis or macropinocytosis. Although migration and extracellular material uptake have been shown to be coupled in some immune cells, the mechanisms involved in such coupling are largely unknown. By combining time-lapse imaging with genetics, we here identify the lysosomal Ca2+ channel Trpml as an essential player in the coupling of cell locomotion and phagocytosis in hemocytes, the Drosophila macrophage-like immune cells. Trpml is needed for both hemocyte migration and phagocytic processing at distinct subcellular localizations: Trpml regulates hemocyte migration by controlling actomyosin contractility at the cell rear, whereas its role in phagocytic processing lies near the phagocytic cup in a myosin-independent fashion. We further highlight that Vamp7 also regulates phagocytic processing and locomotion but uses pathways distinct from those of Trpml. Our results suggest that multiple mechanisms may have emerged during evolution to couple phagocytic processing to cell migration and facilitate space exploration by immune cells.
Collapse
Affiliation(s)
| | - Floris Bosveld
- Institut Curie, PSL Research University, Sorbonne Universités, UPMC Univ Paris 06, Centre National de la Recherche Scientifique UMR 3215, Institut National de la Santé et de la Recherche Médicale U934, Paris, France
| | - Yohanns A Bellaïche
- Institut Curie, PSL Research University, Sorbonne Universités, UPMC Univ Paris 06, Centre National de la Recherche Scientifique UMR 3215, Institut National de la Santé et de la Recherche Médicale U934, Paris, France
| | - Ana-María Lennon-Duménil
- Institut Curie, PSL Research University, Institut National de la Santé et de la Recherche Médicale U932 Immunité et Cancer, Paris, France
| | - Álvaro Glavic
- Centro de Regulación del Genoma, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
31
|
Kelley CA, Triplett O, Mallick S, Burkewitz K, Mair WB, Cram EJ. FLN-1/filamin is required to anchor the actomyosin cytoskeleton and for global organization of sub-cellular organelles in a contractile tissue. Cytoskeleton (Hoboken) 2020; 77:379-398. [PMID: 32969593 DOI: 10.1002/cm.21633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 01/01/2023]
Abstract
Actomyosin networks are organized in space, direction, size, and connectivity to produce coordinated contractions across cells. We use the C. elegans spermatheca, a tube composed of contractile myoepithelial cells, to study how actomyosin structures are organized. FLN-1/filamin is required for the formation and stabilization of a regular array of parallel, contractile, actomyosin fibers in this tissue. Loss of fln-1 results in the detachment of actin fibers from the basal surface, which then accumulate along the cell junctions and are stabilized by spectrin. In addition, actin and myosin are captured at the nucleus by the linker of nucleoskeleton and cytoskeleton complex (LINC) complex, where they form large foci. Nuclear positioning and morphology, distribution of the endoplasmic reticulum and the mitochondrial network are also disrupted. These results demonstrate that filamin is required to prevent large actin bundle formation and detachment, to prevent excess nuclear localization of actin and myosin, and to ensure correct positioning of organelles.
Collapse
Affiliation(s)
- Charlotte A Kelley
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Olivia Triplett
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Samyukta Mallick
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Kristopher Burkewitz
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts, USA.,Department of Cell and Developmental Biology, Vanderbilt School of Medicine, Nashville, Tennessee, USA
| | - William B Mair
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Erin J Cram
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
32
|
Singh SK, Sinha S, Padhan J, Jangde N, Ray R, Rai V. MYH9 suppresses melanoma tumorigenesis, metastasis and regulates tumor microenvironment. Med Oncol 2020; 37:88. [PMID: 32902730 DOI: 10.1007/s12032-020-01413-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/22/2020] [Indexed: 10/23/2022]
Abstract
Non-muscle myosin IIA heavy chain (MYH9) has been implicated in many physiological and pathological functions including cell adhesion, polarity, motility to cancer. However, its role in melanoma remains unexplored. The aim of our study was to evaluate the role of MYH9 in melanoma tumor development and metastasis and further to find out the potential underlying mechanisms. In this study, we evaluated the in vitro migratory and invasive properties and in vivo tumor development and metastasis in C57BL/6 mice by silencing MYH9 in B16F10 melanoma cells. Knocking down MYH9 enhanced migration and invasiveness of B16F10 cells in vitro. Furthermore, MYH9 silencing accelerated tumor growth and metastasis in melanoma subcutaneous and intravenous mouse models. Next, oncogenes analysis revealed epithelial-mesenchymal transition and Erk signaling pathway are being regulated with MYH9 expression. Finally, MYH9 silencing in B16F10 cells modulates the tumor microenvironment by manipulating the leukocytes and macrophages infiltration in tumors. These findings established the opposing role of MYH9 as a tumor suppressor in melanoma suggesting specific MYH9 based approaches in therapeutics.
Collapse
Affiliation(s)
- Satyendra Kumar Singh
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, 751023, India
| | - Sunita Sinha
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, 751023, India.,Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Jyotirmayee Padhan
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, 751023, India
| | - Nitish Jangde
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, 751023, India
| | - Rashmi Ray
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, 751023, India
| | - Vivek Rai
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, 751023, India.
| |
Collapse
|
33
|
Min Q, Parkinson DB, Dun XP. Migrating Schwann cells direct axon regeneration within the peripheral nerve bridge. Glia 2020; 69:235-254. [PMID: 32697392 DOI: 10.1002/glia.23892] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022]
Abstract
Schwann cells within the peripheral nervous system possess a remarkable regenerative potential. Current research shows that peripheral nerve-associated Schwann cells possess the capacity to promote repair of multiple tissues including peripheral nerve gap bridging, skin wound healing, digit tip repair as well as tooth regeneration. One of the key features of the specialized repair Schwann cells is that they become highly motile. They not only migrate into the area of damaged tissue and become a key component of regenerating tissue but also secrete signaling molecules to attract macrophages, support neuronal survival, promote axonal regrowth, activate local mesenchymal stem cells, and interact with other cell types. Currently, the importance of migratory Schwann cells in tissue regeneration is most evident in the case of a peripheral nerve transection injury. Following nerve transection, Schwann cells from both proximal and distal nerve stumps migrate into the nerve bridge and form Schwann cell cords to guide axon regeneration. The formation of Schwann cell cords in the nerve bridge is key to successful peripheral nerve repair following transection injury. In this review, we first examine nerve bridge formation and the behavior of Schwann cell migration in the nerve bridge, and then discuss how migrating Schwann cells direct regenerating axons into the distal nerve. We also review the current understanding of signals that could activate Schwann cell migration and signals that Schwann cells utilize to direct axon regeneration. Understanding the molecular mechanism of Schwann cell migration could potentially offer new therapeutic strategies for peripheral nerve repair.
Collapse
Affiliation(s)
- Qing Min
- School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei Province, People's Republic of China
| | - David B Parkinson
- Peninsula Medical School, Faculty of Health, Plymouth University, Plymouth, Devon, UK
| | - Xin-Peng Dun
- School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei Province, People's Republic of China
- Peninsula Medical School, Faculty of Health, Plymouth University, Plymouth, Devon, UK
- The Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| |
Collapse
|
34
|
Abedrabbo M, Ravid S. Scribble, Lgl1, and myosin II form a complex in vivo to promote directed cell migration. Mol Biol Cell 2020; 31:2234-2248. [PMID: 32697665 PMCID: PMC7550706 DOI: 10.1091/mbc.e19-11-0657] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Scribble (Scrib) and Lethal giant larvae 1 (Lgl1) are conserved polarity proteins that play important roles in different forms of cell polarity. The roles of Scrib and Lgl1 in apical-basal cell polarity have been studied extensively, but little is known about their roles in the cell polarity of migrating cells. Furthermore, the effect of Scrib and Lgl1 interaction on cell polarity is largely unknown. In this study, we show that Scrib, through its leucine-rich repeat domain, forms a complex in vivo with Lgl1. Scrib also forms a complex with myosin II, and Scrib, Lgl1, and myosin II colocalize at the leading edge of migrating cells. The cellular localization and the cytoskeletal association of Scrib and Lgl1 are interdependent, as depletion of either protein affects its counterpart. In addition, depletion of either Scrib or Lgl1 disrupts the cellular localization of myosin II. We show that depletion of either Scrib or Lgl1 affects cell adhesion through the inhibition of focal adhesion disassembly. Finally, we show that Scrib and Lgl1 are required for proper cell polarity of migrating cells. These results provide new insights into the mechanism regulating the cell polarity of migrating cells by Scrib, Lgl1, and myosin II.
Collapse
Affiliation(s)
- Maha Abedrabbo
- Department of Biochemistry and Molecular Biology, The Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Shoshana Ravid
- Department of Biochemistry and Molecular Biology, The Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| |
Collapse
|
35
|
Chen W, Wang W, Sun X, Xie S, Xu X, Liu M, Yang C, Li M, Zhang W, Liu W, Wang L, Zhou T, Yang Y. NudCL2 regulates cell migration by stabilizing both myosin-9 and LIS1 with Hsp90. Cell Death Dis 2020; 11:534. [PMID: 32665550 PMCID: PMC7360774 DOI: 10.1038/s41419-020-02739-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cell migration plays pivotal roles in many biological processes; however, its underlying mechanism remains unclear. Here, we find that NudC-like protein 2 (NudCL2), a cochaperone of heat shock protein 90 (Hsp90), modulates cell migration by stabilizing both myosin-9 and lissencephaly protein 1 (LIS1). Either knockdown or knockout of NudCL2 significantly increases single-cell migration, but has no significant effect on collective cell migration. Immunoprecipitation-mass spectrometry and western blotting analyses reveal that NudCL2 binds to myosin-9 in mammalian cells. Depletion of NudCL2 not only decreases myosin-9 protein levels, but also results in actin disorganization. Ectopic expression of myosin-9 efficiently reverses defects in actin disorganization and single-cell migration in cells depleted of NudCL2. Interestingly, knockdown of myosin-9 increases both single and collective cell migration. Depletion of LIS1, a NudCL2 client protein, suppresses both single and collective cell migration, which exhibits the opposite effect compared with myosin-9 depletion. Co-depletion of myosin-9 and LIS1 promotes single-cell migration, resembling the phenotype caused by NudCL2 depletion. Furthermore, inhibition of Hsp90 ATPase activity also reduces the Hsp90-interacting protein myosin-9 stability and increases single-cell migration. Forced expression of Hsp90 efficiently reverses myosin-9 protein instability and the defects induced by NudCL2 depletion, but not vice versa. Taken together, these data suggest that NudCL2 plays an important role in the precise regulation of cell migration by stabilizing both myosin-9 and LIS1 via Hsp90 pathway.
Collapse
Affiliation(s)
- Wenwen Chen
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Wei Wang
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China
| | - Xiaoxia Sun
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Shanshan Xie
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Xiaoyang Xu
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Min Liu
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Chunxia Yang
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Min Li
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Wen Zhang
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Wei Liu
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Liangjing Wang
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
| | - Tianhua Zhou
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
- The Cancer Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, 310003, China.
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.
| | - Yuehong Yang
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
36
|
Coelho NM, Wang A, Petrovic P, Wang Y, Lee W, McCulloch CA. MRIP Regulates the Myosin IIA Activity and DDR1 Function to Enable Collagen Tractional Remodeling. Cells 2020; 9:cells9071672. [PMID: 32664526 PMCID: PMC7407560 DOI: 10.3390/cells9071672] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/18/2022] Open
Abstract
DDR1 is a collagen adhesion-mechanoreceptor expressed in fibrotic lesions. DDR1 mediates non-muscle myosin IIA (NMIIA)-dependent collagen remodeling. We discovered that the myosin phosphatase Rho-interacting protein (MRIP), is enriched in DDR1-NMIIA adhesions on collagen. MRIP regulates RhoA- and myosin phosphatase-dependent myosin activity. We hypothesized that MRIP regulates DDR1-NMIIA interactions to enable cell migration and collagen tractional remodeling. After deletion of MRIP in β1-integrin null cells expressing DDR1, in vitro wound closure, collagen realignment, and contraction were reduced. Cells expressing DDR1 and MRIP formed larger and more abundant DDR1 clusters on collagen than cells cultured on fibronectin or cells expressing DDR1 but null for MRIP or cells expressing a non-activating DDR1 mutant. Deletion of MRIP reduced DDR1 autophosphorylation and blocked myosin light chain-dependent contraction. Deletion of MRIP did not disrupt the association of DDR1 with NMIIA. We conclude that MRIP regulates NMIIA-dependent DDR1 cluster growth and activation. Accordingly, MRIP may provide a novel drug target for dysfunctional DDR1-related collagen tractional remodeling in fibrosis.
Collapse
|
37
|
Kelley CA, De Henau S, Bell L, Dansen TB, Cram EJ. Redox signaling modulates Rho activity and tissue contractility in the Caenorhabditis elegans spermatheca. Mol Biol Cell 2020; 31:1486-1497. [PMID: 32374641 PMCID: PMC7359568 DOI: 10.1091/mbc.e20-04-0236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Actomyosin-based contractility in smooth muscle and nonmuscle cells is regulated by signaling through the small GTPase Rho and by calcium-activated pathways. We use the myoepithelial cells of the Caenorhabditis elegans spermatheca to study the mechanisms of coordinated myosin activation in vivo. Here, we show that redox signaling modulates RHO-1/Rho activity in this contractile tissue. Exogenously added as well as endogenously generated hydrogen peroxide decreases spermathecal contractility by inhibition of RHO-1, which depends on a conserved cysteine in its nucleotide binding site (C20). Further, we identify an endogenous gradient of H2O2 across the spermathecal tissue, which depends on the activity of cytosolic superoxide dismutase, SOD-1. Collectively, we show that SOD-1-mediated H2O2 production regulates the redox environment and fine tunes Rho activity across the spermatheca through oxidation of RHO-1 C20.
Collapse
Affiliation(s)
| | - Sasha De Henau
- Center for Molecular Medicine, Molecular Cancer Research Section, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Liam Bell
- Department of Biology, Northeastern University, Boston, MA 02115
| | - Tobias B Dansen
- Center for Molecular Medicine, Molecular Cancer Research Section, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Erin J Cram
- Department of Biology, Northeastern University, Boston, MA 02115
| |
Collapse
|
38
|
Non-Muscle Myosin 2A (NM2A): Structure, Regulation and Function. Cells 2020; 9:cells9071590. [PMID: 32630196 PMCID: PMC7408548 DOI: 10.3390/cells9071590] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/30/2022] Open
Abstract
Non-muscle myosin 2A (NM2A) is a motor cytoskeletal enzyme with crucial importance from the early stages of development until adulthood. Due to its capacity to convert chemical energy into force, NM2A powers the contraction of the actomyosin cytoskeleton, required for proper cell division, adhesion and migration, among other cellular functions. Although NM2A has been extensively studied, new findings revealed that a lot remains to be discovered concerning its spatiotemporal regulation in the intracellular environment. In recent years, new functions were attributed to NM2A and its activity was associated to a plethora of illnesses, including neurological disorders and infectious diseases. Here, we provide a concise overview on the current knowledge regarding the structure, the function and the regulation of NM2A. In addition, we recapitulate NM2A-associated diseases and discuss its potential as a therapeutic target.
Collapse
|
39
|
Lecarpentier Y, Kindler V, Krokidis X, Bochaton-Piallat ML, Claes V, Hébert JL, Vallée A, Schussler O. Statistical Mechanics of Non-Muscle Myosin IIA in Human Bone Marrow-Derived Mesenchymal Stromal Cells Seeded in a Collagen Scaffold: A Thermodynamic Near-Equilibrium Linear System Modified by the Tripeptide Arg-Gly-Asp (RGD). Cells 2020; 9:E1510. [PMID: 32575851 PMCID: PMC7349514 DOI: 10.3390/cells9061510] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 11/25/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) were obtained from human bone marrow and amplified in cultures supplemented with human platelet lysate. Once semi-confluent, cells were seeded in solid collagen scaffolds that were rapidly colonized by the cells generating a 3D cell scaffold. Here, they acquired a myofibroblast phenotype and when exposed to appropriate chemical stimulus, developed tension and cell shortening, similar to those of striated and smooth muscle cells. Myofibroblasts contained a molecular motor-the non-muscle myosin type IIA (NMMIIA) whose crossbridge (CB) kinetics are dramatically slow compared with striated and smooth muscle myosins. Huxley's equations were used to determine the molecular mechanical properties of NMMIIA. Thank to the great number of NMMIIA molecules, we determined the statistical mechanics (SM) of MSCs, using the grand canonical ensemble which made it possible to calculate various thermodynamic entities such as the chemical affinity, statistical entropy, internal energy, thermodynamic flow, thermodynamic force, and entropy production rate. The linear relationship observed between the thermodynamic force and the thermodynamic flow allowed to establish that MSC-laden in collagen scaffolds were in a near-equilibrium stationary state (affinity ≪ RT), MSCs were also seeded in solid collagen scaffolds functionalized with the tripeptide Arg-Gly-Asp (RGD). This induced major changes in NMMIIA SM particularly by increasing the rate of entropy production. In conclusion, collagen scaffolds laden with MSCs can be viewed as a non-muscle contractile bioengineered tissue operating in a near-equilibrium linear regime, whose SM could be substantially modified by the RGD peptide.
Collapse
Affiliation(s)
- Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l’Est Francilien, 77104 Meaux, France;
| | - Vincent Kindler
- Department of Specialties in Medicine, Hematology Service, Geneva University Hospital, Faculty of Medicine, 1200 Geneva, Switzerland;
| | - Xénophon Krokidis
- Centre de Recherche Clinique, Grand Hôpital de l’Est Francilien, 77104 Meaux, France;
| | - Marie-Luce Bochaton-Piallat
- Department of Pathology and Immunology, Geneva University Hospital, Faculty of Medicine, 1200 Geneva, Switzerland;
| | - Victor Claes
- Department of Pharmaceutical Sciences, University of Antwerp, 2000 Wilrijk, Belgium;
| | - Jean-Louis Hébert
- Institut de Cardiologie, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France;
| | - Alexandre Vallée
- Diagnosis and Therapeutic Center, Hypertension and Cardiovascular Prevention Unit, Hôtel-Dieu Hospital, AP-HP, Paris-Descartes University, 75004 Paris, France;
| | - Olivier Schussler
- Department of Cardiovascular Surgery, Research Laboratory, Geneva University Hospital, Faculty of Medicine, 1200 Geneva, Switzerland;
- Department of Thoracic surgery, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
40
|
Alimohamadi H, Smith AS, Nowak RB, Fowler VM, Rangamani P. Non-uniform distribution of myosin-mediated forces governs red blood cell membrane curvature through tension modulation. PLoS Comput Biol 2020; 16:e1007890. [PMID: 32453720 PMCID: PMC7274484 DOI: 10.1371/journal.pcbi.1007890] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 06/05/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022] Open
Abstract
The biconcave disk shape of the mammalian red blood cell (RBC) is unique to the RBC and is vital for its circulatory function. Due to the absence of a transcellular cytoskeleton, RBC shape is determined by the membrane skeleton, a network of actin filaments cross-linked by spectrin and attached to membrane proteins. While the physical properties of a uniformly distributed actin network interacting with the lipid bilayer membrane have been assumed to control RBC shape, recent experiments reveal that RBC biconcave shape also depends on the contractile activity of nonmuscle myosin IIA (NMIIA) motor proteins. Here, we use the classical Helfrich-Canham model for the RBC membrane to test the role of heterogeneous force distributions along the membrane and mimic the contractile activity of sparsely distributed NMIIA filaments. By incorporating this additional contribution to the Helfrich-Canham energy, we find that the RBC biconcave shape depends on the ratio of forces per unit volume in the dimple and rim regions of the RBC. Experimental measurements of NMIIA densities at the dimple and rim validate our prediction that (a) membrane forces must be non-uniform along the RBC membrane and (b) the force density must be larger in the dimple than the rim to produce the observed membrane curvatures. Furthermore, we predict that RBC membrane tension and the orientation of the applied forces play important roles in regulating this force-shape landscape. Our findings of heterogeneous force distributions on the plasma membrane for RBC shape maintenance may also have implications for shape maintenance in different cell types. The spectrin-actin network of the membrane skeleton plays an important role in controlling specialized cell membrane morphology. In the paradigmatic red blood cell (RBC), where actin filaments are present exclusively in the membrane skeleton, recent experiments reveal that nonmuscle myosin IIA (NMIIA) motor contractility maintains the RBC biconcave disk shape. In this study, we have identified criteria for micron-scale distributions of NMIIA forces at the membrane required to maintain the biconcave disk shape of an RBC in the resting condition. Supported by experimental measurements of RBC NMIIA distribution, we showed that a heterogeneous force distribution with a larger force density at the dimple is able to capture the experimentally observed biconcave morphology of an RBC with better accuracy compared to previous models that did not consider the heterogeneity in the force distribution. Furthermore, we showed that the biconcave geometry of the RBC is closely regulated by the effective membrane tension and the direction of applied forces on the membrane. These findings can be generalized to any force-mediated membrane shape, providing insight into the role of actomyosin forces in prescribing and maintaining the morphology of different cell types.
Collapse
Affiliation(s)
- Haleh Alimohamadi
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, United States of America
| | - Alyson S. Smith
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Roberta B. Nowak
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Velia M. Fowler
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States of America
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
41
|
Banerjee A, Kulkarni S, Mukherjee A. Herpes Simplex Virus: The Hostile Guest That Takes Over Your Home. Front Microbiol 2020; 11:733. [PMID: 32457704 PMCID: PMC7221137 DOI: 10.3389/fmicb.2020.00733] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 03/30/2020] [Indexed: 12/15/2022] Open
Abstract
Alpha (α)-herpesviruses (HSV-1 and HSV-2), like other viruses, are obligate intracellular parasites. They hijack the cellular machinery to survive and replicate through evading the defensive responses by the host. The viral genome of herpes simplex viruses (HSVs) contains viral genes, the products of which are destined to exploit the host apparatus for their own existence. Cellular modulations begin from the entry point itself. The two main gateways that the virus has to penetrate are the cell membrane and the nuclear membrane. Changes in the cell membrane are triggered when the glycoproteins of HSV interact with the surface receptors of the host cell, and from here, the components of the cytoskeleton take over. The rearrangement in the cytoskeleton components help the virus to enter as well as transport to the nucleus and back to the cell membrane to spread out to the other cells. The entire carriage process is also mediated by the motor proteins of the kinesin and dynein superfamily and is directed by the viral tegument proteins. Also, the virus captures the cell’s most efficient cargo carrying system, the endoplasmic reticulum (ER)–Golgi vesicular transport machinery for egress to the cell membrane. For these reasons, the host cell has its own checkpoints where the normal functions are halted once a danger is sensed. However, a cell may be prepared for the adversities from an invading virus, and it is simply commendable that the virus has the antidote to these cellular strategies as well. The HSV viral proteins are capable of limiting the use of the transcriptional and translational tools for the cell itself, so that its own transcription and translation pathways remain unhindered. HSV prefers to constrain any self-destruction process of the cell—be it autophagy in the lysosome or apoptosis by the mitochondria, so that it can continue to parasitize the cell for its own survival. This review gives a detailed account of the significance of compartmentalization during HSV pathogenesis. It also highlights the undiscovered areas in the HSV cell biology research which demand attention for devising improved therapeutics against the infection.
Collapse
Affiliation(s)
- Anwesha Banerjee
- Division of Virology, Indian Council of Medical Research-National AIDS Research Institute, Pune, India
| | - Smita Kulkarni
- Division of Virology, Indian Council of Medical Research-National AIDS Research Institute, Pune, India
| | - Anupam Mukherjee
- Division of Virology, Indian Council of Medical Research-National AIDS Research Institute, Pune, India
| |
Collapse
|
42
|
Yoneda A, Minomi K, Tamura Y. HSP47 promotes metastasis of breast cancer by interacting with myosin IIA via the unfolded protein response transducer IRE1α. Oncogene 2020; 39:4519-4537. [PMID: 32366908 DOI: 10.1038/s41388-020-1311-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 12/21/2022]
Abstract
Breast cancer (BC) is an aggressive cancer that is a leading cause of cancer-associated death in women worldwide. Although increased expression of heat shock protein 47 (HSP47), a collagen-specific chaperone, is associated with the high malignancy of BC, its role in BC remains largely unclear. Here we show that a small population of high-invasive BC cells expresses HSP47 and that HSP47-positive high-invasive BC cells have a high metastatic potential that is completely abolished by disruption of HSP47. HSP47 interacts with non-muscle myosin IIA (NMIIA) via the unfolded protein response transducer IRE1α, resulting in enhancement of the metastatic potential of high-invasive BC cells by augmenting the contractile force of actin filaments. Ablation of NMIIA abrogates the metastatic potential of HSP47-positive high-invasive BC cells. We further show that forced expression of NMIIA confers a high metastatic potential on low-invasive BC cells in which HSP47 but not NMIIA is expressed. Overall, our study indicates that HSP47 acts as a stimulator for metastasis of BC cells and suggest that HSP47 may be a candidate for a therapeutic target against BC.
Collapse
Affiliation(s)
- Akihiro Yoneda
- Department of Molecular Therapeutics, Center for Food & Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Sapporo, 001-0021, Japan.
| | - Kenjiro Minomi
- Department of Molecular Therapeutics, Center for Food & Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Sapporo, 001-0021, Japan.,Research & Development Department, Nucleic Acid Medicine Business Division, Nitto Denko Corporation, Sapporo, 001-0021, Japan
| | - Yasuaki Tamura
- Department of Molecular Therapeutics, Center for Food & Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Sapporo, 001-0021, Japan
| |
Collapse
|
43
|
Li X, Xie S, Qian L, Cai C, Bi H, Cui W. Identification of genes related to skeletal muscle growth and development by integrated analysis of transcriptome and proteome in myostatin-edited Meishan pigs. J Proteomics 2020; 213:103628. [DOI: 10.1016/j.jprot.2019.103628] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/08/2019] [Accepted: 12/22/2019] [Indexed: 12/29/2022]
|
44
|
Abstract
Many proteins in cells and in the extracellular matrix assemble into force-bearing networks, and some proteins clearly transduce mechanical stimuli into biochemical signals. Although structural mechanisms remain poorly understood, the designs of such proteins enable mechanical forces to either inhibit or facilitate interactions of protein domains with other proteins, including small molecules and enzymes, including proteases and kinases. Here, we review some of the structural proteins and processes that exhibit distinct modes of force-dependent signal conversion.
Collapse
Affiliation(s)
- Karanvir Saini
- Molecular and Cell Biophysics Lab , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Dennis E Discher
- Molecular and Cell Biophysics Lab , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| |
Collapse
|
45
|
Vallée A, Lecarpentier Y. TGF-β in fibrosis by acting as a conductor for contractile properties of myofibroblasts. Cell Biosci 2019; 9:98. [PMID: 31827764 PMCID: PMC6902440 DOI: 10.1186/s13578-019-0362-3] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 11/30/2019] [Indexed: 12/21/2022] Open
Abstract
Myofibroblasts are non-muscle contractile cells that play a key physiologically role in organs such as the stem villi of the human placenta during physiological pregnancy. They are able to contract and relax in response to changes in the volume of the intervillous chamber. Myofibroblasts have also been observed in several diseases and are involved in wound healing and the fibrotic processes affecting several organs, such as the liver, lungs, kidneys and heart. During the fibrotic process, tissue retraction rather than contraction is correlated with collagen synthesis in the extracellular matrix, leading to irreversible fibrosis and, finally, apoptosis of myofibroblasts. The molecular motor of myofibroblasts is the non-muscle type IIA and B myosin (NMMIIA and NMMIIB). Fibroblast differentiation into myofibroblasts is largely governed by the transforming growth factor-β1 (TGF-β1). This system controls the canonical WNT/β-catenin pathway in a positive manner, and PPARγ in a negative manner. The WNT/β-catenin pathway promotes fibrosis, while PPARγ prevents it. This review focuses on the contractile properties of myofibroblasts and the conductor, TGF-β1, which together control the opposing interplay between PPARγ and the canonical WNT/β-catenin pathway.
Collapse
Affiliation(s)
- Alexandre Vallée
- 1Délégation à la Recherche Clinique (DRCI), Hôpital Foch, Suresnes, France.,DACTIM-MIS, Laboratoire de Mathématiques et Applications (LMA), CNRS, UMR 7348, Université de Poitiers, CHU de Poitiers, Poitiers, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| |
Collapse
|
46
|
Lecarpentier Y, Kindler V, Bochaton-Piallat ML, Sakic A, Claes V, Hébert JL, Vallée A, Schussler O. Tripeptide Arg-Gly-Asp (RGD) modifies the molecular mechanical properties of the non-muscle myosin IIA in human bone marrow-derived myofibroblasts seeded in a collagen scaffold. PLoS One 2019; 14:e0222683. [PMID: 31574082 PMCID: PMC6772000 DOI: 10.1371/journal.pone.0222683] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/04/2019] [Indexed: 01/21/2023] Open
Abstract
Mesenchymal stem cells (MSCs) were obtained from human bone marrow and amplified in cultures supplemented with human platelet lysate in order to generate myofibroblasts. When MSCs were seeded in solid collagen scaffolds, they differentiated into myofibroblasts that were observed to strongly bind to the substrate, forming a 3D cell scaffold network that developed tension and shortening after KCl stimulation. Moreover, MSC-laden scaffolds recapitulated the Frank-Starling mechanism so that active tension increased in response to increases in the initial length of the contractile system. This constituted a bioengineering tissue that exhibited the contractile properties observed in both striated and smooth muscles. By using the A. F. Huxley formalism, we determined the myosin crossbridge (CB) kinetics of attachment (f1) and detachment (g1 and g2), maximum myosin ATPase activity, molar myosin concentration, unitary CB force and maximum CB efficiency. CB kinetics were dramatically slow, characterizing the non-muscle myosin type IIA (NMMIIA) present in myofibroblasts. When MSCs were seeded in solid collagen scaffolds functionalized with Arg-Gly-Asp (RGD), contractility increased and CB kinetics were modified, whereas the unitary NMMIIA-CB force and maximum CB efficiency did not change. In conclusion, we provided a non-muscle bioengineering tissue whose molecular mechanical characteristics of NMMIIA were very close to those of a non-muscle contractile tissue such as the human placenta.
Collapse
Affiliation(s)
- Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l’Est Francilien, Meaux, France
- * E-mail:
| | - Vincent Kindler
- Department of Specialties in Medicine, Hematology Service, Geneva University Hospital, Switzerland Faculty of Medicine, Geneva, Switzerland
| | - Marie-Luce Bochaton-Piallat
- Department of Pathology and Immunology, Centre Médical Universitaire Geneva, Faculty of Medicine, Geneva, Switzerland
| | - Antonija Sakic
- Department of Pathology and Immunology, Centre Médical Universitaire Geneva, Faculty of Medicine, Geneva, Switzerland
| | - Victor Claes
- Department of Pharmaceutical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Jean-Louis Hébert
- Institut de Cardiologie, Hôpital de la Pitié-Salpétrière, Paris, France
| | - Alexandre Vallée
- Paris-Descartes University, Diagnosis and Therapeutic Center, Hypertension and Cardiovascular Prevention Unit, Hôtel-Dieu Hospital, Paris, France
- DRCI (Délégation à la Recherche Clinique et Industrielle) Hôpital Foch, Suresnes, France
| | - Olivier Schussler
- Department of Cardiovascular Surgery, Research Laboratory, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
47
|
Carmena A. Non-muscle myosin II activation: adding a classical touch to ROCK. Small GTPases 2019; 12:161-166. [PMID: 31552778 DOI: 10.1080/21541248.2019.1671148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Non-muscle myosin II molecules are actin-binding proteins with ATPase activity, this latter capacity providing the energy required for actin filament cross-linking and contraction. The activation of these molecular motors relies on direct phosphorylation at conserved sites through different protein kinases, including the Rho-associated coiled coil-containing kinase (ROCK). In the light of some recent results found in our lab, we comment on the necessity of additional regulatory mechanisms to control the subcellular distribution of non-muscle myosin II proteins to ensure their full activation.
Collapse
Affiliation(s)
- Ana Carmena
- Developmental Neurobiology Department, Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas/Universidad Miguel Hernández, Alicante, Spain
| |
Collapse
|
48
|
Heuzé ML, Sankara Narayana GHN, D'Alessandro J, Cellerin V, Dang T, Williams DS, Van Hest JC, Marcq P, Mège RM, Ladoux B. Myosin II isoforms play distinct roles in adherens junction biogenesis. eLife 2019; 8:46599. [PMID: 31486768 PMCID: PMC6756789 DOI: 10.7554/elife.46599] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 09/05/2019] [Indexed: 12/27/2022] Open
Abstract
Adherens junction (AJ) assembly under force is essential for many biological processes like epithelial monolayer bending, collective cell migration, cell extrusion and wound healing. The acto-myosin cytoskeleton acts as a major force-generator during the de novo formation and remodeling of AJ. Here, we investigated the role of non-muscle myosin II isoforms (NMIIA and NMIIB) in epithelial junction assembly. NMIIA and NMIIB differentially regulate biogenesis of AJ through association with distinct actin networks. Analysis of junction dynamics, actin organization, and mechanical forces of control and knockdown cells for myosins revealed that NMIIA provides the mechanical tugging force necessary for cell-cell junction reinforcement and maintenance. NMIIB is involved in E-cadherin clustering, maintenance of a branched actin layer connecting E-cadherin complexes and perijunctional actin fibres leading to the building-up of anisotropic stress. These data reveal unanticipated complementary functions of NMIIA and NMIIB in the biogenesis and integrity of AJ.
Collapse
Affiliation(s)
- Mélina L Heuzé
- Institut Jacques Monod, Université de Paris and CNRS UMR 7592, Paris, France
| | | | - Joseph D'Alessandro
- Institut Jacques Monod, Université de Paris and CNRS UMR 7592, Paris, France
| | - Victor Cellerin
- Institut Jacques Monod, Université de Paris and CNRS UMR 7592, Paris, France
| | - Tien Dang
- Institut Jacques Monod, Université de Paris and CNRS UMR 7592, Paris, France
| | - David S Williams
- Department of Chemistry, College of Science, Swansea University, Swansea, United Kingdom
| | - Jan Cm Van Hest
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Philippe Marcq
- Laboratoire Physique et Mécanique des Milieux Hétérogènes, Sorbonne Université and CNRS UMR 7636, Paris, France
| | - René-Marc Mège
- Institut Jacques Monod, Université de Paris and CNRS UMR 7592, Paris, France
| | - Benoit Ladoux
- Institut Jacques Monod, Université de Paris and CNRS UMR 7592, Paris, France
| |
Collapse
|
49
|
Hou G, Xue B, Li L, Nan Y, Zhang L, Li K, Zhao Q, Hiscox JA, Stewart JP, Wu C, Wang J, Zhou EM. Direct Interaction Between CD163 N-Terminal Domain and MYH9 C-Terminal Domain Contributes to Porcine Reproductive and Respiratory Syndrome Virus Internalization by Permissive Cells. Front Microbiol 2019; 10:1815. [PMID: 31447818 PMCID: PMC6691103 DOI: 10.3389/fmicb.2019.01815] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 07/23/2019] [Indexed: 12/16/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has a highly restricted tropism for cells of the monocyte-macrophage lineage, including porcine alveolar macrophages (PAMs). PRRSV entry into permissive cells involves several mediators in addition to two required host cell receptors, CD163 and MYH9. It is unknown whether CD163 directly interacts and/or cooperates with MYH9 to facilitate PRRSV infection. In this study, CD163 and MYH9 were co-immunoprecipitated from PAMs regardless of PRRSV infection status. Further truncation analysis indicated that the CD163 N-terminal region, containing scavenger receptor cysteine-rich domains 1 to 4 (SRCR1-4), directly interacts with the MYH9 C-terminal domain region without involvement of other adaptor proteins. Meanwhile, non-permissive HEK293T cells that stably expressed truncated swine CD163 SRCR1-4 domain did not support virus attachment. However, virus attachment to cells stably expressing SRCR5-CT domain was demonstrated to occur without appreciable virus internalization. The involvement of the SRCR1-4 domain in virus internalization was further demonstrated by the fact that incubation of recombinant SRCR1-4 protein with PAMs abolished subsequent virus internalization by permissive cells. These results demonstrated that CD163 SRCR1-4 interacts with the MYH9 C–terminal domain to facilitate PRRSV virion internalization in permissive cells, thus expanding our understanding of PRRSV cell-invasion mechanisms.
Collapse
Affiliation(s)
- Gaopeng Hou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Biyun Xue
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Liangliang Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yuchen Nan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Lu Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Kuokuo Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Julian A Hiscox
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - James P Stewart
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Chunyan Wu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jingfei Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - En-Min Zhou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
50
|
Smith AS, Pal K, Nowak RB, Demenko A, Zaninetti C, Da Costa L, Favier R, Pecci A, Fowler VM. MYH9-related disease mutations cause abnormal red blood cell morphology through increased myosin-actin binding at the membrane. Am J Hematol 2019; 94:667-677. [PMID: 30916803 PMCID: PMC6510596 DOI: 10.1002/ajh.25472] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 01/29/2023]
Abstract
MYH9-related disease (MYH9-RD) is a rare, autosomal dominant disorder caused by mutations in MYH9, the gene encoding the actin-activated motor protein non-muscle myosin IIA (NMIIA). MYH9-RD patients suffer from bleeding syndromes, progressive kidney disease, deafness, and/or cataracts, but the impact of MYH9 mutations on other NMIIA-expressing tissues remains unknown. In human red blood cells (RBCs), NMIIA assembles into bipolar filaments and binds to actin filaments (F-actin) in the spectrin-F-actin membrane skeleton to control RBC biconcave disk shape and deformability. Here, we tested the effects of MYH9 mutations in different NMIIA domains (motor, coiled-coil rod, or non-helical tail) on RBC NMIIA function. We found that MYH9-RD does not cause clinically significant anemia and that patient RBCs have normal osmotic deformability as well as normal membrane skeleton composition and micron-scale distribution. However, analysis of complete blood count data and peripheral blood smears revealed reduced hemoglobin content and elongated shapes, respectively, of MYH9-RD RBCs. Patients with mutations in the NMIIA motor domain had the highest numbers of elongated RBCs. Patients with mutations in the motor domain also had elevated association of NMIIA with F-actin at the RBC membrane. Our findings support a central role for motor domain activity in NMIIA regulation of RBC shape and define a new sub-clinical phenotype of MYH9-RD.
Collapse
Affiliation(s)
- Alyson S. Smith
- Department of Molecular Medicine, The Scripps Research
Institute, La Jolla, CA 92037
| | - Kasturi Pal
- Department of Molecular Medicine, The Scripps Research
Institute, La Jolla, CA 92037
| | - Roberta B. Nowak
- Department of Molecular Medicine, The Scripps Research
Institute, La Jolla, CA 92037
| | - Anastasiya Demenko
- Department of Molecular Medicine, The Scripps Research
Institute, La Jolla, CA 92037
| | - Carlo Zaninetti
- Department of Internal Medicine, IRCCS Policlinico San
Matteo Foundation and University of Pavia, Pavia, Italy
| | - Lydie Da Costa
- AP-HP, Service d’Hématologie Biologique,
Hôpital R. Debré, Paris F-75019, France; Université Paris 7,
Sorbonne Paris Cité, Paris F-75010, France; INSERM U1134, INTS, F-75015,
France; Laboratoire d’Excellence GR-Ex, France
| | - Remi Favier
- Assistance Publique-Hôpitaux de Paris, Armand
Trousseau Children Hospital, French Reference Center for platelet disorders, Paris,
75012, France
| | - Alessandro Pecci
- Department of Internal Medicine, IRCCS Policlinico San
Matteo Foundation and University of Pavia, Pavia, Italy
| | - Velia M. Fowler
- Department of Molecular Medicine, The Scripps Research
Institute, La Jolla, CA 92037
- Department of Biological Sciences, University of Delaware, Newark, DE 19711
| |
Collapse
|