51
|
Urizar-Arenaza I, Benedicto A, Perez-Valle A, Osinalde N, Akimov V, Muñoa-Hoyos I, Rodriguez JA, Asumendi A, Boyano MD, Blagoev B, Kratchmarova I, Subiran N. The multifunctional role of SPANX-A/D protein subfamily in the promotion of pro-tumoural processes in human melanoma. Sci Rep 2021; 11:3583. [PMID: 33574425 PMCID: PMC7878863 DOI: 10.1038/s41598-021-83169-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/27/2021] [Indexed: 12/14/2022] Open
Abstract
Human sperm protein associated with the nucleus on the X chromosome (SPANX) genes encode a protein family (SPANX-A, -B, -C and -D), whose expression is limited to the testis and spermatozoa in normal tissues and various tumour cells. SPANX-A/D proteins have been detected in metastatic melanoma cells, but their contribution to cancer development and the underlying molecular mechanisms of skin tumourigenesis remain unknown. Combining functional and proteomic approaches, the present work describes the presence of SPANX-A/D in primary and metastatic human melanoma cells and how it promotes pro-tumoural processes such as cell proliferation, motility and migration. We provide insights into the molecular features of skin tumourigenesis, describing for the first time a multifunctional role of the SPANX-A/D protein family in nuclear function, energy metabolism and cell survival, considered key hallmarks of cancer. A better comprehension of the SPANX-A/D protein subfamily and its molecular mechanisms will help to describe new aspects of tumour cell biology and develop new therapeutic targets and tumour-directed pharmacological drugs for skin tumours.
Collapse
Affiliation(s)
- Itziar Urizar-Arenaza
- Department of Physiology, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain. .,Biocruces Bizkaia Health Research Institute, Bizkaia, Spain.
| | - Aitor Benedicto
- Department of Cell Biology and Histology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Arantza Perez-Valle
- Department of Cell Biology and Histology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Nerea Osinalde
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Vyacheslav Akimov
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Iraia Muñoa-Hoyos
- Department of Physiology, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain.,Biocruces Bizkaia Health Research Institute, Bizkaia, Spain
| | - Jose Antonio Rodriguez
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Aintzane Asumendi
- Department of Cell Biology and Histology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Maria Dolores Boyano
- Department of Cell Biology and Histology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Irina Kratchmarova
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Nerea Subiran
- Department of Physiology, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain. .,Biocruces Bizkaia Health Research Institute, Bizkaia, Spain.
| |
Collapse
|
52
|
Ghosh S, Cuevas VC, Seelbinder B, Neu CP. Image-Based Elastography of Heterochromatin and Euchromatin Domains in the Deforming Cell Nucleus. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006109. [PMID: 33448065 PMCID: PMC7869959 DOI: 10.1002/smll.202006109] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/29/2020] [Indexed: 05/21/2023]
Abstract
Chromatin of the eukaryotic cell nucleus comprises microscopically dense heterochromatin and loose euchromatin domains, each with distinct transcriptional ability and roles in cellular mechanotransduction. While recent methods are developed to characterize the mechanics of nucleus, measurement of intranuclear mechanics remains largely unknown. Here, the development of "nuclear elastography," which combines microscopic imaging and computational modeling to quantify the relative elasticity of the heterochromatin and euchromatin domains, is described. Using contracting murine embryonic cardiomyocytes, nuclear elastography reveals that the heterochromatin is almost four times stiffer than the euchromatin at peak deformation. The relative elasticity between the two domains changes rapidly during the active deformation of the cardiomyocyte in the normal physiological condition but progresses more slowly in cells cultured in a mechanically stiff environment, although the relative stiffness at peak deformation does not change. Further, it is found that the disruption of the Klarsicht, ANC-1, Syne Homology domain of the Linker of Nucleoskeleton and Cytoskeleton complex compromises the intranuclear elasticity distribution resulting in elastically similar heterochromatin and euchromatin. These results provide insight into the elastography dynamics of heterochromatin and euchromatin domains and provide a noninvasive framework to further investigate the mechanobiological function of subcellular and subnuclear domains limited only by the spatiotemporal resolution of the acquired images.
Collapse
Affiliation(s)
- Soham Ghosh
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| | - Victor Crespo Cuevas
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| | - Benjamin Seelbinder
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| | - Corey P. Neu
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| |
Collapse
|
53
|
Stachecka J, Kolodziejski PA, Noak M, Szczerbal I. Alteration of active and repressive histone marks during adipogenic differentiation of porcine mesenchymal stem cells. Sci Rep 2021; 11:1325. [PMID: 33446668 PMCID: PMC7809488 DOI: 10.1038/s41598-020-79384-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/07/2020] [Indexed: 01/01/2023] Open
Abstract
A characteristic spatial distribution of the main chromatin fractions is observed in most mammalian cell nuclei, with euchromatin localized in the interior and heterochromatin at the nuclear periphery. It has been shown that interactions of heterochromatin with the nuclear lamina are necessary to establish this conventional architecture. Adipocytes are specific cells in which a reduction in lamin A/C expression is observed. We hypothesize that the loss of lamin A/C during adipogenic differentiation of mesenchymal stem cells (MSCs) may be associated with the reorganization of the main classes of chromatin in the nucleus. Thus, in this study, we examine the abundance and nuclear distribution of selected heterochromatin (H3K9me3, H3K27me3 and H4K20me3) and euchromatin (H4K8ac, H3K4me3 and H3K9ac) histone marks during in vitro adipogenesis, using the pig as a model organism. We found that not only did the expression of lamin A/C decrease in our differentiation system, but so did the expression of lamin B receptor (LBR). The level of two heterochromatin marks, H3K27me3 and H4K20me3, increased during differentiation, while no changes were observed for H3K9me3. The levels of two euchromatin histone marks, H4K8ac and H3K9ac, were significantly higher in adipocytes than in undifferentiated cells, while the level of H3K4me3 did not change significantly. The spatial distribution of all the examined histone marks altered during in vitro adipogenesis. H3K27me3 and H4K20me3 moved towards the nuclear periphery and H3K9me3 localized preferentially in the intermediate part of adipocyte nuclei. The euchromatin marks H3K9ac and H3K4me3 preferentially occupied the peripheral part of the adipocyte nuclei, while H4K8ac was more evenly distributed in the nuclei of undifferentiated and differentiated cells. Analysis of the nuclear distribution of repetitive sequences has shown their clustering and relocalization toward nuclear periphery during differentiation. Our study shows that dynamic changes in the abundance and nuclear distribution of active and repressive histone marks take place during adipocyte differentiation. Nuclear reorganization of heterochromatin histone marks may allow the maintenance of the nuclear morphology of the adipocytes, in which reduced expression of lamin A/C and LBR is observed.
Collapse
Affiliation(s)
- Joanna Stachecka
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637, Poznan, Poland
| | - Pawel A Kolodziejski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Wolynska 35, 60-637, Poznan, Poland
| | - Magdalena Noak
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637, Poznan, Poland
| | - Izabela Szczerbal
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637, Poznan, Poland.
| |
Collapse
|
54
|
Gensbittel V, Kräter M, Harlepp S, Busnelli I, Guck J, Goetz JG. Mechanical Adaptability of Tumor Cells in Metastasis. Dev Cell 2020; 56:164-179. [PMID: 33238151 DOI: 10.1016/j.devcel.2020.10.011] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/18/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022]
Abstract
The most dangerous aspect of cancer lies in metastatic progression. Tumor cells will successfully form life-threatening metastases when they undergo sequential steps along a journey from the primary tumor to distant organs. From a biomechanics standpoint, growth, invasion, intravasation, circulation, arrest/adhesion, and extravasation of tumor cells demand particular cell-mechanical properties in order to survive and complete the metastatic cascade. With metastatic cells usually being softer than their non-malignant counterparts, high deformability for both the cell and its nucleus is thought to offer a significant advantage for metastatic potential. However, it is still unclear whether there is a finely tuned but fixed mechanical state that accommodates all mechanical features required for survival throughout the cascade or whether tumor cells need to dynamically refine their properties and intracellular components at each new step encountered. Here, we review the various mechanical requirements successful cancer cells might need to fulfill along their journey and speculate on the possibility that they dynamically adapt their properties accordingly. The mechanical signature of a successful cancer cell might actually be its ability to adapt to the successive microenvironmental constraints along the different steps of the journey.
Collapse
Affiliation(s)
- Valentin Gensbittel
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Sébastien Harlepp
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Ignacio Busnelli
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany.
| | - Jacky G Goetz
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
| |
Collapse
|
55
|
Michel JB. Phylogenic Determinants of Cardiovascular Frailty, Focus on Hemodynamics and Arterial Smooth Muscle Cells. Physiol Rev 2020; 100:1779-1837. [DOI: 10.1152/physrev.00022.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The evolution of the circulatory system from invertebrates to mammals has involved the passage from an open system to a closed in-parallel system via a closed in-series system, accompanying the increasing complexity and efficiency of life’s biological functions. The archaic heart enables pulsatile motion waves of hemolymph in invertebrates, and the in-series circulation in fish occurs with only an endothelium, whereas mural smooth muscle cells appear later. The present review focuses on evolution of the circulatory system. In particular, we address how and why this evolution took place from a closed, flowing, longitudinal conductance at low pressure to a flowing, highly pressurized and bifurcating arterial compartment. However, although arterial pressure was the latest acquired hemodynamic variable, the general teleonomy of the evolution of species is the differentiation of individual organ function, supported by specific fueling allowing and favoring partial metabolic autonomy. This was achieved via the establishment of an active contractile tone in resistance arteries, which permitted the regulation of blood supply to specific organ activities via its localized function-dependent inhibition (active vasodilation). The global resistance to viscous blood flow is the peripheral increase in frictional forces caused by the tonic change in arterial and arteriolar radius, which backscatter as systemic arterial blood pressure. Consequently, the arterial pressure gradient from circulating blood to the adventitial interstitium generates the unidirectional outward radial advective conductance of plasma solutes across the wall of conductance arteries. This hemodynamic evolution was accompanied by important changes in arterial wall structure, supported by smooth muscle cell functional plasticity, including contractility, matrix synthesis and proliferation, endocytosis and phagocytosis, etc. These adaptive phenotypic shifts are due to epigenetic regulation, mainly related to mechanotransduction. These paradigms actively participate in cardio-arterial pathologies such as atheroma, valve disease, heart failure, aneurysms, hypertension, and physiological aging.
Collapse
|
56
|
Anderson JR, Phelan MM, Foddy L, Clegg PD, Peffers MJ. Ex Vivo Equine Cartilage Explant Osteoarthritis Model: A Metabolomics and Proteomics Study. J Proteome Res 2020; 19:3652-3667. [PMID: 32701294 PMCID: PMC7476031 DOI: 10.1021/acs.jproteome.0c00143] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
![]()
Osteoarthritis is an age-related
degenerative musculoskeletal disease
characterized by loss of articular cartilage, synovitis, and subchondral
bone sclerosis. Osteoarthritis pathogenesis is yet to be fully elucidated
with no osteoarthritis-specific biomarkers in clinical use. Ex vivo equine cartilage explants (n =
5) were incubated in tumor necrosis factor-α (TNF-α)/interleukin-1β
(IL-1β)-supplemented culture media for 8 days, with the media
removed and replaced at 2, 5, and 8 days. Acetonitrile metabolite
extractions of 8 day cartilage explants and media samples at all time
points underwent one-dimensional (1D) 1H nuclear magnetic
resonance metabolomic analysis, with media samples also undergoing
mass spectrometry proteomic analysis. Within the cartilage, glucose
and lysine were elevated following TNF-α/IL-1β treatment,
while adenosine, alanine, betaine, creatine, myo-inositol, and uridine
decreased. Within the culture media, 4, 4, and 6 differentially abundant
metabolites and 154, 138, and 72 differentially abundant proteins
were identified at 1–2, 3–5, and 6–8 days, respectively,
including reduced alanine and increased isoleucine, enolase 1, vimentin,
and lamin A/C following treatment. Nine potential novel osteoarthritis
neopeptides were elevated in the treated media. Implicated pathways
were dominated by those involved in cellular movement. Our innovative
study has provided insightful information on early osteoarthritis
pathogenesis, enabling potential translation for clinical markers
and possible new therapeutic targets.
Collapse
Affiliation(s)
- James R Anderson
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, U.K
| | - Marie M Phelan
- NMR Metabolomics Facility, Technology Directorate & Department of Biochemistry & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, U.K
| | - Laura Foddy
- School of Veterinary Science, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool L69 3GH, U.K
| | - Peter D Clegg
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, U.K
| | - Mandy J Peffers
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, U.K
| |
Collapse
|
57
|
Hernandez PA, Jacobsen TD, Chahine NO. Actomyosin contractility confers mechanoprotection against TNFα-induced disruption of the intervertebral disc. SCIENCE ADVANCES 2020; 6:eaba2368. [PMID: 32875103 PMCID: PMC7438088 DOI: 10.1126/sciadv.aba2368] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 07/08/2020] [Indexed: 06/11/2023]
Abstract
Inflammation triggers degradation of intervertebral disc extracellular matrix (ECM), a hallmark of disc degeneration that contributes to back pain. Mechanosensitive nucleus pulposus cells are responsible for ECM production, yet the impact of a proinflammatory microenvironment on cell mechanobiology is unknown. Using gain- and loss-of-function approaches, we show that tumor necrosis factor-α (TNFα)-induced inflammation alters cell morphology and biophysical properties (circularity, contractility, cell stiffness, and hydraulic permeability) in a mechanism dependent on actomyosin contractility in a three-dimensional (3D) culture. We found that RhoA activation rescued cells from TNFα-induced mechanobiological disruption. Using a novel explant-in-hydrogel culture system, we demonstrate that nuclear factor kappa-B nuclear translocation and transcription are mechanosensitive, and its downstream effects on ECM degradation are regulated by actomyosin contractility. Results define a scaling relationship between circularity, contractility, and hydraulic permeability that is conserved from healthy to inflammatory microenvironments and is indicative of cell mechanobiological control across scales in 3D.
Collapse
Affiliation(s)
- Paula A. Hernandez
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Timothy D. Jacobsen
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Nadeen O. Chahine
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Orthopedic Surgery, Columbia University, New York, NY, USA
| |
Collapse
|
58
|
Domingues C, Geraldo AM, Anjo SI, Matos A, Almeida C, Caramelo I, Lopes-da-Silva JA, Paiva A, Carvalho J, Pires das Neves R, Manadas B, Grãos M. Cofilin-1 Is a Mechanosensitive Regulator of Transcription. Front Cell Dev Biol 2020; 8:678. [PMID: 32903827 PMCID: PMC7438942 DOI: 10.3389/fcell.2020.00678] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
The mechanical properties of the extracellular environment are interrogated by cells and integrated through mechanotransduction. Many cellular processes depend on actomyosin-dependent contractility, which is influenced by the microenvironment’s stiffness. Here, we explored the influence of substrate stiffness on the proteome of proliferating undifferentiated human umbilical cord-matrix mesenchymal stem/stromal cells. The relative abundance of several proteins changed significantly by expanding cells on soft (∼3 kPa) or stiff substrates (GPa). Many such proteins are associated with the regulation of the actin cytoskeleton, a major player of mechanotransduction and cell physiology in response to mechanical cues. Specifically, Cofilin-1 levels were elevated in cells cultured on soft comparing with stiff substrates. Furthermore, Cofilin-1 was de-phosphorylated (active) and present in the nuclei of cells kept on soft substrates, in contrast with phosphorylated (inactive) and widespread distribution in cells on stiff. Soft substrates promoted Cofilin-1-dependent increased RNA transcription and faster RNA polymerase II-mediated transcription elongation. Cofilin-1 is part of a novel mechanism linking mechanotransduction and transcription.
Collapse
Affiliation(s)
- Catarina Domingues
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - A Margarida Geraldo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Sandra Isabel Anjo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - André Matos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Polytechnic Institute of Coimbra, Coimbra College of Agriculture, Coimbra, Portugal
| | - Cláudio Almeida
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Polytechnic Institute of Coimbra, Coimbra College of Agriculture, Coimbra, Portugal
| | - Inês Caramelo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | | | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, Coimbra, Portugal
| | - João Carvalho
- Centro de Física da Universidade de Coimbra (CFisUC), Department of Physics, University of Coimbra, Coimbra, Portugal
| | - Ricardo Pires das Neves
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - Bruno Manadas
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Mário Grãos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal.,Biocant, Technology Transfer Association, Cantanhede, Portugal
| |
Collapse
|
59
|
Piccus R, Brayson D. The nuclear envelope: LINCing tissue mechanics to genome regulation in cardiac and skeletal muscle. Biol Lett 2020; 16:20200302. [PMID: 32634376 DOI: 10.1098/rsbl.2020.0302] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Regulation of the genome is viewed through the prism of gene expression, DNA replication and DNA repair as controlled through transcription, chromatin compartmentalisation and recruitment of repair factors by enzymes such as DNA polymerases, ligases, acetylases, methylases and cyclin-dependent kinases. However, recent advances in the field of muscle cell physiology have also shown a compelling role for 'outside-in' biophysical control of genomic material through mechanotransduction. The crucial hub that transduces these biophysical signals is called the Linker of Nucleoskeleton and Cytoskeleton (LINC). This complex is embedded across the nuclear envelope, which separates the nucleus from the cytoplasm. How the LINC complex operates to mechanically regulate the many functions of DNA is becoming increasingly clear, and recent advances have provided exciting insight into how this occurs in cells from mechanically activated tissues such as skeletal and cardiac muscle. Nevertheless, there are still some notable shortcomings in our understanding of these processes and resolving these will likely help us understand how muscle diseases manifest at the level of the genome.
Collapse
Affiliation(s)
- Rachel Piccus
- Centre for Human and Applied Physiological Sciences, King's College London, London SE1 1UL, UK
| | - Daniel Brayson
- School of Cardiovascular Medicine and Sciences, King's College London, London SE5 9NU, UK.,Molecular Neurosciences, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
60
|
Ghosh S, Seelbinder B, Henderson JT, Watts RD, Scott AK, Veress AI, Neu CP. Deformation Microscopy for Dynamic Intracellular and Intranuclear Mapping of Mechanics with High Spatiotemporal Resolution. Cell Rep 2020; 27:1607-1620.e4. [PMID: 31042484 PMCID: PMC8769958 DOI: 10.1016/j.celrep.2019.04.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 01/10/2019] [Accepted: 04/01/2019] [Indexed: 12/14/2022] Open
Abstract
Structural heterogeneity is a hallmark of living cells that drives local mechanical properties and dynamic cellular responses. However, the robust quantification of intracellular mechanics is lacking from conventional methods. Here, we describe the development of deformation microscopy, which leverages conventional imaging and an automated hyperelastic warping algorithm to investigate strain history, deformation dynamics, and changes in structural heterogeneity within the interior of cells and cell nuclei. Using deformation microscopy, we found that partial or complete disruption of LINC complexes in cardiomyocytes in vitro and lamin A/C deficiency in myocytes in vivo abrogate dominant tensile loading in the nuclear interior. We also found that cells cultured on stiff substrates or in hyperosmotic conditions displayed abnormal strain burden and asymmetries at interchromatin regions, which are associated with active transcription. Deformation microscopy represents a foundational approach toward intracellular elastography, with the potential utility to provide mechanistic and quantitative insights in diverse mechanobiological applications. Ghosh et al. show that deformation microscopy, a technique based on image analysis and mechanics, reveals deformation dynamics and structural heterogeneity changes for several applications and at multiple scales, including tissues, cells, and nuclei. They reveal how the disruption of nuclear proteins and pathological conditions abrogate mechanical strain in the nuclear interior.
Collapse
Affiliation(s)
- Soham Ghosh
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Benjamin Seelbinder
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Jonathan T Henderson
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Ryan D Watts
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Adrienne K Scott
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Alexander I Veress
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Corey P Neu
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
61
|
Serna-Márquez N, Rodríguez-Hernández A, Ayala-Reyes M, Martínez-Hernández LO, Peña-Rico MÁ, Carretero-Ortega J, Hautefeuille M, Vázquez-Victorio G. Fibrillar Collagen Type I Participates in the Survival and Aggregation of Primary Hepatocytes Cultured on Soft Hydrogels. Biomimetics (Basel) 2020; 5:E30. [PMID: 32630500 PMCID: PMC7345357 DOI: 10.3390/biomimetics5020030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Liver is an essential organ that carries out multiple functions such as glycogen storage, the synthesis of plasma proteins, and the detoxification of xenobiotics. Hepatocytes are the parenchyma that sustain almost all the functions supported by this organ. Hepatocytes and non-parenchymal cells respond to the mechanical alterations that occur in the extracellular matrix (ECM) caused by organogenesis and regenerating processes. Rearrangements of the ECM modify the composition and mechanical properties that result in specific dedifferentiation programs inside the hepatic cells. Quiescent hepatocytes are embedded in the soft ECM, which contains an important concentration of fibrillar collagens in combination with a basement membrane-associated matrix (BM). This work aims to evaluate the role of fibrillar collagens and BM on actin cytoskeleton organization and the function of rat primary hepatocytes cultured on soft elastic polyacrylamide hydrogels (PAA HGs). We used rat tail collagen type I and Matrigel® as references of fibrillar collagens and BM respectively and mixed different percentages of collagen type I in combination with BM. We also used peptides obtained from decellularized liver matrices (dECM). Remarkably, hepatocytes showed a poor adhesion in the absence of collagen on soft PAA HGs. We demonstrated that collagen type I inhibited apoptosis and activated extracellular signal-regulated kinases 1/2 (ERK1/2) in primary hepatocytes cultured on soft hydrogels. Epidermal growth factor (EGF) was not able to rescue cell viability in conjugated BM but affected cell aggregation in soft PAA HGs conjugated with combinations of different proportions of collagen and BM. Interestingly, actin cytoskeleton was localized and preserved close to plasma membrane (cortical actin) and proximal to intercellular ducts (canaliculi-like structures) in soft conditions; however, albumin protein expression was not preserved, even though primary hepatocytes did not remodel their actin cytoskeleton significantly in soft conditions. This investigation highlights the important role of fibrillar collagens on soft hydrogels for the maintenance of survival and aggregation of the hepatocytes. Data suggest evaluating the conditions that allow the establishment of optimal biomimetic environments for physiology and cell biology studies, where the phenotype of primary cells may be preserved for longer periods of time.
Collapse
Affiliation(s)
- Nathalia Serna-Márquez
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Adriana Rodríguez-Hernández
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Marisol Ayala-Reyes
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Lorena Omega Martínez-Hernández
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Instituto de Biotecnología, Universidad del Papaloapan, Tuxtepec CP 68301, Oaxaca, Mexico;
| | - Miguel Ángel Peña-Rico
- Instituto de Biotecnología, Universidad del Papaloapan, Tuxtepec CP 68301, Oaxaca, Mexico;
| | - Jorge Carretero-Ortega
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Mathieu Hautefeuille
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico
| | - Genaro Vázquez-Victorio
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico
| |
Collapse
|
62
|
Alcorta-Sevillano N, Macías I, Rodríguez CI, Infante A. Crucial Role of Lamin A/C in the Migration and Differentiation of MSCs in Bone. Cells 2020; 9:cells9061330. [PMID: 32466483 PMCID: PMC7348862 DOI: 10.3390/cells9061330] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/17/2022] Open
Abstract
Lamin A/C, intermediate filament proteins from the nuclear lamina encoded by the LMNA gene, play a central role in mediating the mechanosignaling of cytoskeletal forces into nucleus. In fact, this mechanotransduction process is essential to ensure the proper functioning of other tasks also mediated by lamin A/C: the structural support of the nucleus and the regulation of gene expression. In this way, lamin A/C is fundamental for the migration and differentiation of mesenchymal stem cells (MSCs), the progenitors of osteoblasts, thus affecting bone homeostasis. Bone formation is a complex process regulated by chemical and mechanical cues, coming from the surrounding extracellular matrix. MSCs respond to signals modulating the expression levels of lamin A/C, and therefore, adapting their nuclear shape and stiffness. To promote cell migration, MSCs need soft nuclei with low lamin A content. Conversely, during osteogenic differentiation, lamin A/C levels are known to be increased. Several LMNA mutations present a negative impact in the migration and osteogenesis of MSCs, affecting bone tissue homeostasis and leading to pathological conditions. This review aims to describe these concepts by discussing the latest state-of-the-art in this exciting area, focusing on the relationship between lamin A/C in MSCs' function and bone tissue from both, health and pathological points of view.
Collapse
|
63
|
Gómez-Domínguez D, Epifano C, de Miguel F, Castaño AG, Vilaplana-Martí B, Martín A, Amarilla-Quintana S, Bertrand AT, Bonne G, Ramón-Azcón J, Rodríguez-Milla MA, Pérez de Castro I. Consequences of Lmna Exon 4 Mutations in Myoblast Function. Cells 2020; 9:cells9051286. [PMID: 32455813 PMCID: PMC7291140 DOI: 10.3390/cells9051286] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/06/2020] [Accepted: 05/16/2020] [Indexed: 02/06/2023] Open
Abstract
Laminopathies are causally associated with mutations on the Lamin A/C gene (LMNA). To date, more than 400 mutations in LMNA have been reported in patients. These mutations are widely distributed throughout the entire gene and are associated with a wide range of phenotypes. Unfortunately, little is known about the mechanisms underlying the effect of the majority of these mutations. This is the case of more than 40 mutations that are located at exon 4. Using CRISPR/Cas9 technology, we generated a collection of Lmna exon 4 mutants in mouse C2C12 myoblasts. These cell models included different types of exon 4 deletions and the presence of R249W mutation, one of the human variants associated with a severe type of laminopathy, LMNA-associated congenital muscular dystrophy (L-CMD). We characterized these clones by measuring their nuclear circularity, myogenic differentiation capacity in 2D and 3D conditions, DNA damage, and levels of p-ERK and p-AKT (phosphorylated Mitogen-Activated Protein Kinase 1/3 and AKT serine/threonine kinase 1). Our results indicated that Lmna exon 4 mutants showed abnormal nuclear morphology. In addition, levels and/or subcellular localization of different members of the lamin and LINC (LInker of Nucleoskeleton and Cytoskeleton) complex were altered in all these mutants. Whereas no significant differences were observed for ERK and AKT activities, the accumulation of DNA damage was associated to the Lmna p.R249W mutant myoblasts. Finally, significant myogenic differentiation defects were detected in the Lmna exon 4 mutants. These results have key implications in the development of future therapeutic strategies for the treatment of laminopathies.
Collapse
Affiliation(s)
- Déborah Gómez-Domínguez
- Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo km2.2, E-28029 Madrid, Spain; (D.G.-D.); (F.d.M.); (B.V.-M.); (A.M.); (M.A.R.-M.)
| | - Carolina Epifano
- Fundación Andrés Marcio, niños contra la laminopatía, C/Núñez de Balboa, 11, E-28001 Madrid, Spain;
| | - Fernando de Miguel
- Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo km2.2, E-28029 Madrid, Spain; (D.G.-D.); (F.d.M.); (B.V.-M.); (A.M.); (M.A.R.-M.)
- Universidad Europea de Madrid, C/ Tajo, s/n, E-28670 Villaviciosa de Odón, Spain
| | - Albert García Castaño
- Institute for Bioengineering of Catalonia (IBEC), C/Baldiri Reixac, 10-12, E-08028 Barcelona, Spain; (A.G.C.); (J.R.-A.)
| | - Borja Vilaplana-Martí
- Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo km2.2, E-28029 Madrid, Spain; (D.G.-D.); (F.d.M.); (B.V.-M.); (A.M.); (M.A.R.-M.)
| | - Alberto Martín
- Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo km2.2, E-28029 Madrid, Spain; (D.G.-D.); (F.d.M.); (B.V.-M.); (A.M.); (M.A.R.-M.)
| | - Sandra Amarilla-Quintana
- Fundación de Investigación HM Hospitales, Plaza del Conde Valle Suchil, 2, E-28015 Madrid, Spain;
| | - Anne T Bertrand
- UMRS 974, Center of Research in Myology, Institut de Myologie, Sorbonne Université, INSERM, 75013 Paris, France; (A.T.B.); (G.B.)
| | - Gisèle Bonne
- UMRS 974, Center of Research in Myology, Institut de Myologie, Sorbonne Université, INSERM, 75013 Paris, France; (A.T.B.); (G.B.)
| | - Javier Ramón-Azcón
- Institute for Bioengineering of Catalonia (IBEC), C/Baldiri Reixac, 10-12, E-08028 Barcelona, Spain; (A.G.C.); (J.R.-A.)
- ICREA-Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| | - Miguel A Rodríguez-Milla
- Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo km2.2, E-28029 Madrid, Spain; (D.G.-D.); (F.d.M.); (B.V.-M.); (A.M.); (M.A.R.-M.)
| | - Ignacio Pérez de Castro
- Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo km2.2, E-28029 Madrid, Spain; (D.G.-D.); (F.d.M.); (B.V.-M.); (A.M.); (M.A.R.-M.)
- Correspondence: ; Tel.: +34-918223188
| |
Collapse
|
64
|
Role of the nuclear membrane protein Emerin in front-rear polarity of the nucleus. Nat Commun 2020; 11:2122. [PMID: 32358486 PMCID: PMC7195445 DOI: 10.1038/s41467-020-15910-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 04/02/2020] [Indexed: 12/03/2022] Open
Abstract
Cell polarity refers to the intrinsic asymmetry of cells, including the orientation of the cytoskeleton. It affects cell shape and structure as well as the distribution of proteins and organelles. In migratory cells, front-rear polarity is essential and dictates movement direction. While the link between the cytoskeleton and nucleus is well-studied, we aim to investigate if front-rear polarity can be transmitted to the nucleus. We show that the knock-down of emerin, an integral protein of the nuclear envelope, abolishes preferential localization of several nuclear proteins. We propose that the frontally biased localization of the endoplasmic reticulum, through which emerin reaches the nuclear envelope, is sufficient to generate its observed bias. In primary emerin-deficient myoblasts, its expression partially rescues the polarity of the nucleus. Our results demonstrate that front-rear cell polarity is transmitted to the nucleus and that emerin is an important determinant of nuclear polarity. During cell migration, cells are polarized with distinct front vs. rear regions but whether and how polarity is transmitted to the nucleus is unclear. Here the authors show that frontally-biased endoplasmic reticulum and the nuclear membrane protein Emerin contribute to front-rear nuclear cell polarity.
Collapse
|
65
|
Abstract
The mechanical reprogramming of fibroblasts, followed by their redifferentiation into rejuvenated fibroblasts in an optimized 3D collagen matrix, made these cells more contractile and more efficient at synthesizing matrix components including laminin, fibronectin, and collagen-IV. Moreover, the rejuvenated fibroblasts obtained through this approach exhibited a decrease in DNA damage. The rejuvenated fibroblasts derived from this method precisely align into tissue architectures, suggesting its potential application as clinical implants in tissue engineering and regenerative medicine. Over the course of the aging process, fibroblasts lose contractility, leading to reduced connective-tissue stiffness. A promising therapeutic avenue for functional rejuvenation of connective tissue is reprogrammed fibroblast replacement, although major hurdles still remain. Toward this, we recently demonstrated that the laterally confined growth of fibroblasts on micropatterned substrates induces stem-cell-like spheroids. In this study, we embedded these partially reprogrammed spheroids in collagen-I matrices of varying densities, mimicking different three-dimensional (3D) tissue constraints. In response to such matrix constraints, these spheroids regained their fibroblastic properties and sprouted to form 3D connective-tissue networks. Interestingly, we found that these differentiated fibroblasts exhibit reduced DNA damage, enhanced cytoskeletal gene expression, and actomyosin contractility. In addition, the rejuvenated fibroblasts show increased matrix protein (fibronectin and laminin) deposition and collagen remodeling compared to the parental fibroblast tissue network. Furthermore, we show that the partially reprogrammed cells have comparatively open chromatin compaction states and may be more poised to redifferentiate into contractile fibroblasts in 3D-collagen matrix. Collectively, our results highlight efficient fibroblast rejuvenation through laterally confined reprogramming, which has important implications in regenerative medicine.
Collapse
|
66
|
Multiple particle tracking analysis in isolated nuclei reveals the mechanical phenotype of leukemia cells. Sci Rep 2020; 10:6707. [PMID: 32317728 PMCID: PMC7174401 DOI: 10.1038/s41598-020-63682-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
The nucleus is fundamentally composed by lamina and nuclear membranes that enclose the chromatin, nucleoskeletal components and suspending nucleoplasm. The functional connections of this network integrate external stimuli into cell signals, including physical forces to mechanical responses of the nucleus. Canonically, the morphological characteristics of the nucleus, as shape and size, have served for pathologists to stratify and diagnose cancer patients; however, novel biophysical techniques must exploit physical parameters to improve cancer diagnosis. By using multiple particle tracking (MPT) technique on chromatin granules, we designed a SURF (Speeded Up Robust Features)-based algorithm to study the mechanical properties of isolated nuclei and in living cells. We have determined the apparent shear stiffness, viscosity and optical density of the nucleus, and how the chromatin structure influences on these biophysical values. Moreover, we used our MPT-SURF analysis to study the apparent mechanical properties of isolated nuclei from patients of acute lymphoblastic leukemia. We found that leukemia cells exhibited mechanical differences compared to normal lymphocytes. Interestingly, isolated nuclei from high-risk leukemia cells showed increased viscosity than their counterparts from normal lymphocytes, whilst nuclei from relapsed-patient's cells presented higher density than those from normal lymphocytes or standard- and high-risk leukemia cells. Taken together, here we presented how MPT-SURF analysis of nuclear chromatin granules defines nuclear mechanical phenotypic features, which might be clinically relevant.
Collapse
|
67
|
Hamczyk MR, Villa-Bellosta R, Quesada V, Gonzalo P, Vidak S, Nevado RM, Andrés-Manzano MJ, Misteli T, López-Otín C, Andrés V. Progerin accelerates atherosclerosis by inducing endoplasmic reticulum stress in vascular smooth muscle cells. EMBO Mol Med 2020; 11:emmm.201809736. [PMID: 30862662 PMCID: PMC6460349 DOI: 10.15252/emmm.201809736] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a rare genetic disorder caused by progerin, a mutant lamin A variant. HGPS patients display accelerated aging and die prematurely, typically from atherosclerosis complications. Recently, we demonstrated that progerin‐driven vascular smooth muscle cell (VSMC) loss accelerates atherosclerosis leading to premature death in apolipoprotein E‐deficient mice. However, the molecular mechanism underlying this process remains unknown. Using a transcriptomic approach, we identify here endoplasmic reticulum stress (ER) and the unfolded protein responses as drivers of VSMC death in two mouse models of HGPS exhibiting ubiquitous and VSMC‐specific progerin expression. This stress pathway was also activated in HGPS patient‐derived cells. Targeting ER stress response with a chemical chaperone delayed medial VSMC loss and inhibited atherosclerosis in both progeria models, and extended lifespan in the VSMC‐specific model. Our results identify a mechanism underlying cardiovascular disease in HGPS that could be targeted in patients. Moreover, these findings may help to understand other vascular diseases associated with VSMC death, and provide insight into aging‐dependent vascular damage related to accumulation of unprocessed toxic forms of lamin A.
Collapse
Affiliation(s)
- Magda R Hamczyk
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain.,Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Ricardo Villa-Bellosta
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Fundación Instituto de Investigación Sanitaria Fundación Jiménez Díaz (FIIS-FJD), Madrid, Spain
| | - Víctor Quesada
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | - Pilar Gonzalo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Sandra Vidak
- Cell Biology of Genomes Group, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Rosa M Nevado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - María J Andrés-Manzano
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Tom Misteli
- Cell Biology of Genomes Group, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain .,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain .,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| |
Collapse
|
68
|
Chromatin and Cytoskeletal Tethering Determine Nuclear Morphology in Progerin-Expressing Cells. Biophys J 2020; 118:2319-2332. [PMID: 32320674 DOI: 10.1016/j.bpj.2020.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/18/2020] [Accepted: 04/03/2020] [Indexed: 12/13/2022] Open
Abstract
The nuclear morphology of eukaryotic cells is determined by the interplay between the lamina forming the nuclear skeleton, the chromatin inside the nucleus, and the coupling with the cytoskeleton. Nuclear alterations are often associated with pathological conditions as in Hutchinson-Gilford progeria syndrome, in which a mutation in the lamin A gene yields an altered form of the protein, named progerin, and an aberrant nuclear shape. Here, we introduce an inducible cellular model of Hutchinson-Gilford progeria syndrome in HeLa cells in which increased progerin expression leads to alterations in the coupling of the lamin shell with cytoskeletal or chromatin tethers as well as with polycomb group proteins. Furthermore, our experiments show that progerin expression leads to enhanced nuclear shape fluctuations in response to cytoskeletal activity. To interpret the experimental results, we introduce a computational model of the cell nucleus that explicitly includes chromatin fibers, the nuclear shell, and coupling with the cytoskeleton. The model allows us to investigate how the geometrical organization of the chromatin-lamin tether affects nuclear morphology and shape fluctuations. In sum, our findings highlight the crucial role played by lamin-chromatin and lamin-cytoskeletal alterations in determining nuclear shape morphology and in affecting cellular functions and gene regulation.
Collapse
|
69
|
Yattah C, Hernandez M, Huang D, Park H, Liao W, Casaccia P. Dynamic Lamin B1-Gene Association During Oligodendrocyte Progenitor Differentiation. Neurochem Res 2020; 45:606-619. [PMID: 32020491 PMCID: PMC7060805 DOI: 10.1007/s11064-019-02941-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/04/2019] [Accepted: 12/19/2019] [Indexed: 12/27/2022]
Abstract
Differentiation of oligodendrocytes (OL) from progenitor cells (OPC) is the result of a unique program of gene expression, which is further regulated by the formation of topological domains of association with the nuclear lamina. In this study, we show that cultured OPC were characterized by progressively declining levels of endogenous Lamin B1 (LMNB1) during differentiation into OL. We then identify the genes dynamically associated to the nuclear lamina component LMNB1 during this transition, using a well established technique called DamID, which is based on the ability of a bacterially-derived deoxyadenosine methylase (Dam), to modify genomic regions in close proximity. We expressed a fusion protein containing Dam and LMNB1 in OPC (OPCLMNB1-Dam) and either kept them proliferating or differentiated them into OL (OLLMNB1-Dam) and identified genes that were dynamically associated to LMNB1 with differentiation. Importantly, we identified Lss, the gene encoding for lanosterol synthase, a key enzyme in cholesterol synthesis, as associated to the nuclear lamina in OLLMNB1-Dam. This finding could at least in part explain the lipid dysregulation previously reported for mouse models of ADLD characterized by persistent LMNB1 expression in oligodendrocytes.
Collapse
Affiliation(s)
- Camila Yattah
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of the City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA
- Graduate Program in Biochemistry, The Graduate Center of The City University of New York, 365 5th Avenue, New York, NY, 10016, USA
| | - Marylens Hernandez
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dennis Huang
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of the City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA
- Graduate Program in Biochemistry, The Graduate Center of The City University of New York, 365 5th Avenue, New York, NY, 10016, USA
| | - HyeJin Park
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of the City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA
| | - Will Liao
- New York Genome Center, New York, NY, 10013, USA
| | - Patrizia Casaccia
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of the City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Graduate Program in Biochemistry and in Biology, The Graduate Center of The City University of New York, 365 5th Avenue, New York, NY, 10016, USA.
| |
Collapse
|
70
|
Ward M, Iskratsch T. Mix and (mis-)match - The mechanosensing machinery in the changing environment of the developing, healthy adult and diseased heart. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118436. [PMID: 30742931 PMCID: PMC7042712 DOI: 10.1016/j.bbamcr.2019.01.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/07/2019] [Accepted: 01/29/2019] [Indexed: 01/01/2023]
Abstract
The composition and the stiffness of cardiac microenvironment change during development and/or in heart disease. Cardiomyocytes (CMs) and their progenitors sense these changes, which decides over the cell fate and can trigger CM (progenitor) proliferation, differentiation, de-differentiation or death. The field of mechanobiology has seen a constant increase in output that also includes a wealth of new studies specific to cardiac or cardiomyocyte mechanosensing. As a result, mechanosensing and transduction in the heart is increasingly being recognised as a main driver of regulating the heart formation and function. Recent work has for instance focused on measuring the molecular, physical and mechanical changes of the cellular environment - as well as intracellular contributors to the passive stiffness of the heart. On the other hand, a variety of new studies shed light into the molecular machinery that allow the cardiomyocytes to sense these properties. Here we want to discuss the recent work on this topic, but also specifically focus on how the different components are regulated at various stages during development, in health or disease in order to highlight changes that might contribute to disease progression and heart failure.
Collapse
Key Words
- cm, cardiomyocytes
- hcm, hypertrophic cardiomyopathy
- dcm, dilated cardiomyopathy
- icm, idiopathic cardiomyopathy
- myh, myosin heavy chain
- tnnt, troponin t
- tnni, troponin i
- afm, atomic force microscope
- mre, magnetic resonance elastography
- swe, ultrasound cardiac shear-wave elastography
- lv, left ventricle
- lox, lysyl oxidase
- loxl, lysyl oxidase like protein
- lh, lysyl hydroxylase
- lys, lysin
- lccs, lysald-derived collagen crosslinks
- hlccs, hylald-derived collagen crosslinks
- pka, protein kinase a
- pkc, protein kinase c
- vash1, vasohibin-1
- svbp, small vasohibin binding protein
- tcp, tubulin carboxypeptidase
- ttl, tubulin tyrosine ligase
- mrtf, myocardin-related transcription factor
- gap, gtpase activating protein
- gef, guanine nucleotide exchange factor
Collapse
Affiliation(s)
- Matthew Ward
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, United Kingdom
| | - Thomas Iskratsch
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, United Kingdom.
| |
Collapse
|
71
|
Zonderland J, Moldero IL, Anand S, Mota C, Moroni L. Dimensionality changes actin network through lamin A/C and zyxin. Biomaterials 2020; 240:119854. [PMID: 32087459 DOI: 10.1016/j.biomaterials.2020.119854] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/08/2020] [Accepted: 02/07/2020] [Indexed: 12/16/2022]
Abstract
Mechanosensing proteins have mainly been investigated in 2D culture platforms, while understanding their regulation in 3D enviroments is critical for tissue engineering. Among mechanosensing proteins, the actin cytoskeleton plays a key role in human mesenchymal stromal cells (hMSCs) activity, but its regulation in 3D tissue engineered scaffolds remains poorly studied. Here, we show that human mesenchymal stromal cells (hMSCs) cultured on 3D electrospun scaffolds made of a stiff material do not form actin stress fibers, contrary to hMSCs on 2D films of the same material. On 3D electrospun and additive manufactured scaffolds, hMSCs also displayed fewer focal adhesions, lower lamin A and C expression and less YAP1 nuclear localization and myosin light chain phosphorylation. Together, this strongly suggests that dimensionality prevents the build-up of cellular tension, even on stiff materials. Knock down of either lamin A and C or zyxin resulted in fewer stress fibers in the cell center. Zyxin knock down reduced lamin A and C expression, but not vice versa, showing that this signal chain starts from the outside of the cell. Lineage commitment was not affected by the lack of these important osteogenic proteins in 3D, as all cells committed to osteogenesis in bi-potential medium. Our study demonstrates that dimensionality changes the actin cytoskeleton through lamin A and C and zyxin, and highlights the difference in the regulation of lineage commitment in 3D enviroments. Together, these results can have important implications for future scaffold design for both stiff- and soft tissue engineering constructs.
Collapse
Affiliation(s)
- Jip Zonderland
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ET, Maastricht, the Netherlands
| | - Ivan Lorenzo Moldero
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ET, Maastricht, the Netherlands
| | - Shivesh Anand
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ET, Maastricht, the Netherlands
| | - Carlos Mota
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ET, Maastricht, the Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ET, Maastricht, the Netherlands.
| |
Collapse
|
72
|
Urciuoli E, Petrini S, D’Oria V, Leopizzi M, Della Rocca C, Peruzzi B. Nuclear Lamins and Emerin Are Differentially Expressed in Osteosarcoma Cells and Scale with Tumor Aggressiveness. Cancers (Basel) 2020; 12:cancers12020443. [PMID: 32069980 PMCID: PMC7073215 DOI: 10.3390/cancers12020443] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 02/10/2020] [Indexed: 12/17/2022] Open
Abstract
The nuclear lamina is essential for the maintenance of nuclear shape and mechanics. Mutations in lamin genes have been identified in a heterogeneous spectrum of human diseases known as “laminopathies” associated with nuclear envelope defects and deregulation of cellular functions. Interestingly, osteosarcoma is the only neoplasm described in the literature in association with laminopathies. This study aims characterized the expression of A-type and B-type lamins and emerin in osteosarcoma, revealing a higher percentage of dysmorphic nuclei in osteosarcoma cells in comparison to normal osteoblasts and all the hallmarks of laminopathic features. Both lamins and emerin were differentially expressed in osteosarcoma cell lines in comparison to normal osteoblasts and correlated with tumor aggressiveness. We analysed lamin A/C expression in a tissue-microarray including osteosarcoma samples with different prognosis, finding a positive correlation between lamin A/C expression and the overall survival of osteosarcoma patients. An inefficient MKL1 nuclear shuttling and actin depolymerization, as well as a reduced expression of pRb and a decreased YAP nuclear content were observed in A-type lamin deficient 143B cells. In conclusion, we described for the first time laminopathic nuclear phenotypes in osteosarcoma cells, providing evidence for an altered lamins and emerin expression and a deregulated nucleoskeleton architecture of this tumor.
Collapse
Affiliation(s)
- Enrica Urciuoli
- Multifactorial Disease and Complex Phenotype Area, Research Center, Bambino Gesù Children’s Hospital, 00165 Rome, Italy;
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Research Center, Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (S.P.); (V.D.)
| | - Valentina D’Oria
- Confocal Microscopy Core Facility, Research Center, Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (S.P.); (V.D.)
| | - Martina Leopizzi
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University, 04100 Latina, Italy; (M.L.); (C.D.R.)
| | - Carlo Della Rocca
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University, 04100 Latina, Italy; (M.L.); (C.D.R.)
| | - Barbara Peruzzi
- Multifactorial Disease and Complex Phenotype Area, Research Center, Bambino Gesù Children’s Hospital, 00165 Rome, Italy;
- Correspondence: ; Tel.: +39-06-6859-2938
| |
Collapse
|
73
|
Moure A, Gomez H. Dual role of the nucleus in cell migration on planar substrates. Biomech Model Mechanobiol 2020; 19:1491-1508. [PMID: 31907682 DOI: 10.1007/s10237-019-01283-6] [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: 07/24/2019] [Accepted: 12/21/2019] [Indexed: 01/09/2023]
Abstract
Cell migration is essential to sustain life. There have been significant advances in the understanding of the mechanisms that control cell crawling, but the role of the nucleus remains poorly understood. The nucleus exerts a tight control of cell migration in 3D environments, but its influence in 2D migration on planar substrates remains unclear. Here, we study the role of the cell nucleus in 2D cell migration using a computational model of fish keratocytes. Our results indicate that the apparently minor role played by the nucleus emerges from two antagonist effects: While the nucleus modifies the spatial distributions of actin and myosin in a way that reduces cell velocity (e.g., the nucleus displaces myosin to the sides and front of the cell), its mechanical connection with the cytoskeleton alters the intracellular stresses promoting cell migration. Overall, the favorable effect of the nucleus-cytoskeleton connection prevails, which may explain why regular cells usually move faster than enucleated cells.
Collapse
Affiliation(s)
- Adrian Moure
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47907, USA.
| | - Hector Gomez
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47907, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47906, USA
| |
Collapse
|
74
|
Koaykul C, Kim MH, Kawahara Y, Yuge L, Kino-Oka M. Maintenance of Neurogenic Differentiation Potential in Passaged Bone Marrow-Derived Human Mesenchymal Stem Cells Under Simulated Microgravity Conditions. Stem Cells Dev 2019; 28:1552-1561. [PMID: 31588849 DOI: 10.1089/scd.2019.0146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) are considered to be able to adapt to environmental changes induced by gravity during cell expansion. In this study, we investigated neurogenic differentiation potential of passaged hMSCs under conventional gravity and simulated microgravity conditions. Immunostaining, quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR), and western blot analysis of neurogenic differentiation markers, neurofilament heavy (NF-H), and microtubule-associated protein 2 (MAP2) revealed that differentiated cells from the cells cultured under simulated microgravity conditions expressed higher neurogenic levels than those from conventional gravity conditions. The levels of NF-H and MAP2 in the cells from simulated microgravity conditions were consistent during passage culture, whereas cells from conventional gravity conditions exhibited a reduction of the neurogenic levels against an increase of their passage number. In growth culture, cells under simulated microgravity conditions showed less apical stress fibers over their nucleus with fewer cells having a polarization of lamin A/C than those under conventional gravity conditions. The ratio of lamin A/C to lamin B expression in the cells under simulated microgravity conditions was constant; however, cells cultured under conventional gravity conditions showed an increase in the lamin ratio during passages. Furthermore, analysis of activating H3K4me3 and repressive H3K27me3 modifications at promoters of neuronal lineage genes indicated that cells passaged under simulated microgravity conditions sustained the methylation during serial cultivation. Nevertheless, the enrichment of H3K27me3 significantly increased in the passaged cells cultured under conventional gravity conditions. These results demonstrated that simulated microgravity-coordinated cytoskeleton-lamin reorganization leads to suppression of histone modification associated with neurogenic differentiation capacity of passaged hMSCs.
Collapse
Affiliation(s)
- Chaiyong Koaykul
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
| | | | - Louis Yuge
- Space Bio-Laboratories Co., Ltd., Hiroshima, Japan.,Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
| |
Collapse
|
75
|
Sneider A, Hah J, Wirtz D, Kim DH. Recapitulation of molecular regulators of nuclear motion during cell migration. Cell Adh Migr 2019; 13:50-62. [PMID: 30261154 PMCID: PMC6527386 DOI: 10.1080/19336918.2018.1506654] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/05/2018] [Accepted: 07/18/2018] [Indexed: 01/12/2023] Open
Abstract
Cell migration is a highly orchestrated cellular event that involves physical interactions of diverse subcellular components. The nucleus as the largest and stiffest organelle in the cell not only maintains genetic functionality, but also actively changes its morphology and translocates through dynamic formation of nucleus-bound contractile stress fibers. Nuclear motion is an active and essential process for successful cell migration and nucleus self-repairs in response to compression and extension forces in complex cell microenvironment. This review recapitulates molecular regulators that are crucial for nuclear motility during cell migration and highlights recent advances in nuclear deformation-mediated rupture and repair processes in a migrating cell.
Collapse
Affiliation(s)
- Alexandra Sneider
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jungwon Hah
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
76
|
Sphingosine 1-Phosphate (S1P)/ S1P Receptor Signaling and Mechanotransduction: Implications for Intrinsic Tissue Repair/Regeneration. Int J Mol Sci 2019; 20:ijms20225545. [PMID: 31703256 PMCID: PMC6888058 DOI: 10.3390/ijms20225545] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 12/16/2022] Open
Abstract
Tissue damage, irrespective from the underlying etiology, destroys tissue structure and, eventually, function. In attempt to achieve a morpho-functional recover of the damaged tissue, reparative/regenerative processes start in those tissues endowed with regenerative potential, mainly mediated by activated resident stem cells. These cells reside in a specialized niche that includes different components, cells and surrounding extracellular matrix (ECM), which, reciprocally interacting with stem cells, direct their cell behavior. Evidence suggests that ECM stiffness represents an instructive signal for the activation of stem cells sensing it by various mechanosensors, able to transduce mechanical cues into gene/protein expression responses. The actin cytoskeleton network dynamic acts as key mechanotransducer of ECM signal. The identification of signaling pathways influencing stem cell mechanobiology may offer therapeutic perspectives in the regenerative medicine field. Sphingosine 1-phosphate (S1P)/S1P receptor (S1PR) signaling, acting as modulator of ECM, ECM-cytoskeleton linking proteins and cytoskeleton dynamics appears a promising candidate. This review focuses on the current knowledge on the contribution of S1P/S1PR signaling in the control of mechanotransduction in stem/progenitor cells. The potential contribution of S1P/S1PR signaling in the mechanobiology of skeletal muscle stem cells will be argued based on the intriguing findings on S1P/S1PR action in this mechanically dynamic tissue.
Collapse
|
77
|
Antmen E, Demirci U, Hasirci V. Amplification of nuclear deformation of breast cancer cells by seeding on micropatterned surfaces to better distinguish their malignancies. Colloids Surf B Biointerfaces 2019; 183:110402. [DOI: 10.1016/j.colsurfb.2019.110402] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/21/2019] [Accepted: 07/26/2019] [Indexed: 12/16/2022]
|
78
|
Argentati C, Morena F, Tortorella I, Bazzucchi M, Porcellati S, Emiliani C, Martino S. Insight into Mechanobiology: How Stem Cells Feel Mechanical Forces and Orchestrate Biological Functions. Int J Mol Sci 2019; 20:E5337. [PMID: 31717803 PMCID: PMC6862138 DOI: 10.3390/ijms20215337] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/23/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022] Open
Abstract
The cross-talk between stem cells and their microenvironment has been shown to have a direct impact on stem cells' decisions about proliferation, growth, migration, and differentiation. It is well known that stem cells, tissues, organs, and whole organisms change their internal architecture and composition in response to external physical stimuli, thanks to cells' ability to sense mechanical signals and elicit selected biological functions. Likewise, stem cells play an active role in governing the composition and the architecture of their microenvironment. Is now being documented that, thanks to this dynamic relationship, stemness identity and stem cell functions are maintained. In this work, we review the current knowledge in mechanobiology on stem cells. We start with the description of theoretical basis of mechanobiology, continue with the effects of mechanical cues on stem cells, development, pathology, and regenerative medicine, and emphasize the contribution in the field of the development of ex-vivo mechanobiology modelling and computational tools, which allow for evaluating the role of forces on stem cell biology.
Collapse
Affiliation(s)
- Chiara Argentati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
| | - Ilaria Tortorella
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
| | - Martina Bazzucchi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
| | - Serena Porcellati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy
| |
Collapse
|
79
|
Vázquez-Victorio G, Peto-Gutiérrez C, Díaz-Bello B, Cano-Jorge M, Pérez-Calixto D, Jiménez-Escobar A, Espinosa-Matías S, Lara Martínez R, Courson R, Malaquin L, Zamarrón-Hernández D, Hautefeuille M. Building a microfluidic cell culture platform with stiffness control using Loctite 3525 glue. LAB ON A CHIP 2019; 19:3512-3525. [PMID: 31544189 DOI: 10.1039/c9lc00649d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The study of mechanotransduction signals and cell response to mechanical properties requires designing culture substrates that possess some, or ideally all, of the following characteristics: (1) biological compatibility and adhesive properties, (2) stiffness control or tunability in a dynamic mode, (3) patternability on the microscale and (4) integrability in microfluidic chips. The most common materials used to address cell mechanotransduction are hydrogels, due to their softness. However, they may be impractical when complex scaffolds are sought and they lack viscous dissipative properties that are very important in mechanobiology. In this work, we show that an off-the-shelf, biocompatible photosensitive glue, Loctite 3525, may be used readily in mechanobiology assays without any special treatment prior to fabrication of cell culture platforms. Despite a high (MPa) stiffness easily tunable by UV exposure time at a fixed dose, 3T3 fibroblasts showed a response to the mechanics of the material similar to that obtained on much softer (kPa) hydrogels. Loctite's viscous dissipation properties indeed seemed to be responsible for such cell mechanical response, as suggested by recent works where more complex two-phase hydrogels were employed. More interestingly, it was possible to stiffen soft Loctite substrates by post-exposing them during cell culture, to observe changes in cell spreading caused by a dynamic stiffness modification. Thanks to Loctite 3525's patternability, micropillars were also fabricated to demonstrate the compatibility with traction force microscopy studies. Finally, the glue was used as an excellent adhesion layer for hydrogels on glass or PDMS, without the need for additional treatment, enabling the easy fabrication of microfluidic chips integrating hydrogels.
Collapse
Affiliation(s)
- Genaro Vázquez-Victorio
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Circuito Exterior S/N, Ciudad Universitaria CP, 04510, Ciudad de México, Mexico.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Gilbert HTJ, Mallikarjun V, Dobre O, Jackson MR, Pedley R, Gilmore AP, Richardson SM, Swift J. Nuclear decoupling is part of a rapid protein-level cellular response to high-intensity mechanical loading. Nat Commun 2019; 10:4149. [PMID: 31515493 PMCID: PMC6742657 DOI: 10.1038/s41467-019-11923-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/26/2019] [Indexed: 12/17/2022] Open
Abstract
Studies of cellular mechano-signaling have often utilized static models that do not fully replicate the dynamics of living tissues. Here, we examine the time-dependent response of primary human mesenchymal stem cells (hMSCs) to cyclic tensile strain (CTS). At low-intensity strain (1 h, 4% CTS at 1 Hz), cell characteristics mimic responses to increased substrate stiffness. As the strain regime is intensified (frequency increased to 5 Hz), we characterize rapid establishment of a broad, structured and reversible protein-level response, even as transcription is apparently downregulated. Protein abundance is quantified coincident with changes to protein conformation and post-translational modification (PTM). Furthermore, we characterize changes to the linker of nucleoskeleton and cytoskeleton (LINC) complex that bridges the nuclear envelope, and specifically to levels and PTMs of Sad1/UNC-84 (SUN) domain-containing protein 2 (SUN2). The result of this regulation is to decouple mechano-transmission between the cytoskeleton and the nucleus, thus conferring protection to chromatin.
Collapse
Affiliation(s)
- Hamish T J Gilbert
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester, M13 9PT, UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK
| | - Venkatesh Mallikarjun
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester, M13 9PT, UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK
| | - Oana Dobre
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester, M13 9PT, UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK
- School of Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Mark R Jackson
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester, M13 9PT, UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK
- Institute of Cancer Sciences, Glasgow, G61 1QH, UK
| | - Robert Pedley
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester, M13 9PT, UK
- Division of Molecular and Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK
| | - Andrew P Gilmore
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester, M13 9PT, UK
- Division of Molecular and Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK
| | - Stephen M Richardson
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK
| | - Joe Swift
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester, M13 9PT, UK.
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK.
| |
Collapse
|
81
|
Chen G, Xia B, Fu Q, Huang X, Wang F, Chen Z, Lv Y. Matrix Mechanics as Regulatory Factors and Therapeutic Targets in Hepatic Fibrosis. Int J Biol Sci 2019; 15:2509-2521. [PMID: 31754325 PMCID: PMC6854372 DOI: 10.7150/ijbs.37500] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/07/2019] [Indexed: 12/12/2022] Open
Abstract
The hallmark of liver fibrosis is excessive extracellular matrix (ECM) synthesis and deposition that improve liver matrix remodeling and stiffening. Increased matrix stiffness is not only a pathological consequence of liver fibrosis in traditional view, but also recognized as a key driver in pathological progression of hepatic fibrosis. Cells can perceive changes in the mechanical characteristics of hepatic matrix and respond by means of mechanical signal transduction pathways to regulate cell behavior. In this review, the authors first classify causes of liver matrix stiffening during fibrotic progression, such as higher degree of collagen cross-linking. The latest advances of the research on the matrix mechanics in regulating activation of HSCs or fibroblasts under two-dimensional (2D) and three-dimensional (3D) microenvironment is also classified and summarized. The mechanical signaling pathways involved in the process of hepatic matrix stiffening, such as YAP-TAZ signaling pathway, are further summarized. Finally, some potential therapeutic concepts and strategies based on matrix mechanics will be detailed. Collectively, these findings reinforce the importance of matrix mechanics in hepatic fibrosis, and underscore the value of clarifying its modulation in hopes of advancing the development of novel therapeutic targets and strategies for hepatic fibrosis.
Collapse
Affiliation(s)
- Guobao Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Bin Xia
- Chongqing Technology and Business University, Chongqing 400067, P. R. China
| | - Qiang Fu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Xiang Huang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Fuping Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Zhongmin Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Yonggang Lv
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, P. R. China
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
82
|
Das A, Barai A, Monteiro M, Kumar S, Sen S. Nuclear softening is essential for protease-independent migration. Matrix Biol 2019; 82:4-19. [DOI: 10.1016/j.matbio.2019.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 01/07/2019] [Accepted: 01/07/2019] [Indexed: 02/08/2023]
|
83
|
Mohammed D, Versaevel M, Bruyère C, Alaimo L, Luciano M, Vercruysse E, Procès A, Gabriele S. Innovative Tools for Mechanobiology: Unraveling Outside-In and Inside-Out Mechanotransduction. Front Bioeng Biotechnol 2019; 7:162. [PMID: 31380357 PMCID: PMC6646473 DOI: 10.3389/fbioe.2019.00162] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/20/2019] [Indexed: 12/26/2022] Open
Abstract
Cells and tissues can sense and react to the modifications of the physico-chemical properties of the extracellular environment (ECM) through integrin-based adhesion sites and adapt their physiological response in a process called mechanotransduction. Due to their critical localization at the cell-ECM interface, transmembrane integrins are mediators of bidirectional signaling, playing a key role in “outside-in” and “inside-out” signal transduction. After presenting the basic conceptual fundamentals related to cell mechanobiology, we review the current state-of-the-art technologies that facilitate the understanding of mechanotransduction signaling pathways. Finally, we highlight innovative technological developments that can help to advance our understanding of the mechanisms underlying nuclear mechanotransduction.
Collapse
Affiliation(s)
- Danahe Mohammed
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Marie Versaevel
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Céline Bruyère
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Laura Alaimo
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Marine Luciano
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Eléonore Vercruysse
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Anthony Procès
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium.,Department of Neurosciences, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Sylvain Gabriele
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| |
Collapse
|
84
|
Chin IL, Hool L, Choi YS. A Review of in vitro Platforms for Understanding Cardiomyocyte Mechanobiology. Front Bioeng Biotechnol 2019; 7:133. [PMID: 31231644 PMCID: PMC6560053 DOI: 10.3389/fbioe.2019.00133] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022] Open
Abstract
Mechanobiology—a cell's interaction with its physical environment—can influence a myriad of cellular processes including how cells migrate, differentiate and proliferate. In many diseases, remodeling of the extracellular matrix (ECM) is observed such as tissue stiffening in rigid scar formation after myocardial infarct. Utilizing knowledge of cell mechanobiology in relation to ECM remodeling during pathogenesis, elucidating the role of the ECM in the progression—and perhaps regression—of disease is a primary focus of the field. Although the importance of mechanical signaling in the cardiac cell is well-appreciated, our understanding of how these signals are sensed and transduced by cardiomyocytes is limited. To overcome this limitation, recently developed tools and resources have provided exciting opportunities to further our understandings by better recapitulating pathological spatiotemporal ECM stiffness changes in an in vitro setting. In this review, we provide an overview of a conventional model of mechanotransduction and present understandings of cardiomyocyte mechanobiology, followed by a review of emerging tools and resources that can be used to expand our knowledge of cardiomyocyte mechanobiology toward more clinically relevant applications.
Collapse
Affiliation(s)
- Ian L Chin
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Livia Hool
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia.,Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
85
|
Sidorenko E, Vartiainen MK. Nucleoskeletal regulation of transcription: Actin on MRTF. Exp Biol Med (Maywood) 2019; 244:1372-1381. [PMID: 31142145 DOI: 10.1177/1535370219854669] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Myocardin-related transcription factor A (MRTF-A) and serum response factor (SRF) form an essential transcriptional complex that regulates the expression of many cytoskeletal genes in response to dynamic changes in the actin cytoskeleton. The nucleoskeleton, a “dynamic network of networks,” consists of numerous proteins that contribute to nuclear shape and to its various functions, including gene expression. In this review, we will discuss recent work that has identified many nucleoskeletal proteins, such as nuclear lamina and lamina-associated proteins, nuclear actin, and the linker of the cytoskeleton and nucleoskeleton complex as important regulators of MRTF-A/SRF transcriptional activity, especially in the context of mechanical control of transcription. Impact statement Regulation of gene expression is a fundamental cellular process that ensures the appropriate response of a cell to its surroundings. Alongside biochemical signals, mechanical cues, such as substrate rigidity, have been recognized as key regulators of gene expression. Nucleoskeletal components play an important role in mechanoresponsive transcription, particularly in controlling the activity of MRTF-A/SRF transcription factors. This ensures that the cell can balance the internal and external mechanical forces by fine-tuning the expression of cytoskeletal genes.
Collapse
Affiliation(s)
- Ekaterina Sidorenko
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Maria K Vartiainen
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| |
Collapse
|
86
|
Ranade D, Pradhan R, Jayakrishnan M, Hegde S, Sengupta K. Lamin A/C and Emerin depletion impacts chromatin organization and dynamics in the interphase nucleus. BMC Mol Cell Biol 2019; 20:11. [PMID: 31117946 PMCID: PMC6532135 DOI: 10.1186/s12860-019-0192-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 04/16/2019] [Indexed: 12/26/2022] Open
Abstract
Background Nuclear lamins are type V intermediate filament proteins that maintain nuclear structure and function. Furthermore, Emerin - an interactor of Lamin A/C, facilitates crosstalk between the cytoskeleton and the nucleus as it also interacts with actin and Nuclear Myosin 1 (NM1). Results Here we show that the depletion of Lamin A/C or Emerin, alters the localization of the nuclear motor protein - Nuclear Myosin 1 (NM1) that manifests as an increase in NM1 foci in the nucleus and are rescued to basal levels upon the combined knockdown of Lamin A/C and Emerin. Furthermore, Lamin A/C-Emerin co-depletion destabilizes cytoskeletal organization as it increases actin stress fibers. This further impinges on nuclear organization, as it enhances chromatin mobility more toward the nuclear interior in Lamin A/C-Emerin co-depleted cells. This enhanced chromatin mobility was restored to basal levels either upon inhibition of Nuclear Myosin 1 (NM1) activity or actin depolymerization. In addition, the combined loss of Lamin A/C and Emerin alters the otherwise highly conserved spatial positions of chromosome territories. Furthermore, knockdown of Lamin A/C or Lamin A/C-Emerin combined, deregulates expression levels of a candidate subset of genes. Amongst these genes, both KLK10 (Chr.19, Lamina Associated Domain (LAD+)) and MADH2 (Chr.18, LAD-) were significantly repressed, while BCL2L12 (Chr.19, LAD-) is de-repressed. These genes differentially reposition with respect to the nuclear envelope. Conclusions Taken together, these studies underscore a remarkable interplay between Lamin A/C and Emerin in modulating cytoskeletal organization of actin and NM1 that impinges on chromatin dynamics and function in the interphase nucleus. Electronic supplementary material The online version of this article (10.1186/s12860-019-0192-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Devika Ranade
- Indian Institute of Science Education and Research (IISER)-Pune, Dr. Homi Bhabha Road, Biology, Room#B-216, 1st Floor, Main Building, Pashan, Pune, Maharashtra, 411008, India
| | - Roopali Pradhan
- Indian Institute of Science Education and Research (IISER)-Pune, Dr. Homi Bhabha Road, Biology, Room#B-216, 1st Floor, Main Building, Pashan, Pune, Maharashtra, 411008, India
| | - Muhunden Jayakrishnan
- Indian Institute of Science Education and Research (IISER)-Pune, Dr. Homi Bhabha Road, Biology, Room#B-216, 1st Floor, Main Building, Pashan, Pune, Maharashtra, 411008, India
| | - Sushmitha Hegde
- Indian Institute of Science Education and Research (IISER)-Pune, Dr. Homi Bhabha Road, Biology, Room#B-216, 1st Floor, Main Building, Pashan, Pune, Maharashtra, 411008, India
| | - Kundan Sengupta
- Indian Institute of Science Education and Research (IISER)-Pune, Dr. Homi Bhabha Road, Biology, Room#B-216, 1st Floor, Main Building, Pashan, Pune, Maharashtra, 411008, India.
| |
Collapse
|
87
|
Freedman BR, Mooney DJ. Biomaterials to Mimic and Heal Connective Tissues. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1806695. [PMID: 30908806 PMCID: PMC6504615 DOI: 10.1002/adma.201806695] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/27/2019] [Indexed: 05/11/2023]
Abstract
Connective tissue is one of the four major types of animal tissue and plays essential roles throughout the human body. Genetic factors, aging, and trauma all contribute to connective tissue dysfunction and motivate the need for strategies to promote healing and regeneration. The goal here is to link a fundamental understanding of connective tissues and their multiscale properties to better inform the design and translation of novel biomaterials to promote their regeneration. Major clinical problems in adipose tissue, cartilage, dermis, and tendon are discussed that inspire the need to replace native connective tissue with biomaterials. Then, multiscale structure-function relationships in native soft connective tissues that may be used to guide material design are detailed. Several biomaterials strategies to improve healing of these tissues that incorporate biologics and are biologic-free are reviewed. Finally, important guidance documents and standards (ASTM, FDA, and EMA) that are important to consider for translating new biomaterials into clinical practice are highligted.
Collapse
Affiliation(s)
- Benjamin R Freedman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
88
|
Song Q, Steuber M, Druzhinin SI, Schönherr H. Tailored Combinatorial Microcompartments through the Self‐Organization of Microobjects: Assembly, Characterization, and Cell Studies. Angew Chem Int Ed Engl 2019; 58:5246-5250. [DOI: 10.1002/anie.201814076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/09/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Qimeng Song
- Physical Chemistry I and Research Center of Micro and Nano-chemistry and Engineering (Cμ)University of Siegen Adolf-Reichwein-Straße 2 57076 Siegen Germany
| | - Marc Steuber
- Physical Chemistry I and Research Center of Micro and Nano-chemistry and Engineering (Cμ)University of Siegen Adolf-Reichwein-Straße 2 57076 Siegen Germany
| | - Sergey I. Druzhinin
- Physical Chemistry I and Research Center of Micro and Nano-chemistry and Engineering (Cμ)University of Siegen Adolf-Reichwein-Straße 2 57076 Siegen Germany
| | - Holger Schönherr
- Physical Chemistry I and Research Center of Micro and Nano-chemistry and Engineering (Cμ)University of Siegen Adolf-Reichwein-Straße 2 57076 Siegen Germany
| |
Collapse
|
89
|
Song Q, Steuber M, Druzhinin SI, Schönherr H. Tailored Combinatorial Microcompartments through the Self‐Organization of Microobjects: Assembly, Characterization, and Cell Studies. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201814076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Qimeng Song
- Physical Chemistry I and Research Center of Micro and Nano-chemistry and Engineering (Cμ)University of Siegen Adolf-Reichwein-Straße 2 57076 Siegen Germany
| | - Marc Steuber
- Physical Chemistry I and Research Center of Micro and Nano-chemistry and Engineering (Cμ)University of Siegen Adolf-Reichwein-Straße 2 57076 Siegen Germany
| | - Sergey I. Druzhinin
- Physical Chemistry I and Research Center of Micro and Nano-chemistry and Engineering (Cμ)University of Siegen Adolf-Reichwein-Straße 2 57076 Siegen Germany
| | - Holger Schönherr
- Physical Chemistry I and Research Center of Micro and Nano-chemistry and Engineering (Cμ)University of Siegen Adolf-Reichwein-Straße 2 57076 Siegen Germany
| |
Collapse
|
90
|
Hah J, Kim DH. Deciphering Nuclear Mechanobiology in Laminopathy. Cells 2019; 8:E231. [PMID: 30862117 PMCID: PMC6468464 DOI: 10.3390/cells8030231] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/23/2019] [Accepted: 03/05/2019] [Indexed: 12/13/2022] Open
Abstract
Extracellular mechanical stimuli are translated into biochemical signals inside the cell via mechanotransduction. The nucleus plays a critical role in mechanoregulation, which encompasses mechanosensing and mechanotransduction. The nuclear lamina underlying the inner nuclear membrane not only maintains the structural integrity, but also connects the cytoskeleton to the nuclear envelope. Lamin mutations, therefore, dysregulate the nuclear response, resulting in abnormal mechanoregulations, and ultimately, disease progression. Impaired mechanoregulations even induce malfunction in nuclear positioning, cell migration, mechanosensation, as well as differentiation. To know how to overcome laminopathies, we need to understand the mechanisms of laminopathies in a mechanobiological way. Recently, emerging studies have demonstrated the varying defects from lamin mutation in cellular homeostasis within mechanical surroundings. Therefore, this review summarizes recent findings highlighting the role of lamins, the architecture of nuclear lamina, and their disease relevance in the context of nuclear mechanobiology. We will also provide an overview of the differentiation of cellular mechanics in laminopathy.
Collapse
Affiliation(s)
- Jungwon Hah
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea.
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea.
| |
Collapse
|
91
|
Henry MP, Hawkins JR, Boyle J, Bridger JM. The Genomic Health of Human Pluripotent Stem Cells: Genomic Instability and the Consequences on Nuclear Organization. Front Genet 2019; 9:623. [PMID: 30719030 PMCID: PMC6348275 DOI: 10.3389/fgene.2018.00623] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are increasingly used for cell-based regenerative therapies worldwide, with embryonic and induced pluripotent stem cells as potential treatments for debilitating and chronic conditions, such as age-related macular degeneration, Parkinson's disease, spinal cord injuries, and type 1 diabetes. However, with the level of genomic anomalies stem cells generate in culture, their safety may be in question. Specifically, hPSCs frequently acquire chromosomal abnormalities, often with gains or losses of whole chromosomes. This review discusses how important it is to efficiently and sensitively detect hPSC aneuploidies, to understand how these aneuploidies arise, consider the consequences for the cell, and indeed the individual to whom aneuploid cells may be administered.
Collapse
Affiliation(s)
- Marianne P Henry
- Advanced Therapies Division, National Institute for Biological Standards and Control, Potters Bar, United Kingdom.,Laboratory of Nuclear and Genomic Health, Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - J Ross Hawkins
- Advanced Therapies Division, National Institute for Biological Standards and Control, Potters Bar, United Kingdom
| | - Jennifer Boyle
- Advanced Therapies Division, National Institute for Biological Standards and Control, Potters Bar, United Kingdom
| | - Joanna M Bridger
- Laboratory of Nuclear and Genomic Health, Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| |
Collapse
|
92
|
González-Cruz RD, Dahl KN, Darling EM. The Emerging Role of Lamin C as an Important LMNA Isoform in Mechanophenotype. Front Cell Dev Biol 2018; 6:151. [PMID: 30450357 PMCID: PMC6224339 DOI: 10.3389/fcell.2018.00151] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/15/2018] [Indexed: 12/17/2022] Open
Abstract
Lamin A and lamin C isoforms of the gene LMNA are major structural and mechanotransductive components of the nuclear lamina. Previous reports have proposed lamin A as the isoform with the most dominant contributions to cellular mechanophenotype. Recently, expression of lamin C has also been shown to strongly correlate to cellular elastic and viscoelastic properties. Nevertheless, LMNA isoforms exist as part of a network that collectively provides structural integrity to the nucleus and their expression is ultimately regulated in a cell-specific manner. Thus, they have importance in mechanotransduction and structural integrity of the nucleus as well as potential candidates for biomarkers of whole-cell mechanophenotype. Therefore, a fuller discussion of lamin isoforms as mechanophenotypic biomarkers should compare both individual and ratiometric isoform contributions toward whole-cell mechanophenotype across different cell types. In this perspective, we discuss the distinctions between the mechanophenotypic correlations of individual and ratiometric lamins A:B1, C:B1, (A + C):B1, and C:A across cells from different lineages, demonstrating that the collective contribution of ratiometric lamin (A + C):B1 isoforms exhibited the strongest correlation to whole-cell stiffness. Additionally, we highlight the potential roles of lamin isoform ratios as indicators of mechanophenotypic change in differentiation and disease to demonstrate that the contributions of individual and collective lamin isoforms can occur as both static and dynamic biomarkers of mechanophenotype.
Collapse
Affiliation(s)
| | - Kris N Dahl
- Department of Chemical Engineering, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Eric M Darling
- Center for Biomedical Engineering, Brown University, Providence, RI, United States.,Department of Molecular Pharmacology, Physiology and Biotechnology, School of Engineering, Department of Orthopaedics, Brown University, Providence, RI, United States
| |
Collapse
|
93
|
Jiang Y, Ji JY. Understanding lamin proteins and their roles in aging and cardiovascular diseases. Life Sci 2018; 212:20-29. [DOI: 10.1016/j.lfs.2018.09.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/12/2018] [Accepted: 09/14/2018] [Indexed: 02/04/2023]
|
94
|
Gerbino A, Procino G, Svelto M, Carmosino M. Role of Lamin A/C Gene Mutations in the Signaling Defects Leading to Cardiomyopathies. Front Physiol 2018; 9:1356. [PMID: 30319452 PMCID: PMC6167438 DOI: 10.3389/fphys.2018.01356] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/07/2018] [Indexed: 01/03/2023] Open
Abstract
Nuclear lamin A/C are crucial components of the intricate protein mesh that underlies the inner nuclear membrane and confers mainly nuclear and cytosolic rigidity. However, throughout the years a number of other key physiological processes have been associated with lamins such as modulation of both genes expression and the activity of signaling mediators. To further solidify its importance in cell physiology, mutations in the lamin A/C gene (LMNA) have been associated to diverse pathological phenotypes with skeletal muscles and the heart being the most affected systems. When affected, the heart develops a wide array of phenotypes spanning from dilated cardiomyopathy with conduction defects to arrhythmogenic right ventricular cardiomyopathy. The surprising large number of cardiac phenotypes reflects the equally large number of specific mutations identified in the LMNA gene. In this review, we underlie how mutations in LMNA can impact the activity and the spatial/temporal organization of signaling mediators and transcription factors. We analyzed the ever-increasing amount of findings collected in LmnaH222P/H222P mice whose cardiomyopathy resemble the most important features of the disease in humans and a number of key evidences from other experimental models. With this mini review, we attempt to combine the newest insights regarding both the pathogenic effects of LMNA mutations in terms of signaling abnormalities and cardiac laminopathies.
Collapse
Affiliation(s)
- Andrea Gerbino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Procino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Maria Svelto
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Monica Carmosino
- Department of Sciences, University of Basilicata, Potenza, Italy
| |
Collapse
|
95
|
Pradhan R, Ranade D, Sengupta K. Emerin modulates spatial organization of chromosome territories in cells on softer matrices. Nucleic Acids Res 2018; 46:5561-5586. [PMID: 29684168 PMCID: PMC6009696 DOI: 10.1093/nar/gky288] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 02/06/2023] Open
Abstract
Cells perceive and relay external mechanical forces into the nucleus through the nuclear envelope. Here we examined the effect of lowering substrate stiffness as a paradigm to address the impact of altered mechanical forces on nuclear structure-function relationships. RNA sequencing of cells on softer matrices revealed significant transcriptional imbalances, predominantly in chromatin associated processes and transcriptional deregulation of human Chromosome 1. Furthermore, 3-Dimensional fluorescence in situ hybridization (3D-FISH) analyses showed a significant mislocalization of Chromosome 1 and 19 Territories (CT) into the nuclear interior, consistent with their transcriptional deregulation. However, CT18 with relatively lower transcriptional dysregulation, also mislocalized into the nuclear interior. Furthermore, nuclear Lamins that regulate chromosome positioning, were mislocalized into the nuclear interior in response to lowered matrix stiffness. Notably, Lamin B2 overexpression retained CT18 near the nuclear periphery in cells on softer matrices. While, cells on softer matrices also activated emerin phosphorylation at a novel Tyr99 residue, the inhibition of which in a phospho-deficient mutant (emerinY99F), selectively retained chromosome 18 and 19 but not chromosome 1 territories at their conserved nuclear locations. Taken together, emerin functions as a key mechanosensor, that modulates the spatial organization of chromosome territories in the interphase nucleus.
Collapse
Affiliation(s)
- Roopali Pradhan
- Biology, Main Building, First Floor, Room#B-216, Indian Institute of Science Education and Research (IISER), Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| | - Devika Ranade
- Biology, Main Building, First Floor, Room#B-216, Indian Institute of Science Education and Research (IISER), Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| | - Kundan Sengupta
- Biology, Main Building, First Floor, Room#B-216, Indian Institute of Science Education and Research (IISER), Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| |
Collapse
|
96
|
Insight into the functional organization of nuclear lamins in health and disease. Curr Opin Cell Biol 2018; 54:72-79. [PMID: 29800922 DOI: 10.1016/j.ceb.2018.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/24/2018] [Accepted: 05/08/2018] [Indexed: 11/20/2022]
Abstract
Lamins are the main component of the nuclear lamina, a protein meshwork at the inner nuclear membrane which primarily provide mechanical stability to the nucleus. Lamins, type V intermediate filament proteins, are also involved in many nuclear activities. Structural analysis of nuclei revealed that lamins form 3.5nm thick filaments often interact with nuclear pore complexes. Mutations in the LMNA gene, encoding A-type lamins, have been associated with at least 15 distinct diseases collectively termed laminopathies, including muscle, metabolic and neurological disorders, and premature aging syndrome. It is unclear how laminopathic mutations lead to such a wide array of diseases, essentially affecting almost all tissues.
Collapse
|
97
|
Wang S, Stoops E, Cp U, Markus B, Reuveny A, Ordan E, Volk T. Mechanotransduction via the LINC complex regulates DNA replication in myonuclei. J Cell Biol 2018; 217:2005-2018. [PMID: 29650775 PMCID: PMC5987719 DOI: 10.1083/jcb.201708137] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 02/08/2018] [Accepted: 03/27/2018] [Indexed: 12/26/2022] Open
Abstract
Nuclear mechanotransduction has been implicated in the control of chromatin organization and gene expression. Wang et al. show that, in Drosophila myofibers, the LINC complex is required for the regulation of DNA replication and synchronized cell-cycle progression in myonuclei. Nuclear mechanotransduction has been implicated in the control of chromatin organization; however, its impact on functional contractile myofibers is unclear. We found that deleting components of the linker of nucleoskeleton and cytoskeleton (LINC) complex in Drosophila melanogaster larval muscles abolishes the controlled and synchronized DNA endoreplication, typical of nuclei across myofibers, resulting in increased and variable DNA content in myonuclei of individual myofibers. Moreover, perturbation of LINC-independent mechanical input after knockdown of β-Integrin in larval muscles similarly led to increased DNA content in myonuclei. Genome-wide RNA-polymerase II occupancy analysis in myofibers of the LINC mutant klar indicated an altered binding profile, including a significant decrease in the chromatin regulator barrier-to-autointegration factor (BAF) and the contractile regulator Troponin C. Importantly, muscle-specific knockdown of BAF led to increased DNA content in myonuclei, phenocopying the LINC mutant phenotype. We propose that mechanical stimuli transmitted via the LINC complex act via BAF to regulate synchronized cell-cycle progression of myonuclei across single myofibers.
Collapse
Affiliation(s)
- Shuoshuo Wang
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Elizabeth Stoops
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Unnikannan Cp
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Barak Markus
- G-INCPM/Mantoux Institute for Bioinformatics, Weizmann Institute of Science, Rehovot, Israel
| | - Adriana Reuveny
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Elly Ordan
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Talila Volk
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
98
|
Lee IN, Dobre O, Richards D, Ballestrem C, Curran JM, Hunt JA, Richardson SM, Swift J, Wong LS. Photoresponsive Hydrogels with Photoswitchable Mechanical Properties Allow Time-Resolved Analysis of Cellular Responses to Matrix Stiffening. ACS APPLIED MATERIALS & INTERFACES 2018; 10:7765-7776. [PMID: 29430919 PMCID: PMC5864053 DOI: 10.1021/acsami.7b18302] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/12/2018] [Indexed: 05/03/2023]
Abstract
As cell function and phenotype can be directed by the mechanical characteristics of the surrounding matrix, hydrogels have become important platforms for cell culture systems, with properties that can be tuned by external stimuli, such as divalent cations, enzymatic treatment, and pH. However, many of these stimuli can directly affect cell behavior, making it difficult to distinguish purely mechanical signaling events. This study reports on the development of a hydrogel that incorporates photoswitchable cross-linkers, which can reversibly alter their stiffness upon irradiation with the appropriate wavelength of light. Furthermore, this study reports the response of bone-marrow-derived mesenchymal stem cells (MSCs) on these hydrogels that were stiffened systematically by irradiation with blue light. The substrates were shown to be noncytotoxic, and crucially MSCs were not affected by blue-light exposure. Time-resolved analysis of cell morphology showed characteristic cell spreading and increased aspect ratios in response to greater substrate stiffness. This hydrogel provides a platform to study mechanosignaling in cells responding to dynamic changes in stiffness, offering a new way to study mechanotransduction signaling pathways and biological processes, with implicit changes to tissue mechanics, such as development, ageing, and fibrosis.
Collapse
Affiliation(s)
- I-Ning Lee
- Manchester Institute
of Biotechnology and School of Chemistry, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
- School
of Engineering, University of Liverpool, Harrison Hughes Building, Liverpool L69 3GH, United Kingdom
| | - Oana Dobre
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology, Medicine and Health, Manchester Academic
Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - David Richards
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology, Medicine and Health, Manchester Academic
Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology, Medicine and Health, Manchester Academic
Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Judith M. Curran
- School
of Engineering, University of Liverpool, Harrison Hughes Building, Liverpool L69 3GH, United Kingdom
| | - John A. Hunt
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom
| | - Stephen M. Richardson
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology, Medicine and Health, Manchester Academic
Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Joe Swift
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology, Medicine and Health, Manchester Academic
Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Lu Shin Wong
- Manchester Institute
of Biotechnology and School of Chemistry, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
99
|
Emerging views of the nucleus as a cellular mechanosensor. Nat Cell Biol 2018; 20:373-381. [PMID: 29467443 DOI: 10.1038/s41556-018-0038-y] [Citation(s) in RCA: 365] [Impact Index Per Article: 52.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/09/2018] [Indexed: 12/14/2022]
Abstract
The ability of cells to respond to mechanical forces is critical for numerous biological processes. Emerging evidence indicates that external mechanical forces trigger changes in nuclear envelope structure and composition, chromatin organization and gene expression. However, it remains unclear if these processes originate in the nucleus or are downstream of cytoplasmic signals. Here we discuss recent findings that support a direct role of the nucleus in cellular mechanosensing and highlight novel tools to study nuclear mechanotransduction.
Collapse
|
100
|
Broders-Bondon F, Nguyen Ho-Bouldoires TH, Fernandez-Sanchez ME, Farge E. Mechanotransduction in tumor progression: The dark side of the force. J Cell Biol 2018; 217:1571-1587. [PMID: 29467174 PMCID: PMC5940296 DOI: 10.1083/jcb.201701039] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 01/19/2018] [Accepted: 02/01/2018] [Indexed: 12/11/2022] Open
Abstract
Broders-Bondon et al. review the pathological mechanical properties of tumor tissues and how abnormal mechanical signals result in oncogenic biochemical signals during tumor progression. Cancer has been characterized as a genetic disease, associated with mutations that cause pathological alterations of the cell cycle, adhesion, or invasive motility. Recently, the importance of the anomalous mechanical properties of tumor tissues, which activate tumorigenic biochemical pathways, has become apparent. This mechanical induction in tumors appears to consist of the destabilization of adult tissue homeostasis as a result of the reactivation of embryonic developmental mechanosensitive pathways in response to pathological mechanical strains. These strains occur in many forms, for example, hypervascularization in late tumors leads to high static hydrodynamic pressure that can promote malignant progression through hypoxia or anomalous interstitial liquid and blood flow. The high stiffness of tumors directly induces the mechanical activation of biochemical pathways enhancing the cell cycle, epithelial–mesenchymal transition, and cell motility. Furthermore, increases in solid-stress pressure associated with cell hyperproliferation activate tumorigenic pathways in the healthy epithelial cells compressed by the neighboring tumor. The underlying molecular mechanisms of the translation of a mechanical signal into a tumor inducing biochemical signal are based on mechanically induced protein conformational changes that activate classical tumorigenic signaling pathways. Understanding these mechanisms will be important for the development of innovative treatments to target such mechanical anomalies in cancer.
Collapse
Affiliation(s)
- Florence Broders-Bondon
- Mechanics and Genetics of Embryonic and Tumor Development Group, Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR168, Inserm, Sorbonne Universities, Paris, France
| | - Thanh Huong Nguyen Ho-Bouldoires
- Mechanics and Genetics of Embryonic and Tumor Development Group, Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR168, Inserm, Sorbonne Universities, Paris, France
| | - Maria-Elena Fernandez-Sanchez
- Mechanics and Genetics of Embryonic and Tumor Development Group, Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR168, Inserm, Sorbonne Universities, Paris, France
| | - Emmanuel Farge
- Mechanics and Genetics of Embryonic and Tumor Development Group, Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR168, Inserm, Sorbonne Universities, Paris, France
| |
Collapse
|