51
|
Brown SJ, Šoltić D, Synowsky SA, Shirran SL, Chilcott E, Shorrock HK, Gillingwater TH, Yáñez-Muñoz RJ, Schneider B, Bowerman M, Fuller HR. AAV9-mediated SMN gene therapy rescues cardiac desmin but not lamin A/C and elastin dysregulation in Smn2B/- spinal muscular atrophy mice. Hum Mol Genet 2023; 32:2950-2965. [PMID: 37498175 PMCID: PMC10549791 DOI: 10.1093/hmg/ddad121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/27/2023] [Accepted: 07/25/2023] [Indexed: 07/28/2023] Open
Abstract
Structural, functional and molecular cardiac defects have been reported in spinal muscular atrophy (SMA) patients and mouse models. Previous quantitative proteomics analyses demonstrated widespread molecular defects in the severe Taiwanese SMA mouse model. Whether such changes are conserved across different mouse models, including less severe forms of the disease, has yet to be established. Here, using the same high-resolution proteomics approach in the less-severe Smn2B/- SMA mouse model, 277 proteins were found to be differentially abundant at a symptomatic timepoint (post-natal day (P) 18), 50 of which were similarly dysregulated in severe Taiwanese SMA mice. Bioinformatics analysis linked many of the differentially abundant proteins to cardiovascular development and function, with intermediate filaments highlighted as an enriched cellular compartment in both datasets. Lamin A/C was increased in the cardiac tissue, whereas another intermediate filament protein, desmin, was reduced. The extracellular matrix (ECM) protein, elastin, was also robustly decreased in the heart of Smn2B/- mice. AAV9-SMN1-mediated gene therapy rectified low levels of survival motor neuron protein and restored desmin levels in heart tissues of Smn2B/- mice. In contrast, AAV9-SMN1 therapy failed to correct lamin A/C or elastin levels. Intermediate filament proteins and the ECM have key roles in cardiac function and their dysregulation may explain cardiac impairment in SMA, especially since mutations in genes encoding these proteins cause other diseases with cardiac aberration. Cardiac pathology may need to be considered in the long-term care of SMA patients, as it is unclear whether currently available treatments can fully rescue peripheral pathology in SMA.
Collapse
Affiliation(s)
- Sharon J Brown
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Darija Šoltić
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Silvia A Synowsky
- BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews, St Andrews KY16 9ST, UK
| | - Sally L Shirran
- BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews, St Andrews KY16 9ST, UK
| | - Ellie Chilcott
- AGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham Hill, Egham, Surrey TW20 0EX, UK
| | - Hannah K Shorrock
- Edinburgh Medical School: Biomedical Sciences, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School: Biomedical Sciences, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Rafael J Yáñez-Muñoz
- AGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham Hill, Egham, Surrey TW20 0EX, UK
| | - Bernard Schneider
- Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1202 Geneva, Switzerland
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Melissa Bowerman
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
- School of Medicine, Keele University, Keele ST5 5BG, UK
| | - Heidi R Fuller
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| |
Collapse
|
52
|
Kuburich NA, Sabapathy T, Demestichas BR, Maddela JJ, den Hollander P, Mani SA. Proactive and reactive roles of TGF-β in cancer. Semin Cancer Biol 2023; 95:120-139. [PMID: 37572731 PMCID: PMC10530624 DOI: 10.1016/j.semcancer.2023.08.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/14/2023]
Abstract
Cancer cells adapt to varying stress conditions to survive through plasticity. Stem cells exhibit a high degree of plasticity, allowing them to generate more stem cells or differentiate them into specialized cell types to contribute to tissue development, growth, and repair. Cancer cells can also exhibit plasticity and acquire properties that enhance their survival. TGF-β is an unrivaled growth factor exploited by cancer cells to gain plasticity. TGF-β-mediated signaling enables carcinoma cells to alter their epithelial and mesenchymal properties through epithelial-mesenchymal plasticity (EMP). However, TGF-β is a multifunctional cytokine; thus, the signaling by TGF-β can be detrimental or beneficial to cancer cells depending on the cellular context. Those cells that overcome the anti-tumor effect of TGF-β can induce epithelial-mesenchymal transition (EMT) to gain EMP benefits. EMP allows cancer cells to alter their cell properties and the tumor immune microenvironment (TIME), facilitating their survival. Due to the significant roles of TGF-β and EMP in carcinoma progression, it is essential to understand how TGF-β enables EMP and how cancer cells exploit this plasticity. This understanding will guide the development of effective TGF-β-targeting therapies that eliminate cancer cell plasticity.
Collapse
Affiliation(s)
- Nick A Kuburich
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Thiru Sabapathy
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Breanna R Demestichas
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Joanna Joyce Maddela
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Petra den Hollander
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Sendurai A Mani
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
53
|
Bastianello G, Foiani M. Mechanisms controlling the mechanical properties of the nuclei. Curr Opin Cell Biol 2023; 84:102222. [PMID: 37619290 DOI: 10.1016/j.ceb.2023.102222] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/26/2023]
Abstract
The mechanical properties of the nucleus influence different cellular and nuclear functions and have relevant implications for several human diseases. The nucleus protects genetic information while acting as a mechano-sensory hub in response to internal and external forces. Cells have evolved mechano-transduction signaling to respond to physical cellular and nuclear perturbations and adopted a multitude of molecular pathways to maintain nuclear shape stability and prevent morphological abnormalities of the nucleus. Here we describe those key biological processes that control nuclear mechanics and discuss emerging perspectives on the mechanobiology of the nucleus as a diagnostic tool and clinical target.
Collapse
Affiliation(s)
- Giulia Bastianello
- IFOM, The FIRC Institute of Molecular Oncology, Milan 20139, Italy; Oncology and Haemato-Oncology Department, University of Milan, Milan 20122, Italy.
| | - Marco Foiani
- IFOM, The FIRC Institute of Molecular Oncology, Milan 20139, Italy; Oncology and Haemato-Oncology Department, University of Milan, Milan 20122, Italy.
| |
Collapse
|
54
|
De Silva NS, Siewiera J, Alkhoury C, Nader GPF, Nadalin F, de Azevedo K, Couty M, Izquierdo HM, Bhargava A, Conrad C, Maurin M, Antoniadou K, Fouillade C, Londono-Vallejo A, Behrendt R, Bertotti K, Serdjebi C, Lanthiez F, Gallwitz L, Saftig P, Herrero-Fernández B, Saez A, González-Granado JM, van Niel G, Boissonnas A, Piel M, Manel N. Nuclear envelope disruption triggers hallmarks of aging in lung alveolar macrophages. NATURE AGING 2023; 3:1251-1268. [PMID: 37723209 DOI: 10.1038/s43587-023-00488-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/16/2023] [Indexed: 09/20/2023]
Abstract
Aging is characterized by gradual immune dysfunction and increased disease risk. Genomic instability is considered central to the aging process, but the underlying mechanisms of DNA damage are insufficiently defined. Cells in confined environments experience forces applied to their nucleus, leading to transient nuclear envelope rupture (NER) and DNA damage. Here, we show that Lamin A/C protects lung alveolar macrophages (AMs) from NER and hallmarks of aging. AMs move within constricted spaces in the lung. Immune-specific ablation of lamin A/C results in selective depletion of AMs and heightened susceptibility to influenza virus-induced pathogenesis and lung cancer growth. Lamin A/C-deficient AMs that persist display constitutive NER marks, DNA damage and p53-dependent senescence. AMs from aged wild-type and from lamin A/C-deficient mice share a lysosomal signature comprising CD63. CD63 is required to limit damaged DNA in macrophages. We propose that NER-induced genomic instability represents a mechanism of aging in AMs.
Collapse
Affiliation(s)
| | - Johan Siewiera
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Chantal Alkhoury
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | | | | | - Kevin de Azevedo
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Mickaël Couty
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Team van Niel, Paris, France
| | | | - Anvita Bhargava
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Cécile Conrad
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Mathieu Maurin
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | | | - Charles Fouillade
- Institut Curie, PSL Research University, Université Paris-Saclay, CNRS, INSERM, UMR3347, U1021, Orsay, France
| | | | - Rayk Behrendt
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | | | | | - François Lanthiez
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Lisa Gallwitz
- Biochemical Institute, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Paul Saftig
- Biochemical Institute, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Beatriz Herrero-Fernández
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Angela Saez
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria (UFV), Pozuelo de Alarcón, Spain
| | - José María González-Granado
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12). Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid. CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Guillaume van Niel
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Team van Niel, Paris, France
| | - Alexandre Boissonnas
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Matthieu Piel
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France
| | - Nicolas Manel
- Institut Curie, PSL Research University, INSERM U932, Paris, France.
| |
Collapse
|
55
|
Saini K, Cho S, Tewari M, Jalil AR, Wang M, Kasznel AJ, Yamamoto K, Chenoweth DM, Discher DE. Pan-tissue scaling of stiffness versus fibrillar collagen reflects contractility-driven strain that inhibits fibril degradation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559759. [PMID: 37808742 PMCID: PMC10557712 DOI: 10.1101/2023.09.27.559759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Polymer network properties such as stiffness often exhibit characteristic power laws in polymer density and other parameters. However, it remains unclear whether diverse animal tissues, composed of many distinct polymers, exhibit such scaling. Here, we examined many diverse tissues from adult mouse and embryonic chick to determine if stiffness ( E tissue ) follows a power law in relation to the most abundant animal protein, Collagen-I, even with molecular perturbations. We quantified fibrillar collagen in intact tissue by second harmonic generation (SHG) imaging and from tissue extracts by mass spectrometry (MS), and collagenase-mediated decreases were also tracked. Pan-tissue power laws for tissue stiffness versus Collagen-I levels measured by SHG or MS exhibit sub-linear scaling that aligns with results from cellularized gels of Collagen-I but not acellular gels. Inhibition of cellular myosin-II based contraction fits the scaling, and combination with inhibitors of matrix metalloproteinases (MMPs) show collagenase activity is strain - not stress- suppressed in tissues, consistent with past studies of gels and fibrils. Beating embryonic hearts and tendons, which differ in both collagen levels and stiffness by >1000-fold, similarly suppressed collagenases at physiological strains of ∼5%, with fiber-orientation regulating degradation. Scaling of E tissue based on 'use-it-or-lose-it' kinetics provides insight into scaling of organ size, microgravity effects, and regeneration processes while suggesting contractility-driven therapeutics.
Collapse
|
56
|
de Freitas Nader GP, García-Arcos JM. Cell migration in dense microenvironments. C R Biol 2023; 346:89-93. [PMID: 37779383 DOI: 10.5802/crbiol.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/29/2023] [Accepted: 08/29/2023] [Indexed: 10/03/2023]
Abstract
The nucleus has been viewed as a passenger during cell migration that functions merely to protect the genome. However, increasing evidence shows that the nucleus is an active organelle, constantly sensing the surrounding environment and translating extracellular mechanical inputs into intracellular signaling. The nuclear envelope has a large membrane reservoir which serves as a buffer for mechanical inputs as it unfolds without increasing its tension. In contrast, when cells cope with mechanical strain, such as migration through solid tumors or dense interstitial spaces, the nuclear envelope folds stretch, increasing nuclear envelope tension and sometimes causing rupture. Different degrees of nuclear envelope tension regulate cellular behaviors and functions, especially in cells that move and grow within dense matrices. The crosstalk between extracellular mechanical inputs and the cell nucleus is a critical component in the modulation of cell function of cells that navigate within packed microenvironments. Moreover, there is a link between regimes of nuclear envelope unfolding and different cellular behaviors, from orchestrated signaling cascades to cellular perturbations and damage.
Collapse
|
57
|
Gunn AL, Yashchenko AI, Dubrulle J, Johnson J, Hatch EM. A high-content screen reveals new regulators of nuclear membrane stability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542944. [PMID: 37398267 PMCID: PMC10312541 DOI: 10.1101/2023.05.30.542944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Nuclear membrane rupture is a physiological response to multiple in vivo processes, such as cell migration, that can cause extensive genome instability and upregulate invasive and inflammatory pathways. However, the underlying molecular mechanisms of rupture are unclear and few regulators have been identified. In this study, we developed a reporter that is size excluded from re-compartmentalization following nuclear rupture events. This allows for robust detection of factors influencing nuclear integrity in fixed cells. We combined this with an automated image analysis pipeline in a high-content siRNA screen to identify new proteins that both increase and decrease nuclear rupture frequency in cancer cells. Pathway analysis identified an enrichment of nuclear membrane and ER factors in our hits and we demonstrate that one of these, the protein phosphatase CTDNEP1, is required for nuclear stability. Further analysis of known rupture contributors, including a newly developed automated quantitative analysis of nuclear lamina gaps, strongly suggests that CTDNEP1 acts in a new pathway. Our findings provide new insights into the molecular mechanism of nuclear rupture and define a highly adaptable program for rupture analysis that removes a substantial barrier to new discoveries in the field.
Collapse
Affiliation(s)
- Amanda L. Gunn
- Divisions of Basic Sciences and Human Biology, The Fred Hutchinson Cancer Center, 1100 Fairview Ave, Seattle, Washington 98109, USA
| | - Artem I. Yashchenko
- Divisions of Basic Sciences and Human Biology, The Fred Hutchinson Cancer Center, 1100 Fairview Ave, Seattle, Washington 98109, USA
| | - Julien Dubrulle
- Cellular Imaging Shared Resource, The Fred Hutchinson Cancer Center, 1100 Fairview Ave, Seattle, Washington 98109, USA
| | - Jodiene Johnson
- Divisions of Basic Sciences and Human Biology, The Fred Hutchinson Cancer Center, 1100 Fairview Ave, Seattle, Washington 98109, USA
| | - Emily M. Hatch
- Divisions of Basic Sciences and Human Biology, The Fred Hutchinson Cancer Center, 1100 Fairview Ave, Seattle, Washington 98109, USA
| |
Collapse
|
58
|
Coscarella IL, Landim-Vieira M, Rastegarpouyani H, Chase PB, Irianto J, Pinto JR. Nucleus Mechanosensing in Cardiomyocytes. Int J Mol Sci 2023; 24:13341. [PMID: 37686151 PMCID: PMC10487505 DOI: 10.3390/ijms241713341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Cardiac muscle contraction is distinct from the contraction of other muscle types. The heart continuously undergoes contraction-relaxation cycles throughout an animal's lifespan. It must respond to constantly varying physical and energetic burdens over the short term on a beat-to-beat basis and relies on different mechanisms over the long term. Muscle contractility is based on actin and myosin interactions that are regulated by cytoplasmic calcium ions. Genetic variants of sarcomeric proteins can lead to the pathophysiological development of cardiac dysfunction. The sarcomere is physically connected to other cytoskeletal components. Actin filaments, microtubules and desmin proteins are responsible for these interactions. Therefore, mechanical as well as biochemical signals from sarcomeric contractions are transmitted to and sensed by other parts of the cardiomyocyte, particularly the nucleus which can respond to these stimuli. Proteins anchored to the nuclear envelope display a broad response which remodels the structure of the nucleus. In this review, we examine the central aspects of mechanotransduction in the cardiomyocyte where the transmission of mechanical signals to the nucleus can result in changes in gene expression and nucleus morphology. The correlation of nucleus sensing and dysfunction of sarcomeric proteins may assist the understanding of a wide range of functional responses in the progress of cardiomyopathic diseases.
Collapse
Affiliation(s)
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Hosna Rastegarpouyani
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
- Institute for Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Prescott Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Jerome Irianto
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Jose Renato Pinto
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
59
|
Wallace M, Zahr H, Perati S, Morsink CD, Johnson LE, Gacita AM, Lai S, Wallrath LL, Benjamin IJ, McNally EM, Kirby TJ, Lammerding J. Nuclear damage in LMNA mutant iPSC-derived cardiomyocytes is associated with impaired lamin localization to the nuclear envelope. Mol Biol Cell 2023; 34:mbcE21100527. [PMID: 37585285 PMCID: PMC10846625 DOI: 10.1091/mbc.e21-10-0527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 07/31/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
The LMNA gene encodes the nuclear envelope proteins Lamins A and C, which comprise a major part of the nuclear lamina, provide mechanical support to the nucleus, and participate in diverse intracellular signaling. LMNA mutations give rise to a collection of diseases called laminopathies, including dilated cardiomyopathy (LMNA-DCM) and muscular dystrophies. Although nuclear deformities are a hallmark of LMNA-DCM, the role of nuclear abnormalities in the pathogenesis of LMNA-DCM remains incompletely understood. Using induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) from LMNA mutant patients and healthy controls, we show that LMNA mutant iPSC-CM nuclei have altered shape or increased size compared to healthy control iPSC-CM nuclei. The LMNA mutation exhibiting the most severe nuclear deformities, R249Q, additionally caused reduced nuclear stiffness and increased nuclear fragility. Importantly, for all cell lines, the degree of nuclear abnormalities corresponded to the degree of Lamin A/C and Lamin B1 mislocalization from the nuclear envelope. The mislocalization was likely due to altered assembly of Lamin A/C. Collectively, these results point to the importance of correct lamin assembly at the nuclear envelope in providing mechanical stability to the nucleus and suggest that defects in nuclear lamina organization may contribute to the nuclear and cellular dysfunction in LMNA-DCM.
Collapse
Affiliation(s)
- Melanie Wallace
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
- Weill Institute for Cell and Molecular Biology, Ithaca, NY 14853
| | - Hind Zahr
- Weill Institute for Cell and Molecular Biology, Ithaca, NY 14853
| | - Shriya Perati
- Weill Institute for Cell and Molecular Biology, Ithaca, NY 14853
| | - Chloé D. Morsink
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, VU Medical Center, 1081 HZ Amsterdam, The Netherlands
| | | | - Anthony M. Gacita
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern Medicine, Chicago, IL 60611
| | - Shuping Lai
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Lori L. Wallrath
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242
| | - Ivor J. Benjamin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern Medicine, Chicago, IL 60611
| | - Tyler J. Kirby
- Weill Institute for Cell and Molecular Biology, Ithaca, NY 14853
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, VU Medical Center, 1081 HZ Amsterdam, The Netherlands
| | - Jan Lammerding
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
- Weill Institute for Cell and Molecular Biology, Ithaca, NY 14853
| |
Collapse
|
60
|
Abdelrahman S, Ge R, Susapto HH, Liu Y, Samkari F, Moretti M, Liu X, Hoehndorf R, Emwas AH, Jaremko M, Rawas RH, Hauser CAE. The Impact of Mechanical Cues on the Metabolomic and Transcriptomic Profiles of Human Dermal Fibroblasts Cultured in Ultrashort Self-Assembling Peptide 3D Scaffolds. ACS NANO 2023; 17:14508-14531. [PMID: 37477873 DOI: 10.1021/acsnano.3c01176] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Cells' interactions with their microenvironment influence their morphological features and regulate crucial cellular functions including proliferation, differentiation, metabolism, and gene expression. Most biological data available are based on in vitro two-dimensional (2D) cellular models, which fail to recapitulate the three-dimensional (3D) in vivo systems. This can be attributed to the lack of cell-matrix interaction and the limitless access to nutrients and oxygen, in contrast to in vivo systems. Despite the emergence of a plethora of 3D matrices to address this challenge, there are few reports offering a proper characterization of these matrices or studying how the cell-matrix interaction influences cellular metabolism in correlation with gene expression. In this study, two tetrameric ultrashort self-assembling peptide sequences, FFIK and FIIK, were used to create in vitro 3D models using well-described human dermal fibroblast cells. The peptide sequences are derived from naturally occurring amino acids that are capable of self-assembling into stable hydrogels without UV or chemical cross-linking. Our results showed that 2D cultured fibroblasts exhibited distinct metabolic and transcriptomic profiles compared to 3D cultured cells. The observed changes in the metabolomic and transcriptomic profiles were closely interconnected and influenced several important metabolic pathways including the TCA cycle, glycolysis, MAPK signaling cascades, and hemostasis. Data provided here may lead to clearer insights into the influence of the surrounding microenvironment on human dermal fibroblast metabolic patterns and molecular mechanisms, underscoring the importance of utilizing efficient 3D in vitro models to study such complex mechanisms.
Collapse
Affiliation(s)
- Sherin Abdelrahman
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Rui Ge
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Hepi H Susapto
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Yang Liu
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Faris Samkari
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Manola Moretti
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Xinzhi Liu
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Robert Hoehndorf
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Computer, Electrical and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955, Saudi Arabia
| | - Abdul-Hamid Emwas
- Core Laboratories, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Mariusz Jaremko
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Ranim H Rawas
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Charlotte A E Hauser
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| |
Collapse
|
61
|
Ivanovska IL, Tobin MP, Bai T, Dooling LJ, Discher DE. Small lipid droplets are rigid enough to indent a nucleus, dilute the lamina, and cause rupture. J Cell Biol 2023; 222:e202208123. [PMID: 37212777 PMCID: PMC10202833 DOI: 10.1083/jcb.202208123] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 03/24/2023] [Accepted: 05/04/2023] [Indexed: 05/23/2023] Open
Abstract
The nucleus in many cell types is a stiff organelle, but fat-filled lipid droplets (FDs) in cytoplasm are seen to indent and displace the nucleus. FDs are phase-separated liquids with a poorly understood interfacial tension γ that determines how FDs interact with other organelles. Here, micron-sized FDs remain spherical as they indent peri-nuclear actomyosin and the nucleus, while causing local dilution of Lamin-B1 independent of Lamin-A,C and sometimes triggering nuclear rupture. Focal accumulation of the cytosolic DNA sensor cGAS at the rupture site is accompanied by sustained mislocalization of DNA repair factors to cytoplasm, increased DNA damage, and delayed cell cycle. Macrophages show FDs and engulfed rigid beads cause similar indentation dilution. Spherical shapes of small FDs indicate a high γ, which we measure for FDs mechanically isolated from fresh adipose tissue as ∼40 mN/m. This value is far higher than that of protein condensates, but typical of oils in water and sufficiently rigid to perturb cell structures including nuclei.
Collapse
Affiliation(s)
- Irena L. Ivanovska
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P. Tobin
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Tianyi Bai
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Lawrence J. Dooling
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennis E. Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
62
|
Danielsson BE, George Abraham B, Mäntylä E, Cabe JI, Mayer CR, Rekonen A, Ek F, Conway DE, Ihalainen TO. Nuclear lamina strain states revealed by intermolecular force biosensor. Nat Commun 2023; 14:3867. [PMID: 37391402 PMCID: PMC10313699 DOI: 10.1038/s41467-023-39563-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/19/2023] [Indexed: 07/02/2023] Open
Abstract
Nuclear lamins have been considered an important structural element of the nucleus. The nuclear lamina is thought both to shield DNA from excessive mechanical forces and to transmit mechanical forces onto the DNA. However, to date there is not yet a technical approach to directly measure mechanical forces on nuclear lamins at the protein level. To overcome this limitation, we developed a nanobody-based intermolecular tension FRET biosensor capable of measuring the mechanical strain of lamin filaments. Using this sensor, we were able to show that the nuclear lamina is subjected to significant force. These forces are dependent on nuclear volume, actomyosin contractility, functional LINC complex, chromatin condensation state, cell cycle, and EMT. Interestingly, large forces were also present on nucleoplasmic lamins, indicating that these lamins may also have an important mechanical role in the nucleus. Overall, we demonstrate that the nanobody-based approach allows construction of biosensors for complex protein structures for mechanobiology studies.
Collapse
Affiliation(s)
- Brooke E Danielsson
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Bobin George Abraham
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Elina Mäntylä
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jolene I Cabe
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Carl R Mayer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Anna Rekonen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Frans Ek
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Daniel E Conway
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Ohio State University and Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA.
| | - Teemu O Ihalainen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland.
| |
Collapse
|
63
|
Antonelli F. 3D Cell Models in Radiobiology: Improving the Predictive Value of In Vitro Research. Int J Mol Sci 2023; 24:10620. [PMID: 37445795 DOI: 10.3390/ijms241310620] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Cancer is intrinsically complex, comprising both heterogeneous cellular composition and extracellular matrix. In vitro cancer research models have been widely used in the past to model and study cancer. Although two-dimensional (2D) cell culture models have traditionally been used for cancer research, they have many limitations, such as the disturbance of interactions between cellular and extracellular environments and changes in cell morphology, polarity, division mechanism, differentiation and cell motion. Moreover, 2D cell models are usually monotypic. This implies that 2D tumor models are ineffective at accurately recapitulating complex aspects of tumor cell growth, as well as their radiation responses. Over the past decade there has been significant uptake of three-dimensional (3D) in vitro models by cancer researchers, highlighting a complementary model for studies of radiation effects on tumors, especially in conjunction with chemotherapy. The introduction of 3D cell culture approaches aims to model in vivo tissue interactions with radiation by positioning itself halfway between 2D cell and animal models, and thus opening up new possibilities in the study of radiation response mechanisms of healthy and tumor tissues.
Collapse
Affiliation(s)
- Francesca Antonelli
- Laboratory of Biomedical Technologies, Division of Health Protection Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), 00123 Rome, Italy
| |
Collapse
|
64
|
Atcha H, Choi YS, Chaudhuri O, Engler AJ. Getting physical: Material mechanics is an intrinsic cell cue. Cell Stem Cell 2023; 30:750-765. [PMID: 37267912 PMCID: PMC10247187 DOI: 10.1016/j.stem.2023.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/30/2023] [Accepted: 05/02/2023] [Indexed: 06/04/2023]
Abstract
Advances in biomaterial science have allowed for unprecedented insight into the ability of material cues to influence stem cell function. These material approaches better recapitulate the microenvironment, providing a more realistic ex vivo model of the cell niche. However, recent advances in our ability to measure and manipulate niche properties in vivo have led to novel mechanobiological studies in model organisms. Thus, in this review, we will discuss the importance of material cues within the cell niche, highlight the key mechanotransduction pathways involved, and conclude with recent evidence that material cues regulate tissue function in vivo.
Collapse
Affiliation(s)
- Hamza Atcha
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Yu Suk Choi
- School of Human Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA.
| |
Collapse
|
65
|
Yin Q, Morris GF, Saito S, Zhuang Y, Thannickal VJ, Jazwinski SM, Lasky JA. Enhanced Expression of a Novel Lamin A/C Splice Variant in Idiopathic Pulmonary Fibrosis Lung. Am J Respir Cell Mol Biol 2023; 68:625-637. [PMID: 36848480 PMCID: PMC10257069 DOI: 10.1165/rcmb.2022-0222oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 02/23/2023] [Indexed: 03/01/2023] Open
Abstract
In idiopathic pulmonary fibrosis (IPF), the normal delicate lung architecture is replaced with rigid extracellular matrix (ECM) as a result of the accumulation of activated myofibroblasts and excessive deposition of ECM. Lamins have a role in fostering mechanosignaling from the ECM to the nucleus. Although there is a growing number of studies on lamins and associated diseases, there are no prior reports linking aberrations in lamins with pulmonary fibrosis. Here, we discovered, through analysis of RNA sequencing data, a novel isoform of lamin A/C that is more highly expressed in IPF compared with control lung. This novel LMNA (lamin A/C) splice variant includes retained introns 10 and 11 and exons 11 and 12 as documented by rapid amplification of cDNA ends. We found that this novel isoform is induced by stiff ECM. To better clarify the specific effects of this novel isoform of lamin A/C and how it may contribute to the pathogenesis of IPF, we transduced the lamin transcript into primary lung fibroblasts and alveolar epithelial cells and found that it impacts several biological effects, including cell proliferation, senescence, cell contraction, and the transition of fibroblasts to myofibroblasts. We also observed that type II epithelial cells and myofibroblasts in the IPF lung exhibited wrinkled nuclei, and this is notable because this has not been previously described and is consistent with laminopathy-mediated cellular effects.
Collapse
Affiliation(s)
- Qinyan Yin
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Department of Medicine
| | | | - Shigeki Saito
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Department of Medicine
| | - Yan Zhuang
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Department of Medicine
| | - Victor J. Thannickal
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana; and
| | - S. Michal Jazwinski
- Tulane Center for Aging, General Internal Medicine & Geriatrics, New Orleans, Louisiana
| | - Joseph A. Lasky
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Department of Medicine
| |
Collapse
|
66
|
Jingyuan L, Yu L, Hong J, Tao W, Kan L, Xiaomei L, Guiqing L, Yujie L. Matrix stiffness induces an invasive-dormant subpopulation via cGAS-STING axis in oral cancer. Transl Oncol 2023; 33:101681. [PMID: 37137218 PMCID: PMC10165441 DOI: 10.1016/j.tranon.2023.101681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/25/2023] [Accepted: 04/22/2023] [Indexed: 05/05/2023] Open
Abstract
OBJECTIVES Dormancy is a crucial machinery for cancer cells to survive hostile microenvironment. It is considered as the major cause of post-treatment relapse and metastases. However, its regulatory mechanism in oral squamous cell carcinoma (OSCC) remains unclear. Here we sought to decipher the impacts of matrix stiffness on OSCC-cell dormancy. MATERIALS AND METHODS Clinicopathological relevance of matrix stiffness in OSCC was analyzed in a 127 patients' cohort. Impacts of stiffness-related mechanical stress (MS) on OSCC-cell behaviors were investigated in vitro and in vivo. Transcriptomic profiling of MS induced dormant cells were performed, following by mechanistic investigations on MS-induced dormancy. The functional relevance of cGAS in OSCC were analyzed through a bioinformatic approach. RESULTS Stiffened matrix correlated with poor survival and post-surgical recurrence in OSCC. Stiffness-related MS induces a dormant subpopulation in OSCC cells, which showed increased drug resistance, enhanced tumor repopulating ability, and an unexpected upregulation of epithelial-mesenchymal transition (EMT) and invasiveness. Mechanistically, MS caused DNA damage, resulted in activation of cGAS-STING signaling. Either blocking of cGAS or STING dramatically impeded the MS-induced production of this invasive-dormant subpopulation. Moreover, cGAS was found being central to the cell-cycle regulation and correlated with poor prognosis in OSCC. DISCUSSION We revealed a previously unsuspected role of cGAS-STING axis in mediating the induction of an invasive-dormant subpopulation in response to mechanical cues. Our findings indicated an adaptive machinery whereby tumor cells survive and escape from harsh microenvironment. Targeting this machinery may be a potential strategy for preventing post-therapeutic recurrence and lymphatic metastasis in OSCC.
Collapse
Affiliation(s)
- Li Jingyuan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Liu Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jiang Hong
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Wang Tao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Li Kan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Lao Xiaomei
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Liao Guiqing
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.
| | - Liang Yujie
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
67
|
Chen P, Mishra S, Levy DL. Nuclear growth and import can be uncoupled. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.537556. [PMID: 37131802 PMCID: PMC10153267 DOI: 10.1101/2023.04.19.537556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
What drives nuclear growth? Studying nuclei assembled in Xenopus egg extract and focusing on importin α/β-mediated nuclear import, we show that, while nuclear growth depends on nuclear import, nuclear growth and import can be uncoupled. Nuclei containing fragmented DNA grew slowly despite exhibiting normal import rates, suggesting nuclear import itself is insufficient to drive nuclear growth. Nuclei containing more DNA grew larger but imported more slowly. Altering chromatin modifications caused nuclei to grow less while still importing to the same extent or to grow larger without increasing nuclear import. Increasing heterochromatin in vivo in sea urchin embryos increased nuclear growth but not import. These data suggest that nuclear import is not the primary driving force for nuclear growth. Instead, live imaging showed that nuclear growth preferentially occurred at sites of high chromatin density and lamin addition, whereas small nuclei lacking DNA exhibited less lamin incorporation. Our hypothesized model is that lamin incorporation and nuclear growth are driven by chromatin mechanical properties, which depend on and can be tuned by nuclear import.
Collapse
Affiliation(s)
- Pan Chen
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Sampada Mishra
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| | - Daniel L. Levy
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
68
|
Yamada S, Ko T, Ito M, Sassa T, Nomura S, Okuma H, Sato M, Imasaki T, Kikkawa S, Zhang B, Yamada T, Seki Y, Fujita K, Katoh M, Kubota M, Hatsuse S, Katagiri M, Hayashi H, Hamano M, Takeda N, Morita H, Takada S, Toyoda M, Uchiyama M, Ikeuchi M, Toyooka K, Umezawa A, Yamanishi Y, Nitta R, Aburatani H, Komuro I. TEAD1 trapping by the Q353R-Lamin A/C causes dilated cardiomyopathy. SCIENCE ADVANCES 2023; 9:eade7047. [PMID: 37058558 PMCID: PMC10104473 DOI: 10.1126/sciadv.ade7047] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 03/09/2023] [Indexed: 06/19/2023]
Abstract
Mutations in the LMNA gene encoding Lamin A and C (Lamin A/C), major components of the nuclear lamina, cause laminopathies including dilated cardiomyopathy (DCM), but the underlying molecular mechanisms have not been fully elucidated. Here, by leveraging single-cell RNA sequencing (RNA-seq), assay for transposase-accessible chromatin using sequencing (ATAC-seq), protein array, and electron microscopy analysis, we show that insufficient structural maturation of cardiomyocytes owing to trapping of transcription factor TEA domain transcription factor 1 (TEAD1) by mutant Lamin A/C at the nuclear membrane underlies the pathogenesis of Q353R-LMNA-related DCM. Inhibition of the Hippo pathway rescued the dysregulation of cardiac developmental genes by TEAD1 in LMNA mutant cardiomyocytes. Single-cell RNA-seq of cardiac tissues from patients with DCM with the LMNA mutation confirmed the dysregulated expression of TEAD1 target genes. Our results propose an intervention for transcriptional dysregulation as a potential treatment of LMNA-related DCM.
Collapse
Affiliation(s)
- Shintaro Yamada
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Toshiyuki Ko
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Masamichi Ito
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Department of Advanced Clinical Science and Therapeutics, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tatsuro Sassa
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Hiromichi Okuma
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Mayuko Sato
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa 230-0045, Japan
| | - Tsuyoshi Imasaki
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Satoshi Kikkawa
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Bo Zhang
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Takanobu Yamada
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Yuka Seki
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kanna Fujita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Manami Katoh
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Masayuki Kubota
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Satoshi Hatsuse
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Mikako Katagiri
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiromu Hayashi
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan
| | - Momoko Hamano
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan
| | - Norifumi Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shuji Takada
- Department of Systems BioMedicine, National Center for Child Health and Development Research Institute, Setagaya-ku, Tokyo 157-8535, Japan
| | - Masashi Toyoda
- Center for Regenerative Medicine, National Center for Child Health and Development Research Institute, Setagaya-ku, Tokyo 157-8535, Japan
| | - Masanobu Uchiyama
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Masashi Ikeuchi
- Division of Biofunctional Restoration, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Kiminori Toyooka
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa 230-0045, Japan
| | - Akihiro Umezawa
- Center for Regenerative Medicine, National Center for Child Health and Development Research Institute, Setagaya-ku, Tokyo 157-8535, Japan
| | - Yoshihiro Yamanishi
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan
| | - Ryo Nitta
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
69
|
Cao Y, Chen S, Lu J, Zhang M, Shi L, Qin J, Lv J, Li D, Ma L, Zhang Y. BPA induces placental trophoblast proliferation inhibition and fetal growth restriction by inhibiting the expression of SRB1. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:60805-60819. [PMID: 37037937 DOI: 10.1007/s11356-023-26850-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/03/2023] [Indexed: 04/12/2023]
Abstract
Bisphenol-A (BPA) is a common environmental toxicant that is known to be associated with fetal growth restriction (FGR). However, the mechanisms of how BPA induce FGR is poorly characterized. We conducted proteomics to identify the abnormal expression of SRB1 in female placental tissues with high BPA-induced FGR and further verified its decreased expression in human placenta and BeWo cells. Next, the effect of BPA on fetal development was further confirmed in pregnant C57BL/6 mice. The expression of SRB1 was consistently downregulated in human FGR placentas, BPA-exposed trophoblasts and mouse placentas. In addition, we found that SRB1 interacted with PCNA, and BPA exposure indirectly reduced the expression of PCNA and further inhibited placental proliferation. In vitro studies showed that BPA exposure reduced the expression of CDK1, CDK2, cyclin B and phosphorylated Rb in placental trophoblast cells, indicating cell cycle arrest after exposure to BPA. In addition, the expression of γ-H2AX and phosphorylated ATM was upregulated in BPA-exposed trophoblasts, indicating increased DNA damage. Our results indicate that BPA-induced FGR is achieved by reducing the expression of SRB1, inhibiting placental proliferation and increasing DNA damage. Our findings not only explain the mechanism of BPA-associated developmental toxicity but also shed light upon developing novel therapeutic targets.
Collapse
Affiliation(s)
- Yuming Cao
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, No 169 of Donghu Road, Wuhan, 430071, Hubei, China
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Gynaecology and Obstetrics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, People's Republic of China
| | - Sihan Chen
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jing Lu
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, No 169 of Donghu Road, Wuhan, 430071, Hubei, China
| | - Ming Zhang
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, No 169 of Donghu Road, Wuhan, 430071, Hubei, China
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lei Shi
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Juling Qin
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jing Lv
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Danyang Li
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ling Ma
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuanzhen Zhang
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, No 169 of Donghu Road, Wuhan, 430071, Hubei, China.
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Department of Gynaecology and Obstetrics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, People's Republic of China.
| |
Collapse
|
70
|
Liu S, Li Y, Hong Y, Wang M, Zhang H, Ma J, Qu K, Huang G, Lu TJ. Mechanotherapy in oncology: Targeting nuclear mechanics and mechanotransduction. Adv Drug Deliv Rev 2023; 194:114722. [PMID: 36738968 DOI: 10.1016/j.addr.2023.114722] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 12/23/2022] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
Mechanotherapy is proposed as a new option for cancer treatment. Increasing evidence suggests that characteristic differences are present in the nuclear mechanics and mechanotransduction of cancer cells compared with those of normal cells. Recent advances in understanding nuclear mechanics and mechanotransduction provide not only further insights into the process of malignant transformation but also useful references for developing new therapeutic approaches. Herein, we present an overview of the alterations of nuclear mechanics and mechanotransduction in cancer cells and highlight their implications in cancer mechanotherapy.
Collapse
Affiliation(s)
- Shaobao Liu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics, Nanjing 210016, PR China
| | - Yuan Li
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuan Hong
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; National Science Foundation Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO 63130, USA
| | - Ming Wang
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Hao Zhang
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics, Nanjing 210016, PR China
| | - Jinlu Ma
- Department of Radiation Oncology, the First Affiliated Hospital, Xian Jiaotong University, Xi'an 710061, PR China
| | - Kai Qu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, Xian Jiaotong University, Xi'an 710061, PR China
| | - Guoyou Huang
- Department of Engineering Mechanics, School of Civil Engineering, Wuhan University, Wuhan 430072, PR China.
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics, Nanjing 210016, PR China.
| |
Collapse
|
71
|
Wang Y, Dobreva G. Epigenetics in LMNA-Related Cardiomyopathy. Cells 2023; 12:cells12050783. [PMID: 36899919 PMCID: PMC10001118 DOI: 10.3390/cells12050783] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/18/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Mutations in the gene for lamin A/C (LMNA) cause a diverse range of diseases known as laminopathies. LMNA-related cardiomyopathy is a common inherited heart disease and is highly penetrant with a poor prognosis. In the past years, numerous investigations using mouse models, stem cell technologies, and patient samples have characterized the phenotypic diversity caused by specific LMNA variants and contributed to understanding the molecular mechanisms underlying the pathogenesis of heart disease. As a component of the nuclear envelope, LMNA regulates nuclear mechanostability and function, chromatin organization, and gene transcription. This review will focus on the different cardiomyopathies caused by LMNA mutations, address the role of LMNA in chromatin organization and gene regulation, and discuss how these processes go awry in heart disease.
Collapse
Affiliation(s)
- Yinuo Wang
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), 68167 Mannheim, Germany
- Correspondence: (Y.W.); (G.D.)
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), 68167 Mannheim, Germany
- Correspondence: (Y.W.); (G.D.)
| |
Collapse
|
72
|
Ma Y, Zhang X, Tang S, Xue L, Wang J, Zhang X. Extended preconditioning on soft matrices directs human mesenchymal stem cell fate via YAP transcriptional activity and chromatin organization. APL Bioeng 2023; 7:016110. [PMID: 36845904 PMCID: PMC9949900 DOI: 10.1063/5.0124424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
Dynamic extracellular matrix (ECM) mechanics plays a crucial role in tissue development and disease progression through regulation of stem cell behavior, differentiation, and fate determination. Periodontitis is a typical case characterized by decreased ECM stiffness within diseased periodontal tissues as well as with irreversible loss of osteogenesis capacity of periodontal tissue-derived human periodontal tissue-derived MSCs (hMSCs) even returning back to a physiological mechanical microenvironment. We hypothesized that the hMSCs extendedly residing in the soft ECM of diseased periodontal tissues may memorize the mechanical information and have further effect on ultimate cell fate besides the current mechanical microenvironment. Using a soft priming and subsequent stiff culture system based on collagen-modified polydimethylsiloxane substrates, we were able to discover that extended preconditioning on soft matrices (e.g., 7 days of exposure) led to approximately one-third decrease in cell spreading, two-third decrease in osteogenic markers (e.g., RUNX2 and OPN) of hMSCs, and one-thirteenth decrease in the production of mineralized nodules. The significant loss of osteogenic ability may attribute to the long-term residing of hMSCs in diseased periodontal tissue featured with reduced stiffness. This is associated with the regulation of transcriptional activity through alterations of subcellular localization of yes-associated protein and nuclear feature-mediated chromatin organization. Collectively, we reconstructed phenomena of irreversible loss of hMSC osteogenesis capacity in diseased periodontal tissues in our system and revealed the critical effect of preconditioning duration on soft matrices as well as the potential mechanisms in determining ultimate hMSC fate.
Collapse
Affiliation(s)
- Yufei Ma
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Xu Zhang
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Shaoxin Tang
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Li Xue
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Center for Mitochondrial Biology and Medicine, School of Life Science and Technology, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technology, Xi'an Key Laboratory for Biomedical Testing and High-End Equipment, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Jing Wang
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Xiaohui Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Center for Mitochondrial Biology and Medicine, School of Life Science and Technology, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technology, Xi'an Key Laboratory for Biomedical Testing and High-End Equipment, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| |
Collapse
|
73
|
Manoj P, Kim JA, Kim S, Li T, Sewani M, Chelu MG, Li N. Sinus node dysfunction: current understanding and future directions. Am J Physiol Heart Circ Physiol 2023; 324:H259-H278. [PMID: 36563014 PMCID: PMC9886352 DOI: 10.1152/ajpheart.00618.2022] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
The sinoatrial node (SAN) is the primary pacemaker of the heart. Normal SAN function is crucial in maintaining proper cardiac rhythm and contraction. Sinus node dysfunction (SND) is due to abnormalities within the SAN, which can affect the heartbeat frequency, regularity, and the propagation of electrical pulses through the cardiac conduction system. As a result, SND often increases the risk of cardiac arrhythmias. SND is most commonly seen as a disease of the elderly given the role of degenerative fibrosis as well as other age-dependent changes in its pathogenesis. Despite the prevalence of SND, current treatment is limited to pacemaker implantation, which is associated with substantial medical costs and complications. Emerging evidence has identified various genetic abnormalities that can cause SND, shedding light on the molecular underpinnings of SND. Identification of these molecular mechanisms and pathways implicated in the pathogenesis of SND is hoped to identify novel therapeutic targets for the development of more effective therapies for this disease. In this review article, we examine the anatomy of the SAN and the pathophysiology and epidemiology of SND. We then discuss in detail the most common genetic mutations correlated with SND and provide our perspectives on future research and therapeutic opportunities in this field.
Collapse
Affiliation(s)
- Pavan Manoj
- School of Public Health, Texas A&M University, College Station, Texas
| | - Jitae A Kim
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Stephanie Kim
- Department of BioSciences, Rice University, Houston, Texas
| | - Tingting Li
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Maham Sewani
- Department of BioSciences, Rice University, Houston, Texas
| | - Mihail G Chelu
- Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Na Li
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
74
|
Wang C, Ramahdita G, Genin G, Huebsch N, Ma Z. Dynamic mechanobiology of cardiac cells and tissues: Current status and future perspective. BIOPHYSICS REVIEWS 2023; 4:011314. [PMID: 37008887 PMCID: PMC10062054 DOI: 10.1063/5.0141269] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/08/2023] [Indexed: 03/31/2023]
Abstract
Mechanical forces impact cardiac cells and tissues over their entire lifespan, from development to growth and eventually to pathophysiology. However, the mechanobiological pathways that drive cell and tissue responses to mechanical forces are only now beginning to be understood, due in part to the challenges in replicating the evolving dynamic microenvironments of cardiac cells and tissues in a laboratory setting. Although many in vitro cardiac models have been established to provide specific stiffness, topography, or viscoelasticity to cardiac cells and tissues via biomaterial scaffolds or external stimuli, technologies for presenting time-evolving mechanical microenvironments have only recently been developed. In this review, we summarize the range of in vitro platforms that have been used for cardiac mechanobiological studies. We provide a comprehensive review on phenotypic and molecular changes of cardiomyocytes in response to these environments, with a focus on how dynamic mechanical cues are transduced and deciphered. We conclude with our vision of how these findings will help to define the baseline of heart pathology and of how these in vitro systems will potentially serve to improve the development of therapies for heart diseases.
Collapse
Affiliation(s)
| | - Ghiska Ramahdita
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | | | | | - Zhen Ma
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
75
|
Elevated Levels of Lamin A Promote HR and NHEJ-Mediated Repair Mechanisms in High-Grade Ovarian Serous Carcinoma Cell Line. Cells 2023; 12:cells12050757. [PMID: 36899893 PMCID: PMC10001195 DOI: 10.3390/cells12050757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Extensive research for the last two decades has significantly contributed to understanding the roles of lamins in the maintenance of nuclear architecture and genome organization which is drastically modified in neoplasia. It must be emphasized that alteration in lamin A/C expression and distribution is a consistent event during tumorigenesis of almost all tissues of human bodies. One of the important signatures of a cancer cell is its inability to repair DNA damage which befalls several genomic events that transform the cells to be sensitive to chemotherapeutic agents. This genomic and chromosomal instability is the most common feature found in cases of high-grade ovarian serous carcinoma. Here, we report elevated levels of lamins in OVCAR3 cells (high-grade ovarian serous carcinoma cell line) in comparison to IOSE (immortalised ovarian surface epithelial cells) and, consequently, altered damage repair machinery in OVCAR3. We have analysed the changes in global gene expression as a sequel to DNA damage induced by etoposide in ovarian carcinoma where lamin A is particularly elevated in expression and reported some differentially expressed genes associated with pathways conferring cellular proliferation and chemoresistance. We hereby establish the role of elevated lamin A in neoplastic transformation in the context of high-grade ovarian serous cancer through a combination of HR and NHEJ mechanisms.
Collapse
|
76
|
Diniz Filho JFS, de Barros AODS, Pijeira MSO, Ricci-Junior E, Midlej V, Baroni MPMA, dos Santos CC, Alencar LMR, Santos-Oliveira R. Ultrastructural Analysis of Cancer Cells Treated with the Radiopharmaceutical Radium Dichloride ([ 223Ra]RaCl 2): Understanding the Effect on Cell Structure. Cells 2023; 12:451. [PMID: 36766793 PMCID: PMC9913731 DOI: 10.3390/cells12030451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/10/2023] [Accepted: 01/30/2023] [Indexed: 02/01/2023] Open
Abstract
The use of alpha-particle (α-particle) radionuclides, especially [223Ra]RaCl2 (radium dichloride), for targeted alpha therapy is steadily increasing. Despite the positive clinical outcomes of this therapy, very little data are available about the effect on the ultrastructure of cells. The purpose of this study was to evaluate the nanomechanical and ultrastructure effect of [223Ra] RaCl2 on cancer cells. To analyze the effect of [223Ra]RaCl2 on tumor cells, human breast cancer cells (lineage MDA-MB-231) were cultured and treated with the radiopharmaceutical at doses of 2 µCi and 0.9 µCi. The effect was evaluated using atomic force microscopy (AFM) and transmission electron microscopy (TEM) combined with Raman spectroscopy. The results showed massive destruction of the cell membrane but preservation of the nucleus membrane. No evidence of DNA alteration was observed. The data demonstrated the formation of lysosomes and phagosomes. These findings help elucidate the main mechanism involved in cell death during α-particle therapy.
Collapse
Affiliation(s)
- Joel Félix Silva Diniz Filho
- Biophysics and Nanosystems Laboratory, Department of Physics, Federal University of Maranhão, São Luis 65065690, MA, Brazil
| | - Aline Oliveira da Silva de Barros
- Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, RJ, Brazil
| | - Martha Sahylí Ortega Pijeira
- Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, RJ, Brazil
| | - Eduardo Ricci-Junior
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro 21941900, RJ, Brazil
| | - Victor Midlej
- Laboratory of Structural Biology, Oswaldo Cruz Institute (FIOCRUZ), Rio de Janeiro 21040900, RJ, Brazil
| | | | - Clenilton Costa dos Santos
- Biophysics and Nanosystems Laboratory, Department of Physics, Federal University of Maranhão, São Luis 65065690, MA, Brazil
| | | | - Ralph Santos-Oliveira
- Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, RJ, Brazil
- Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, State University of Rio de Janeiro, Rio de Janeiro 23070200, RJ, Brazil
| |
Collapse
|
77
|
Chen R, Buchmann S, Kroth A, Arias-Loza AP, Kohlhaas M, Wagner N, Grüner G, Nickel A, Cirnu A, Williams T, Maack C, Ergün S, Frantz S, Gerull B. Mechanistic Insights of the LEMD2 p.L13R Mutation and Its Role in Cardiomyopathy. Circ Res 2023; 132:e43-e58. [PMID: 36656972 DOI: 10.1161/circresaha.122.321929] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Nuclear envelope proteins play an important role in the pathogenesis of hereditary cardiomyopathies. Recently, a new form of arrhythmic cardiomyopathy caused by a homozygous mutation (p.L13R) in the inner nuclear membrane protein LEMD2 was discovered. The aim was to unravel the molecular mechanisms of mutant LEMD2 in the pathogenesis of cardiomyopathy. METHODS We generated a Lemd2 p.L13R knock-in mouse model and a corresponding cell model via CRISPR/Cas9 technology and investigated the cardiac phenotype as well as cellular and subcellular mechanisms of nuclear membrane rupture and repair. RESULTS Knock-in mice developed a cardiomyopathy with predominantly endocardial fibrosis, left ventricular dilatation, and systolic dysfunction. Electrocardiograms displayed pronounced ventricular arrhythmias and conduction disease. A key finding of knock-in cardiomyocytes on ultrastructural level was a significant increase in nuclear membrane invaginations and decreased nuclear circularity. Furthermore, increased DNA damage and premature senescence were detected as the underlying cause of fibrotic and inflammatory remodeling. As the p.L13R mutation is located in the Lap2/Emerin/Man1 (LEM)-domain, we observed a disrupted interaction between mutant LEMD2 and BAF (barrier-to-autointegration factor), which is required to initiate the nuclear envelope rupture repair process. To mimic increased mechanical stress with subsequent nuclear envelope ruptures, we investigated mutant HeLa-cells upon electrical stimulation and increased stiffness. Here, we demonstrated impaired nuclear envelope rupture repair capacity, subsequent cytoplasmic leakage of the DNA repair factor KU80 along with increased DNA damage, and recruitment of the cGAS (cyclic GMP-AMP synthase) to the nuclear membrane and micronuclei. CONCLUSIONS We show for the first time that the Lemd2 p.L13R mutation in mice recapitulates human dilated cardiomyopathy with fibrosis and severe ventricular arrhythmias. Impaired nuclear envelope rupture repair capacity resulted in increased DNA damage and activation of the cGAS/STING/IFN pathway, promoting premature senescence. Hence, LEMD2 is a new player inthe disease group of laminopathies.
Collapse
Affiliation(s)
- Ruping Chen
- Department of Cardiovascular Genetics, Comprehensive Heart Failure Center (R.C., S.B., A.K., G.G., A.C., T.W., B.G.), University Hospital Würzburg, Germany
- Department of Medicine I (R.C., T.W., C.M., S.F., B.G.), University Hospital Würzburg, Germany
| | - Simone Buchmann
- Department of Cardiovascular Genetics, Comprehensive Heart Failure Center (R.C., S.B., A.K., G.G., A.C., T.W., B.G.), University Hospital Würzburg, Germany
| | - Amos Kroth
- Department of Cardiovascular Genetics, Comprehensive Heart Failure Center (R.C., S.B., A.K., G.G., A.C., T.W., B.G.), University Hospital Würzburg, Germany
| | - Anahi-Paula Arias-Loza
- Department of Nuclear Medicine, Comprehensive Heart Failure Center (A.-P.A.-L.), University Hospital Würzburg, Germany
| | - Michael Kohlhaas
- Department of Translational Research, Comprehensive Heart Failure Center (M.K., A.N., C.M.), University Hospital Würzburg, Germany
| | - Nicole Wagner
- Institute of Anatomy and Cell Biology, University of Würzburg, Germany (N.W., S.E.)
| | - Gianna Grüner
- Department of Cardiovascular Genetics, Comprehensive Heart Failure Center (R.C., S.B., A.K., G.G., A.C., T.W., B.G.), University Hospital Würzburg, Germany
| | - Alexander Nickel
- Department of Translational Research, Comprehensive Heart Failure Center (M.K., A.N., C.M.), University Hospital Würzburg, Germany
| | - Alexandra Cirnu
- Department of Cardiovascular Genetics, Comprehensive Heart Failure Center (R.C., S.B., A.K., G.G., A.C., T.W., B.G.), University Hospital Würzburg, Germany
| | - Tatjana Williams
- Department of Cardiovascular Genetics, Comprehensive Heart Failure Center (R.C., S.B., A.K., G.G., A.C., T.W., B.G.), University Hospital Würzburg, Germany
- Department of Medicine I (R.C., T.W., C.M., S.F., B.G.), University Hospital Würzburg, Germany
| | - Christoph Maack
- Department of Medicine I (R.C., T.W., C.M., S.F., B.G.), University Hospital Würzburg, Germany
- Department of Translational Research, Comprehensive Heart Failure Center (M.K., A.N., C.M.), University Hospital Würzburg, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, University of Würzburg, Germany (N.W., S.E.)
| | - Stefan Frantz
- Department of Medicine I (R.C., T.W., C.M., S.F., B.G.), University Hospital Würzburg, Germany
- Comprehensive Heart Failure Center (S.F.), University Hospital Würzburg, Germany
| | - Brenda Gerull
- Department of Cardiovascular Genetics, Comprehensive Heart Failure Center (R.C., S.B., A.K., G.G., A.C., T.W., B.G.), University Hospital Würzburg, Germany
- Department of Medicine I (R.C., T.W., C.M., S.F., B.G.), University Hospital Würzburg, Germany
| |
Collapse
|
78
|
Scott NR, Parekh SH. A-type lamins involvement in transport and implications in cancer? Nucleus 2022; 13:221-235. [PMID: 36109835 PMCID: PMC9481127 DOI: 10.1080/19491034.2022.2118418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Nuclear lamins and transport are intrinsically linked, but their relationship is yet to be fully unraveled. A multitude of complex, coupled interactions between lamins and nucleoporins (Nups), which mediate active transport into and out of the nucleus, combined with well documented dysregulation of lamins in many cancers, suggests that lamins and nuclear transport may play a pivotal role in carcinogenesis and the preservation of cancer. Changes of function related to lamin/Nup activity can principally lead to DNA damage, further increasing the genetic diversity within a tumor, which could lead to the reduction the effectiveness of antineoplastic treatments. This review discusses and synthesizes different connections of lamins to nuclear transport and offers a number of outlook questions, the answers to which could reveal a new perspective on the connection of lamins to molecular transport of cancer therapeutics, in addition to their established role in nuclear mechanics.
Collapse
Affiliation(s)
- Nicholas R. Scott
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Sapun H. Parekh
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
79
|
Lammerding J, Engler AJ, Kamm R. Mechanobiology of the cell nucleus. APL Bioeng 2022; 6:040401. [PMID: 36536804 PMCID: PMC9759352 DOI: 10.1063/5.0135299] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Affiliation(s)
- Jan Lammerding
- Meinig School of Biomedical Engineering and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853-6007, USA,Authors to whom correspondence should be addressed:; ; and
| | - Adam J. Engler
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, California 92093-0412, USA,Authors to whom correspondence should be addressed:; ; and
| | - Roger Kamm
- Department of Biological Engineering and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA,Authors to whom correspondence should be addressed:; ; and
| |
Collapse
|
80
|
Pfeifer CR, Tobin MP, Cho S, Vashisth M, Dooling LJ, Vazquez LL, Ricci-De Lucca EG, Simon KT, Discher DE. Gaussian curvature dilutes the nuclear lamina, favoring nuclear rupture, especially at high strain rate. Nucleus 2022; 13:129-143. [PMID: 35293271 PMCID: PMC8928808 DOI: 10.1080/19491034.2022.2045726] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Nuclear rupture has long been associated with deficits or defects in lamins, with recent results also indicating a role for actomyosin stress, but key physical determinants of rupture remain unclear. Here, lamin-B filaments stably interact with the nuclear membrane at sites of low Gaussian curvature yet dilute at high curvature to favor rupture, whereas lamin-A depletion requires high strain-rates. Live-cell imaging of lamin-B1 gene-edited cancer cells is complemented by fixed-cell imaging of rupture in: iPS-derived progeria patients cells, cells within beating chick embryo hearts, and cancer cells with multi-site rupture after migration through small pores. Data fit a model of stiff filaments that detach from a curved surface.Rupture is modestly suppressed by inhibiting myosin-II and by hypotonic stress, which slow the strain-rates. Lamin-A dilution and rupture probability indeed increase above a threshold rate of nuclear pulling. Curvature-sensing mechanisms of proteins at plasma membranes, including Piezo1, might thus apply at nuclear membranes.Summary statement: High nuclear curvature drives lamina dilution and nuclear envelope rupture even when myosin stress is inhibited. Stiff filaments generally dilute from sites of high Gaussian curvature, providing mathematical fits of experiments.
Collapse
Affiliation(s)
- Charlotte R. Pfeifer
- Physical Sciences Oncology Center at Penn (PSOC@penn), University of Pennsylvania, Philadelphia, PA, USA,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA,Graduate Group/Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P. Tobin
- Physical Sciences Oncology Center at Penn (PSOC@penn), University of Pennsylvania, Philadelphia, PA, USA,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA,Graduate Group/Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Sangkyun Cho
- Physical Sciences Oncology Center at Penn (PSOC@penn), University of Pennsylvania, Philadelphia, PA, USA,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Manasvita Vashisth
- Physical Sciences Oncology Center at Penn (PSOC@penn), University of Pennsylvania, Philadelphia, PA, USA,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Lawrence J. Dooling
- Physical Sciences Oncology Center at Penn (PSOC@penn), University of Pennsylvania, Philadelphia, PA, USA,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Lizeth Lopez Vazquez
- Physical Sciences Oncology Center at Penn (PSOC@penn), University of Pennsylvania, Philadelphia, PA, USA,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Emma G. Ricci-De Lucca
- Physical Sciences Oncology Center at Penn (PSOC@penn), University of Pennsylvania, Philadelphia, PA, USA,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Keiann T. Simon
- Physical Sciences Oncology Center at Penn (PSOC@penn), University of Pennsylvania, Philadelphia, PA, USA,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennis E. Discher
- Physical Sciences Oncology Center at Penn (PSOC@penn), University of Pennsylvania, Philadelphia, PA, USA,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA,Graduate Group/Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, USA,Graduate Group/Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA,CONTACT Dennis E. Discher Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
81
|
Cheedipudi SM, Asghar S, Marian AJ. Genetic Ablation of the DNA Damage Response Pathway Attenuates Lamin-Associated Dilated Cardiomyopathy in Mice. JACC Basic Transl Sci 2022; 7:1232-1245. [PMID: 36644279 PMCID: PMC9831927 DOI: 10.1016/j.jacbts.2022.06.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 11/07/2022]
Abstract
Hereditary dilated cardiomyopathy (DCM) is a primary disease of cardiac myocytes caused by mutations in genes encoding proteins with a diverse array of functions. Mutations in the LMNA gene, encoding the nuclear envelope protein lamin A/C, are the second most common causes of DCM. The phenotype is characterized by progressive cardiac dysfunction, leading to refractory heart failure, myocardial fibrosis, cardiac arrhythmias, and sudden cardiac death. The molecular pathogenesis of DCM caused by the LMNA mutations is not well known. The LMNA protein is involved in nuclear membrane stability. It is also a guardian of the genome involved in the processing of the topoisomerases at the transcriptionally active domain and the repair of double-stranded DNA breaks (DSBs). Deletion of the mouse Lmna gene in cardiac myocytes leads to premature death, DCM, myocardial fibrosis, and apoptosis. The phenotype is associated with increased expression of the cytosolic DNA sensor cyclic GMP-AMP synthase (CGAS) and activation of the DNA damage response (DDR) pathway. Genetic blockade of the DDR pathway, upon knockout of the Mb21d1 gene encoding CGAS, prolonged survival, improved cardiac function, partially restored levels of molecular markers of heart failure, and attenuated myocardial apoptosis and fibrosis in the LMNA-deficient mice. The findings indicate that targeting the CGAS/DDR pathway might be beneficial in the treatment of DCM caused by mutations in the LMNA gene.
Collapse
Affiliation(s)
- Sirisha M. Cheedipudi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Saman Asghar
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Ali J. Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
82
|
Brayson D, Shanahan CM. Lamin A precursor localizes to the Z-disc of sarcomeres in the heart and is dynamically regulated in muscle cell differentiation. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210490. [PMID: 36189817 PMCID: PMC9527914 DOI: 10.1098/rstb.2021.0490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/08/2022] [Indexed: 11/12/2022] Open
Abstract
The lamin A precursor, prelamin A, requires extensive processing to yield mature lamin A and effect its primary function as a structural filament of the nucleoskeleton. When processing is perturbed, nuclear accumulation of prelamin A is toxic and causes laminopathic diseases such as Hutchinson-Gilford progeria syndrome and cardiomyopathy. However, the physiological role of prelamin A is largely unknown and we sought to identify novel insights about this. Using rodent heart tissue, primary cells and the C2C12 model of myofibrillogenesis, we investigated the expression and localization patterns of prelamin A in heart and skeletal muscle cells. We found that endogenous prelamin A was detectable in mouse heart localized to the sarcomere in both adult mouse heart and isolated neonatal rat cardiomyocytes. We investigated the regulation of prelamin A in C2C12 myofibrillogenesis and found it was dynamically regulated and organized into striations upon myofibril formation, colocalizing with the Z-disc protein α-actinin. These data provide evidence that prelamin A is a component of the sarcomere, underpinning a physiological purpose for unprocessed prelamin A. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- Daniel Brayson
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
- School of Cardiovascular Medicine and Sciences, King's College London BHF Centre for Research Excellence, London, UK
| | - Catherine M. Shanahan
- School of Cardiovascular Medicine and Sciences, King's College London BHF Centre for Research Excellence, London, UK
| |
Collapse
|
83
|
Goult BT, von Essen M, Hytönen VP. The mechanical cell - the role of force dependencies in synchronising protein interaction networks. J Cell Sci 2022; 135:283155. [PMID: 36398718 PMCID: PMC9845749 DOI: 10.1242/jcs.259769] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The role of mechanical signals in the proper functioning of organisms is increasingly recognised, and every cell senses physical forces and responds to them. These forces are generated both from outside the cell or via the sophisticated force-generation machinery of the cell, the cytoskeleton. All regions of the cell are connected via mechanical linkages, enabling the whole cell to function as a mechanical system. In this Review, we define some of the key concepts of how this machinery functions, highlighting the critical requirement for mechanosensory proteins, and conceptualise the coupling of mechanical linkages to mechanochemical switches that enables forces to be converted into biological signals. These mechanical couplings provide a mechanism for how mechanical crosstalk might coordinate the entire cell, its neighbours, extending into whole collections of cells, in tissues and in organs, and ultimately in the coordination and operation of entire organisms. Consequently, many diseases manifest through defects in this machinery, which we map onto schematics of the mechanical linkages within a cell. This mapping approach paves the way for the identification of additional linkages between mechanosignalling pathways and so might identify treatments for diseases, where mechanical connections are affected by mutations or where individual force-regulated components are defective.
Collapse
Affiliation(s)
- Benjamin T. Goult
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, Kent, UK,Authors for correspondence (; )
| | - Magdaléna von Essen
- Faculty of Medicine and Health Technology, Tampere University, FI-33100 Tampere, Finland
| | - Vesa P. Hytönen
- Faculty of Medicine and Health Technology, Tampere University, FI-33100 Tampere, Finland,Fimlab Laboratories, FI-33520 Tampere, Finland,Authors for correspondence (; )
| |
Collapse
|
84
|
Lamin A/C-dependent chromatin architecture safeguards naïve pluripotency to prevent aberrant cardiovascular cell fate and function. Nat Commun 2022; 13:6663. [PMID: 36333314 PMCID: PMC9636150 DOI: 10.1038/s41467-022-34366-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
Tight control of cell fate choices is crucial for normal development. Here we show that lamin A/C plays a key role in chromatin organization in embryonic stem cells (ESCs), which safeguards naïve pluripotency and ensures proper cell fate choices during cardiogenesis. We report changes in chromatin compaction and localization of cardiac genes in Lmna-/- ESCs resulting in precocious activation of a transcriptional program promoting cardiomyocyte versus endothelial cell fate. This is accompanied by premature cardiomyocyte differentiation, cell cycle withdrawal and abnormal contractility. Gata4 is activated by lamin A/C loss and Gata4 silencing or haploinsufficiency rescues the aberrant cardiovascular cell fate choices induced by lamin A/C deficiency. We uncover divergent functions of lamin A/C in naïve pluripotent stem cells and cardiomyocytes, which have distinct contributions to the transcriptional alterations of patients with LMNA-associated cardiomyopathy. We conclude that disruption of lamin A/C-dependent chromatin architecture in ESCs is a primary event in LMNA loss-of-function cardiomyopathy.
Collapse
|
85
|
Fenelon KD, Thomas E, Samani M, Zhu M, Tao H, Sun Y, McNeill H, Hopyan S. Transgenic force sensors and software to measure force transmission across the mammalian nuclear envelope in vivo. Biol Open 2022; 11:bio059656. [PMID: 36350289 PMCID: PMC9672859 DOI: 10.1242/bio.059656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 01/28/2025] Open
Abstract
Nuclear mechanotransduction is a growing field with exciting implications for the regulation of gene expression and cellular function. Mechanical signals may be transduced to the nuclear interior biochemically or physically through connections between the cell surface and chromatin. To define mechanical stresses upon the nucleus in physiological settings, we generated transgenic mouse strains that harbour FRET-based tension sensors or control constructs in the outer and inner aspects of the nuclear envelope. We knocked-in a published esprin-2G sensor to measure tensions across the LINC complex and generated a new sensor that links the inner nuclear membrane to chromatin. To mitigate challenges inherent to fluorescence lifetime analysis in vivo, we developed software (FLIMvivo) that markedly improves the fitting of fluorescence decay curves. In the mouse embryo, the sensors responded to cytoskeletal relaxation and stretch applied by micro-aspiration. They reported organ-specific differences and a spatiotemporal tension gradient along the proximodistal axis of the limb bud, raising the possibility that mechanical mechanisms coregulate pattern formation. These mouse strains and software are potentially valuable tools for testing and refining mechanotransduction hypotheses in vivo.
Collapse
Affiliation(s)
- Kelli D. Fenelon
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Evan Thomas
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Mohammad Samani
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Min Zhu
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Hirotaka Tao
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Helen McNeill
- Department of Developmental Biology, Washington University, St. Louis, MO 63110, USA
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Orthopaedic Surgery, Hospital for Sick Children and University of Toronto, ON M5G 1X8, Canada
| |
Collapse
|
86
|
Pathological Nuclear Hallmarks in Dentate Granule Cells of Alzheimer’s Patients: A Biphasic Regulation of Neurogenesis. Int J Mol Sci 2022; 23:ijms232112873. [PMID: 36361662 PMCID: PMC9654738 DOI: 10.3390/ijms232112873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/29/2022] Open
Abstract
The dentate gyrus (DG) of the human hippocampus is a complex and dynamic structure harboring mature and immature granular neurons in diverse proliferative states. While most mammals show persistent neurogenesis through adulthood, human neurogenesis is still under debate. We found nuclear alterations in granular cells in autopsied human brains, detected by immunohistochemistry. These alterations differ from those reported in pyramidal neurons of the hippocampal circuit. Aging and early AD chromatin were clearly differentiated by the increased epigenetic markers H3K9me3 (heterochromatin suppressive mark) and H3K4me3 (transcriptional euchromatin mark). At early AD stages, lamin B2 was redistributed to the nucleoplasm, indicating cell-cycle reactivation, probably induced by hippocampal nuclear pathology. At intermediate and late AD stages, higher lamin B2 immunopositivity in the perinucleus suggests fewer immature neurons, less neurogenesis, and fewer adaptation resources to environmental factors. In addition, senile samples showed increased nuclear Tau interacting with aged chromatin, likely favoring DNA repair and maintaining genomic stability. However, at late AD stages, the progressive disappearance of phosphorylated Tau forms in the nucleus, increased chromatin disorganization, and increased nuclear autophagy support a model of biphasic neurogenesis in AD. Therefore, designing therapies to alleviate the neuronal nuclear pathology might be the only pathway to a true rejuvenation of brain circuits.
Collapse
|
87
|
Kervella M, Jahier M, Meli AC, Muchir A. Genome organization in cardiomyocytes expressing mutated A-type lamins. Front Cell Dev Biol 2022; 10:1030950. [PMID: 36274847 PMCID: PMC9585167 DOI: 10.3389/fcell.2022.1030950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiomyopathy is a myocardial disorder, in which the heart muscle is structurally and functionally abnormal, often leading to heart failure. Dilated cardiomyopathy is characterized by a compromised left ventricular function and contributes significantly to the heart failure epidemic, which represents a staggering clinical and public health problem worldwide. Gene mutations have been identified in 35% of patients with dilated cardiomyopathy. Pathogenic variants in LMNA, encoding nuclear A-type lamins, are one of the major causative causes of dilated cardiomyopathy (i.e. CardioLaminopathy). A-type lamins are type V intermediate filament proteins, which are the main components of the nuclear lamina. The nuclear lamina is connected to the cytoskeleton on one side, and to the chromatin on the other side. Among the models proposed to explain how CardioLaminopathy arises, the “chromatin model” posits an effect of mutated A-type lamins on the 3D genome organization and thus on the transcription activity of tissue-specific genes. Chromatin contacts with the nuclear lamina via specific genomic regions called lamina-associated domains lamina-associated domains. These LADs play a role in the chromatin organization and gene expression regulation. This review focuses on the identification of LADs and chromatin remodeling in cardiac muscle cells expressing mutated A-type lamins and discusses the methods and relevance of these findings in disease.
Collapse
Affiliation(s)
- Marie Kervella
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Maureen Jahier
- Sorbonne Université, INSERM U974, Institute of Myology, Center of Research in Myology, Paris, France
| | - Albano C. Meli
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Antoine Muchir
- Sorbonne Université, INSERM U974, Institute of Myology, Center of Research in Myology, Paris, France
- *Correspondence: Antoine Muchir,
| |
Collapse
|
88
|
Song Y, Soto J, Chen B, Hoffman T, Zhao W, Zhu N, Peng Q, Liu L, Ly C, Wong PK, Wang Y, Rowat AC, Kurdistani SK, Li S. Transient nuclear deformation primes epigenetic state and promotes cell reprogramming. NATURE MATERIALS 2022; 21:1191-1199. [PMID: 35927431 PMCID: PMC9529815 DOI: 10.1038/s41563-022-01312-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 06/14/2022] [Indexed: 05/22/2023]
Abstract
Cell reprogramming has wide applications in tissue regeneration, disease modelling and personalized medicine. In addition to biochemical cues, mechanical forces also contribute to the modulation of the epigenetic state and a variety of cell functions through distinct mechanisms that are not fully understood. Here we show that millisecond deformation of the cell nucleus caused by confinement into microfluidic channels results in wrinkling and transient disassembly of the nuclear lamina, local detachment of lamina-associated domains in chromatin and a decrease of histone methylation (histone H3 lysine 9 trimethylation) and DNA methylation. These global changes in chromatin at the early stage of cell reprogramming boost the conversion of fibroblasts into neurons and can be partially reproduced by inhibition of histone H3 lysine 9 and DNA methylation. This mechanopriming approach also triggers macrophage reprogramming into neurons and fibroblast conversion into induced pluripotent stem cells, being thus a promising mechanically based epigenetic state modulation method for cell engineering.
Collapse
Affiliation(s)
- Yang Song
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer Soto
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Binru Chen
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Tyler Hoffman
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Weikang Zhao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Ninghao Zhu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Qin Peng
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
| | - Longwei Liu
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
| | - Chau Ly
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Pak Kin Wong
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Yingxiao Wang
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
| | - Amy C Rowat
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Department of Integrative Biology & Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Siavash K Kurdistani
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Song Li
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
89
|
La Torre M, Merigliano C, Maccaroni K, Chojnowski A, Goh WI, Giubettini M, Vernì F, Capanni C, Rhodes D, Wright G, Burke B, Soddu S, Burla R, Saggio I. Combined alteration of lamin and nuclear morphology influences the localization of the tumor-associated factor AKTIP. J Exp Clin Cancer Res 2022; 41:273. [PMID: 36096808 PMCID: PMC9469526 DOI: 10.1186/s13046-022-02480-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022] Open
Abstract
Background Lamins, key nuclear lamina components, have been proposed as candidate risk biomarkers in different types of cancer but their accuracy is still debated. AKTIP is a telomeric protein with the property of being enriched at the nuclear lamina. AKTIP has similarity with the tumor susceptibility gene TSG101. AKTIP deficiency generates genome instability and, in p53−/− mice, the reduction of the mouse counterpart of AKTIP induces the exacerbation of lymphomas. Here, we asked whether the distribution of AKTIP is altered in cancer cells and whether this is associated with alterations of lamins. Methods We performed super-resolution imaging, quantification of lamin expression and nuclear morphology on HeLa, MCF7, and A549 tumor cells, and on non-transformed fibroblasts from healthy donor and HGPS (LMNA c.1824C > T p.Gly608Gly) and EDMD2 (LMNA c.775 T > G) patients. As proof of principle model combining a defined lamin alteration with a tumor cell setting, we produced HeLa cells exogenously expressing the HGPS lamin mutant progerin that alters nuclear morphology. Results In HeLa cells, AKTIP locates at less than 0.5 µm from the nuclear rim and co-localizes with lamin A/C. As compared to HeLa, there is a reduced co-localization of AKTIP with lamin A/C in both MCF7 and A549. Additionally, MCF7 display lower amounts of AKTIP at the rim. The analyses in non-transformed fibroblasts show that AKTIP mislocalizes in HGPS cells but not in EDMD2. The integrated analysis of lamin expression, nuclear morphology, and AKTIP topology shows that positioning of AKTIP is influenced not only by lamin expression, but also by nuclear morphology. This conclusion is validated by progerin-expressing HeLa cells in which nuclei are morphologically altered and AKTIP is mislocalized. Conclusions Our data show that the combined alteration of lamin and nuclear morphology influences the localization of the tumor-associated factor AKTIP. The results also point to the fact that lamin alterations per se are not predictive of AKTIP mislocalization, in both non-transformed and tumor cells. In more general terms, this study supports the thesis that a combined analytical approach should be preferred to predict lamin-associated changes in tumor cells. This paves the way of next translational evaluation to validate the use of this combined analytical approach as risk biomarker. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02480-5.
Collapse
|
90
|
Han T, Jiang Y, Wang X, Deng S, Hu Y, Jin Q, Long D, Liu K. 3D matrix promotes cell dedifferentiation into colorectal cancer stem cells via integrin/cytoskeleton/glycolysis signaling. Cancer Sci 2022; 113:3826-3837. [PMID: 36052705 DOI: 10.1111/cas.15548] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/12/2022] [Accepted: 07/20/2022] [Indexed: 11/29/2022] Open
Abstract
The potential for tumor occurrence triggered by cancer stem cells (CSCs) has emerged as a significant challenge for human colorectal cancer therapy. However, the underlying mechanism of CSC development remains controversial. Our study provided evidence that the bulk of tumor cells could dedifferentiate to CSCs and reacquire CSCs-like phenotypes if cultured in the presence of extracellular matrix reagents, such as Matrigel and fibrin gels. In these 3D gels, CD133- colorectal cancer cells can regain tumorigenic potential and stem-like phenotypes. Mechanistically, the 3D extracellular matrix could mediate cytoskeletal F-actin bundling through biomechanical force associated receptors integrin β1 (ITGB1), contributing to the release of E3 ligase Tripartite motif protein 11 (TRIM11) from cytoskeleton and degradation of the glycolytic rate-limiting enzyme phosphofructokinase (PFK). Consequently, PFK inhibition resulted in enhanced glycolysis and upregulation of hypoxia-inducible factor 1 (HIF1α), thereby promoting the reprogramming of stem cell transcription factors and facilitating tumor progression in patients. This study provided novel insights into the role of the extracellular matrix in the regulation of CSC dedifferentiation in a cytoskeleton/glycolysis-dependent manner.
Collapse
Affiliation(s)
- Tong Han
- Department of General Surgery, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Yuhong Jiang
- Department of General Surgery, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaobo Wang
- Department of General Surgery, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuangya Deng
- Department of General Surgery, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Yongjun Hu
- Department of General Surgery, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Qianqian Jin
- Department of General Surgery, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Dongju Long
- Department of General Surgery, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Kuijie Liu
- Department of General Surgery, the Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
91
|
Li Y, Wong IY, Guo M. Reciprocity of Cell Mechanics with Extracellular Stimuli: Emerging Opportunities for Translational Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107305. [PMID: 35319155 PMCID: PMC9463119 DOI: 10.1002/smll.202107305] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/20/2022] [Indexed: 06/14/2023]
Abstract
Human cells encounter dynamic mechanical cues in healthy and diseased tissues, which regulate their molecular and biophysical phenotype, including intracellular mechanics as well as force generation. Recent developments in bio/nanomaterials and microfluidics permit exquisitely sensitive measurements of cell mechanics, as well as spatiotemporal control over external mechanical stimuli to regulate cell behavior. In this review, the mechanobiology of cells interacting bidirectionally with their surrounding microenvironment, and the potential relevance for translational medicine are considered. Key fundamental concepts underlying the mechanics of living cells as well as the extracelluar matrix are first introduced. Then the authors consider case studies based on 1) microfluidic measurements of nonadherent cell deformability, 2) cell migration on micro/nano-topographies, 3) traction measurements of cells in three-dimensional (3D) matrix, 4) mechanical programming of organoid morphogenesis, as well as 5) active mechanical stimuli for potential therapeutics. These examples highlight the promise of disease diagnosis using mechanical measurements, a systems-level understanding linking molecular with biophysical phenotype, as well as therapies based on mechanical perturbations. This review concludes with a critical discussion of these emerging technologies and future directions at the interface of engineering, biology, and medicine.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, Hubei, 430074, China
| | - Ian Y Wong
- School of Engineering, Center for Biomedical Engineering, Joint Program in Cancer Biology, Brown University, 184 Hope St Box D, Providence, RI, 02912, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
92
|
Bell ES, Shah P, Zuela-Sopilniak N, Kim D, Varlet AA, Morival JL, McGregor AL, Isermann P, Davidson PM, Elacqua JJ, Lakins JN, Vahdat L, Weaver VM, Smolka MB, Span PN, Lammerding J. Low lamin A levels enhance confined cell migration and metastatic capacity in breast cancer. Oncogene 2022; 41:4211-4230. [PMID: 35896617 PMCID: PMC9925375 DOI: 10.1038/s41388-022-02420-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 02/07/2023]
Abstract
Aberrations in nuclear size and shape are commonly used to identify cancerous tissue. However, it remains unclear whether the disturbed nuclear structure directly contributes to the cancer pathology or is merely a consequence of other events occurring during tumorigenesis. Here, we show that highly invasive and proliferative breast cancer cells frequently exhibit Akt-driven lower expression of the nuclear envelope proteins lamin A/C, leading to increased nuclear deformability that permits enhanced cell migration through confined environments that mimic interstitial spaces encountered during metastasis. Importantly, increasing lamin A/C expression in highly invasive breast cancer cells reflected gene expression changes characteristic of human breast tumors with higher LMNA expression, and specifically affected pathways related to cell-ECM interactions, cell metabolism, and PI3K/Akt signaling. Further supporting an important role of lamins in breast cancer metastasis, analysis of lamin levels in human breast tumors revealed a significant association between lower lamin A levels, Akt signaling, and decreased disease-free survival. These findings suggest that downregulation of lamin A/C in breast cancer cells may influence both cellular physical properties and biochemical signaling to promote metastatic progression.
Collapse
Affiliation(s)
- Emily S. Bell
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY,Current address: Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA
| | - Pragya Shah
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | | | - Dongsung Kim
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Alice-Anais Varlet
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Julien L.P. Morival
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Alexandra L. McGregor
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Philipp Isermann
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | | | - Joshua J. Elacqua
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Jonathan N. Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Linda Vahdat
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA,Helen Diller Cancer Center, Department of Bioengineering and Therapeutic Sciences, and Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA
| | - Marcus B. Smolka
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Paul N. Span
- Department of Radiation Oncology, Radiotherapy & OncoImmunology laboratory, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA. .,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
93
|
Kalukula Y, Stephens AD, Lammerding J, Gabriele S. Mechanics and functional consequences of nuclear deformations. Nat Rev Mol Cell Biol 2022; 23:583-602. [PMID: 35513718 PMCID: PMC9902167 DOI: 10.1038/s41580-022-00480-z] [Citation(s) in RCA: 201] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2022] [Indexed: 02/08/2023]
Abstract
As the home of cellular genetic information, the nucleus has a critical role in determining cell fate and function in response to various signals and stimuli. In addition to biochemical inputs, the nucleus is constantly exposed to intrinsic and extrinsic mechanical forces that trigger dynamic changes in nuclear structure and morphology. Emerging data suggest that the physical deformation of the nucleus modulates many cellular and nuclear functions. These functions have long been considered to be downstream of cytoplasmic signalling pathways and dictated by gene expression. In this Review, we discuss an emerging perspective on the mechanoregulation of the nucleus that considers the physical connections from chromatin to nuclear lamina and cytoskeletal filaments as a single mechanical unit. We describe key mechanisms of nuclear deformations in time and space and provide a critical review of the structural and functional adaptive responses of the nucleus to deformations. We then consider the contribution of nuclear deformations to the regulation of important cellular functions, including muscle contraction, cell migration and human disease pathogenesis. Collectively, these emerging insights shed new light on the dynamics of nuclear deformations and their roles in cellular mechanobiology.
Collapse
Affiliation(s)
- Yohalie Kalukula
- University of Mons, Soft Matter and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, CIRMAP, Place du Parc, 20 B-7000 Mons, Belgium
| | - Andrew D. Stephens
- Biology Department, University of Massachusetts Amherst, Amherst, MA, USA
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Sylvain Gabriele
- University of Mons, Soft Matter and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, CIRMAP, Place du Parc, 20 B-7000 Mons, Belgium
| |
Collapse
|
94
|
Catarinella G, Nicoletti C, Bracaglia A, Procopio P, Salvatori I, Taggi M, Valle C, Ferri A, Canipari R, Puri PL, Latella L. SerpinE1 drives a cell-autonomous pathogenic signaling in Hutchinson-Gilford progeria syndrome. Cell Death Dis 2022; 13:737. [PMID: 36028501 PMCID: PMC9418244 DOI: 10.1038/s41419-022-05168-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 01/21/2023]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare, fatal disease caused by Lamin A mutation, leading to altered nuclear architecture, loss of peripheral heterochromatin and deregulated gene expression. HGPS patients eventually die by coronary artery disease and cardiovascular alterations. Yet, how deregulated transcriptional networks at the cellular level impact on the systemic disease phenotype is currently unclear. A genome-wide analysis of gene expression in cultures of primary HGPS fibroblasts identified SerpinE1, also known as Plasminogen Activator Inhibitor (PAI-1), as central gene that propels a cell-autonomous pathogenic signaling from the altered nuclear lamina. Indeed, siRNA-mediated downregulation and pharmacological inhibition of SerpinE1 by TM5441 could revert key pathological features of HGPS in patient-derived fibroblasts, including re-activation of cell cycle progression, reduced DNA damage signaling, decreased expression of pro-fibrotic genes and recovery of mitochondrial defects. These effects were accompanied by the correction of nuclear abnormalities. These data point to SerpinE1 as a novel potential effector and target for therapeutic interventions in HGPS pathogenesis.
Collapse
Affiliation(s)
| | - Chiara Nicoletti
- grid.479509.60000 0001 0163 8573Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA
| | - Andrea Bracaglia
- grid.417778.a0000 0001 0692 3437IRCCS Fondazione Santa Lucia, Rome, Italy ,grid.6530.00000 0001 2300 0941PhD Program in Cellular and Molecular Biology, Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Paola Procopio
- grid.417778.a0000 0001 0692 3437IRCCS Fondazione Santa Lucia, Rome, Italy ,grid.10253.350000 0004 1936 9756Present Address: BPC, Pharmakologisches Institut, Philipps-Universität Marburg, Marburg, Germany
| | - Illari Salvatori
- grid.417778.a0000 0001 0692 3437IRCCS Fondazione Santa Lucia, Rome, Italy ,grid.7841.aDepartment of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Marilena Taggi
- grid.7841.aDAHFMO, Unit of Histology and Medical Embryology, Sapienza, University of Rome, Rome, Italy
| | - Cristiana Valle
- grid.417778.a0000 0001 0692 3437IRCCS Fondazione Santa Lucia, Rome, Italy ,grid.5326.20000 0001 1940 4177Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| | - Alberto Ferri
- grid.417778.a0000 0001 0692 3437IRCCS Fondazione Santa Lucia, Rome, Italy ,grid.5326.20000 0001 1940 4177Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| | - Rita Canipari
- grid.7841.aDAHFMO, Unit of Histology and Medical Embryology, Sapienza, University of Rome, Rome, Italy
| | - Pier Lorenzo Puri
- grid.479509.60000 0001 0163 8573Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA
| | - Lucia Latella
- grid.417778.a0000 0001 0692 3437IRCCS Fondazione Santa Lucia, Rome, Italy ,grid.5326.20000 0001 1940 4177Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| |
Collapse
|
95
|
Leong EL, Khaing NT, Cadot B, Hong WL, Kozlov S, Werner H, Wong ESM, Stewart CL, Burke B, Lee YL. Nesprin-1 LINC complexes recruit microtubule cytoskeleton proteins and drive pathology in Lmna-mutant striated muscle. Hum Mol Genet 2022; 32:177-191. [PMID: 35925868 PMCID: PMC9840208 DOI: 10.1093/hmg/ddac179] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 01/19/2023] Open
Abstract
Mutations in LMNA, the gene encoding A-type lamins, cause laminopathies-diseases of striated muscle and other tissues. The aetiology of laminopathies has been attributed to perturbation of chromatin organization or structural weakening of the nuclear envelope (NE) such that the nucleus becomes more prone to mechanical damage. The latter model requires a conduit for force transmission to the nucleus. NE-associated Linker of Nucleoskeleton and Cytoskeleton (LINC) complexes are one such pathway. Using clustered regularly interspaced short palindromic repeats to disrupt the Nesprin-1 KASH (Klarsicht, ANC-1, Syne Homology) domain, we identified this LINC complex protein as the predominant NE anchor for microtubule cytoskeleton components, including nucleation activities and motor complexes, in mouse cardiomyocytes. Loss of Nesprin-1 LINC complexes resulted in loss of microtubule cytoskeleton proteins at the nucleus and changes in nuclear morphology and positioning in striated muscle cells, but with no overt physiological defects. Disrupting the KASH domain of Nesprin-1 suppresses Lmna-linked cardiac pathology, likely by reducing microtubule cytoskeleton activities at the nucleus. Nesprin-1 LINC complexes thus represent a potential therapeutic target for striated muscle laminopathies.
Collapse
Affiliation(s)
| | | | - Bruno Cadot
- Sorbonne Université, INSERM U974, Institut de Myologie, GH Pitié Salpêtrière, 47 Boulevard de l’Hôpital, Paris 75013, France
| | - Wei Liang Hong
- Institute of Medical Biology, Agency for Science Technology and Research (ASTAR), 8A Biomedical Grove, Level 6 Immunos, Singapore 138648, Singapore,ASTAR Skin Research Labs (ASRL), Agency for Science Technology and Research (ASTAR), 8A Biomedical Grove, Level 6 Immunos, Singapore 138648, Singapore
| | - Serguei Kozlov
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Hendrikje Werner
- Institute of Medical Biology, Agency for Science Technology and Research (ASTAR), 8A Biomedical Grove, Level 6 Immunos, Singapore 138648, Singapore,ASTAR Skin Research Labs (ASRL), Agency for Science Technology and Research (ASTAR), 8A Biomedical Grove, Level 6 Immunos, Singapore 138648, Singapore
| | - Esther Sook Miin Wong
- Institute of Medical Biology, Agency for Science Technology and Research (ASTAR), 8A Biomedical Grove, Level 6 Immunos, Singapore 138648, Singapore,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Level 5 Immunos, Singapore 138648, Singapore
| | - Colin L Stewart
- To whom correspondence should be addressed. Colin L. Stewart, ; Brian Burke, ; Yin Loon Lee,
| | - Brian Burke
- To whom correspondence should be addressed. Colin L. Stewart, ; Brian Burke, ; Yin Loon Lee,
| | - Yin Loon Lee
- To whom correspondence should be addressed. Colin L. Stewart, ; Brian Burke, ; Yin Loon Lee,
| |
Collapse
|
96
|
Dooling LJ, Saini K, Anlaş AA, Discher DE. Tissue mechanics coevolves with fibrillar matrisomes in healthy and fibrotic tissues. Matrix Biol 2022; 111:153-188. [PMID: 35764212 PMCID: PMC9990088 DOI: 10.1016/j.matbio.2022.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/16/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022]
Abstract
Fibrillar proteins are principal components of extracellular matrix (ECM) that confer mechanical properties to tissues. Fibrosis can result from wound repair in nearly every tissue in adults, and it associates with increased ECM density and crosslinking as well as increased tissue stiffness. Such fibrotic tissues are a major biomedical challenge, and an emerging view posits that the altered mechanical environment supports both synthetic and contractile myofibroblasts in a state of persistent activation. Here, we review the matrisome in several fibrotic diseases, as well as normal tissues, with a focus on physicochemical properties. Stiffness generally increases with the abundance of fibrillar collagens, the major constituent of ECM, with similar mathematical trends for fibrosis as well as adult tissues from soft brain to stiff bone and heart development. Changes in expression of other core matrisome and matrisome-associated proteins or proteoglycans contribute to tissue stiffening in fibrosis by organizing collagen, crosslinking ECM, and facilitating adhesion of myofibroblasts. Understanding how ECM composition and mechanics coevolve during fibrosis can lead to better models and help with antifibrotic therapies.
Collapse
Affiliation(s)
- Lawrence J Dooling
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Karanvir Saini
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Alişya A Anlaş
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Dennis E Discher
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA.
| |
Collapse
|
97
|
Chen M, Ma Y, Chang W. SARS-CoV-2 and the Nucleus. Int J Biol Sci 2022; 18:4731-4743. [PMID: 35874947 PMCID: PMC9305274 DOI: 10.7150/ijbs.72482] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/20/2022] [Indexed: 11/22/2022] Open
Abstract
The ongoing COVID-19 pandemic is caused by an RNA virus, SARS-CoV-2. The genome of SARS-CoV-2 lacks a nuclear phase in its life cycle and is replicated in the cytoplasm. However, interfering with nuclear trafficking using pharmacological inhibitors greatly reduces virus infection and virus replication of other coronaviruses is blocked in enucleated cells, suggesting a critical role of the nucleus in virus infection. Here, we summarize the alternations of nuclear pathways caused by SARS-CoV-2, including nuclear translocation pathways, innate immune responses, mRNA metabolism, epigenetic mechanisms, DNA damage response, cytoskeleton regulation, and nuclear rupture. We consider how these alternations contribute to virus replication and discuss therapeutic treatments that target these pathways, focusing on small molecule drugs that are being used in clinical studies.
Collapse
Affiliation(s)
- Mengqi Chen
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Yue Ma
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wakam Chang
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
98
|
Molecular genetic mechanisms of dilated cardiomyopathy. Curr Opin Genet Dev 2022; 76:101959. [PMID: 35870234 DOI: 10.1016/j.gde.2022.101959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/20/2022]
Abstract
Heart failure (HF) is a rapidly growing cardiovascular condition with a prevalence of ~40 million individuals worldwide [1]. While HF can be caused by acquired conditions such as myocardial infarctions and viruses [2], the genetic basis for HF is rapidly emerging particularly for dilated cardiomyopathy (DCM) that is the most prevalent HF type. In this review, insights from the rapid expansion in next-generation sequencing technologies applied in the HF clinic are merged with recent functional genomics studies to provide a contemporary view of DCM molecular genetics.
Collapse
|
99
|
Yang Q, Ciebiera M, Bariani MV, Ali M, Elkafas H, Boyer TG, Al-Hendy A. Comprehensive Review of Uterine Fibroids: Developmental Origin, Pathogenesis, and Treatment. Endocr Rev 2022; 43:678-719. [PMID: 34741454 PMCID: PMC9277653 DOI: 10.1210/endrev/bnab039] [Citation(s) in RCA: 158] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Indexed: 11/24/2022]
Abstract
Uterine fibroids are benign monoclonal neoplasms of the myometrium, representing the most common tumors in women worldwide. To date, no long-term or noninvasive treatment option exists for hormone-dependent uterine fibroids, due to the limited knowledge about the molecular mechanisms underlying the initiation and development of uterine fibroids. This paper comprehensively summarizes the recent research advances on uterine fibroids, focusing on risk factors, development origin, pathogenetic mechanisms, and treatment options. Additionally, we describe the current treatment interventions for uterine fibroids. Finally, future perspectives on uterine fibroids studies are summarized. Deeper mechanistic insights into tumor etiology and the complexity of uterine fibroids can contribute to the progress of newer targeted therapies.
Collapse
Affiliation(s)
- Qiwei Yang
- Qiwei Yang, Ph.D. Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave, M167, Billings, Chicago, IL 60637, USA.
| | - Michal Ciebiera
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, ul. Cegłowska 80, 01-809, Warsaw, Poland
| | | | - Mohamed Ali
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Hoda Elkafas
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Pharmacology and Toxicology, Egyptian Drug Authority, formerly National Organization for Drug Control and Research, Cairo 35521, Egypt
| | - Thomas G Boyer
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Ayman Al-Hendy
- Correspondence: Ayman Al-Hendy, MD, Ph.D. Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave, N112, Peck Pavilion, Chicago, IL 60637. USA.
| |
Collapse
|
100
|
Sengupta D, Sengupta K. Lamin A and telomere maintenance in aging: Two to Tango. Mutat Res 2022; 825:111788. [PMID: 35687934 DOI: 10.1016/j.mrfmmm.2022.111788] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 03/28/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
Lamin proteins which constitute the nuclear lamina in almost all higher eukaryotes, are mainly of two types A & B encoded by LMNA and LMNB1/B2 genes respectively. While lamin A remains the principal product of LMNA gene, variants like lamin C, C2 and A∆10 are also formed as alternate splice products. Role of lamin A in aging has been highlighted in recent times due to its association with progeroid or premature aging syndromes which is classified as a type of laminopathy. Progeria caused by accelerated accumulation of lamin A Δ50 or progerin occurs due to a mutation in this LMNA gene leading to defects in post translational modification of lamin A. One of the most common and severe symptoms of progeroid laminopathy is accelerated cellular senescence or aging along with bone resorption, muscle weakness, lipodystrophy and cardiovascular disorders. On the other hand, progerin accumulation and telomere dysfunction merge as common traits in the process of chronological aging. Two major hallmarks of physiological aging in humans include loss of genomic integrity and telomere attrition which can result from defective laminar organization leading to deformed nuclear architecture and culminates into replicative senescence. This also adversely affects epigenetic landscape, mitochondrial dysfunction and several signalling pathways like DNA repair, mTOR, MAPK, TGFβ. In this review, we discuss the telomere-lamina interplay in the context of physiological aging and progeria.
Collapse
Affiliation(s)
- Duhita Sengupta
- Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, West Bengal, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Kaushik Sengupta
- Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, West Bengal, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|