1
|
Nguyen TD, Winek MA, Rao MK, Dhyani SP, Lee MY. Nuclear envelope components in vascular mechanotransduction: emerging roles in vascular health and disease. Nucleus 2025; 16:2453752. [PMID: 39827403 DOI: 10.1080/19491034.2025.2453752] [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: 10/08/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
The vascular network, uniquely sensitive to mechanical changes, translates biophysical forces into biochemical signals for vessel function. This process relies on the cell's architectural integrity, enabling uniform responses to physical stimuli. Recently, the nuclear envelope (NE) has emerged as a key regulator of vascular cell function. Studies implicate nucleoskeletal elements (e.g. nuclear lamina) and the linker of nucleoskeleton and cytoskeleton (LINC) complex in force transmission, emphasizing nucleo-cytoskeletal communication in mechanotransduction. The nuclear pore complex (NPC) and its component proteins (i.e. nucleoporins) also play roles in cardiovascular disease (CVD) progression. We herein summarize evidence on the roles of nuclear lamina proteins, LINC complex members, and nucleoporins in endothelial and vascular cell mechanotransduction. Numerous studies attribute NE components in cytoskeletal-related cellular behaviors to insinuate dysregulation of nucleocytoskeletal feedback and nucleocytoplasmic transport as a mechanism of endothelial and vascular dysfunction, and hence implications for aging and vascular pathophysiology.
Collapse
Affiliation(s)
- Tung D Nguyen
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
- The Center for Cardiovascular Research, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Michael A Winek
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Mihir K Rao
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Shaiva P Dhyani
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Monica Y Lee
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
- The Center for Cardiovascular Research, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Park JE, Jo J, Xu K, Lee SA, Han SB, Lee Y, Cho WK, Li B, Kim SH, Kim DH. Attenuated Nuclear Tension Regulates Progerin-Induced Mechanosensitive Nuclear Wrinkling and Chromatin Remodeling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2502375. [PMID: 40344643 DOI: 10.1002/advs.202502375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/17/2025] [Indexed: 05/11/2025]
Abstract
Hutchinson-Gilford progeria syndrome, caused by a mutation in the LMNA gene, leads to increased levels of truncated prelamin A, progerin, in the nuclear membrane. The accumulation of progerin results in defective nuclear morphology and is associated with altered expression of linker of the nucleoskeleton and cytoskeleton complex proteins, which are critical for nuclear signal transduction via molecular coupling between the extranuclear cytoskeleton and lamin-associated nuclear envelope. However, the molecular mechanisms underlying progerin accumulation-induced nuclear deformation and its effects on intranuclear chromosomal organization remain unclear. Here, the spatiotemporal evolution of nuclear wrinkles is analyzed in response to variations in substrate stiffness using a doxycycline-inducible progerin expression system. It is found that cytoskeletal tension regulates the onset of progerin-induced nuclear envelope wrinkling and that the molecular interaction between SUN1 and LMNA controls the actomyosin-dependent attenuation of nuclear tension. Genome-wide analysis of chromatin accessibility and gene expression further suggests that an imbalance in force between the intra- and extranuclear spaces induces nuclear deformation, which specifically regulates progeria-associated gene expression via modification of mechanosensitive signaling pathways. The findings highlight the crucial role of nuclear lamin-cytoskeletal connectivity in bridging nuclear mechanotransduction and the biological aging process.
Collapse
Affiliation(s)
- Ji-Eun Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Juhyeon Jo
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Kun Xu
- Department of Engineering Mechanics, Tsinghua University, Beijing, 100084, China
| | - Sun-Ah Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Seong-Beom Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - YigJi Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea
| | - Won-Ki Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea
| | - Bo Li
- Department of Engineering Mechanics, Tsinghua University, Beijing, 100084, China
| | - Soo Hyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
- Department of Integrative Energy Engineering, College of Engineering, Korea University, Seoul, 02841, South Korea
| |
Collapse
|
3
|
Paganelli F, Poli A, Truocchio S, Martelli AM, Palumbo C, Lattanzi G, Chiarini F. At the nucleus of cancer: how the nuclear envelope controls tumor progression. MedComm (Beijing) 2025; 6:e70073. [PMID: 39866838 PMCID: PMC11758262 DOI: 10.1002/mco2.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 01/28/2025] Open
Abstract
Historically considered downstream effects of tumorigenesis-arising from changes in DNA content or chromatin organization-nuclear alterations have long been seen as mere prognostic markers within a genome-centric model of cancer. However, recent findings have placed the nuclear envelope (NE) at the forefront of tumor progression, highlighting its active role in mediating cellular responses to mechanical forces. Despite significant progress, the precise interplay between NE components and cancer progression remains under debate. In this review, we provide a comprehensive and up-to-date overview of how changes in NE composition affect nuclear mechanics and facilitate malignant transformation, grounded in the latest molecular and functional studies. We also review recent research that uses advanced technologies, including artificial intelligence, to predict malignancy risk and treatment outcomes by analyzing nuclear morphology. Finally, we discuss how progress in understanding nuclear mechanics has paved the way for mechanotherapy-a promising cancer treatment approach that exploits the mechanical differences between cancerous and healthy cells. Shifting the perspective on NE alterations from mere diagnostic markers to potential therapeutic targets, this review calls for further investigation into the evolving role of the NE in cancer, highlighting the potential for innovative strategies to transform conventional cancer therapies.
Collapse
Affiliation(s)
- Francesca Paganelli
- Department of Biomedical and Neuromotor SciencesAlma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Alessandro Poli
- IFOM ETS ‐ The AIRC Institute of Molecular OncologyMilanItaly
| | - Serena Truocchio
- Department of Biomedical and Neuromotor SciencesAlma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Alberto M. Martelli
- Department of Biomedical and Neuromotor SciencesAlma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Carla Palumbo
- Department of BiomedicalMetabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza”Unit of BolognaBolognaItaly
- IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Francesca Chiarini
- Department of BiomedicalMetabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| |
Collapse
|
4
|
Chen H, Yang J, Yang Q, Jia Y, Guo X. Protein prenylation in mechanotransduction: implications for disease and therapy. Trends Pharmacol Sci 2025; 46:163-179. [PMID: 39818521 DOI: 10.1016/j.tips.2024.12.008] [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: 11/06/2024] [Revised: 12/05/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
The process by which cells translate external mechanical cues into intracellular biochemical signals involves intricate mechanisms that remain unclear. In recent years, research into post-translational modifications (PTMs) has offered valuable insights into this field, spotlighting protein prenylation as a crucial mechanism in cellular mechanotransduction and various human diseases. Protein prenylation, which involves the covalent attachment of isoprenoid groups to specific substrate proteins, profoundly affects the functions of key mechanotransduction proteins such as Rho, Ras, and lamins. This review provides the first comprehensive examination of the connections between prenylation and mechanotransduction, exploring both the mechanistic details and its impact on mechanosensitive cellular behaviors. We further highlight recent evidence linking protein prenylation to diseases associated with disrupted mechanical homeostasis, and outline emerging targeted therapeutic strategies.
Collapse
Affiliation(s)
- Heng Chen
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Jian Yang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qingzhen Yang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yuanbo Jia
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P.R. China; Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an 710004, P.R. China; TFX Group-Xi'an Jiaotong University Institute of Life Health, Xi'an 710049, P.R. China.
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
5
|
Kono Y, Pack CG, Ichikawa T, Komatsubara A, Adam SA, Miyazawa K, Rolas L, Nourshargh S, Medalia O, Goldman RD, Fukuma T, Kimura H, Shimi T. Roles of the lamin A-specific tail region in the localization to sites of nuclear envelope rupture. PNAS NEXUS 2024; 3:pgae527. [PMID: 39677369 PMCID: PMC11645434 DOI: 10.1093/pnasnexus/pgae527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024]
Abstract
The nuclear lamina (NL) lines the nuclear envelope (NE) to maintain nuclear structure in metazoan cells. The major NL components, the nuclear lamins contribute to the protection against NE rupture induced by mechanical stress. Lamin A (LA) and a short form of the splicing variant lamin C (LC) are diffused from the nucleoplasm to sites of NE rupture in immortalized mouse embryonic fibroblasts (MEFs). LA localization to the rupture sites is significantly slow and weak compared with LC, but the underlying mechanism remains unknown. In this study, wild-type (WT), Hutchinson-Gilford Progeria syndrome (HGPS) knock-in MEFs expressing progerin (PG, an LA mutant lacking the second proteolytic cleavage site), and LA/C-knockout MEFs transiently and heterogeneously expressing LA/C WTs and mutants fused to mEmerald are examined before and after NE rupture induced by single-cell compression and laser microirradiation. The farnesylation at the CaaX motif of unprocessed LA and the inhibition of the second proteolytic cleavage decrease the nucleoplasmic pool and slow the localization to the rupture sites in a long-time window (60-70 min) after the induction of NE rupture. Our data could explain the defective repair of NE rupture in HGPS through the farnesylation at the CaaX motif of unprocessed progerin. In addition, unique segments in LA-specific tail region cooperate with each other to inhibit the rapid accumulation within a short-time window (3 min) that is also observed with LC.
Collapse
Affiliation(s)
- Yohei Kono
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
- Faculty of Frontier Engineering, Institute of Science and Engineering, Kanazawa University, Kanazawa 920-1192, Japan
| | - Chan-Gi Pack
- Convergence Medicine Research Center, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Korea
- Department of Biomedical Engineering, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Takehiko Ichikawa
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
| | - Arata Komatsubara
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Stephen A Adam
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611 USA
| | - Keisuke Miyazawa
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
- Faculty of Frontier Engineering, Institute of Science and Engineering, Kanazawa University, Kanazawa 920-1192, Japan
| | - Loïc Rolas
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Sussan Nourshargh
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Zurich 8057, Switzerland
| | - Robert D Goldman
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611 USA
| | - Takeshi Fukuma
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
- Faculty of Frontier Engineering, Institute of Science and Engineering, Kanazawa University, Kanazawa 920-1192, Japan
| | - Hiroshi Kimura
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Takeshi Shimi
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
| |
Collapse
|
6
|
Shores KL, Truskey GA. Mechanotransduction of the vasculature in Hutchinson-Gilford Progeria Syndrome. Front Physiol 2024; 15:1464678. [PMID: 39239311 PMCID: PMC11374724 DOI: 10.3389/fphys.2024.1464678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature aging disorder that causes severe cardiovascular disease, resulting in the death of patients in their teenage years. The disease pathology is caused by the accumulation of progerin, a mutated form of the nuclear lamina protein, lamin A. Progerin binds to the inner nuclear membrane, disrupting nuclear integrity, and causes severe nuclear abnormalities and changes in gene expression. This results in increased cellular inflammation, senescence, and overall dysfunction. The molecular mechanisms by which progerin induces the disease pathology are not fully understood. Progerin's detrimental impact on nuclear mechanics and the role of the nucleus as a mechanosensor suggests dysfunctional mechanotransduction could play a role in HGPS. This is especially relevant in cells exposed to dynamic, continuous mechanical stimuli, like those of the vasculature. The endothelial (ECs) and smooth muscle cells (SMCs) within arteries rely on physical forces produced by blood flow to maintain function and homeostasis. Certain regions within arteries produce disturbed flow, leading to an impaired transduction of mechanical signals, and a reduction in cellular function, which also occurs in HGPS. In this review, we discuss the mechanics of nuclear mechanotransduction, how this is disrupted in HGPS, and what effect this has on cell health and function. We also address healthy responses of ECs and SMCs to physiological mechanical stimuli and how these responses are impaired by progerin accumulation.
Collapse
Affiliation(s)
- Kevin L Shores
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
7
|
Turkmen AM, Saik NO, Ullman KS. The dynamic nuclear envelope: resilience in health and dysfunction in disease. Curr Opin Cell Biol 2023; 85:102230. [PMID: 37660480 PMCID: PMC10843620 DOI: 10.1016/j.ceb.2023.102230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023]
Abstract
The canonical appearance of the nucleus depends on constant adaptation and remodeling of the nuclear envelope in response to changing biomechanical forces and metabolic demands. Dynamic events at the nuclear envelope play a vital role in supporting key nuclear functions as well as conferring plasticity to this organelle. Moreover, imbalance of these dynamic processes is emerging as a central feature of disease etiology. This review focuses on recent advances that shed light on the myriad events at the nuclear envelope that contribute to resilience and flexibility in nuclear architecture.
Collapse
Affiliation(s)
- Ayse M Turkmen
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Natasha O Saik
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Katharine S Ullman
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
8
|
Jebane C, Varlet AA, Karnat M, Hernandez- Cedillo LM, Lecchi A, Bedu F, Desgrouas C, Vigouroux C, Vantyghem MC, Viallat A, Rupprecht JF, Helfer E, Badens C. Enhanced cell viscosity: A new phenotype associated with lamin A/C alterations. iScience 2023; 26:107714. [PMID: 37701573 PMCID: PMC10494210 DOI: 10.1016/j.isci.2023.107714] [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: 05/03/2023] [Revised: 07/13/2023] [Accepted: 08/22/2023] [Indexed: 09/14/2023] Open
Abstract
Lamin A/C is a well-established key contributor to nuclear stiffness and its role in nucleus mechanical properties has been extensively studied. However, its impact on whole-cell mechanics has been poorly addressed, particularly concerning measurable physical parameters. In this study, we combined microfluidic experiments with theoretical analyses to quantitatively estimate the whole-cell mechanical properties. This allowed us to characterize the mechanical changes induced in cells by lamin A/C alterations and prelamin A accumulation resulting from atazanavir treatment or lipodystrophy-associated LMNA R482W pathogenic variant. Our results reveal a distinctive increase in long-time viscosity as a signature of cells affected by lamin A/C alterations. Furthermore, they show that the whole-cell response to mechanical stress is driven not only by the nucleus but also by the nucleo-cytoskeleton links and the microtubule network. The enhanced cell viscosity assessed with our microfluidic assay could serve as a valuable diagnosis marker for lamin-related diseases.
Collapse
Affiliation(s)
- Cécile Jebane
- Aix Marseille Univ, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| | | | - Marc Karnat
- Aix Marseille Univ, Université de Toulon, CNRS, CPT, Turing Centre for Living Systems, Marseille, France
| | | | | | | | | | - Corinne Vigouroux
- Assistance Publique–Hôpitaux de Paris (AP-HP), Saint-Antoine Hospital, National Reference Centre for Rares diseases of Insulin-Secretion and Insulin-Sensitivity (PRISIS), Department of Endocrinology, Paris, France
- Sorbonne University, Saint-Antoine Research Centre, Inserm UMR_S938, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Marie-Christine Vantyghem
- Endocrinology, Diabetology and Metabolism Department, Inserm U1190, EGID, Lille University Hospital, Lille, France
| | - Annie Viallat
- Aix Marseille Univ, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| | - Jean-François Rupprecht
- Aix Marseille Univ, Université de Toulon, CNRS, CPT, Turing Centre for Living Systems, Marseille, France
| | - Emmanuèle Helfer
- Aix Marseille Univ, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| | - Catherine Badens
- Aix Marseille Univ, INSERM, MMG, Marseille, France
- AP-HM, Laboratoire de Biochimie, Marseille, France
| |
Collapse
|
9
|
Walther BK, Sears AP, Mojiri A, Avazmohammadi R, Gu J, Chumakova OV, Pandian NKR, Dominic A, Martiel JL, Yazdani SK, Cooke JP, Ohayon J, Pettigrew RI. Disrupted Stiffness Ratio Alters Nuclear Mechanosensing. MATTER 2023; 6:3608-3630. [PMID: 37937235 PMCID: PMC10627551 DOI: 10.1016/j.matt.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
The ability of endothelial cells to sense and respond to dynamic changes in blood flow is critical for vascular homeostasis and cardiovascular health. The mechanical and geometric properties of the nuclear and cytoplasmic compartments affect mechanotransduction. We hypothesized that alterations to these parameters have resulting mechanosensory consequences. Using atomic force microscopy and mathematical modeling, we assessed how the nuclear and cytoplasmic compartment stiffnesses modulate shear stress transfer to the nucleus within aging endothelial cells. Our computational studies revealed that the critical parameter controlling shear transfer is not the individual mechanics of these compartments, but the stiffness ratio between them. Replicatively aged cells had a reduced stiffness ratio, attenuating shear transfer, while the ratio was not altered in a genetic model of accelerated aging. We provide a theoretical framework suggesting that dysregulation of the shear stress response can be uniquely imparted by relative mechanical changes in subcellular compartments.
Collapse
Affiliation(s)
- Brandon K. Walther
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
- Texas A&M University, Department of Biomedical Engineering, College Station, TX 77843, USA
| | - Adam P. Sears
- Texas A&M University, Department of Biomedical Engineering, College Station, TX 77843, USA
- Houston Methodist Hospital, Houston, TX 77030, USA
| | - Anahita Mojiri
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Reza Avazmohammadi
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
- Texas A&M University, Department of Biomedical Engineering, College Station, TX 77843, USA
- Texas A&M University, Department of Mechanical Engineering, College Station, TX 77843, USA
| | - Jianhua Gu
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Olga V. Chumakova
- University of Texas Health Science Center, Department of Integrative Biology and Pharmacology, Houston, TX 77030, USA
| | | | - Abishai Dominic
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | | | - Saami K. Yazdani
- Wake Forest University, Department of Engineering, Winston-Salem, NC 27101, USA
| | - John P. Cooke
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
- Texas A&M University, Department of Biomedical Engineering, College Station, TX 77843, USA
| | - Jacques Ohayon
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
- University Grenoble Alpes, CNRS, TIMC UMR 5525, 38000 Grenoble, France
- Savoie Mont-Blanc University, Polytech Annecy-Chambéry, 73376 Le Bourget du Lac, France
| | - Roderic I. Pettigrew
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
- Texas A&M University, Department of Biomedical Engineering, College Station, TX 77843, USA
- Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
10
|
Yue X, Cui J, Sun Z, Liu L, Li Y, Shao L, Feng Q, Wang Z, Hambright WS, Cui Y, Huard J, Mu Y, Mu X. Nuclear softening mediated by Sun2 suppression delays mechanical stress-induced cellular senescence. Cell Death Discov 2023; 9:167. [PMID: 37198162 DOI: 10.1038/s41420-023-01467-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/28/2023] [Accepted: 05/05/2023] [Indexed: 05/19/2023] Open
Abstract
Nuclear decoupling and softening are the main cellular mechanisms to resist mechanical stress-induced nuclear/DNA damage, however, its molecular mechanisms remain much unknown. Our recent study of Hutchinson-Gilford progeria syndrome (HGPS) disease revealed the role of nuclear membrane protein Sun2 in mediating nuclear damages and cellular senescence in progeria cells. However, the potential role of Sun2 in mechanical stress-induced nuclear damage and its correlation with nuclear decoupling and softening is still not clear. By applying cyclic mechanical stretch to mesenchymal stromal cells (MSCs) of WT and Zmpset24-/- mice (Z24-/-, a model for HGPS), we observed much increased nuclear damage in Z24-/- MSCs, which also featured elevated Sun2 expression, RhoA activation, F-actin polymerization and nuclear stiffness, indicating the compromised nuclear decoupling capacity. Suppression of Sun2 with siRNA effectively reduced nuclear/DNA damages caused by mechanical stretch, which was mediated by increased nuclear decoupling and softening, and consequently improved nuclear deformability. Our results reveal that Sun2 is greatly involved in mediating mechanical stress-induced nuclear damage by regulating nuclear mechanical properties, and Sun2 suppression can be a novel therapeutic target for treating progeria aging or aging-related diseases.
Collapse
Affiliation(s)
- Xianlin Yue
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jie Cui
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zewei Sun
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lei Liu
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ying Li
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Liwei Shao
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qi Feng
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ziyue Wang
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - William S Hambright
- Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO, USA
| | - Yan Cui
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Johnny Huard
- Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO, USA
| | - Yanling Mu
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Xiaodong Mu
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
11
|
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: 198] [Impact Index Per Article: 66.0] [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
|
12
|
Ueda N, Maekawa M, Matsui TS, Deguchi S, Takata T, Katahira J, Higashiyama S, Hieda M. Inner Nuclear Membrane Protein, SUN1, is Required for Cytoskeletal Force Generation and Focal Adhesion Maturation. Front Cell Dev Biol 2022; 10:885859. [PMID: 35663386 PMCID: PMC9157646 DOI: 10.3389/fcell.2022.885859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/02/2022] [Indexed: 12/20/2022] Open
Abstract
The linker of nucleoskeleton and cytoskeleton (LINC) complex is composed of the inner nuclear membrane-spanning SUN proteins and the outer nuclear membrane-spanning nesprin proteins. The LINC complex physically connects the nucleus and plasma membrane via the actin cytoskeleton to perform diverse functions including mechanotransduction from the extracellular environment to the nucleus. Mammalian somatic cells express two principal SUN proteins, namely SUN1 and SUN2. We have previously reported that SUN1, but not SUN2, is essential for directional cell migration; however, the underlying mechanism remains elusive. Because the balance between adhesive force and traction force is critical for cell migration, in the present study, we focused on focal adhesions (FAs) and the actin cytoskeleton. We observed that siRNA-mediated SUN1 depletion did not affect the recruitment of integrin β1, one of the ubiquitously expressed focal adhesion molecules, to the plasma membrane. Consistently, SUN1-depleted cells normally adhered to extracellular matrix proteins, including collagen, fibronectin, laminin, and vitronectin. In contrast, SUN1 depletion reduced the activation of integrin β1. Strikingly, the depletion of SUN1 interfered with the incorporation of vinculin into the focal adhesions, whereas no significant differences in the expression of vinculin were observed between wild-type and SUN1-depleted cells. In addition, SUN1 depletion suppressed the recruitment of zyxin to nascent focal adhesions. These data indicate that SUN1 is involved in the maturation of focal adhesions. Moreover, disruption of the SUN1-containing LINC complex abrogates the actin cytoskeleton and generation of intracellular traction force, despite the presence of SUN2. Thus, a physical link between the nucleus and cytoskeleton through SUN1 is required for the proper organization of actin, thereby suppressing the incorporation of vinculin and zyxin into focal adhesions and the activation of integrin β1, both of which are dependent on traction force. This study provides insights into a previously unappreciated signaling pathway from the nucleus to the cytoskeleton, which is in the opposite direction to the well-known mechanotransduction pathways from the extracellular matrix to the nucleus.
Collapse
Affiliation(s)
- Nanami Ueda
- Department of Medical Technology, Ehime Prefectural University of Health Sciences, Tobe, Japan
| | - Masashi Maekawa
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Matsuyama, Japan
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | | | - Shinji Deguchi
- Division of Bioengineering, Osaka University, Toyonaka, Japan
| | - Tomoyo Takata
- Department of Medical Technology, Ehime Prefectural University of Health Sciences, Tobe, Japan
| | - Jun Katahira
- Department of Veterinary Sciences, Osaka Prefecture University, Sakai, Japan
| | - Shigeki Higashiyama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Matsuyama, Japan
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Japan
- Department of Oncogenesis and Growth Regulation, Osaka International Cancer Institute, Osaka, Japan
| | - Miki Hieda
- Department of Medical Technology, Ehime Prefectural University of Health Sciences, Tobe, Japan
- *Correspondence: Miki Hieda,
| |
Collapse
|
13
|
Razin SV, Ulianov SV. Genome-Directed Cell Nucleus Assembly. BIOLOGY 2022; 11:biology11050708. [PMID: 35625436 PMCID: PMC9138775 DOI: 10.3390/biology11050708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/02/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Speckles and other nuclear bodies, the nucleolus and perinucleolar zone, transcription/replication factories and the lamina-associated compartment, serve as a structural basis for various genomic functions. In turn, genome activity and specific chromatin 3D organization directly impact the integrity of intranuclear assemblies, initiating/facilitating their formation and dictating their composition. Thus, the large-scale nucleus structure and genome activity mutually influence each other. The cell nucleus is frequently considered a compartment in which the genome is placed to protect it from external forces. Here, we discuss the evidence demonstrating that the cell nucleus should be considered, rather, as structure built around the folded genome. Decondensing chromosomes provide a scaffold for the assembly of the nuclear envelope after mitosis, whereas genome activity directs the assembly of various nuclear compartments, including nucleolus, speckles and transcription factories. Abstract The cell nucleus is frequently considered a cage in which the genome is placed to protect it from various external factors. Inside the nucleus, many functional compartments have been identified that are directly or indirectly involved in implementing genomic DNA’s genetic functions. For many years, it was assumed that these compartments are assembled on a proteinaceous scaffold (nuclear matrix), which provides a structural milieu for nuclear compartmentalization and genome folding while simultaneously offering some rigidity to the cell nucleus. The results of research in recent years have made it possible to consider the cell nucleus from a different angle. From the “box” in which the genome is placed, the nucleus has become a kind of mobile exoskeleton, which is formed around the packaged genome, under the influence of transcription and other processes directly related to the genome activity. In this review, we summarize the main arguments in favor of this point of view by analyzing the mechanisms that mediate cell nucleus assembly and support its resistance to mechanical stresses.
Collapse
Affiliation(s)
- Sergey V. Razin
- Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
- Correspondence: or
| | - Sergey V. Ulianov
- Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
14
|
Vahabikashi A, Adam SA, Medalia O, Goldman RD. Nuclear lamins: Structure and function in mechanobiology. APL Bioeng 2022; 6:011503. [PMID: 35146235 PMCID: PMC8810204 DOI: 10.1063/5.0082656] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
Nuclear lamins are type V intermediate filament proteins that polymerize into complex filamentous meshworks at the nuclear periphery and in less structured forms throughout the nucleoplasm. Lamins interact with a wide range of nuclear proteins and are involved in numerous nuclear and cellular functions. Within the nucleus, they play roles in chromatin organization and gene regulation, nuclear shape, size, and mechanics, and the organization and anchorage of nuclear pore complexes. At the whole cell level, they are involved in the organization of the cytoskeleton, cell motility, and mechanotransduction. The expression of different lamin isoforms has been associated with developmental progression, differentiation, and tissue-specific functions. Mutations in lamins and their binding proteins result in over 15 distinct human diseases, referred to as laminopathies. The laminopathies include muscular (e.g., Emery-Dreifuss muscular dystrophy and dilated cardiomyopathy), neurological (e.g., microcephaly), and metabolic (e.g., familial partial lipodystrophy) disorders as well as premature aging diseases (e.g., Hutchinson-Gilford Progeria and Werner syndromes). How lamins contribute to the etiology of laminopathies is still unknown. In this review article, we summarize major recent findings on the structure, organization, and multiple functions of lamins in nuclear and more global cellular processes.
Collapse
Affiliation(s)
- Amir Vahabikashi
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Stephen A. Adam
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Robert D. Goldman
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| |
Collapse
|
15
|
Danielsson BE, Peters HC, Bathula K, Spear LM, Noll NA, Dahl KN, Conway DE. Progerin-expressing endothelial cells are unable to adapt to shear stress. Biophys J 2022; 121:620-628. [PMID: 34999130 PMCID: PMC8873939 DOI: 10.1016/j.bpj.2022.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/17/2021] [Accepted: 01/05/2022] [Indexed: 11/19/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature aging disease caused by a single-point mutation in the lamin A gene, resulting in a truncated and farnesylated form of lamin A. This mutant lamin A protein, known as progerin, accumulates at the periphery of the nuclear lamina, resulting in both an abnormal nuclear morphology and nuclear stiffening. Patients with HGPS experience rapid onset of atherosclerosis, with death from heart attack or stroke as teenagers. Progerin expression has been shown to cause dysfunction in both vascular smooth muscle cells and endothelial cells (ECs). In this study, we examined how progerin-expressing endothelial cells adapt to fluid shear stress, the principal mechanical force from blood flow. We compared the response to shear stress for progerin-expressing, wild-type lamin A overexpressing, and control endothelial cells to physiological levels of fluid shear stress. Additionally, we also knocked down ZMPSTE24 in endothelial cells, which results in increased farnesylation of lamin A and similar phenotypes to HGPS. Our results showed that endothelial cells either overexpressing progerin or with ZMPSTE24 knockdown were unable to adapt to shear stress, experiencing significant cell loss at a longer duration of exposure to shear stress (3 days). Endothelial cells overexpressing wild-type lamin A also exhibited similar impairments in adaptation to shear stress, including similar levels of cell loss. Quantification of nuclear morphology showed that progerin-expressing endothelial cells had similar nuclear abnormalities in both static and shear conditions. Treatment of progerin-expressing cells and ZMPSTE24 KD cells with lonafarnib and methystat, drugs previously shown to improve HGPS nuclear morphology, resulted in improvements in adaptation to shear stress. Additionally, the prealignment of cells to shear stress before progerin-expression prevented cell loss. Our results demonstrate that changes in nuclear lamins can affect the ability of endothelial cells to properly adapt to shear stress.
Collapse
Affiliation(s)
- Brooke E Danielsson
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Hannah C Peters
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Kranthi Bathula
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Lindsay M Spear
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Natalie A Noll
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Kris N Dahl
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania; Department, Thornton Tomasetti, New York City, New York
| | - Daniel E Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
16
|
Transient nuclear lamin A/C accretion aids in recovery from vapor nanobubble-induced permeabilisation of the plasma membrane. Cell Mol Life Sci 2022; 79:23. [PMID: 34984553 PMCID: PMC8727414 DOI: 10.1007/s00018-021-04099-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/23/2021] [Accepted: 12/15/2021] [Indexed: 01/14/2023]
Abstract
Vapor nanobubble (VNB) photoporation is a physical method for intracellular delivery that has gained significant interest in the past decade. It has successfully been used to introduce molecular cargo of diverse nature into different cell types with high throughput and minimal cytotoxicity. For translational purposes, it is important to understand whether and how photoporation affects cell homeostasis. To obtain a comprehensive view on the transcriptional rewiring that takes place after VNB photoporation, we performed a longitudinal shotgun RNA-sequencing experiment. Six hours after photoporation, we found a marked upregulation of LMNA transcripts as well as their protein products, the A-type lamins. At the same time point, we observed a significant increase in several heterochromatin marks, suggesting a global stiffening of the nucleus. These molecular features vanished 24 h after photoporation. Since VNB-induced chromatin condensation was prolonged in LMNA knockout cells, A-type lamins may be required for restoring the nucleus to its original state. Selective depletion of A-type lamins reduced cell viability after VNB photoporation, while pharmacological stimulation of LMNA transcription increased the percentage of successfully transfected cells that survived after photoporation. Therefore, our results suggest that cells respond to VNB photoporation by temporary upregulation of A-type lamins to facilitate their recovery.
Collapse
|
17
|
Hobson CM, Falvo MR, Superfine R. A survey of physical methods for studying nuclear mechanics and mechanobiology. APL Bioeng 2021; 5:041508. [PMID: 34849443 PMCID: PMC8604565 DOI: 10.1063/5.0068126] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/20/2021] [Indexed: 12/23/2022] Open
Abstract
It is increasingly appreciated that the cell nucleus is not only a home for DNA but also a complex material that resists physical deformations and dynamically responds to external mechanical cues. The molecules that confer mechanical properties to nuclei certainly contribute to laminopathies and possibly contribute to cellular mechanotransduction and physical processes in cancer such as metastasis. Studying nuclear mechanics and the downstream biochemical consequences or their modulation requires a suite of complex assays for applying, measuring, and visualizing mechanical forces across diverse length, time, and force scales. Here, we review the current methods in nuclear mechanics and mechanobiology, placing specific emphasis on each of their unique advantages and limitations. Furthermore, we explore important considerations in selecting a new methodology as are demonstrated by recent examples from the literature. We conclude by providing an outlook on the development of new methods and the judicious use of the current techniques for continued exploration into the role of nuclear mechanobiology.
Collapse
Affiliation(s)
| | - Michael R. Falvo
- Department of Physics and Astronomy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Richard Superfine
- Department of Applied Physical Science, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
18
|
Kim PH, Chen NY, Heizer PJ, Tu Y, Weston TA, Fong JLC, Gill NK, Rowat AC, Young SG, Fong LG. Nuclear membrane ruptures underlie the vascular pathology in a mouse model of Hutchinson-Gilford progeria syndrome. JCI Insight 2021; 6:151515. [PMID: 34423791 PMCID: PMC8409987 DOI: 10.1172/jci.insight.151515] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/01/2021] [Indexed: 12/18/2022] Open
Abstract
The mutant nuclear lamin protein (progerin) produced in Hutchinson-Gilford progeria syndrome (HGPS) results in loss of arterial smooth muscle cells (SMCs), but the mechanism has been unclear. We found that progerin induces repetitive nuclear membrane (NM) ruptures, DNA damage, and cell death in cultured SMCs. Reducing lamin B1 expression and exposing cells to mechanical stress - to mirror conditions in the aorta - triggered more frequent NM ruptures. Increasing lamin B1 protein levels had the opposite effect, reducing NM ruptures and improving cell survival. Remarkably, raising lamin B1 levels increased nuclear compliance in cells and was able to offset the increased nuclear stiffness caused by progerin. In mice, lamin B1 expression in aortic SMCs is normally very low, and in mice with a targeted HGPS mutation (LmnaG609G), levels of lamin B1 decrease further with age while progerin levels increase. Those observations suggest that NM ruptures might occur in aortic SMCs in vivo. Indeed, studies in LmnaG609G mice identified NM ruptures in aortic SMCs, along with ultrastructural abnormalities in the cell nucleus that preceded SMC loss. Our studies identify NM ruptures in SMCs as likely causes of vascular pathology in HGPS.
Collapse
Affiliation(s)
- Paul H. Kim
- Department of Medicine
- Department of Bioengineering
| | - Natalie Y. Chen
- Department of Medicine
- Department of Integrative Biology and Physiology, and
| | | | | | | | | | | | - Amy C. Rowat
- Department of Bioengineering
- Department of Integrative Biology and Physiology, and
| | - Stephen G. Young
- Department of Medicine
- Department of Human Genetics, UCLA, Los Angeles, California, USA
| | | |
Collapse
|
19
|
Danielsson BE, Tieu KV, Bathula K, Armiger TJ, Vellala PS, Taylor RE, Dahl KN, Conway DE. Lamin microaggregates lead to altered mechanotransmission in progerin-expressing cells. Nucleus 2021; 11:194-204. [PMID: 32816594 PMCID: PMC7529416 DOI: 10.1080/19491034.2020.1802906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The nuclear lamina is a meshwork of intermediate filament proteins, and lamin A is the primary mechanical protein. An altered splicing of lamin A, known as progerin, causes the disease Hutchinson-Gilford progeria syndrome. Progerin-expressing cells have altered nuclear shapes and stiffened nuclear lamina with microaggregates of progerin. Here, progerin microaggregate inclusions in the lamina are shown to lead to cellular and multicellular dysfunction. We show with Comsol simulations that stiffened inclusions causes redistribution of normally homogeneous forces, and this redistribution is dependent on the stiffness difference and relatively independent of inclusion size. We also show mechanotransmission changes associated with progerin expression in cells under confinement and cells under external forces. Endothelial cells expressing progerin do not align properly with patterning. Fibroblasts expressing progerin do not align properly to applied cyclic force. Combined, these studies show that altered nuclear lamina mechanics and microstructure impacts cytoskeletal force transmission through the cell.
Collapse
Affiliation(s)
- Brooke E Danielsson
- Department of Biomedical Engineering, Virginia Commonwealth University , Richmond, VA, USA
| | - Katie V Tieu
- Department of Biomedical Engineering, Virginia Commonwealth University , Richmond, VA, USA
| | - Kranthidhar Bathula
- Department of Biomedical Engineering, Virginia Commonwealth University , Richmond, VA, USA
| | - Travis J Armiger
- Chemical Engineering, Carnegie Mellon University , Pittsburgh, PA, USA
| | - Pragna S Vellala
- Department of Biomedical Engineering, Carnegie Mellon University , Pittsburgh, PA , USA
| | - Rebecca E Taylor
- Department of Biomedical Engineering, Carnegie Mellon University , Pittsburgh, PA , USA.,Department of Mechanical Engineering, Carnegie Mellon University , Pittsburgh, PA, USA
| | - Kris Noel Dahl
- Chemical Engineering, Carnegie Mellon University , Pittsburgh, PA, USA.,Department of Biomedical Engineering, Carnegie Mellon University , Pittsburgh, PA , USA
| | - Daniel E Conway
- Department of Biomedical Engineering, Virginia Commonwealth University , Richmond, VA, USA
| |
Collapse
|
20
|
von Kleeck R, Castagnino P, Roberts E, Talwar S, Ferrari G, Assoian RK. Decreased vascular smooth muscle contractility in Hutchinson-Gilford Progeria Syndrome linked to defective smooth muscle myosin heavy chain expression. Sci Rep 2021; 11:10625. [PMID: 34012019 PMCID: PMC8134495 DOI: 10.1038/s41598-021-90119-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/06/2021] [Indexed: 01/12/2023] Open
Abstract
Children with Hutchinson-Gilford Progeria Syndrome (HGPS) suffer from multiple cardiovascular pathologies due to the expression of progerin, a mutant form of the nuclear envelope protein Lamin A. Progerin expression has a dramatic effect on arterial smooth muscle cells (SMCs) and results in decreased viability and increased arterial stiffness. However, very little is known about how progerin affects SMC contractility. Here, we studied the LaminAG609G/G609G mouse model of HGPS and found reduced arterial contractility at an early age that correlates with a decrease in smooth muscle myosin heavy chain (SM-MHC) mRNA and protein expression. Traction force microscopy on isolated SMCs from these mice revealed reduced force generation compared to wild-type controls; this effect was phenocopied by depletion of SM-MHC in WT SMCs and overcome by ectopic expression of SM-MHC in HGPS SMCs. Arterial SM-MHC levels are also reduced with age in wild-type mice and humans, suggesting a common defect in arterial contractility in HGPS and normal aging.
Collapse
Affiliation(s)
- Ryan von Kleeck
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Paola Castagnino
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute of Translational Medicine and Therapeutics at University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Emilia Roberts
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute of Translational Medicine and Therapeutics at University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shefali Talwar
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Giovanni Ferrari
- Departments of Surgery and Biomedical Engineering, Columbia University, New York, NY, 10032, USA
| | - Richard K Assoian
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute of Translational Medicine and Therapeutics at University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
21
|
Molecular and Cellular Mechanisms Driving Cardiovascular Disease in Hutchinson-Gilford Progeria Syndrome: Lessons Learned from Animal Models. Cells 2021; 10:cells10051157. [PMID: 34064612 PMCID: PMC8151355 DOI: 10.3390/cells10051157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/29/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disease that recapitulates many symptoms of physiological aging and precipitates death. Patients develop severe vascular alterations, mainly massive vascular smooth muscle cell loss, vessel stiffening, calcification, fibrosis, and generalized atherosclerosis, as well as electrical, structural, and functional anomalies in the heart. As a result, most HGPS patients die of myocardial infarction, heart failure, or stroke typically during the first or second decade of life. No cure exists for HGPS, and therefore it is of the utmost importance to define the mechanisms that control disease progression in order to develop new treatments to improve the life quality of patients and extend their lifespan. Since the discovery of the HGPS-causing mutation, several animal models have been generated to study multiple aspects of the syndrome and to analyze the contribution of different cell types to the acquisition of the HGPS-associated cardiovascular phenotype. This review discusses current knowledge about cardiovascular features in HGPS patients and animal models and the molecular and cellular mechanisms through which progerin causes cardiovascular disease.
Collapse
|
22
|
Parreira MT, Lavrenyuk K, Sanches JM, Dahl KN. A single stiffened nucleus alters cell dynamics and coherence in a monolayer. Cytoskeleton (Hoboken) 2021; 78:277-283. [PMID: 33837677 DOI: 10.1002/cm.21660] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 01/18/2023]
Abstract
Force transmission throughout a monolayer is the result of complex interactions between cells. Monolayer adaptation to force imbalances such as singular stiffened cells provides insight into the initiation of disease and fibrosis. Here, NRK-52E cells transfected with ∆50LA, which significantly stiffens the nucleus. These stiffened cells were sparsely placed in a monolayer of normal NRK-52E cells. Through morphometric analysis and temporal tracking, the impact of the singular stiffened cells shows a pivotal role in mechanoresponse of the monolayer. A method for a detailed analysis of the spatial aspect and temporal progression of the nuclear boundary was developed and used to achieve a full description of the phenotype and dynamics of the monolayers under study. Our findings reveal that cells are highly sensitive to the presence of mechanically impaired neighbors, leading to generalized loss of coordination in collective cell migration, but without seemingly affecting the potential for nuclear lamina fluctuations of neighboring cells. Reduced translocation in neighboring cells appears to be compensated by an increase in nuclear rotation and dynamic variation of shape, suggesting a "frustration" of cells and maintenance of motor activity. Interestingly, some characteristics of the behavior of these cells appear to be dependent on the distance to a ∆50LA cell, pointing to compensatory behavior in response to force transmission imbalances in a monolayer. These insights may suggest the long-range impacts of single cell defects related to tissue dysfunction.
Collapse
Affiliation(s)
- Maria Teresa Parreira
- Institute for Systems and Robotics - Lisboa and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Kirill Lavrenyuk
- Molecular Biophysics and Structural Biology, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - João M Sanches
- Institute for Systems and Robotics - Lisboa and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Kris Noel Dahl
- Molecular Biophysics and Structural Biology, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, Pennsylvania, USA.,Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
23
|
Cell stretchers and the LINC complex in mechanotransduction. Arch Biochem Biophys 2021; 702:108829. [PMID: 33716002 DOI: 10.1016/j.abb.2021.108829] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/23/2021] [Accepted: 03/07/2021] [Indexed: 02/07/2023]
Abstract
How cells respond to mechanical forces from the surrounding environment is critical for cell survival and function. The LINC complex is a central component in the mechanotransduction pathway that transmits mechanical information from the cell surface to the nucleus. Through LINC complex functionality, the nucleus is able to respond to mechanical stress by altering nuclear structure, chromatin organization, and gene expression. The use of specialized devices that apply mechanical strain to cells have been central to investigating how mechanotransduction occurs, how cells respond to mechanical stress, and the role of the LINC complexes in these processes. A large variety of designs have been reported for these devices, with the most common type being cell stretchers. Here we highlight some of the salient features of cell stretchers and suggest some key parameters that should be considered when using these devices. We provide a brief overview of how the LINC complexes contribute to the cellular responses to mechanical strain. And finally, we suggest that stretchers may be a useful tool to study aging.
Collapse
|
24
|
Cenni V, Capanni C, Mattioli E, Schena E, Squarzoni S, Bacalini MG, Garagnani P, Salvioli S, Franceschi C, Lattanzi G. Lamin A involvement in ageing processes. Ageing Res Rev 2020; 62:101073. [PMID: 32446955 DOI: 10.1016/j.arr.2020.101073] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 03/05/2020] [Accepted: 04/11/2020] [Indexed: 12/29/2022]
Abstract
Lamin A, a main constituent of the nuclear lamina, is the major splicing product of the LMNA gene, which also encodes lamin C, lamin A delta 10 and lamin C2. Involvement of lamin A in the ageing process became clear after the discovery that a group of progeroid syndromes, currently referred to as progeroid laminopathies, are caused by mutations in LMNA gene. Progeroid laminopathies include Hutchinson-Gilford Progeria, Mandibuloacral Dysplasia, Atypical Progeria and atypical-Werner syndrome, disabling and life-threatening diseases with accelerated ageing, bone resorption, lipodystrophy, skin abnormalities and cardiovascular disorders. Defects in lamin A post-translational maturation occur in progeroid syndromes and accumulated prelamin A affects ageing-related processes, such as mTOR signaling, epigenetic modifications, stress response, inflammation, microRNA activation and mechanosignaling. In this review, we briefly describe the role of these pathways in physiological ageing and go in deep into lamin A-dependent mechanisms that accelerate the ageing process. Finally, we propose that lamin A acts as a sensor of cell intrinsic and environmental stress through transient prelamin A accumulation, which triggers stress response mechanisms. Exacerbation of lamin A sensor activity due to stably elevated prelamin A levels contributes to the onset of a permanent stress response condition, which triggers accelerated ageing.
Collapse
Affiliation(s)
- Vittoria Cenni
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Cristina Capanni
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisabetta Mattioli
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisa Schena
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Stefano Squarzoni
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy; Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge, University Hospital, Stockholm, Sweden
| | - Stefano Salvioli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy; Interdepartmental Center Alma Mater Research Institute on Global Challenges and Climate Changes, University of Bologna, Bologna, Italy
| | - Claudio Franceschi
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| |
Collapse
|
25
|
Mu X, Tseng C, Hambright WS, Matre P, Lin C, Chanda P, Chen W, Gu J, Ravuri S, Cui Y, Zhong L, Cooke JP, Niedernhofer LJ, Robbins PD, Huard J. Cytoskeleton stiffness regulates cellular senescence and innate immune response in Hutchinson-Gilford Progeria Syndrome. Aging Cell 2020; 19:e13152. [PMID: 32710480 PMCID: PMC7431831 DOI: 10.1111/acel.13152] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 03/10/2020] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is caused by the accumulation of mutant prelamin A (progerin) in the nuclear lamina, resulting in increased nuclear stiffness and abnormal nuclear architecture. Nuclear mechanics are tightly coupled to cytoskeletal mechanics via lamin A/C. However, the role of cytoskeletal/nuclear mechanical properties in mediating cellular senescence and the relationship between cytoskeletal stiffness, nuclear abnormalities, and senescent phenotypes remain largely unknown. Here, using muscle-derived mesenchymal stromal/stem cells (MSCs) from the Zmpste24-/- (Z24-/- ) mouse (a model for HGPS) and human HGPS fibroblasts, we investigated the mechanical mechanism of progerin-induced cellular senescence, involving the role and interaction of mechanical sensors RhoA and Sun1/2 in regulating F-actin cytoskeleton stiffness, nuclear blebbing, micronuclei formation, and the innate immune response. We observed that increased cytoskeletal stiffness and RhoA activation in progeria cells were directly coupled with increased nuclear blebbing, Sun2 expression, and micronuclei-induced cGAS-Sting activation, part of the innate immune response. Expression of constitutively active RhoA promoted, while the inhibition of RhoA/ROCK reduced cytoskeletal stiffness, Sun2 expression, the innate immune response, and cellular senescence. Silencing of Sun2 expression by siRNA also repressed RhoA activation, cytoskeletal stiffness and cellular senescence. Treatment of Zmpste24-/- mice with a RhoA inhibitor repressed cellular senescence and improved muscle regeneration. These results reveal novel mechanical roles and correlation of cytoskeletal/nuclear stiffness, RhoA, Sun2, and the innate immune response in promoting aging and cellular senescence in HGPS progeria.
Collapse
Affiliation(s)
- Xiaodong Mu
- Department of Molecular Physiology and BiophysicsBaylor College of MedicineHoustonTexas
- Department of Orthopaedic SurgeryMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexas
- Shandong First Medical University & Shandong Academy of Medical SciencesJi'nanChina
| | - Chieh Tseng
- Department of Orthopaedic SurgeryMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexas
| | - William S. Hambright
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColorado
| | - Polina Matre
- Department of Cardiovascular SciencesHouston Methodist Research InstituteHoustonTexas
| | - Chih‐Yi Lin
- Department of Orthopaedic SurgeryMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexas
| | - Palas Chanda
- Department of Cardiovascular SciencesHouston Methodist Research InstituteHoustonTexas
| | - Wanqun Chen
- Department of Orthopaedic SurgeryMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexas
- Shandong First Medical University & Shandong Academy of Medical SciencesJi'nanChina
| | - Jianhua Gu
- Electron Microscopy CoreHouston Methodist Research InstituteHoustonTexas
| | - Sudheer Ravuri
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColorado
| | - Yan Cui
- Department of Orthopaedic SurgeryMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexas
| | - Ling Zhong
- Department of Orthopaedic SurgeryMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexas
| | - John P. Cooke
- Department of Cardiovascular SciencesHouston Methodist Research InstituteHoustonTexas
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and BiophysicsUniversity of MinnesotaMinneapolisMinnesota
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and BiophysicsUniversity of MinnesotaMinneapolisMinnesota
| | - Johnny Huard
- Department of Orthopaedic SurgeryMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexas
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColorado
| |
Collapse
|
26
|
Towards delineating the chain of events that cause premature senescence in the accelerated aging syndrome Hutchinson-Gilford progeria (HGPS). Biochem Soc Trans 2020; 48:981-991. [PMID: 32539085 PMCID: PMC7329345 DOI: 10.1042/bst20190882] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 11/17/2022]
Abstract
The metazoan nucleus is equipped with a meshwork of intermediate filament proteins called the A- and B-type lamins. Lamins lie beneath the inner nuclear membrane and serve as a nexus to maintain the architectural integrity of the nucleus, chromatin organization, DNA repair and replication and to regulate nucleocytoplasmic transport. Perturbations or mutations in various components of the nuclear lamina result in a large spectrum of human diseases collectively called laminopathies. One of the most well-characterized laminopathies is Hutchinson-Gilford progeria (HGPS), a rare segmental premature aging syndrome that resembles many features of normal human aging. HGPS patients exhibit alopecia, skin abnormalities, osteoporosis and succumb to cardiovascular complications in their teens. HGPS is caused by a mutation in LMNA, resulting in a mutated form of lamin A, termed progerin. Progerin expression results in a myriad of cellular phenotypes including abnormal nuclear morphology, loss of peripheral heterochromatin, transcriptional changes, DNA replication defects, DNA damage and premature cellular senescence. A key challenge is to elucidate how these different phenotypes are causally and mechanistically linked. In this mini-review, we highlight some key findings and present a model on how progerin-induced phenotypes may be temporally and mechanistically linked.
Collapse
|
27
|
Zhang J, Alisafaei F, Nikolić M, Nou XA, Kim H, Shenoy VB, Scarcelli G. Nuclear Mechanics within Intact Cells Is Regulated by Cytoskeletal Network and Internal Nanostructures. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1907688. [PMID: 32243075 PMCID: PMC7799396 DOI: 10.1002/smll.201907688] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/05/2020] [Accepted: 03/05/2020] [Indexed: 05/11/2023]
Abstract
The mechanical properties of the cellular nucleus are extensively studied as they play a critical role in important processes, such as cell migration, gene transcription, and stem cell differentiation. While the mechanical properties of the isolated nucleus have been tested, there is a lack of measurements about the mechanical behavior of the nucleus within intact cells and specifically about the interplay of internal nuclear components with the intracellular microenvironment, because current testing methods are based on contact and only allow studying the nucleus after isolation from a cell or disruption of cytoskeleton. Here, all-optical Brillouin microscopy and 3D chemomechanical modeling are used to investigate the regulation of nuclear mechanics in physiological conditions. It is observed that the nuclear modulus can be modulated by epigenetic regulation targeting internal nuclear nanostructures such as lamin A/C and chromatin. It is also found that nuclear modulus is strongly regulated by cytoskeletal behavior through a robust mechanism conserved in different culturing conditions. Given the active role of cytoskeletal modulation in nearly all cell functions, this work will enable to reveal highly relevant mechanisms of nuclear mechanical regulations in physiological and pathological conditions.
Collapse
Affiliation(s)
- Jitao Zhang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Farid Alisafaei
- Department of Materials Science and Engineering, School of Engineering and Applied Science, University of Pennsylvania, PA, 19104, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, PA, 19104, USA
| | - Miloš Nikolić
- Maryland Biophysics Program, University of Maryland, College Park, MD 20742, USA
| | - Xuefei A. Nou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Hanyoup Kim
- Canon U.S. Life Sciences, Inc., 9800 Medical Center Drive, Suite C-120, Rockville, MD 20850, USA
| | - Vivek B. Shenoy
- Department of Materials Science and Engineering, School of Engineering and Applied Science, University of Pennsylvania, PA, 19104, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, PA, 19104, USA
| | - Giuliano Scarcelli
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Maryland Biophysics Program, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
28
|
Stephens AD. Chromatin rigidity provides mechanical and genome protection. Mutat Res 2020; 821:111712. [PMID: 32590202 PMCID: PMC8186544 DOI: 10.1016/j.mrfmmm.2020.111712] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/05/2020] [Accepted: 06/15/2020] [Indexed: 12/31/2022]
Abstract
The nucleus is the organelle in the cell that contains the genome and its associate proteins which is collectively called chromatin. New work has shown that chromatin and its compaction level, dictated largely through histone modification state, provides rigidity to protect and stabilize the nucleus. Alterations in chromatin, its mechanics, and downstream loss of nuclear shape and stability are hallmarks of human disease. Weakened nuclear mechanics and abnormal morphology have been shown to cause rupturing of the nucleus which results in nuclear dysfunction including DNA damage. Thus, the rigidity provided by chromatin to maintain nuclear mechanical stability also provides its own protection from DNA damage via compartmentalization maintenance.
Collapse
Affiliation(s)
- Andrew D Stephens
- Biology Department, University of Massachusetts Amherst, Amherst, MA, 01003, United States.
| |
Collapse
|
29
|
Chromatin and Cytoskeletal Tethering Determine Nuclear Morphology in Progerin-Expressing Cells. Biophys J 2020; 118:2319-2332. [PMID: 32320674 DOI: 10.1016/j.bpj.2020.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/18/2020] [Accepted: 04/03/2020] [Indexed: 12/13/2022] Open
Abstract
The nuclear morphology of eukaryotic cells is determined by the interplay between the lamina forming the nuclear skeleton, the chromatin inside the nucleus, and the coupling with the cytoskeleton. Nuclear alterations are often associated with pathological conditions as in Hutchinson-Gilford progeria syndrome, in which a mutation in the lamin A gene yields an altered form of the protein, named progerin, and an aberrant nuclear shape. Here, we introduce an inducible cellular model of Hutchinson-Gilford progeria syndrome in HeLa cells in which increased progerin expression leads to alterations in the coupling of the lamin shell with cytoskeletal or chromatin tethers as well as with polycomb group proteins. Furthermore, our experiments show that progerin expression leads to enhanced nuclear shape fluctuations in response to cytoskeletal activity. To interpret the experimental results, we introduce a computational model of the cell nucleus that explicitly includes chromatin fibers, the nuclear shell, and coupling with the cytoskeleton. The model allows us to investigate how the geometrical organization of the chromatin-lamin tether affects nuclear morphology and shape fluctuations. In sum, our findings highlight the crucial role played by lamin-chromatin and lamin-cytoskeletal alterations in determining nuclear shape morphology and in affecting cellular functions and gene regulation.
Collapse
|
30
|
Wintner O, Hirsch‐Attas N, Schlossberg M, Brofman F, Friedman R, Kupervaser M, Kitsberg D, Buxboim A. A Unified Linear Viscoelastic Model of the Cell Nucleus Defines the Mechanical Contributions of Lamins and Chromatin. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1901222. [PMID: 32328409 PMCID: PMC7175345 DOI: 10.1002/advs.201901222] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 01/22/2020] [Indexed: 05/26/2023]
Abstract
The cell nucleus is constantly subjected to externally applied forces. During metazoan evolution, the nucleus has been optimized to allow physical deformability while protecting the genome under load. Aberrant nucleus mechanics can alter cell migration across narrow spaces in cancer metastasis and immune response and disrupt nucleus mechanosensitivity. Uncovering the mechanical roles of lamins and chromatin is imperative for understanding the implications of physiological forces on cells and nuclei. Lamin-knockout and -rescue fibroblasts and probed nucleus response to physiologically relevant stresses are generated. A minimal viscoelastic model is presented that captures dynamic resistance across different cell types, lamin composition, phosphorylation states, and chromatin condensation. The model is conserved at low and high loading and is validated by micropipette aspiration and nanoindentation rheology. A time scale emerges that separates between dominantly elastic and dominantly viscous regimes. While lamin-A and lamin-B1 contribute to nucleus stiffness, viscosity is specified mostly by lamin-A. Elastic and viscous association of lamin-B1 and lamin-A is supported by transcriptional and proteomic profiling analyses. Chromatin decondensation quantified by electron microscopy softens the nucleus unless lamin-A is expressed. A mechanical framework is provided for assessing nucleus response to applied forces in health and disease.
Collapse
Affiliation(s)
- Oren Wintner
- Department of Cell and Developmental BiologyThe Alexander Silberman Institute of Life SciencesThe Hebrew University of JerusalemJerusalem9190401Israel
- Alexander Grass Center for BioengineeringThe Rachel and Selim Benin School of Computer Science and EngineeringJerusalem9190416Israel
| | - Nivi Hirsch‐Attas
- Department of Cell and Developmental BiologyThe Alexander Silberman Institute of Life SciencesThe Hebrew University of JerusalemJerusalem9190401Israel
| | - Miriam Schlossberg
- Department of Cell and Developmental BiologyThe Alexander Silberman Institute of Life SciencesThe Hebrew University of JerusalemJerusalem9190401Israel
| | - Fani Brofman
- Department of Cell and Developmental BiologyThe Alexander Silberman Institute of Life SciencesThe Hebrew University of JerusalemJerusalem9190401Israel
| | - Roy Friedman
- Alexander Grass Center for BioengineeringThe Rachel and Selim Benin School of Computer Science and EngineeringJerusalem9190416Israel
| | - Meital Kupervaser
- The de Botton Institute for Protein ProfilingThe Nancy and Stephen Grand Israel National Center for Personalized MedicineWeizmann Institute of ScienceRehovot7610001Israel
| | - Danny Kitsberg
- Department of Cell and Developmental BiologyThe Alexander Silberman Institute of Life SciencesThe Hebrew University of JerusalemJerusalem9190401Israel
| | - Amnon Buxboim
- Department of Cell and Developmental BiologyThe Alexander Silberman Institute of Life SciencesThe Hebrew University of JerusalemJerusalem9190401Israel
- Alexander Grass Center for BioengineeringThe Rachel and Selim Benin School of Computer Science and EngineeringJerusalem9190416Israel
| |
Collapse
|
31
|
Song Y, Soto J, Chen B, Yang L, Li S. Cell engineering: Biophysical regulation of the nucleus. Biomaterials 2020; 234:119743. [PMID: 31962231 DOI: 10.1016/j.biomaterials.2019.119743] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/02/2019] [Accepted: 12/25/2019] [Indexed: 12/12/2022]
Abstract
Cells live in a complex and dynamic microenvironment, and a variety of microenvironmental cues can regulate cell behavior. In addition to biochemical signals, biophysical cues can induce not only immediate intracellular responses, but also long-term effects on phenotypic changes such as stem cell differentiation, immune cell activation and somatic cell reprogramming. Cells respond to mechanical stimuli via an outside-in and inside-out feedback loop, and the cell nucleus plays an important role in this process. The mechanical properties of the nucleus can directly or indirectly modulate mechanotransduction, and the physical coupling of the cell nucleus with the cytoskeleton can affect chromatin structure and regulate the epigenetic state, gene expression and cell function. In this review, we will highlight the recent progress in nuclear biomechanics and mechanobiology in the context of cell engineering, tissue remodeling and disease development.
Collapse
Affiliation(s)
- Yang Song
- Department of Bioengineering, University of California, Los Angeles, CA, USA; School of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Jennifer Soto
- Department of Bioengineering, University of California, Los Angeles, CA, USA
| | - Binru Chen
- Department of Bioengineering, University of California, Los Angeles, CA, USA
| | - Li Yang
- School of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, CA, USA; Department of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
32
|
Stephens AD, Banigan EJ, Marko JF. Chromatin's physical properties shape the nucleus and its functions. Curr Opin Cell Biol 2019; 58:76-84. [PMID: 30889417 PMCID: PMC6692209 DOI: 10.1016/j.ceb.2019.02.006] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/06/2019] [Accepted: 02/20/2019] [Indexed: 12/13/2022]
Abstract
The cell nucleus encloses, organizes, and protects the genome. Chromatin maintains nuclear mechanical stability and shape in coordination with lamins and the cytoskeleton. Abnormal nuclear shape is a diagnostic marker for human diseases, and it can cause nuclear dysfunction. Chromatin mechanics underlies this link, as alterations to chromatin and its physical properties can disrupt or rescue nuclear shape. The cell can regulate nuclear shape through mechanotransduction pathways that sense and respond to extracellular cues, thus modulating chromatin compaction and rigidity. These findings reveal how chromatin's physical properties can regulate cellular function and drive abnormal nuclear morphology and dysfunction in disease.
Collapse
Affiliation(s)
- Andrew D Stephens
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, United States.
| | - Edward J Banigan
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - John F Marko
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, United States; Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208, United States.
| |
Collapse
|
33
|
Osmanagic-Myers S, Kiss A, Manakanatas C, Hamza O, Sedlmayer F, Szabo PL, Fischer I, Fichtinger P, Podesser BK, Eriksson M, Foisner R. Endothelial progerin expression causes cardiovascular pathology through an impaired mechanoresponse. J Clin Invest 2019; 129:531-545. [PMID: 30422822 PMCID: PMC6355303 DOI: 10.1172/jci121297] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 11/06/2018] [Indexed: 01/09/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging disorder characterized by accelerated cardiovascular disease with extensive fibrosis. It is caused by a mutation in LMNA leading to expression of truncated prelamin A (progerin) in the nucleus. To investigate the contribution of the endothelium to cardiovascular HGPS pathology, we generated an endothelium-specific HGPS mouse model with selective endothelial progerin expression. Transgenic mice develop interstitial myocardial and perivascular fibrosis and left ventricular hypertrophy associated with diastolic dysfunction and premature death. Endothelial cells show impaired shear stress response and reduced levels of endothelial nitric oxide synthase (eNOS) and NO. On the molecular level, progerin impairs nucleocytoskeletal coupling in endothelial cells through changes in mechanoresponsive components at the nuclear envelope, increased F-actin/G-actin ratios, and deregulation of mechanoresponsive myocardin-related transcription factor-A (MRTFA). MRTFA binds to the Nos3 promoter and reduces eNOS expression, thereby mediating a profibrotic paracrine response in fibroblasts. MRTFA inhibition rescues eNOS levels and ameliorates the profibrotic effect of endothelial cells in vitro. Although this murine model lacks the key anatomical feature of vascular smooth muscle cell loss seen in HGPS patients, our data show that progerin-induced impairment of mechanosignaling in endothelial cells contributes to excessive fibrosis and cardiovascular disease in HGPS patients.
Collapse
Affiliation(s)
- Selma Osmanagic-Myers
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna and University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Attila Kiss
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Christina Manakanatas
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna and University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Ouafa Hamza
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Franziska Sedlmayer
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna and University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Petra L. Szabo
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Irmgard Fischer
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna and University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Petra Fichtinger
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna and University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Bruno K. Podesser
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Maria Eriksson
- Department of Biosciences and Nutrition, Karolinska Institutet, NEO, Huddinge, Sweden
| | - Roland Foisner
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna and University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
34
|
González-Cruz RD, Dahl KN, Darling EM. The Emerging Role of Lamin C as an Important LMNA Isoform in Mechanophenotype. Front Cell Dev Biol 2018; 6:151. [PMID: 30450357 PMCID: PMC6224339 DOI: 10.3389/fcell.2018.00151] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/15/2018] [Indexed: 12/17/2022] Open
Abstract
Lamin A and lamin C isoforms of the gene LMNA are major structural and mechanotransductive components of the nuclear lamina. Previous reports have proposed lamin A as the isoform with the most dominant contributions to cellular mechanophenotype. Recently, expression of lamin C has also been shown to strongly correlate to cellular elastic and viscoelastic properties. Nevertheless, LMNA isoforms exist as part of a network that collectively provides structural integrity to the nucleus and their expression is ultimately regulated in a cell-specific manner. Thus, they have importance in mechanotransduction and structural integrity of the nucleus as well as potential candidates for biomarkers of whole-cell mechanophenotype. Therefore, a fuller discussion of lamin isoforms as mechanophenotypic biomarkers should compare both individual and ratiometric isoform contributions toward whole-cell mechanophenotype across different cell types. In this perspective, we discuss the distinctions between the mechanophenotypic correlations of individual and ratiometric lamins A:B1, C:B1, (A + C):B1, and C:A across cells from different lineages, demonstrating that the collective contribution of ratiometric lamin (A + C):B1 isoforms exhibited the strongest correlation to whole-cell stiffness. Additionally, we highlight the potential roles of lamin isoform ratios as indicators of mechanophenotypic change in differentiation and disease to demonstrate that the contributions of individual and collective lamin isoforms can occur as both static and dynamic biomarkers of mechanophenotype.
Collapse
Affiliation(s)
| | - Kris N Dahl
- Department of Chemical Engineering, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Eric M Darling
- Center for Biomedical Engineering, Brown University, Providence, RI, United States.,Department of Molecular Pharmacology, Physiology and Biotechnology, School of Engineering, Department of Orthopaedics, Brown University, Providence, RI, United States
| |
Collapse
|
35
|
Cho S, Abbas A, Irianto J, Ivanovska IL, Xia Y, Tewari M, Discher DE. Progerin phosphorylation in interphase is lower and less mechanosensitive than lamin-A,C in iPS-derived mesenchymal stem cells. Nucleus 2018; 9:230-245. [PMID: 29619860 PMCID: PMC5973135 DOI: 10.1080/19491034.2018.1460185] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Interphase phosphorylation of lamin-A,C depends dynamically on a cell's microenvironment, including the stiffness of extracellular matrix. However, phosphorylation dynamics is poorly understood for diseased forms such as progerin, a permanently farnesylated mutant of LMNA that accelerates aging of stiff and mechanically stressed tissues. Here, fine-excision alignment mass spectrometry (FEA-MS) is developed to quantify progerin and its phosphorylation levels in patient iPS cells differentiated to mesenchymal stem cells (MSCs). The stoichiometry of total A-type lamins (including progerin) versus B-type lamins measured for Progeria iPS-MSCs prove similar to that of normal MSCs, with total A-type lamins more abundant than B-type lamins. However, progerin behaves more like farnesylated B-type lamins in mechanically-induced segregation from nuclear blebs. Phosphorylation of progerin at multiple sites in iPS-MSCs cultured on rigid plastic is also lower than that of normal lamin-A and C. Reduction of nuclear tension upon i) cell rounding/detachment from plastic, ii) culture on soft gels, and iii) inhibition of actomyosin stress increases phosphorylation and degradation of lamin-C > lamin-A > progerin. Such mechano-sensitivity diminishes, however, with passage as progerin and DNA damage accumulate. Lastly, transcription-regulating retinoids exert equal effects on both diseased and normal A-type lamins, suggesting a differential mechano-responsiveness might best explain the stiff tissue defects in Progeria.
Collapse
Affiliation(s)
- Sangkyun Cho
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Amal Abbas
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Jerome Irianto
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Irena L. Ivanovska
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuntao Xia
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Manu Tewari
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennis E. Discher
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA,CONTACT Dennis E. Discher , University of Pennsylvania, 129 Towne Bldg, Philadelphia, PA 19104
| |
Collapse
|
36
|
Pitrez PR, Estronca L, Vazão H, Egesipe AL, Le Corf A, Navarro C, Lévy N, De Sandre-Giovannoli A, Nissan X, Ferreira L. Substrate Topography Modulates Cell Aging on a Progeria Cell Model. ACS Biomater Sci Eng 2018; 4:1498-1504. [PMID: 33445307 DOI: 10.1021/acsbiomaterials.8b00224] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Aging is characterized by a progressive accumulation of cellular damage, which leads to impaired function. Little is known whether substrates can influence cell aging. This is of utmost importance in the development of medical devices that are in contact with human tissue for long periods of time. To address this question, we have used an accelerated aging cell model derived from Hutchinson-Gilford Progeria Syndrome (HGPS) induced pluripotent stem cells (iPSCs). Our results show that HGPS-iPSC smooth muscle cells (SMCs) have an increased aging profile in substrates with specific micropatterns than in flat ones. This is characterized by an up-regulation in the expression of progerin, β-galactosidase, annexin 3 and 5, and caspase 9. Signs of cell aging are also observed in SMCs without HGPS cultured in substrates with specific microtopographies. It is further showed that specific micropatterned substrates induce cell aging by triggering a DNA damage program likely by the disruption between cyto- and nucleoskeleton.
Collapse
Affiliation(s)
- Patricia R Pitrez
- Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine, Polo I first Floor, 3004-504 Coimbra, Portugal.,Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra, Portugal
| | - Luís Estronca
- Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine, Polo I first Floor, 3004-504 Coimbra, Portugal.,Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra, Portugal
| | - Helena Vazão
- Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine, Polo I first Floor, 3004-504 Coimbra, Portugal
| | - Anne-Laure Egesipe
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic diseases, Genopole Campus 1, 5 rue Henri Desbruères, 91030 Evry Cedex, France
| | - Amélie Le Corf
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic diseases, Genopole Campus 1, 5 rue Henri Desbruères, 91030 Evry Cedex, France
| | - Claire Navarro
- INSERM, GMGF UMR_S 910, Aix Marseille Université, 13385 Marseille, France
| | - Nicolas Lévy
- INSERM, GMGF UMR_S 910, Aix Marseille Université, 13385 Marseille, France.,Molecular Genetics Laboratory, Department of Medical Genetics, La Timone Children's Hospital, APHM, 13385 Marseille, France
| | - Annachiara De Sandre-Giovannoli
- INSERM, GMGF UMR_S 910, Aix Marseille Université, 13385 Marseille, France.,Molecular Genetics Laboratory, Department of Medical Genetics, La Timone Children's Hospital, APHM, 13385 Marseille, France
| | - Xavier Nissan
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic diseases, Genopole Campus 1, 5 rue Henri Desbruères, 91030 Evry Cedex, France
| | - Lino Ferreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine, Polo I first Floor, 3004-504 Coimbra, Portugal.,Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra, Portugal
| |
Collapse
|
37
|
Del Campo L, Hamczyk MR, Andrés V, Martínez-González J, Rodríguez C. Mechanisms of vascular aging: What can we learn from Hutchinson-Gilford progeria syndrome? CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2018; 30:120-132. [PMID: 29602596 DOI: 10.1016/j.arteri.2017.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 01/07/2023]
Abstract
Aging is the main risk factor for cardiovascular disease (CVD). The increased prevalence of CVD is partly due to the global increase in life expectancy. In this context, it is essential to identify the mechanisms by which aging induces CVD, with the ultimate aim of reducing its incidence. Both atherosclerosis and heart failure significantly contribute to age-associated CVD morbidity and mortality. Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder caused by the synthesis of progerin, which is noted for accelerated aging and CVD. This mutant form of prelamin A induces generalised atherosclerosis, vascular calcification, and cardiac electrophysiological abnormalities, leading to premature aging and death, mainly due to myocardial infarction and stroke. This review discusses the main vascular structural and functional abnormalities during physiological and premature aging, as well as the mechanisms involved in the exacerbated CVD and accelerated aging induced by the accumulation of progerin and prelamin A. Both proteins are expressed in non-HGPS individuals, and physiological aging shares many features of progeria. Research into HGPS could therefore shed light on novel mechanisms involved in the physiological aging of the cardiovascular system.
Collapse
Affiliation(s)
- Lara Del Campo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, España; CIBER de Enfermedades Cardiovasculares (CIBERCV), España
| | - Magda R Hamczyk
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, España; CIBER de Enfermedades Cardiovasculares (CIBERCV), España
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, España; CIBER de Enfermedades Cardiovasculares (CIBERCV), España.
| | - José Martínez-González
- CIBER de Enfermedades Cardiovasculares (CIBERCV), España; Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), IIB-Sant Pau, Barcelona, España
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), España; Institut de Recerca del Hospital de la Santa Creu i Sant Pau-Programa ICCC, IIB-Sant Pau, Barcelona, España.
| | | |
Collapse
|
38
|
Stephens AD, Liu PZ, Banigan EJ, Almassalha LM, Backman V, Adam SA, Goldman RD, Marko JF. Chromatin histone modifications and rigidity affect nuclear morphology independent of lamins. Mol Biol Cell 2018; 29:220-233. [PMID: 29142071 PMCID: PMC5909933 DOI: 10.1091/mbc.e17-06-0410] [Citation(s) in RCA: 233] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 10/17/2017] [Accepted: 11/08/2017] [Indexed: 01/29/2023] Open
Abstract
Nuclear shape and architecture influence gene localization, mechanotransduction, transcription, and cell function. Abnormal nuclear morphology and protrusions termed "blebs" are diagnostic markers for many human afflictions including heart disease, aging, progeria, and cancer. Nuclear blebs are associated with both lamin and chromatin alterations. A number of prior studies suggest that lamins dictate nuclear morphology, but the contributions of altered chromatin compaction remain unclear. We show that chromatin histone modification state dictates nuclear rigidity, and modulating it is sufficient to both induce and suppress nuclear blebs. Treatment of mammalian cells with histone deacetylase inhibitors to increase euchromatin or histone methyltransferase inhibitors to decrease heterochromatin results in a softer nucleus and nuclear blebbing, without perturbing lamins. Conversely, treatment with histone demethylase inhibitors increases heterochromatin and chromatin nuclear rigidity, which results in reduced nuclear blebbing in lamin B1 null nuclei. Notably, increased heterochromatin also rescues nuclear morphology in a model cell line for the accelerated aging disease Hutchinson-Gilford progeria syndrome caused by mutant lamin A, as well as cells from patients with the disease. Thus, chromatin histone modification state is a major determinant of nuclear blebbing and morphology via its contribution to nuclear rigidity.
Collapse
Affiliation(s)
- Andrew D Stephens
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Patrick Z Liu
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Edward J Banigan
- Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Luay M Almassalha
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208
| | - Vadim Backman
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208
| | - Stephen A Adam
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Robert D Goldman
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - John F Marko
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208.,Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208
| |
Collapse
|
39
|
Pecorari I, Borin D, Sbaizero O. A Perspective on the Experimental Techniques for Studying Lamins. Cells 2017; 6:E33. [PMID: 28994747 PMCID: PMC5755493 DOI: 10.3390/cells6040033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/01/2017] [Accepted: 10/05/2017] [Indexed: 01/29/2023] Open
Abstract
Lamins are type V intermediate filaments that collectively form a meshwork underneath the inner nuclear membrane, called nuclear lamina. Furthermore, they are also present in the nucleoplasm. Lamins are experiencing a growing interest, since a wide range of diseases are induced by mutations in the gene coding for A-type lamins, globally known as laminopathies. Moreover, it has been demonstrated that lamins are involved in other pathological conditions, like cancer. The role of lamins has been studied from several perspectives, exploiting different techniques and procedures. This multidisciplinary approach has contributed to resolving the unique features of lamins and has provided a thorough insight in their role in living organisms. Yet, there are still many unanswered questions, which constantly generate research in the field. The present work is aimed to review some interesting experimental techniques performed so far to study lamins. Scientists can take advantage of this collection for their novel investigations, being aware of the already pursued and consolidated methodologies. Hopefully, advances in these research directions will provide insights to achieve better diagnostic procedures and effective therapeutic options.
Collapse
Affiliation(s)
- Ilaria Pecorari
- Department of Engineering and Architecture, University of Trieste, Via Valerio 10, 34127 Trieste, Italy.
| | - Daniele Borin
- Department of Engineering and Architecture, University of Trieste, Via Valerio 10, 34127 Trieste, Italy.
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, Via Valerio 10, 34127 Trieste, Italy.
| |
Collapse
|
40
|
Hamczyk MR, del Campo L, Andrés V. Aging in the Cardiovascular System: Lessons from Hutchinson-Gilford Progeria Syndrome. Annu Rev Physiol 2017; 80:27-48. [PMID: 28934587 DOI: 10.1146/annurev-physiol-021317-121454] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aging, the main risk factor for cardiovascular disease (CVD), is becoming progressively more prevalent in our societies. A better understanding of how aging promotes CVD is therefore urgently needed to develop new strategies to reduce disease burden. Atherosclerosis and heart failure contribute significantly to age-associated CVD-related morbimortality. CVD and aging are both accelerated in patients suffering from Hutchinson-Gilford progeria syndrome (HGPS), a rare genetic disorder caused by the prelamin A mutant progerin. Progerin causes extensive atherosclerosis and cardiac electrophysiological alterations that invariably lead to premature aging and death. This review summarizes the main structural and functional alterations to the cardiovascular system during physiological and premature aging and discusses the mechanisms underlying exaggerated CVD and aging induced by prelamin A and progerin. Because both proteins are expressed in normally aging non-HGPS individuals, and most hallmarks of normal aging occur in progeria, research on HGPS can identify mechanisms underlying physiological aging.
Collapse
Affiliation(s)
- Magda R Hamczyk
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; .,CIBER de Enfermedades Cardiovasculares (CIBER-CV), 28029 Madrid, Spain
| | - Lara del Campo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; .,CIBER de Enfermedades Cardiovasculares (CIBER-CV), 28029 Madrid, Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; .,CIBER de Enfermedades Cardiovasculares (CIBER-CV), 28029 Madrid, Spain
| |
Collapse
|
41
|
Zhang J, Nou XA, Kim H, Scarcelli G. Brillouin flow cytometry for label-free mechanical phenotyping of the nucleus. LAB ON A CHIP 2017; 17:663-670. [PMID: 28102402 PMCID: PMC5310767 DOI: 10.1039/c6lc01443g] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The mechanical properties of the nucleus are closely related to many cellular functions; thus, measuring nuclear mechanical properties is crucial to our understanding of cell biomechanics and could lead to intrinsic biophysical contrast mechanisms to classify cells. Although many technologies have been developed to characterize cell stiffness, they generally require contact with the cell and thus cannot provide direct information on nuclear mechanical properties. In this work, we developed a flow cytometry technique based on an all-optical measurement to measure nuclear mechanical properties by integrating Brillouin spectroscopy with microfluidics. Brillouin spectroscopy probes the mechanical properties of material via light scattering, so it is inherently label-free, non-contact, and non-invasive. Using a measuring beam spot of submicron size, we can measure several regions within each cell as they flow, which enables us to classify cell populations based on their nuclear mechanical signatures at a throughput of ∼200 cells per hour. We show that Brillouin cytometry has sufficient sensitivity to detect physiologically-relevant changes in nuclear stiffness by probing the effect of drug-induced chromatin decondensation.
Collapse
Affiliation(s)
- Jitao Zhang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA.
| | - Xuefei A Nou
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA.
| | - Hanyoup Kim
- Canon U.S. Life Sciences, Inc., 9800 Medical Center Drive, Suite C-120, Rockville, MD 20850, USA
| | - Giuliano Scarcelli
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA.
| |
Collapse
|
42
|
Stephens AD, Banigan EJ, Adam SA, Goldman RD, Marko JF. Chromatin and lamin A determine two different mechanical response regimes of the cell nucleus. Mol Biol Cell 2017; 28:1984-1996. [PMID: 28057760 PMCID: PMC5541848 DOI: 10.1091/mbc.e16-09-0653] [Citation(s) in RCA: 310] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/04/2016] [Accepted: 12/29/2016] [Indexed: 02/02/2023] Open
Abstract
The cell nucleus must continually resist and respond to intercellular and intracellular mechanical forces to transduce mechanical signals and maintain proper genome organization and expression. Altered nuclear mechanics is associated with many human diseases, including heart disease, progeria, and cancer. Chromatin and nuclear envelope A-type lamin proteins are known to be key nuclear mechanical components perturbed in these diseases, but their distinct mechanical contributions are not known. Here we directly establish the separate roles of chromatin and lamin A/C and show that they determine two distinct mechanical regimes via micromanipulation of single isolated nuclei. Chromatin governs response to small extensions (<3 μm), and euchromatin/heterochromatin levels modulate the stiffness. In contrast, lamin A/C levels control nuclear strain stiffening at large extensions. These results can be understood through simulations of a polymeric shell and cross-linked polymer interior. Our results provide a framework for understanding the differential effects of chromatin and lamin A/C in cell nuclear mechanics and their alterations in disease.
Collapse
Affiliation(s)
- Andrew D Stephens
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Edward J Banigan
- Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208
| | - Stephen A Adam
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Robert D Goldman
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - John F Marko
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208.,Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208
| |
Collapse
|
43
|
Zhang H, Sun L, Wang K, Wu D, Trappio M, Witting C, Cao K. Loss of H3K9me3 Correlates with ATM Activation and Histone H2AX Phosphorylation Deficiencies in Hutchinson-Gilford Progeria Syndrome. PLoS One 2016; 11:e0167454. [PMID: 27907109 PMCID: PMC5131972 DOI: 10.1371/journal.pone.0167454] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/14/2016] [Indexed: 12/20/2022] Open
Abstract
Compelling evidence suggests that defective DNA damage response (DDR) plays a key role in the premature aging phenotypes in Hutchinson-Gilford progeria syndrome (HGPS). Studies document widespread alterations in histone modifications in HGPS cells, especially, the global loss of histone H3 trimethylated on lysine 9 (H3K9me3). In this study, we explore the potential connection(s) between H3K9me3 loss and the impaired DDR in HGPS. When cells are exposed to a DNA-damaging agent Doxorubicin (Dox), double strand breaks (DSBs) are generated that result in the phosphorylation of histone H2A variant H2AX (gammaH2AX) within an hour. We find that the intensities of gammaH2AX foci appear significantly weaker in the G0/G1 phase HGPS cells compared to control cells. This reduction is associated with a delay in the recruitment of essential DDR factors. We further demonstrate that ataxia-telangiectasia mutated (ATM) is responsible for the amplification of gammaH2AX signals at DSBs during G0/G1 phase, and its activation is inhibited in the HGPS cells that display significant loss of H3K9me3. Moreover, methylene (MB) blue treatment, which is known to save heterochromatin loss in HGPS, restores H3K9me3, stimulates ATM activity, increases gammaH2AX signals and rescues deficient DDR. In summary, this study demonstrates an early DDR defect of attenuated gammaH2AX signals in G0/G1 phase HGPS cells and provides a plausible connection between H3K9me3 loss and DDR deficiency.
Collapse
Affiliation(s)
- Haoyue Zhang
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
| | - Linlin Sun
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
| | - Kun Wang
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States of America
| | - Di Wu
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
| | - Mason Trappio
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
| | - Celeste Witting
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
- * E-mail:
| |
Collapse
|
44
|
Hasturk O, Sivas A, Karasozen B, Demirci U, Hasirci N, Hasirci V. Quantification of Type, Timing, and Extent of Cell Body and Nucleus Deformations Caused by the Dimensions and Hydrophilicity of Square Prism Micropillars. Adv Healthc Mater 2016; 5:2972-2982. [PMID: 27925459 DOI: 10.1002/adhm.201600857] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/21/2016] [Indexed: 01/30/2023]
Abstract
Novel digital analysis strategies are developed for the quantification of changes in the cytoskeletal and nuclear morphologies of mesenchymal stem cells cultured on micropillars. Severe deformations of nucleus and distinct conformational changes of cell body ranging from extensive elongation to branching are visualized and quantified. These deformations are caused mainly by the dimensions and hydrophilicity of the micropillars.
Collapse
Affiliation(s)
- Onur Hasturk
- Graduate Department of Biotechnology; Middle East Technical University (METU); Ankara 06800 Turkey
- BIOMATEN; Center of Excellence in Biomaterials and Tissue Engineering; Middle East Technical University (METU); Ankara 06800 Turkey
| | - Abdullah Sivas
- Institute of Applied Mathematics; Middle East Technical University (METU); Ankara 06800 Turkey
| | - Bulent Karasozen
- Institute of Applied Mathematics; Middle East Technical University (METU); Ankara 06800 Turkey
| | - Utkan Demirci
- Bio-Acoustic-MEMs in Medicine (BAMM) Laboratory; Stanford School of Medicine; Palo Alto CA 94394 USA
| | - Nesrin Hasirci
- Graduate Department of Biotechnology; Middle East Technical University (METU); Ankara 06800 Turkey
- BIOMATEN; Center of Excellence in Biomaterials and Tissue Engineering; Middle East Technical University (METU); Ankara 06800 Turkey
- Department of Chemistry; Middle East Technical University (METU); Ankara 06800 Turkey
| | - Vasif Hasirci
- Graduate Department of Biotechnology; Middle East Technical University (METU); Ankara 06800 Turkey
- BIOMATEN; Center of Excellence in Biomaterials and Tissue Engineering; Middle East Technical University (METU); Ankara 06800 Turkey
- Department of Biological Sciences; Middle East Technical University (METU); Ankara 06800 Turkey
| |
Collapse
|
45
|
The mammalian LINC complex regulates genome transcriptional responses to substrate rigidity. Sci Rep 2016; 6:38063. [PMID: 27905489 PMCID: PMC5131312 DOI: 10.1038/srep38063] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 11/03/2016] [Indexed: 12/14/2022] Open
Abstract
Mechanical integration of the nucleus with the extracellular matrix (ECM) is established by linkage between the cytoskeleton and the nucleus. This integration is hypothesized to mediate sensing of ECM rigidity, but parsing the function of nucleus-cytoskeleton linkage from other mechanisms has remained a central challenge. Here we took advantage of the fact that the LINC (linker of nucleoskeleton and cytoskeleton) complex is a known molecular linker of the nucleus to the cytoskeleton, and asked how it regulates the sensitivity of genome-wide transcription to substratum rigidity. We show that gene mechanosensitivity is preserved after LINC disruption, but reversed in direction. Combined with myosin inhibition studies, we identify genes that depend on nuclear tension for their regulation. We also show that LINC disruption does not attenuate nuclear shape sensitivity to substrate rigidity. Our results show for the first time that the LINC complex facilitates mechano-regulation of expression across the genome.
Collapse
|
46
|
Spagnol ST, Armiger TJ, Dahl KN. Mechanobiology of Chromatin and the Nuclear Interior. Cell Mol Bioeng 2016; 9:268-276. [PMID: 28163791 PMCID: PMC5289645 DOI: 10.1007/s12195-016-0444-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 05/03/2016] [Indexed: 02/06/2023] Open
Abstract
The view of the cell nucleus has evolved from an isolated, static organelle to a dynamic structure integrated with other mechanical elements of the cell. Both dynamics and integration appear to contribute to a mechanical regulation of genome expression. Here, we review physical structures inside the nucleus at different length scales and the dynamic reorganization modulated by cellular forces. First, we discuss nuclear organization focusing on self-assembly and disassembly of DNA structures and various nuclear bodies. We then discuss the importance of connections from the chromatin fiber through the nuclear envelope to the rest of the cell as they relate to mechanobiology. Finally, we discuss how cell stimulation, both chemical and physical, can alter nuclear structures and ultimately cellular function in healthy cells and in some model diseases. The view of chromatin and nuclear bodies as mechanical entities integrated with force generation from the cytoskeleton combines polymer physics with cell biology and medicine.
Collapse
Affiliation(s)
- Stephen T. Spagnol
- Department of Chemical Engineering, Carnegie Mellon University, 5000 Forbes Ave., Pittsburgh, PA 15213, USA
| | - Travis J. Armiger
- Department of Chemical Engineering, Carnegie Mellon University, 5000 Forbes Ave., Pittsburgh, PA 15213, USA
| | - Kris Noel Dahl
- Department of Chemical Engineering, Carnegie Mellon University, 5000 Forbes Ave., Pittsburgh, PA 15213, USA
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, USA
| |
Collapse
|