1
|
Yu H, Dou S, Wang H, Sun Y, Qu J, Liu T, Liu X, Wei C, Gao H. Role of m 6A methyltransferase METTL3 in Keratoconus pathogenesis. Exp Eye Res 2024:110207. [PMID: 39681235 DOI: 10.1016/j.exer.2024.110207] [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: 09/23/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
Keratoconus (KC) is the most common ectatic corneal disease with unknown pathogenesis. This study aimed to investigate the role of methyltransferase-like enzyme 3 (METTL3) in KC pathogenesis. In the present study, we examined the levels of METTL3 and other N6-methyladenosine (m6A) modification-related proteins in KC samples and human stromal keratocyte (HTK) cells stimulated by mechanical stretch (MS) using Western blotting and immunohistochemistry. The level of m6A RNA methylation was quantified using the m6A RNA methylation assay kit. Genetic (Mettl3 knockdown mice) and pharmacological (STM2457) approaches were employed to investigate the effect of METTL3 on the expression of metalloproteinases (MMPs) in MS-treated corneal stromal cells (CSCs) via Western blotting and real-time polymerase chain reaction. Moreover, YAP signaling activity was assessed to explore the relationship between METTL3 and MMPs in MS-treated CSCs. Increased expression of METTL3 and decreased expression of METTL14, WTAP, and YTHDF2 were detected in KC samples and MS-stimulated HTK cells. Correspondingly, the m6A levels in KC specimens and MS-stimulated CSCs were significantly higher than those in controls but were significantly reduced when METTL3 activity was genetically and pharmacologically blocked. Inhibition of METTL3 significantly reduced the expression of MMP1 and MMP3 in mechanically stretched CSCs and reduced YAP activity. Furthermore, pharmacologically inhibiting YAP signaling in MS-stimulated HTK cells significantly reduced MMP1 and MMP3 expression. Our findings highlight the pathogenic role of METTL3 in KC. Further investigation is required to investigate the underlying mechanism.
Collapse
Affiliation(s)
- Huimin Yu
- Medical College, Qingdao University, Qingdao, 266071, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, 266071, China
| | - Shengqian Dou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, 266071, China; School of Ophthalmology, Shandong First Medical University, Jinan, 250000, Shandong, China
| | - Huijin Wang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, 266071, China
| | - Yaru Sun
- Eye Hospital of Shandong First Medical University, Eye Institute of Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Junpeng Qu
- Medical College, Qingdao University, Qingdao, 266071, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, 266071, China
| | - Ting Liu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, 266071, China
| | - Xiaoxue Liu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, 266071, China; School of Ophthalmology, Shandong First Medical University, Jinan, 250000, Shandong, China
| | - Chao Wei
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, 266071, China; School of Ophthalmology, Shandong First Medical University, Jinan, 250000, Shandong, China.
| | - Hua Gao
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, 266071, China; Eye Hospital of Shandong First Medical University, Eye Institute of Shandong First Medical University, Jinan, 250021, Shandong, China; School of Ophthalmology, Shandong First Medical University, Jinan, 250000, Shandong, China; School of Public Health, Shandong First Medical University, Jinan, 250000, Shandong, China.
| |
Collapse
|
2
|
Wang TC, Abolghasemzade S, McKee BP, Singh I, Pendyala K, Mohajeri M, Patel H, Shaji A, Kersey AL, Harsh K, Kaur S, Dollahon CR, Chukkapalli S, Lele PP, Conway DE, Gaharwar AK, Dickinson RB, Lele TP. Matrix stiffness drives drop like nuclear deformation and lamin A/C tension-dependent YAP nuclear localization. Nat Commun 2024; 15:10151. [PMID: 39578439 PMCID: PMC11584751 DOI: 10.1038/s41467-024-54577-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Extracellular matrix (ECM) stiffness influences cancer cell fate by altering gene expression. Previous studies suggest that stiffness-induced nuclear deformation may regulate gene expression through YAP nuclear localization. We investigated the role of the nuclear lamina in this process. We show that the nuclear lamina exhibits mechanical threshold behavior: once unwrinkled, the nuclear lamina is inextensible. A computational model predicts that the unwrinkled lamina is under tension, which is confirmed using a lamin tension sensor. Laminar unwrinkling is caused by nuclear flattening during cell spreading on stiff ECM. Knockdown of lamin A/C eliminates nuclear surface tension and decreases nuclear YAP localization. These findings show that nuclear deformation in cells conforms to the nuclear drop model and reveal a role for lamin A/C tension in controlling YAP localization in cancer cells.
Collapse
Affiliation(s)
- Ting-Ching Wang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Samere Abolghasemzade
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Brendan P McKee
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Ishita Singh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Kavya Pendyala
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Mohammad Mohajeri
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Hailee Patel
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Aakansha Shaji
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Anna L Kersey
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Kajol Harsh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Simran Kaur
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Christina R Dollahon
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Sasanka Chukkapalli
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Pushkar P Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Daniel E Conway
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, USA
| | - Richard B Dickinson
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - Tanmay P Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA.
| |
Collapse
|
3
|
Ayushman M, Mikos G, Tong X, Sinha S, Lopez-Fuentes E, Jones S, Cai PC, Lee HP, Morrison AJ, Spakowitz A, Heilshorn SC, Sweet-Cordero A, Yang F. Cell tumbling enhances stem cell differentiation in hydrogels via nuclear mechanotransduction. NATURE MATERIALS 2024:10.1038/s41563-024-02038-0. [PMID: 39487316 DOI: 10.1038/s41563-024-02038-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 09/30/2024] [Indexed: 11/04/2024]
Abstract
Cells can deform their local niche in three dimensions via whole-cell movements such as spreading, migration or volume expansion. These behaviours, occurring over hours to days, influence long-term cell fates including differentiation. Here we report a whole-cell movement that occurs in sliding hydrogels at the minutes timescale, termed cell tumbling, characterized by three-dimensional cell dynamics and hydrogel deformation elicited by heightened seconds-to-minutes-scale cytoskeletal and nuclear activity. Studies inhibiting or promoting the cell tumbling of mesenchymal stem cells show that this behaviour enhances differentiation into chondrocytes. Further, it is associated with a decrease in global chromatin accessibility, which is required for enhanced differentiation. Cell tumbling also occurs during differentiation into other lineages and its differentiation-enhancing effects are validated in various hydrogel platforms. Our results establish that cell tumbling is an additional regulator of stem cell differentiation, mediated by rapid niche deformation and nuclear mechanotransduction.
Collapse
Affiliation(s)
- Manish Ayushman
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Georgios Mikos
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Xinming Tong
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sauradeep Sinha
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Eunice Lopez-Fuentes
- Division of Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Sarah Jones
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Pamela C Cai
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Hung-Pang Lee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Andrew Spakowitz
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Alejandro Sweet-Cordero
- Division of Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Fan Yang
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
4
|
Sun C, Zhao Y, Guo L, Qiu J, Peng Q. The interplay between histone modifications and nuclear lamina in genome regulation. J Genet Genomics 2024:S1673-8527(24)00277-7. [PMID: 39426590 DOI: 10.1016/j.jgg.2024.10.005] [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: 06/19/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Gene expression is regulated by chromatin architecture and epigenetic remodeling in cell homeostasis and pathologies. Histone modifications act as the key factors to modulate the chromatin accessibility. Different histone modifications are strongly associated with the localization of chromatin. Heterochromatin primarily localizes at the nuclear periphery, where it interacts with lamina proteins to suppress gene expression. In this review, we summarize the potential bridges that have regulatory functions of histone modifications in chromatin organization and transcriptional regulation at the nuclear periphery. We use lamina-associated domains (LADs) as examples to elucidate the biological roles of the interactions between histone modifications and nuclear lamina in cell differentiation and development. In the end, we highlight the technologies that are currently used to identify and visualize histone modifications and LADs, which could provide spatiotemporal information for understanding their regulatory functions in gene expression and discovering new targets for diseases.
Collapse
Affiliation(s)
- Chang Sun
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China; Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Barcelona University, Barcelona, Spain
| | - Yanjing Zhao
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China; Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Liping Guo
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, 400030, China.
| | - Qin Peng
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| |
Collapse
|
5
|
Vahidi M, Rizkalla AS, Mequanint K. Extracellular Matrix-Surrogate Advanced Functional Composite Biomaterials for Tissue Repair and Regeneration. Adv Healthc Mater 2024; 13:e2401218. [PMID: 39036851 DOI: 10.1002/adhm.202401218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/13/2024] [Indexed: 07/23/2024]
Abstract
Native tissues, comprising multiple cell types and extracellular matrix components, are inherently composites. Mimicking the intricate structure, functionality, and dynamic properties of native composite tissues represents a significant frontier in biomaterials science and tissue engineering research. Biomimetic composite biomaterials combine the benefits of different components, such as polymers, ceramics, metals, and biomolecules, to create tissue-template materials that closely simulate the structure and functionality of native tissues. While the design of composite biomaterials and their in vitro testing are frequently reviewed, there is a considerable gap in whole animal studies that provides insight into the progress toward clinical translation. Herein, we provide an insightful critical review of advanced composite biomaterials applicable in several tissues. The incorporation of bioactive cues and signaling molecules into composite biomaterials to mimic the native microenvironment is discussed. Strategies for the spatiotemporal release of growth factors, cytokines, and extracellular matrix proteins are elucidated, highlighting their role in guiding cellular behavior, promoting tissue regeneration, and modulating immune responses. Advanced composite biomaterials design challenges, such as achieving optimal mechanical properties, improving long-term stability, and integrating multifunctionality into composite biomaterials and future directions, are discussed. We believe that this manuscript provides the reader with a timely perspective on composite biomaterials.
Collapse
Affiliation(s)
- Milad Vahidi
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, N6A5B9, Canada
| | - Amin S Rizkalla
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, N6A5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, N6A5B9, Canada
| | - Kibret Mequanint
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, N6A5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, N6A5B9, Canada
| |
Collapse
|
6
|
Combémorel N, Cavell N, Tyser RCV. Early heart development: examining the dynamics of function-form emergence. Biochem Soc Trans 2024; 52:1579-1589. [PMID: 38979619 DOI: 10.1042/bst20230546] [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: 12/01/2023] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/10/2024]
Abstract
During early embryonic development, the heart undergoes a remarkable and complex transformation, acquiring its iconic four-chamber structure whilst concomitantly contracting to maintain its essential function. The emergence of cardiac form and function involves intricate interplays between molecular, cellular, and biomechanical events, unfolding with precision in both space and time. The dynamic morphological remodelling of the developing heart renders it particularly vulnerable to congenital defects, with heart malformations being the most common type of congenital birth defect (∼35% of all congenital birth defects). This mini-review aims to give an overview of the morphogenetic processes which govern early heart formation as well as the dynamics and mechanisms of early cardiac function. Moreover, we aim to highlight some of the interplay between these two processes and discuss how recent findings and emerging techniques/models offer promising avenues for future exploration. In summary, the developing heart is an exciting model to gain fundamental insight into the dynamic relationship between form and function, which will augment our understanding of cardiac congenital defects and provide a blueprint for potential therapeutic strategies to treat disease.
Collapse
Affiliation(s)
- Noémie Combémorel
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, U.K
| | - Natasha Cavell
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, U.K
| | - Richard C V Tyser
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, U.K
| |
Collapse
|
7
|
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
|
8
|
Gao K, Xie Y, Xu F, Peng Q, Fu L, Wang G, Qiu J. Silk fibroin promotes H3K9me3 expression and chromatin reorganization to regulate endothelial cell proliferation. APL Bioeng 2024; 8:026115. [PMID: 38827498 PMCID: PMC11143938 DOI: 10.1063/5.0203858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/07/2024] [Indexed: 06/04/2024] Open
Abstract
Silk fibroin (SF), which is extensively utilized in tissue engineering and vascular grafts for enhancing vascular regeneration, has not been thoroughly investigated for its epigenetic effects on endothelial cells (EC). This study employed RNA sequencing analysis to evaluate the activation of histone modification regulatory genes in EC treated with SF. Subsequent investigations revealed elevated H3K9me3 levels in SF-treated EC, as evidenced by immunofluorescence and western blot analysis. The study utilized H2B-eGFP endothelial cells to demonstrate that SF treatment results in the accumulation of H2B-marked chromatin in the nuclear inner cavities of EC. Inhibition of H3K9me3 levels by a histone deacetylase inhibitor TSA decreased cell proliferation. Furthermore, the activation of the MAPK signaling pathway using chromium picolinate decreased the proliferative activity and H3K9me3 level in SF-treated EC. SF also appeared to enhance cell growth and proliferation by modulating the H3K9me3 level and reorganizing chromatin, particularly after oxidative stress induced by H2O2 treatment. In summary, these findings indicate that SF promotes EC proliferation by increasing the H3K9me3 level even under stress conditions.
Collapse
Affiliation(s)
- Kaixiang Gao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yafan Xie
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Fangning Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Qin Peng
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Li Fu
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Guixue Wang
- Authors to whom correspondence should be addressed:; ; and . Tel.: 023-65102507
| | - Juhui Qiu
- Authors to whom correspondence should be addressed:; ; and . Tel.: 023-65102507
| |
Collapse
|
9
|
Bougaran P, Bautch VL. Life at the crossroads: the nuclear LINC complex and vascular mechanotransduction. Front Physiol 2024; 15:1411995. [PMID: 38831796 PMCID: PMC11144885 DOI: 10.3389/fphys.2024.1411995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
Vascular endothelial cells line the inner surface of all blood vessels, where they are exposed to polarized mechanical forces throughout their lifespan. Both basal substrate interactions and apical blood flow-induced shear stress regulate blood vessel development, remodeling, and maintenance of vascular homeostasis. Disruption of these interactions leads to dysfunction and vascular pathologies, although how forces are sensed and integrated to affect endothelial cell behaviors is incompletely understood. Recently the endothelial cell nucleus has emerged as a prominent force-transducing organelle that participates in vascular mechanotransduction, via communication to and from cell-cell and cell-matrix junctions. The LINC complex, composed of SUN and nesprin proteins, spans the nuclear membranes and connects the nuclear lamina, the nuclear envelope, and the cytoskeleton. Here we review LINC complex involvement in endothelial cell mechanotransduction, describe unique and overlapping functions of each LINC complex component, and consider emerging evidence that two major SUN proteins, SUN1 and SUN2, orchestrate a complex interplay that extends outward to cell-cell and cell-matrix junctions and inward to interactions within the nucleus and chromatin. We discuss these findings in relation to vascular pathologies such as Hutchinson-Gilford progeria syndrome, a premature aging disorder with cardiovascular impairment. More knowledge of LINC complex regulation and function will help to understand how the nucleus participates in endothelial cell force sensing and how dysfunction leads to cardiovascular disease.
Collapse
Affiliation(s)
- Pauline Bougaran
- Department of Biology, The University of North Carolina, Chapel Hill, NC, United States
| | - Victoria L. Bautch
- Department of Biology, The University of North Carolina, Chapel Hill, NC, United States
- McAllister Heart Institute, The University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
10
|
Sen B, Xie Z, Thomas MD, Pattenden SG, Howard S, McGrath C, Styner M, Uzer G, Furey TS, Rubin J. Nuclear actin structure regulates chromatin accessibility. Nat Commun 2024; 15:4095. [PMID: 38750021 PMCID: PMC11096319 DOI: 10.1038/s41467-024-48580-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 05/07/2024] [Indexed: 05/18/2024] Open
Abstract
Polymerized β-actin may provide a structural basis for chromatin accessibility and actin transport into the nucleus can guide mesenchymal stem cell (MSC) differentiation. Using MSC, we show that using CK666 to inhibit Arp2/3 directed secondary actin branching results in decreased nuclear actin structure, and significantly alters chromatin access measured with ATACseq at 24 h. The ATAC-seq results due to CK666 are distinct from those caused by cytochalasin D (CytoD), which enhances nuclear actin structure. In addition, nuclear visualization shows Arp2/3 inhibition decreases pericentric H3K9me3 marks. CytoD, alternatively, induces redistribution of H3K27me3 marks centrally. Such alterations in chromatin landscape are consistent with differential gene expression associated with distinctive differentiation patterns. Further, knockdown of the non-enzymatic monomeric actin binding protein, Arp4, leads to extensive chromatin unpacking, but only a modest increase in transcription, indicating an active role for actin-Arp4 in transcription. These data indicate that dynamic actin remodeling can regulate chromatin interactions.
Collapse
Affiliation(s)
- Buer Sen
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Zhihui Xie
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Michelle D Thomas
- Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Samantha G Pattenden
- Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sean Howard
- Department of Mechanical and Biomedical Engineering, Boise State University, Boise, ID, USA
| | - Cody McGrath
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Maya Styner
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Gunes Uzer
- Department of Mechanical and Biomedical Engineering, Boise State University, Boise, ID, USA
| | - Terrence S Furey
- Departments of Genetics and Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Janet Rubin
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
11
|
Sarıyer OS, Erbaş A. Polymer physics view of peripheral chromatin: de Gennes' self-similar carpet. Phys Rev E 2024; 109:054403. [PMID: 38907468 DOI: 10.1103/physreve.109.054403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/09/2024] [Indexed: 06/24/2024]
Abstract
Using scaling arguments to model peripheral chromatin localized near the inner surface of the nuclear envelope (NE) as a flexible polymer chain, we discuss the structural properties of the peripheral chromatin composed of alternating lamin-associated domains (LADs) and inter-LADs. Modeling the attraction of LADs to NE by de Gennes' self-similar carpet, which treats the chromatin layer as a polymer fractal, explains two major experimental observations. (i) The high density of chromatin close to the nuclear periphery decays to a constant density as the distance to the periphery increases. (ii) Due to the decreasing mesh size towards the nuclear periphery, the chromatin carpet inside NE excludes molecules (via nonspecific interactions) above a threshold size that depends on the distance from the nuclear periphery.
Collapse
Affiliation(s)
- Ozan S Sarıyer
- Pîrî Reis University, School of Arts and Sciences, Tuzla 34940, Istanbul, Turkey
| | - Aykut Erbaş
- UNAM National Nanotechnology Research Center and Institute of Materials Science & Nanotechnology, Bilkent University, Ankara 06800, Turkey and University of Silesia, Institute of Physics, 41-500 Katowice, Poland
| |
Collapse
|
12
|
Biggs LC, Miroshnikova YA. Nuclear mechanotransduction on skin stem cell fate regulation. Curr Opin Cell Biol 2024; 87:102328. [PMID: 38340567 DOI: 10.1016/j.ceb.2024.102328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
Mammalian skin is a highly dynamic and regenerative organ that has long been recognized as a mechanically active composite of tissues withstanding daily compressive and tensile forces that arise from body movement. Importantly, cell- and tissue-scale mechanical signals are critical regulators of skin morphogenesis and homeostasis. These signals are sensed at the cellular periphery and transduced by mechanosensitive proteins within the plasma membrane to the cytoskeletal networks, and eventually into the nucleus to regulate chromatin organization and gene expression. The role of each of these nodes in producing a coherent mechanoresponse at both cell- and tissue-scales is emerging. Here we focus on the key cytoplasmic and nuclear mechanosensitive structures that are critical for the mammalian skin development and homeostatic maintenance. We propose that the mechanical state of the skin, in particular of its nuclear compartment, is a critical rheostat that fine-tunes developmental and homeostatic processes essential for the proper function of the organ.
Collapse
Affiliation(s)
- Leah C Biggs
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany.
| | - Yekaterina A Miroshnikova
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
13
|
Otsuka A, Minami K, Higashi K, Kawaguchi A, Tamura S, Ide S, Hendzel MJ, Kurokawa K, Maeshima K. Chromatin organization and behavior in HRAS-transformed mouse fibroblasts. Chromosoma 2024; 133:135-148. [PMID: 38400910 DOI: 10.1007/s00412-024-00817-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/24/2024] [Accepted: 02/05/2024] [Indexed: 02/26/2024]
Abstract
In higher eukaryotic cells, a string of nucleosomes, where long genomic DNA is wrapped around core histones, are rather irregularly folded into a number of condensed chromatin domains, which have been revealed by super-resolution imaging and Hi-C technologies. Inside these domains, nucleosomes fluctuate and locally behave like a liquid. The behavior of chromatin may be highly related to DNA transaction activities such as transcription and repair, which are often upregulated in cancer cells. To investigate chromatin behavior in cancer cells and compare those of cancer and non-cancer cells, we focused on oncogenic-HRAS (Gly12Val)-transformed mouse fibroblasts CIRAS-3 cells and their parental 10T1/2 cells. CIRAS-3 cells are tumorigenic and highly metastatic. First, we found that HRAS-induced transformation altered not only chromosome structure, but also nuclear morphology in the cell. Using single-nucleosome imaging/tracking in live cells, we demonstrated that nucleosomes are locally more constrained in CIRAS-3 cells than in 10T1/2 cells. Consistently, heterochromatin marked with H3K27me3 was upregulated in CIRAS-3 cells. Finally, Hi-C analysis showed enriched interactions of the B-B compartment in CIRAS-3 cells, which likely represents transcriptionally inactive chromatin. Increased heterochromatin may play an important role in cell migration, as they have been reported to increase during metastasis. Our study also suggests that single-nucleosome imaging provides new insights into how local chromatin is structured in living cells.
Collapse
Affiliation(s)
- Aoi Otsuka
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Shizuoka, 411-8540, Japan
| | - Katsuhiko Minami
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Shizuoka, 411-8540, Japan
| | - Koichi Higashi
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Shizuoka, 411-8540, Japan
- Genome Evolution Laboratory, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan
| | - Akane Kawaguchi
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Shizuoka, 411-8540, Japan
- Molecular Life History Laboratory, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan
| | - Sachiko Tamura
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan
| | - Satoru Ide
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Shizuoka, 411-8540, Japan
| | - Michael J Hendzel
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ken Kurokawa
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Shizuoka, 411-8540, Japan
- Genome Evolution Laboratory, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan
| | - Kazuhiro Maeshima
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan.
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Shizuoka, 411-8540, Japan.
| |
Collapse
|
14
|
Swiatlowska P, Iskratsch T. Cardiovascular Mechano-Epigenetics: Force-Dependent Regulation of Histone Modifications and Gene Regulation. Cardiovasc Drugs Ther 2024; 38:215-222. [PMID: 36653625 PMCID: PMC10959834 DOI: 10.1007/s10557-022-07422-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/28/2022] [Indexed: 01/19/2023]
Abstract
The local mechanical microenvironment impacts on the cell behavior. In the cardiovascular system, cells in both the heart and the vessels are exposed to continuous blood flow, blood pressure, stretching forces, and changing extracellular matrix stiffness. The force-induced signals travel all the way to the nucleus regulating epigenetic changes such as chromatin dynamics and gene expression. Mechanical cues are needed at the very early stage for a faultless embryological development, while later in life, aberrant mechanical signaling can lead to a range of pathologies, including diverse cardiovascular diseases. Hence, an investigation of force-generated epigenetic alteration at different time scales is needed to understand fully the phenotypic changes in disease onset and progression. That being so, cardiovascular mechano-epigenetics emerges as an attractive field of study. Given the rapid advances in this emergent field of research, this short review aims to provide an analysis of the state of knowledge of force-induced epigenetic changes in the cardiovascular field.
Collapse
Affiliation(s)
- Pamela Swiatlowska
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Thomas Iskratsch
- School of Engineering and Materials Science, Queen Mary University of London, London, UK.
| |
Collapse
|
15
|
Lee GE, Bang G, Byun J, Lee CJ, Chen W, Jeung D, An HJ, Kang HC, Lee JY, Lee HS, Hong YS, Kim DJ, Keniry M, Kim JY, Choi JS, Fanto M, Cho SJ, Kim KD, Cho YY. Dysregulated CREB3 cleavage at the nuclear membrane induces karyoptosis-mediated cell death. Exp Mol Med 2024; 56:686-699. [PMID: 38480902 PMCID: PMC10985101 DOI: 10.1038/s12276-024-01195-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/17/2023] [Accepted: 01/02/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer cells often exhibit resistance to apoptotic cell death, but they may be vulnerable to other types of cell death. Elucidating additional mechanisms that govern cancer cell death is crucial for developing new therapies. Our research identified cyclic AMP-responsive element-binding protein 3 (CREB3) as a crucial regulator and initiator of a unique cell death mechanism known as karyoptosis. This process is characterized by nuclear shrinkage, deformation, and the loss of nuclear components following nuclear membrane rupture. We found that the N-terminal domain (aa 1-230) of full-length CREB3 (CREB3-FL), which is anchored to the nuclear inner membrane (INM), interacts with lamins and chromatin DNA. This interaction maintains a balance between the outward force exerted by tightly packed DNA and the inward constraining force, thereby preserving INM integrity. Under endoplasmic reticulum (ER) stress, aberrant cleavage of CREB3-FL at the INM leads to abnormal accumulation of the cleaved form of CREB3 (CREB3-CF). This accumulation disrupts the attachment of CREB3-FL to the INM, resulting in sudden rupture of the nuclear membrane and the onset of karyoptosis. Proteomic studies revealed that CREB3-CF overexpression induces a DNA damage response akin to that caused by UVB irradiation, which is associated with cellular senescence in cancer cells. These findings demonstrated that the dysregulation of CREB3-FL cleavage is a key factor in karyoptotic cell death. Consequently, these findings suggest new therapeutic strategies in cancer treatment that exploit the process of karyoptosis.
Collapse
Affiliation(s)
- Ga-Eun Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Geul Bang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, Cheongju-si, Chungbuk, 28119, Republic of Korea
| | - Jiin Byun
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Cheol-Jung Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- Research Center for Materials Analysis, Korea Basic Science Institute, Daejeon, 34133, Republic of Korea
| | - Weidong Chen
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Dohyun Jeung
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Hyun-Jung An
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Han Chang Kang
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Joo Young Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Hye Suk Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Young-Soo Hong
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Chungbuk, 28116, Republic of Korea
| | - Dae Joon Kim
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, 78504, USA
| | - Megan Keniry
- Department of Biology, University of Texas Rio Grande Valley, Edinburg, TX, 78539, USA
| | - Jin Young Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, Cheongju-si, Chungbuk, 28119, Republic of Korea
| | - Jin-Sung Choi
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Manolis Fanto
- Department of Basic and Clinical Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Sung-Jun Cho
- University of Minnesota, Department of Medicine, 420 Delaware St SE, MMC 284, Minneapolis, MN, 55455, USA
| | - Kwang-Dong Kim
- Division of Applied Life Science (BK21 four), PMBBRC, Gyeongsang National University, Jinju, 52828, Korea
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea.
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea.
| |
Collapse
|
16
|
Sobo JM, Alagna NS, Sun SX, Wilson KL, Reddy KL. Lamins: The backbone of the nucleocytoskeleton interface. Curr Opin Cell Biol 2024; 86:102313. [PMID: 38262116 DOI: 10.1016/j.ceb.2023.102313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/25/2024]
Abstract
The nuclear lamina (NL) is a crucial component of the inner nuclear membrane (INM) and consists of lamin filaments and associated proteins. Lamins are type V intermediate filament proteins essential for maintaining the integrity and mechanical properties of the nucleus. In human cells, 'B-type' lamins (lamin B1 and lamin B2) are ubiquitously expressed, while 'A-type' lamins (lamin A, lamin C, and minor isoforms) are expressed in a tissue- and development-specific manner. Lamins homopolymerize to form filaments that localize primarily near the INM, but A-type lamins also localize to and function in the nucleoplasm. Lamins play central roles in the assembly, structure, positioning, and mechanics of the nucleus, modulating cell signaling and influencing development, differentiation, and other activities. This review highlights recent findings on the structure and regulation of lamin filaments, providing insights into their multifaceted functions, including their role as "mechanosensors", delving into the emerging significance of lamin filaments as vital links between cytoskeletal and nuclear structures, chromatin organization, and the genome.
Collapse
Affiliation(s)
- Joan M Sobo
- Department of Biological Chemistry, Center for Epigenetics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Nicholas S Alagna
- Department of Biological Chemistry, Center for Epigenetics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sean X Sun
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Katherine L Wilson
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Karen L Reddy
- Department of Biological Chemistry, Center for Epigenetics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
17
|
Kaonis S, Aboellail Z, Forman J, Ghosh S. High-Throughput Multiparametric Quantification of Mechanics Driven Heterogeneity in Mesenchymal Stromal Cell Population. Adv Biol (Weinh) 2024; 8:e2300318. [PMID: 37840408 DOI: 10.1002/adbi.202300318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Indexed: 10/17/2023]
Abstract
Mesenchymal stromal or stem cells (MSCs) are one of the most promising candidates for a myriad of cell therapy applications. Despite showing promise in numerous preclinical and clinical studies, MSC-based therapy is not yet a reality for regenerative medicine due to its suboptimal outcome at the clinical endpoint. The mechanical environment is a critical determinant of MSC gene expression and function. This study reports that MSC population becomes phenotypically heterogenous and commits to an unwanted osteoprogenitor pathway when it experiences an abnormal mechanically stiff environment, compared to its native softer environment. A method is developed to measure the heterogeneity using nuclear shape, chromatin state, and CD73 marker. Heterogeneity is shown to be associated with a larger spread in the nuclear shape parameters and a smaller spread in the chromatin openness. Subsequently, intervention strategies are investigated to create a more homogeneous MSC population. Culturing MSCs on soft surfaces or inhibiting actomyosin on stiff surfaces can make them more homogeneous, while inhibiting YAP, Runx2, and actin polymerization helps maintain but does not fully homogenize them. This study offers insights for cell and tissue engineers, aiding in the design of optimal conditions and materials for MSC culture, ultimately enhancing their therapeutic potential.
Collapse
Affiliation(s)
- Samantha Kaonis
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
| | - Zack Aboellail
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
| | - Jack Forman
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
| | - Soham Ghosh
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Mechanical Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
- Cell and Molecular Biology, Colorado State University, 1050 Libbie Coy Way, Fort Collins, CO, 80524, USA
| |
Collapse
|
18
|
Mei Y, Feng X, Jin Y, Kang R, Wang X, Zhao D, Ghosh S, Neu CP, Avril S. Cell nucleus elastography with the adjoint-based inverse solver. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 242:107827. [PMID: 37801883 DOI: 10.1016/j.cmpb.2023.107827] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/09/2023] [Accepted: 09/22/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND AND OBJECTIVES The mechanics of the nucleus depends on cellular structures and architecture, and impact a number of diseases. Nuclear mechanics is yet rather complex due to heterogeneous distribution of dense heterochromatin and loose euchromatin domains, giving rise to spatially variable stiffness properties. METHODS In this study, we propose to use the adjoint-based inverse solver to identify for the first time the nonhomogeneous elastic property distribution of the nucleus. Inputs of the inverse solver are deformation fields measured with microscopic imaging in contracting cardiomyocytes. RESULTS The feasibility of the proposed method is first demonstrated using simulated data. Results indicate accurate identification of the assumed heterochromatin region, with a maximum relative error of less than 5%. We also investigate the influence of unknown Poisson's ratio on the reconstruction and find that variations of the Poisson's ratio in the range [0.3-0.5] result in uncertainties of less than 15% in the identified stiffness. Finally, we apply the inverse solver on actual deformation fields acquired within the nuclei of two cardiomyocytes. The obtained results are in good agreement with the density maps obtained from microscopy images. CONCLUSIONS Overall, the proposed approach shows great potential for nuclear elastography, with promising value for emerging fields of mechanobiology and mechanogenetics.
Collapse
Affiliation(s)
- Yue Mei
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116023, China; International Research Center for Computational Mechanics, Dalian University of Technology, Dalian 116023, China; Ningbo Institute of Dalian University of Technology, No. 26 Yucai Road, Jiangbei District, Ningbo 315016, China
| | - Xuan Feng
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116023, China; International Research Center for Computational Mechanics, Dalian University of Technology, Dalian 116023, China
| | - Yun Jin
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116023, China; International Research Center for Computational Mechanics, Dalian University of Technology, Dalian 116023, China
| | - Rongyao Kang
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116023, China; International Research Center for Computational Mechanics, Dalian University of Technology, Dalian 116023, China
| | - XinYu Wang
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116023, China; International Research Center for Computational Mechanics, Dalian University of Technology, Dalian 116023, China
| | - Dongmei Zhao
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116023, China; International Research Center for Computational Mechanics, Dalian University of Technology, Dalian 116023, China
| | - Soham Ghosh
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States of America
| | - Corey P Neu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States of America; Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, United States of America; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, United States of America
| | - Stephane Avril
- Mines Saint-Étienne, Univ Jean Monnet, INSERM, U 1059 Sainbiose, F - 42023, Saint-Étienne, France.
| |
Collapse
|
19
|
Serdyukova K, Swearingen AR, Coradin M, Nevo M, Tran H, Bajric E, Brumbaugh J. Leveraging dominant-negative histone H3 K-to-M mutations to study chromatin during differentiation and development. Development 2023; 150:dev202169. [PMID: 37846748 PMCID: PMC10617616 DOI: 10.1242/dev.202169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Histone modifications are associated with regulation of gene expression that controls a vast array of biological processes. Often, these associations are drawn by correlating the genomic location of a particular histone modification with gene expression or phenotype; however, establishing a causal relationship between histone marks and biological processes remains challenging. Consequently, there is a strong need for experimental approaches to directly manipulate histone modifications. A class of mutations on the N-terminal tail of histone H3, lysine-to-methionine (K-to-M) mutations, was identified as dominant-negative inhibitors of histone methylation at their respective and specific residues. The dominant-negative nature of K-to-M mutants makes them a valuable tool for studying the function of specific methylation marks on histone H3. Here, we review recent applications of K-to-M mutations to understand the role of histone methylation during development and homeostasis. We highlight important advantages and limitations that require consideration when using K-to-M mutants, particularly in a developmental context.
Collapse
Affiliation(s)
- Ksenia Serdyukova
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Alison R. Swearingen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mariel Coradin
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mika Nevo
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Huong Tran
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Emir Bajric
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Justin Brumbaugh
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
20
|
Wang X, Agrawal V, Dunton CL, Liu Y, Virk RKA, Patel PA, Carter L, Pujadas EM, Li Y, Jain S, Wang H, Ni N, Tsai HM, Rivera-Bolanos N, Frederick J, Roth E, Bleher R, Duan C, Ntziachristos P, He TC, Reid RR, Jiang B, Subramanian H, Backman V, Ameer GA. Chromatin reprogramming and bone regeneration in vitro and in vivo via the microtopography-induced constriction of cell nuclei. Nat Biomed Eng 2023; 7:1514-1529. [PMID: 37308586 PMCID: PMC10804399 DOI: 10.1038/s41551-023-01053-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/10/2023] [Indexed: 06/14/2023]
Abstract
Topographical cues on cells can, through contact guidance, alter cellular plasticity and accelerate the regeneration of cultured tissue. Here we show how changes in the nuclear and cellular morphologies of human mesenchymal stromal cells induced by micropillar patterns via contact guidance influence the conformation of the cells' chromatin and their osteogenic differentiation in vitro and in vivo. The micropillars impacted nuclear architecture, lamin A/C multimerization and 3D chromatin conformation, and the ensuing transcriptional reprogramming enhanced the cells' responsiveness to osteogenic differentiation factors and decreased their plasticity and off-target differentiation. In mice with critical-size cranial defects, implants with micropillar patterns inducing nuclear constriction altered the cells' chromatin conformation and enhanced bone regeneration without the need for exogenous signalling molecules. Our findings suggest that medical device topographies could be designed to facilitate bone regeneration via chromatin reprogramming.
Collapse
Affiliation(s)
- Xinlong Wang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Vasundhara Agrawal
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Cody L Dunton
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Yugang Liu
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Ranya K A Virk
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Priyam A Patel
- Quantitative Data Science Core, Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lucas Carter
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Emily M Pujadas
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Yue Li
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Surbhi Jain
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Hao Wang
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Na Ni
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Hsiu-Ming Tsai
- Department of Radiology, The University of Chicago, Chicago, IL, USA
| | - Nancy Rivera-Bolanos
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Jane Frederick
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Eric Roth
- Department of Materials Sciences and Engineering, Northwestern University, Evanston, IL, USA
| | - Reiner Bleher
- Department of Materials Sciences and Engineering, Northwestern University, Evanston, IL, USA
| | - Chongwen Duan
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Panagiotis Ntziachristos
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
- Simpson Querrey Center for Epigenetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Tong Chuan He
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Russell R Reid
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL, USA
| | - Bin Jiang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hariharan Subramanian
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Vadim Backman
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA.
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| | - Guillermo A Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA.
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA.
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Chemistry of Life Process Institute, Northwestern University, Chicago, IL, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
- Simpson Querrey Institute for Bionanotechnology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
21
|
Negri ML, D'Annunzio S, Vitali G, Zippo A. May the force be with you: Nuclear condensates function beyond transcription control: Potential nongenetic functions of nuclear condensates in physiological and pathological conditions. Bioessays 2023; 45:e2300075. [PMID: 37530178 DOI: 10.1002/bies.202300075] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/03/2023] [Accepted: 07/13/2023] [Indexed: 08/03/2023]
Abstract
Over the past decade, research has revealed biomolecular condensates' relevance in diverse cellular functions. Through a phase separation process, they concentrate macromolecules in subcompartments shaping the cellular organization and physiology. In the nucleus, biomolecular condensates assemble relevant biomolecules that orchestrate gene expression. We here hypothesize that chromatin condensates can also modulate the nongenetic functions of the genome, including the nuclear mechanical properties. The importance of chromatin condensates is supported by the genetic evidence indicating that mutations in their members are causative of a group of rare Mendelian diseases named chromatinopathies (CPs). Despite a broad spectrum of clinical features and the perturbations of the epigenetic machinery characterizing the CPs, recent findings highlighted negligible changes in gene expression. These data argue in favor of possible noncanonical functions of chromatin condensates in regulating the genome's spatial organization and, consequently, the nuclear mechanics. In this review, we discuss how condensates may impact nuclear mechanical properties, thus affecting the cellular response to mechanical cues and, eventually, cell fate and identity. Chromatin condensates organize macromolecules in the nucleus orchestrating the transcription regulation and mutations in their members are responsible for rare diseases named chromatinopathies. We argue that chromatin condensates, in concert with the nuclear lamina, may also govern the nuclear mechanical properties affecting the cellular response to external cues.
Collapse
Affiliation(s)
- Maria Luce Negri
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Sarah D'Annunzio
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Giulia Vitali
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Alessio Zippo
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| |
Collapse
|
22
|
Woodhams LG, Guo J, Schuftan D, Boyle JJ, Pryse KM, Elson EL, Huebsch N, Genin GM. Virtual blebbistatin: A robust and rapid software approach to motion artifact removal in optical mapping of cardiomyocytes. Proc Natl Acad Sci U S A 2023; 120:e2212949120. [PMID: 37695908 PMCID: PMC10515162 DOI: 10.1073/pnas.2212949120] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 07/31/2023] [Indexed: 09/13/2023] Open
Abstract
Fluorescent reporters of cardiac electrophysiology provide valuable information on heart cell and tissue function. However, motion artifacts caused by cardiac muscle contraction interfere with accurate measurement of fluorescence signals. Although drugs such as blebbistatin can be applied to stop cardiac tissue from contracting by uncoupling calcium-contraction, their usage prevents the study of excitation-contraction coupling and, as we show, impacts cellular structure. We therefore developed a robust method to remove motion computationally from images of contracting cardiac muscle and to map fluorescent reporters of cardiac electrophysiological activity onto images of undeformed tissue. When validated on cardiomyocytes derived from human induced pluripotent stem cells (iPSCs), in both monolayers and engineered tissues, the method enabled efficient and robust reduction of motion artifact. As with pharmacologic approaches using blebbistatin for motion removal, our algorithm improved the accuracy of optical mapping, as demonstrated by spatial maps of calcium transient decay. However, unlike pharmacologic motion removal, our computational approach allowed direct analysis of calcium-contraction coupling. Results revealed calcium-contraction coupling to be more uniform across cells within engineered tissues than across cells in monolayer culture. The algorithm shows promise as a robust and accurate tool for optical mapping studies of excitation-contraction coupling in heart tissue.
Collapse
Affiliation(s)
- Louis G Woodhams
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, St. Louis, MO 63130
| | - Jingxuan Guo
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, St. Louis, MO 63130
| | - David Schuftan
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO 63130
| | - John J Boyle
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO 63130
| | - Kenneth M Pryse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Elliot L Elson
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in Saint Louis, St. Louis, MO 63130
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO 63130
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in Saint Louis, St. Louis, MO 63130
| | - Guy M Genin
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, St. Louis, MO 63130
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in Saint Louis, St. Louis, MO 63130
| |
Collapse
|
23
|
Donnaloja F, Raimondi MT, Messa L, Barzaghini B, Carnevali F, Colombo E, Mazza D, Martinelli C, Boeri L, Rey F, Cereda C, Osellame R, Cerullo G, Carelli S, Soncini M, Jacchetti E. 3D photopolymerized microstructured scaffolds influence nuclear deformation, nucleo/cytoskeletal protein organization, and gene regulation in mesenchymal stem cells. APL Bioeng 2023; 7:036112. [PMID: 37692376 PMCID: PMC10491463 DOI: 10.1063/5.0153215] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023] Open
Abstract
Mechanical stimuli from the extracellular environment affect cell morphology and functionality. Recently, we reported that mesenchymal stem cells (MSCs) grown in a custom-made 3D microscaffold, the Nichoid, are able to express higher levels of stemness markers. In fact, the Nichoid is an interesting device for autologous MSC expansion in clinical translation and would appear to regulate gene activity by altering intracellular force transmission. To corroborate this hypothesis, we investigated mechanotransduction-related nuclear mechanisms, and we also treated spread cells with a drug that destroys the actin cytoskeleton. We observed a roundish nuclear shape in MSCs cultured in the Nichoid and correlated the nuclear curvature with the import of transcription factors. We observed a more homogeneous euchromatin distribution in cells cultured in the Nichoid with respect to the Flat sample, corresponding to a standard glass coverslip. These results suggest a different gene regulation, which we confirmed by an RNA-seq analysis that revealed the dysregulation of 1843 genes. We also observed a low structured lamina mesh, which, according to the implemented molecular dynamic simulations, indicates reduced damping activity, thus supporting the hypothesis of low intracellular force transmission. Also, our investigations regarding lamin expression and spatial organization support the hypothesis that the gene dysregulation induced by the Nichoid is mainly related to a reduction in force transmission. In conclusion, our findings revealing the Nichoid's effects on MSC behavior is a step forward in the control of stem cells via mechanical manipulation, thus paving the way to new strategies for MSC translation to clinical applications.
Collapse
Affiliation(s)
- Francesca Donnaloja
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | | | - Bianca Barzaghini
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Federica Carnevali
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | | | - Davide Mazza
- Istituto Scientifico Ospedale San Raffaele, Centro di Imaging Sperimentale, Milan, Italy
| | - Chiara Martinelli
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Lucia Boeri
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Federica Rey
- Pediatric Research Center “Romeo ed Enrica Invernizzi,” Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Cristina Cereda
- Center of Functional Genomic and Rare Diseases, “V. Buzzi” Children's Hospital, 20154 Milan, Italy
| | - Roberto Osellame
- Institute for Photonics and Nanotechnologies—CNR, and Physics Department, Politecnico di Milano, Milan, Italy
| | - Giulio Cerullo
- Institute for Photonics and Nanotechnologies—CNR, and Physics Department, Politecnico di Milano, Milan, Italy
| | | | - Monica Soncini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| |
Collapse
|
24
|
严 然, 陈 祥, 张 译, 王 梦, 李 顺, 刘 贻. [Advances in cell nuclear mechanobiology and its regulation mechanisms]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2023; 40:617-624. [PMID: 37666750 PMCID: PMC10477395 DOI: 10.7507/1001-5515.202304036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/09/2023] [Indexed: 09/06/2023]
Abstract
As an important intracellular genetic and regulatory center, the nucleus is not only a terminal effector of intracellular biochemical signals, but also has a significant impact on cell function and phenotype through direct or indirect regulation of nuclear mechanistic cues after the cell senses and responds to mechanical stimuli. The nucleus relies on chromatin-nuclear membrane-cytoskeleton infrastructure to couple signal transduction, and responds to these mechanical stimuli in the intracellular and extracellular physical microenvironments. Changes in the morphological structure of the nucleus are the most intuitive manifestation of this mechanical response cascades and are the basis for the direct response of the nucleus to mechanical stimuli. Based on such relationships of the nucleus with cell behavior and phenotype, abnormal nuclear morphological changes are widely used in clinical practice as disease diagnostic tools. This review article highlights the latest advances in how nuclear morphology responds and adapts to mechanical stimuli. Additionally, this article will shed light on the factors that mechanically regulate nuclear morphology as well as the tumor physio-pathological processes involved in nuclear morphology and the underlying mechanobiological mechanisms. It provides new insights into the mechanisms that nuclear mechanics regulates disease development and its use as a potential target for diagnosis and treatment.
Collapse
Affiliation(s)
- 然 严
- 电子科技大学 生命科学与技术学院(成都 610054)School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
- 成都中医药大学附属医院(成都 610072)Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, P. R. China
| | - 祥燕 陈
- 电子科技大学 生命科学与技术学院(成都 610054)School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
| | - 译兮 张
- 电子科技大学 生命科学与技术学院(成都 610054)School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
| | - 梦 王
- 电子科技大学 生命科学与技术学院(成都 610054)School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
| | - 顺 李
- 电子科技大学 生命科学与技术学院(成都 610054)School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
| | - 贻尧 刘
- 电子科技大学 生命科学与技术学院(成都 610054)School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
- 成都中医药大学附属医院(成都 610072)Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, P. R. China
| |
Collapse
|
25
|
Almeida JA, Mathur J, Lee YL, Sarker B, Pathak A. Mechanically primed cells transfer memory to fibrous matrices for invasion across environments of distinct stiffness and dimensionality. Mol Biol Cell 2023; 34:ar54. [PMID: 36696158 PMCID: PMC10208097 DOI: 10.1091/mbc.e22-10-0469] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/04/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Cells sense and migrate across mechanically dissimilar environments throughout development and disease progression. However, it remains unclear whether mechanical memory of past environments empowers cells to navigate new, three-dimensional extracellular matrices. Here, we show that cells previously primed on stiff, compared with soft, matrices generate a higher level of forces to remodel collagen fibers and promote invasion. This priming advantage persists in dense or stiffened collagen. We explain this memory-dependent, cross-environment cell invasion through a lattice-based model wherein stiff-primed cellular forces remodel collagen and minimize energy required for future cell invasion. According to our model, cells transfer their mechanical memory to the matrix via collagen alignment and tension, and this remodeled matrix informs future cell invasion. Thus, memory-laden cells overcome mechanosensing of softer or challenging future environments via a cell-matrix transfer of memory. Consistent with model predictions, depletion of yes-associated protein destabilizes the cellular memory required for collagen remodeling before invasion. We release tension in collagen fibers via laser ablation and disable fiber remodeling by lysyl-oxidase inhibition, both of which disrupt cell-to-matrix transfer of memory and hamper cross-environment invasion. These results have implications for cancer, fibrosis, and aging, where a potential cell-to-matrix transfer of mechanical memory of cells may generate a prolonged cellular response.
Collapse
Affiliation(s)
- José A. Almeida
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
| | - Jairaj Mathur
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, MO 63130
| | - Ye Lim Lee
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
| | - Bapi Sarker
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, MO 63130
| | - Amit Pathak
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, MO 63130
| |
Collapse
|
26
|
Shah PP, Santini GT, Shen KM, Jain R. InterLINCing Chromatin Organization and Mechanobiology in Laminopathies. Curr Cardiol Rep 2023; 25:307-314. [PMID: 37052760 PMCID: PMC10185580 DOI: 10.1007/s11886-023-01853-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2023] [Indexed: 04/14/2023]
Abstract
PURPOSE OF REVIEW In this review, we explore the chromatin-related consequences of laminopathy-linked mutations through the lens of mechanotransduction. RECENT FINDINGS Multiple studies have highlighted the role of the nuclear lamina in maintaining the integrity of the nucleus. The lamina also has a critical role in 3D genome organization. Mutations in lamina proteins associated with various laminopathies result in the loss of organization of DNA at the nuclear periphery. However, it remains unclear if or how these two aspects of lamin function are connected. Recent data suggests that unlinking the cytoskeleton from the nuclear lamina may be beneficial to slow progress of deleterious phenotypes observed in laminopathies. In this review, we highlight emerging data that suggest interlinked chromatin- and mechanical biology-related pathways are interconnected in the pathogenesis of laminopathies.
Collapse
Affiliation(s)
- Parisha P. Shah
- Departments of Medicine and Cell and Developmental Biology, Penn Cardiovascular Institute, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
- Smilow Center for Translational Research, 09-184, 3400 Civic Center Blvd., Philadelphia, PA 19104 USA
| | - Garrett T. Santini
- Departments of Medicine and Cell and Developmental Biology, Penn Cardiovascular Institute, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Kaitlyn M. Shen
- Departments of Medicine and Cell and Developmental Biology, Penn Cardiovascular Institute, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Rajan Jain
- Departments of Medicine and Cell and Developmental Biology, Penn Cardiovascular Institute, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
- Smilow Center for Translational Research, 09-101, 3400 Civic Center Blvd., Philadelphia, PA 19104 USA
| |
Collapse
|
27
|
Scott AK, Casas E, Schneider SE, Swearingen AR, Van Den Elzen CL, Seelbinder B, Barthold JE, Kugel JF, Stern JL, Foster KJ, Emery NC, Brumbaugh J, Neu CP. Mechanical memory stored through epigenetic remodeling reduces cell therapeutic potential. Biophys J 2023; 122:1428-1444. [PMID: 36871159 PMCID: PMC10147835 DOI: 10.1016/j.bpj.2023.03.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/31/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Understanding how cells remember previous mechanical environments to influence their fate, or mechanical memory, informs the design of biomaterials and therapies in medicine. Current regeneration therapies, such as cartilage regeneration procedures, require 2D cell expansion processes to achieve large cell populations critical for the repair of damaged tissues. However, the limit of mechanical priming for cartilage regeneration procedures before inducing long-term mechanical memory following expansion processes is unknown, and mechanisms defining how physical environments influence the therapeutic potential of cells remain poorly understood. Here, we identify a threshold to mechanical priming separating reversible and irreversible effects of mechanical memory. After 16 population doublings in 2D culture, expression levels of tissue-identifying genes in primary cartilage cells (chondrocytes) are not recovered when transferred to 3D hydrogels, while expression levels of these genes were recovered for cells only expanded for eight population doublings. Additionally, we show that the loss and recovery of the chondrocyte phenotype correlates with a change in chromatin architecture, as shown by structural remodeling of the trimethylation of H3K9. Efforts to disrupt the chromatin architecture by suppressing or increasing levels of H3K9me3 reveal that only with increased levels of H3K9me3 did the chromatin architecture of the native chondrocyte phenotype partially return, along with increased levels of chondrogenic gene expression. These results further support the connection between the chondrocyte phenotype and chromatin architecture, and also reveal the therapeutic potential of inhibitors of epigenetic modifiers as disruptors of mechanical memory when large numbers of phenotypically suitable cells are required for regeneration procedures.
Collapse
Affiliation(s)
- Adrienne K Scott
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Eduard Casas
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, Colorado
| | - Stephanie E Schneider
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Alison R Swearingen
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, Colorado
| | - Courtney L Van Den Elzen
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, Boulder, Colorado
| | - Benjamin Seelbinder
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Jeanne E Barthold
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Jennifer F Kugel
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado
| | - Josh Lewis Stern
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado; Biochemistry and Molecular Genetics, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kyla J Foster
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado
| | - Nancy C Emery
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, Boulder, Colorado
| | - Justin Brumbaugh
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, Colorado
| | - Corey P Neu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado; Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado; BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado.
| |
Collapse
|
28
|
Scott AK, Rafuse M, Neu CP. Mechanically induced alterations in chromatin architecture guide the balance between cell plasticity and mechanical memory. Front Cell Dev Biol 2023; 11:1084759. [PMID: 37143893 PMCID: PMC10151697 DOI: 10.3389/fcell.2023.1084759] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 04/07/2023] [Indexed: 05/06/2023] Open
Abstract
Phenotypic plasticity, or adaptability, of a cell determines its ability to survive and function within changing cellular environments. Changes in the mechanical environment, ranging from stiffness of the extracellular matrix (ECM) to physical stress such as tension, compression, and shear, are critical environmental cues that influence phenotypic plasticity and stability. Furthermore, an exposure to a prior mechanical signal has been demonstrated to play a fundamental role in modulating phenotypic changes that persist even after the mechanical stimulus is removed, creating stable mechanical memories. In this mini review, our objective is to highlight how the mechanical environment alters both phenotypic plasticity and stable memories through changes in chromatin architecture, mainly focusing on examples in cardiac tissue. We first explore how cell phenotypic plasticity is modulated in response to changes in the mechanical environment, and then connect the changes in phenotypic plasticity to changes in chromatin architecture that reflect short-term and long-term memories. Finally, we discuss how elucidating the mechanisms behind mechanically induced chromatin architecture that lead to cell adaptations and retention of stable mechanical memories could uncover treatment methods to prevent mal-adaptive permanent disease states.
Collapse
Affiliation(s)
- Adrienne K. Scott
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States
| | - Michael Rafuse
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States
| | - Corey P. Neu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, United States
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, United States
| |
Collapse
|
29
|
Mongelli A, Panunzi S, Nesta M, Gottardi Zamperla M, Atlante S, Barbi V, Mongiardini V, Ferraro F, De Martino S, Cis L, Re A, Maltese S, Bachetti T, La Rovere MT, Martelli F, Pesce M, Nanni S, Massetti M, Pontecorvi A, Farsetti A, Gaetano C. Distinguishable DNA methylation defines a cardiac-specific epigenetic clock. Clin Epigenetics 2023; 15:53. [PMID: 36991505 PMCID: PMC10053964 DOI: 10.1186/s13148-023-01467-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 03/18/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND The present study investigates whether epigenetic differences emerge in the heart of patients undergoing cardiac surgery for an aortic valvular replacement (AVR) or coronary artery bypass graft (CABG). An algorithm is also established to determine how the pathophysiological condition might influence the human biological cardiac age. RESULTS Blood samples and cardiac auricles were collected from patients who underwent cardiac procedures: 94 AVR and 289 CABG. The CpGs from three independent blood-derived biological clocks were selected to design a new blood- and the first cardiac-specific clocks. Specifically, 31 CpGs from six age-related genes, ELOVL2, EDARADD, ITGA2B, ASPA, PDE4C, and FHL2, were used to construct the tissue-tailored clocks. The best-fitting variables were combined to define new cardiac- and blood-tailored clocks validated through neural network analysis and elastic regression. In addition, telomere length (TL) was measured by qPCR. These new methods revealed a similarity between chronological and biological age in the blood and heart; the average TL was significantly higher in the heart than in the blood. In addition, the cardiac clock discriminated well between AVR and CABG and was sensitive to cardiovascular risk factors such as obesity and smoking. Moreover, the cardiac-specific clock identified an AVR patient's subgroup whose accelerated bioage correlated with the altered ventricular parameters, including left ventricular diastolic and systolic volume. CONCLUSION This study reports on applying a method to evaluate the cardiac biological age revealing epigenetic features that separate subgroups of AVR and CABG.
Collapse
Affiliation(s)
- A Mongelli
- Laboratorio di Epigenetica, Istituti Clinici Scientifici (ICS) Maugeri IRCCS, 27100, Pavia, Italy
- Center for Translational and Experimental Cardiology (CTEC), University of Zurich, 8952, Schlieren, Switzerland
| | - S Panunzi
- National Research Council (CNR)-IASI, 00185, Rome, Italy
| | - M Nesta
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - M Gottardi Zamperla
- Laboratorio di Epigenetica, Istituti Clinici Scientifici (ICS) Maugeri IRCCS, 27100, Pavia, Italy
| | - S Atlante
- Laboratorio di Epigenetica, Istituti Clinici Scientifici (ICS) Maugeri IRCCS, 27100, Pavia, Italy
| | - V Barbi
- Laboratorio di Epigenetica, Istituti Clinici Scientifici (ICS) Maugeri IRCCS, 27100, Pavia, Italy
| | - V Mongiardini
- Laboratorio di Epigenetica, Istituti Clinici Scientifici (ICS) Maugeri IRCCS, 27100, Pavia, Italy
- Molecular Medicine, Istituto Italiano di Tecnologia, Genoa, Italy
| | - F Ferraro
- Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - S De Martino
- Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - L Cis
- Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - A Re
- National Research Council (CNR)-IASI, 00185, Rome, Italy
- Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - S Maltese
- National Research Council (CNR)-IRIB, 90146, Palermo, Italy
| | - T Bachetti
- Direzione Scientifica Centrale ICS Maugeri IRCCS, Pavia, Italy
| | - M T La Rovere
- Dipartimento di Cardiologia ICS Maugeri and Direzione Scientifica ICS Maugeri Montescano IRCCS, Pavia, Italy
| | - F Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - M Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - S Nanni
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - M Massetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - A Pontecorvi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - A Farsetti
- National Research Council (CNR)-IASI, 00185, Rome, Italy.
| | - C Gaetano
- Laboratorio di Epigenetica, Istituti Clinici Scientifici (ICS) Maugeri IRCCS, 27100, Pavia, Italy.
| |
Collapse
|
30
|
Amiad Pavlov D, Unnikannan CP, Lorber D, Bajpai G, Olender T, Stoops E, Reuveny A, Safran S, Volk T. The LINC Complex Inhibits Excessive Chromatin Repression. Cells 2023; 12:932. [PMID: 36980273 PMCID: PMC10047284 DOI: 10.3390/cells12060932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
The Linker of Nucleoskeleton and Cytoskeleton (LINC) complex transduces nuclear mechanical inputs suggested to control chromatin organization and gene expression; however, the underlying mechanism is currently unclear. We show here that the LINC complex is needed to minimize chromatin repression in muscle tissue, where the nuclei are exposed to significant mechanical inputs during muscle contraction. To this end, the genomic binding profiles of Polycomb, Heterochromatin Protein1 (HP1a) repressors, and of RNA-Pol II were studied in Drosophila larval muscles lacking functional LINC complex. A significant increase in the binding of Polycomb and parallel reduction of RNA-Pol-II binding to a set of muscle genes was observed. Consistently, enhanced tri-methylated H3K9 and H3K27 repressive modifications and reduced chromatin activation by H3K9 acetylation were found. Furthermore, larger tri-methylated H3K27me3 repressive clusters, and chromatin redistribution from the nuclear periphery towards nuclear center, were detected in live LINC mutant larval muscles. Computer simulation indicated that the observed dissociation of the chromatin from the nuclear envelope promotes growth of tri-methylated H3K27 repressive clusters. Thus, we suggest that by promoting chromatin-nuclear envelope binding, the LINC complex restricts the size of repressive H3K27 tri-methylated clusters, thereby limiting the binding of Polycomb transcription repressor, directing robust transcription in muscle fibers.
Collapse
Affiliation(s)
- Daria Amiad Pavlov
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | | | - Dana Lorber
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Gaurav Bajpai
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tsviya Olender
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Elizabeth Stoops
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Adriana Reuveny
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Samuel Safran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Talila Volk
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
31
|
Lin Y, Zhang F, Chen S, Zhu X, Jiao J, Zhang Y, Li Z, Lin J, Ma B, Chen M, Wang PY, Cui C. Binary Colloidal Crystals Promote Cardiac Differentiation of Human Pluripotent Stem Cells via Nuclear Accumulation of SETDB1. ACS NANO 2023; 17:3181-3193. [PMID: 36655945 PMCID: PMC9933589 DOI: 10.1021/acsnano.3c00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 01/17/2023] [Indexed: 06/17/2023]
Abstract
Biophysical cues can facilitate the cardiac differentiation of human pluripotent stem cells (hPSCs), yet the mechanism is far from established. One of the binary colloidal crystals, composed of 5 μm Si and 400 nm poly(methyl methacrylate) particles named 5PM, has been applied as a substrate for hPSCs cultivation and cardiac differentiation. In this study, cell nucleus, cytoskeleton, and epigenetic states of human induced pluripotent stem cells on the 5PM were analyzed using atomic force microscopy, molecular biology assays, and the assay for transposase-accessible chromatin sequencing (ATAC-seq). Cells were more spherical with stiffer cell nuclei on the 5PM compared to the flat control. ATAC-seq revealed that chromatin accessibility decreased on the 5PM, caused by the increased entry of histone lysine methyltransferase SETDB1 into the cell nuclei and the amplified level of histone H3K9me3 modification. Reducing cytoskeleton tension using a ROCK inhibitor attenuated the nuclear accumulation of SETDB1 on the 5PM, indicating that the effect is cytoskeleton-dependent. In addition, the knockdown of SETDB1 reversed the promotive effects of the 5PM on cardiac differentiation, demonstrating that biophysical cue-induced cytoskeletal tension, cell nucleus deformation, and then SETDB1 accumulation are critical outside-in signal transformations in cardiac differentiation. Human embryonic stem cells showed similar results, indicating that the biophysical impact of the 5PM surfaces on cardiac differentiation could be universal. These findings contribute to our understanding of material-assistant hPSC differentiation, which benefits materiobiology and stem cell bioengineering.
Collapse
Affiliation(s)
- Yongping Lin
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
- Department
of Cardiology, The Affiliated Taizhou People’s Hospital of
Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou225300, Jiangsu, China
| | - Feng Zhang
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
| | - Shaojie Chen
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
| | - Xiyu Zhu
- Department
of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical
School, Nanjing210000, China
| | - Jincheng Jiao
- State
Key Laboratory of Bioelectronics, School of Biological Science and
Medical Engineering, Southeast University, Nanjing210000, Jiangsu, China
| | - Yike Zhang
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
| | - Zhaomin Li
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
| | - Jiao Lin
- Shenzhen
Key Laboratory of Biomimetic Materials and Cellular Immunomodulation,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen518000, Guangdong, China
| | - Biao Ma
- State
Key Laboratory of Bioelectronics, School of Biological Science and
Medical Engineering, Southeast University, Nanjing210000, Jiangsu, China
| | - Minglong Chen
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
- Department
of Cardiology, The Affiliated Taizhou People’s Hospital of
Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou225300, Jiangsu, China
- Key
Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative
Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing210000, Jiangsu, China
| | - Peng-Yuan Wang
- Oujiang
Laboratory; Key Laboratory of Alzheimer’s Disease of Zhejiang
Province, Institute of Aging, Wenzhou Medical
University, Wenzhou325000, Zhejiang, China
| | - Chang Cui
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
| |
Collapse
|
32
|
Guo Y, Mofrad MRK, Tepole AB. On modeling the multiscale mechanobiology of soft tissues: Challenges and progress. BIOPHYSICS REVIEWS 2022; 3:031303. [PMID: 38505274 PMCID: PMC10903412 DOI: 10.1063/5.0085025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 07/12/2022] [Indexed: 03/21/2024]
Abstract
Tissues grow and remodel in response to mechanical cues, extracellular and intracellular signals experienced through various biological events, from the developing embryo to disease and aging. The macroscale response of soft tissues is typically nonlinear, viscoelastic anisotropic, and often emerges from the hierarchical structure of tissues, primarily their biopolymer fiber networks at the microscale. The adaptation to mechanical cues is likewise a multiscale phenomenon. Cell mechanobiology, the ability of cells to transform mechanical inputs into chemical signaling inside the cell, and subsequent regulation of cellular behavior through intra- and inter-cellular signaling networks, is the key coupling at the microscale between the mechanical cues and the mechanical adaptation seen macroscopically. To fully understand mechanics of tissues in growth and remodeling as observed at the tissue level, multiscale models of tissue mechanobiology are essential. In this review, we summarize the state-of-the art modeling tools of soft tissues at both scales, the tissue level response, and the cell scale mechanobiology models. To help the interested reader become more familiar with these modeling frameworks, we also show representative examples. Our aim here is to bring together scientists from different disciplines and enable the future leap in multiscale modeling of tissue mechanobiology.
Collapse
Affiliation(s)
- Yifan Guo
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Mohammad R. K. Mofrad
- Departments of Bioengineering and Mechanical Engineering, University of California Berkeley, Berkeley, California 94720, USA
| | - Adrian Buganza Tepole
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| |
Collapse
|
33
|
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: 144] [Impact Index Per Article: 72.0] [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
|
34
|
Jones ML, Dahl KN, Lele TP, Conway DE, Shenoy V, Ghosh S, Szczesny SE. The Elephant in the Cell: Nuclear Mechanics and Mechanobiology. J Biomech Eng 2022; 144:080802. [PMID: 35147160 PMCID: PMC8990742 DOI: 10.1115/1.4053797] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/28/2022] [Indexed: 11/08/2022]
Abstract
The 2021 Summer Biomechanics, Bioengineering, and Biotransport Conference (SB3C) featured a workshop titled "The Elephant in the Room: Nuclear Mechanics and Mechanobiology." The goal of this workshop was to provide a perspective from experts in the field on the current understanding of nuclear mechanics and its role in mechanobiology. This paper reviews the major themes and questions discussed during the workshop, including historical context on the initial methods of measuring the mechanical properties of the nucleus and classifying the primary structures dictating nuclear mechanics, physical plasticity of the nucleus, the emerging role of the linker of nucleoskeleton and cytoskeleton (LINC) complex in coupling the nucleus to the cytoplasm and driving the behavior of individual cells and multicellular assemblies, and the computational models currently in use to investigate the mechanisms of gene expression and cell signaling. Ongoing questions and controversies, along with promising future directions, are also discussed.
Collapse
Affiliation(s)
| | - Kris Noel Dahl
- Department of Chemical Engineering, Carnegie Mellon University, Doherty Hall, 5000 Forbes Avenue, Pittsburgh, PA 15213; Forensics Department, Thornton Tomasetti, 120 Broadway 15th Floor, New York City, NY 10271
| | - Tanmay P. Lele
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX 77840; Department of Chemical Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX 77840; Department of Translational Medical Sciences, Texas A&M University, 101 Bizzell Street, College Station, TX 77840
| | - Daniel E. Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, 601 West Main Street, P.O. Box 843068, Richmond, VA 23284
| | - Vivek Shenoy
- Materials Science and Engineering Bioengineering, University of Pennsylvania, 3231 Walnut Street, Philadelphia, PA 19104; Mechanical Engineering and Applied Mechanics, University of Pennsylvania, 3231 Walnut Street, Philadelphia, PA 19104; Center for Engineering Mechanobiology, University of Pennsylvania, 3231 Walnut Street, Philadelphia, PA 19104
| | - Soham Ghosh
- Department of Mechanical Engineering, School of Biomedical Engineering, Translational Medicine Institute, Colorado State University, 400 Isotope Drive, Fort Collins, CO 80521
| | - Spencer E. Szczesny
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802; Department of Orthopaedics and Rehabilitation, Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
35
|
Rhodes ADY, Duran-Mota JA, Oliva N. Current progress in bionanomaterials to modulate the epigenome. Biomater Sci 2022; 10:5081-5091. [PMID: 35880652 DOI: 10.1039/d2bm01027e] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recent advances in genomics during the 1990s have made it possible to study and identify genetic and epigenetic responses of cells and tissues to various drugs and environmental factors. This has accelerated the number of targets available to treat a range of diseases from cancer to wound healing disorders. Equally interesting is the understanding of how bio- and nanomaterials alter gene expression through epigenetic mechanisms, and whether they have the potential to elicit a positive therapeutic response without requiring additional biomolecule delivery. In fact, from a cell's perspective, a biomaterial is nothing more than an environmental factor, and so it has the power to epigenetically modulate gene expression of cells in contact with it. Understanding these epigenetic interactions between biomaterials and cells will open new avenues in the development of technologies that can not only provide biological signals (i.e. drugs, growth factors) necessary for therapy and regeneration, but also intimately interact with cells to promote the expression of genes of interest. This review article aims to summarise the current state-of-the-art and progress on the development of bio- and nanomaterials to modulate the epigenome.
Collapse
Affiliation(s)
- Anna D Y Rhodes
- Department of Bioengineering, Imperial College London, London W12 0BZ, UK.
| | - Jose Antonio Duran-Mota
- Department of Bioengineering, Imperial College London, London W12 0BZ, UK. .,Materials Engineering Group (GEMAT), IQS Barcelona, Barcelona 08017, Spain
| | - Nuria Oliva
- Department of Bioengineering, Imperial College London, London W12 0BZ, UK.
| |
Collapse
|
36
|
Liu W, Padhi A, Zhang X, Narendran J, Anastasio MA, Nain AS, Irudayaraj J. Dynamic Heterochromatin States in Anisotropic Nuclei of Cells on Aligned Nanofibers. ACS NANO 2022; 16:10754-10767. [PMID: 35803582 PMCID: PMC9332347 DOI: 10.1021/acsnano.2c02660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The cancer cell nucleus deforms as it invades the interstitial spaces in tissues and the tumor microenvironment. While alteration of the chromatin structure in a deformed nucleus is expected and documented, the chromatin structure in the nuclei of cells on aligned matrices has not been elucidated. In this work we elucidate the spatiotemporal organization of heterochromatin in the elongated nuclei of cells on aligned nanofibers with stimulated emission depletion nanoscopy and fluorescence correlation spectroscopy. We show that the anisotropy of nuclei is sufficient to drive H3K9me3-heterochromatin alterations, with enhanced H3K9me3 nanocluster compaction and aggregation states that otherwise are indistinguishable from diffraction-limited microscopy. We interrogated the higher-order heterochromatin structures within major chromatin compartments in anisotropic nuclei and discovered a wider spatial dispersion of nanodomain clusters in the nucleoplasm and condensed larger nanoclusters near the periphery and pericentromeric heterochromatin. Upon examining the spatiotemporal dynamics of heterochromatin in anisotropic nuclei, we observed reduced mobility of the constitutive heterochromatin mark H3K9me3 and the associated heterochromatin protein 1 (HP1α) at the nucleoplasm and periphery regions, correlating with increased viscosity and changes in gene expression. Since heterochromatin remodeling is crucial to genome integrity, our results reveal an unconventional H3K9me3 heterochromatin distribution, providing cues to an altered chromatin state due to perturbations of the nuclei in aligned fiber configurations.
Collapse
Affiliation(s)
- Wenjie Liu
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, 1102 Everitt Lab, 1406 W. Green Street, Urbana, Illinois 61801, United States
- Biomedical
Research Center, Mills Breast Cancer Institute, Cancer Center at Illinois,
Micro and Nanotechnology Laboratory, Beckman
Institute, Carl Woese Institute for Genomic Biology, Urbana, Illinois 61801, United States
| | - Abinash Padhi
- Department
of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Xiaohui Zhang
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, 1102 Everitt Lab, 1406 W. Green Street, Urbana, Illinois 61801, United States
| | - Jairaj Narendran
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, 1102 Everitt Lab, 1406 W. Green Street, Urbana, Illinois 61801, United States
| | - Mark A. Anastasio
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, 1102 Everitt Lab, 1406 W. Green Street, Urbana, Illinois 61801, United States
| | - Amrinder S. Nain
- Department
of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Joseph Irudayaraj
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, 1102 Everitt Lab, 1406 W. Green Street, Urbana, Illinois 61801, United States
- Biomedical
Research Center, Mills Breast Cancer Institute, Cancer Center at Illinois,
Micro and Nanotechnology Laboratory, Beckman
Institute, Carl Woese Institute for Genomic Biology, Urbana, Illinois 61801, United States
| |
Collapse
|
37
|
Bowers SL, Meng Q, Molkentin JD. Fibroblasts orchestrate cellular crosstalk in the heart through the ECM. NATURE CARDIOVASCULAR RESEARCH 2022; 1:312-321. [PMID: 38765890 PMCID: PMC11101212 DOI: 10.1038/s44161-022-00043-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/02/2022] [Indexed: 05/22/2024]
Abstract
Cell communication is needed for organ function and stress responses, especially in the heart. Cardiac fibroblasts, cardiomyocytes, immune cells, and endothelial cells comprise the major cell types in ventricular myocardium that together coordinate all functional processes. Critical to this cellular network is the non-cellular extracellular matrix (ECM) that provides structure and harbors growth factors and other signaling proteins that affect cell behavior. The ECM is not only produced and modified by cells within the myocardium, largely cardiac fibroblasts, it also acts as an avenue for communication among all myocardial cells. In this Review, we discuss how the development of therapeutics to combat cardiac diseases, specifically fibrosis, relies on a deeper understanding of how the cardiac ECM is intertwined with signaling processes that underlie cellular activation and behavior.
Collapse
Affiliation(s)
| | | | - Jeffery D. Molkentin
- Cincinnati Children’s Hospital, Division of Molecular Cardiovascular Biology; University of Cincinnati, Department of Pediatrics, Cincinnati, OH
| |
Collapse
|
38
|
Unique Astrocyte Cytoskeletal and Nuclear Morphology in a Three-Dimensional Tissue-Engineered Rostral Migratory Stream. NEUROGLIA (BASEL, SWITZERLAND) 2022; 3:41-60. [PMID: 36776937 PMCID: PMC9910099 DOI: 10.3390/neuroglia3010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neural precursor cells (NPCs) are generated in the subventricular zone (SVZ) and travel through the rostral migratory stream (RMS) to replace olfactory bulb interneurons in the brains of most adult mammals. Following brain injury, SVZ-derived NPCs can divert from the RMS and migrate toward injured brain regions but arrive in numbers too low to promote functional recovery without experimental intervention. Our lab has biofabricated a "living scaffold" that replicates the structural and functional features of the endogenous RMS. This tissue-engineered rostral migratory stream (TE-RMS) is a new regenerative medicine strategy designed to facilitate stable and sustained NPC delivery into neuron-deficient brain regions following brain injury or neurodegenerative disease and an in vitro tool to investigate the mechanisms of neuronal migration and cell-cell communication. We have previously shown that the TE-RMS replicates the basic structure and protein expression of the endogenous RMS and can direct immature neuronal migration in vitro and in vivo. Here, we further describe profound morphological changes that occur following precise physical manipulation and subsequent self-assembly of astrocytes into the TE-RMS, including significant cytoskeletal rearrangement and nuclear elongation. The unique cytoskeletal and nuclear architecture of TE-RMS astrocytes mimics astrocytes in the endogenous rat RMS. Advanced imaging techniques reveal the unique morphology of TE-RMS cells that has yet to be described of astrocytes in vitro. The TE-RMS offers a novel platform to elucidate astrocyte cytoskeletal and nuclear dynamics and their relationship to cell behavior and function.
Collapse
|
39
|
|