1
|
Choi JC. Perinuclear organelle trauma at the nexus of cardiomyopathy pathogenesis arising from loss of function LMNA mutation. Nucleus 2025; 16:2449500. [PMID: 39789731 PMCID: PMC11730615 DOI: 10.1080/19491034.2024.2449500] [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: 10/31/2024] [Revised: 12/22/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Over the past 25 years, nuclear envelope (NE) perturbations have been reported in various experimental models with mutations in the LMNA gene. Although the hypothesis that NE perturbations from LMNA mutations are a fundamental feature of striated muscle damage has garnered wide acceptance, the molecular sequalae provoked by the NE damage and how they underlie disease pathogenesis such as cardiomyopathy (LMNA cardiomyopathy) remain poorly understood. We recently shed light on one such consequence, by employing a cardiomyocyte-specific Lmna deletion in vivo in the adult heart. We observed extensive NE perturbations prior to cardiac function deterioration with collateral damage in the perinuclear space. The Golgi is particularly affected, leading to cytoprotective stress responses that are likely disrupted by the progressive deterioration of the Golgi itself. In this review, we discuss the etiology of LMNA cardiomyopathy with perinuclear 'organelle trauma' as the nexus between NE damage and disease pathogenesis.
Collapse
Affiliation(s)
- Jason C. Choi
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Nair A, Khanna J, Kler J, Ragesh R, Sengupta K. Nuclear envelope and chromatin choreography direct cellular differentiation. Nucleus 2025; 16:2449520. [PMID: 39943681 PMCID: PMC11834525 DOI: 10.1080/19491034.2024.2449520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025] Open
Abstract
The nuclear envelope plays an indispensable role in the spatiotemporal organization of chromatin and transcriptional regulation during the intricate process of cell differentiation. This review outlines the distinct regulatory networks between nuclear envelope proteins, transcription factors and epigenetic modifications in controlling the expression of cell lineage-specific genes during differentiation. Nuclear lamina with its associated nuclear envelope proteins organize heterochromatin via Lamina-Associated Domains (LADs), proximal to the nuclear periphery. Since nuclear lamina is mechanosensitive, we critically examine the impact of extracellular forces on differentiation outcomes. The nuclear envelope is spanned by nuclear pore complexes which, in addition to their central role in transport, are associated with chromatin organization. Furthermore, mutations in the nuclear envelope proteins disrupt differentiation, resulting in developmental disorders. Investigating the underlying nuclear envelope controlled regulatory mechanisms of chromatin remodelling during lineage commitment will accelerate our fundamental understanding of developmental biology and regenerative medicine.
Collapse
Affiliation(s)
- Anjitha Nair
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Jayati Khanna
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Jashan Kler
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Rohith Ragesh
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Kundan Sengupta
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| |
Collapse
|
3
|
Zhou H, Liu R, Xu Y, Fan J, Liu X, Chen L, Wei Q. Viscoelastic mechanics of living cells. Phys Life Rev 2025; 53:91-116. [PMID: 40043484 DOI: 10.1016/j.plrev.2025.02.004] [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: 02/11/2025] [Accepted: 02/25/2025] [Indexed: 05/18/2025]
Abstract
In cell mechanotransduction, cells respond to external forces or to perceive mechanical properties of their supporting substrates by remodeling themselves. This ability is endowed by modulating cells' viscoelastic properties, which dominates over various complex cellular processes. The viscoelasticity of living cells, a concept adapted from rheology, exhibits substantially spatial and temporal variability. This review aims not only to discuss the rheological properties of cells but also to clarify the complexity of cellular rheology, emphasizing its dependence on both the size scales and time scales of the measurements. Like typical viscoelastic materials, the storage and loss moduli of cells often exhibit robust power-law rheological characteristics with respect to loading frequency. This intrinsic feature is consistent across cell types and is attributed to internal structures, such as cytoskeleton, cortex, cytoplasm and nucleus, all of which contribute to the complexity of cellular rheology. Moreover, the rheological properties of cells are dynamic and play a crucial role in various cellular and tissue functions. In this review, we focus on elucidating time- and size-dependent aspects of cell rheology, the origins of intrinsic rheological properties and how these properties adapt to cellular functions, with the goal of interpretation of rheology into the language of cell biology.
Collapse
Affiliation(s)
- Hui Zhou
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Ruye Liu
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Yizhou Xu
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Jierui Fan
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Xinyue Liu
- Shanghai Institute of Applied Mathematics and Mechanics, School of Mechanics and Engineering Science, Shanghai University, Shanghai, 200072, China
| | - Longquan Chen
- School of Physics, University of Electronic Science and Technology of China, Chengdu 611731, China.
| | - Qiang Wei
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
4
|
Pancheri NM, Daw JT, Ditton D, Schiele NR, Birks S, Uzer G, Jones CL, Penney BT, Theodossiou SK. The LINC Complex Regulates Tendon Elastic Modulus, Collagen Crimp, and Lateral Expansion During Early Postnatal Development. J Orthop Res 2025; 43:1090-1100. [PMID: 40089904 DOI: 10.1002/jor.26069] [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] [Received: 04/01/2024] [Revised: 02/02/2025] [Accepted: 02/27/2025] [Indexed: 03/17/2025]
Abstract
There is limited understanding of how mechanical signals regulate tendon development. The nucleus has emerged as a major regulator of cellular mechanosensation via the linker of nucleoskeleton and cytoskeleton (LINC) protein complex. Specific roles of LINC in tenogenesis have not been explored. In this study, we investigate how LINC regulates tendon development by disabling LINC-mediated mechanosensing via dominant negative (dn) overexpression of the Klarsicht, ANC-1, and Syne Homology (KASH) domain, which is necessary for LINC to function. We hypothesized that LINC regulates mechanotransduction in developing tendons and that disabling LINC would impact tendon's mechanical properties and structure in a mouse model of dnKASH. We used Achilles tendon (AT) and tail tendon (TT) as representative energy-storing and positional tendons, respectively. Mechanical testing at postnatal day 10 showed that disabling the LINC complex via dnKASH significantly impacted tendon mechanical properties and cross-sectional area and that the effects differed between ATs and TTs. Collagen crimp distance was also impacted in dnKASH tendons and was significantly decreased in ATs and increased in TTs. Overall, we show that disruption to the LINC complex specifically impacts tendon mechanics and collagen crimp structure, with unique responses between an energy-storing and limb-positioning tendon. This suggests that nuclear mechanotransduction through LINC plays a role in regulating tendon formation during neonatal development.
Collapse
Affiliation(s)
| | - Jordan T Daw
- Mechanical & Biomedical Engineering, Boise State University, Boise, Idaho, USA
| | - Destinee Ditton
- Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| | - Nathan R Schiele
- Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| | - Scott Birks
- Mechanical & Biomedical Engineering, Boise State University, Boise, Idaho, USA
| | - Gunes Uzer
- Mechanical & Biomedical Engineering, Boise State University, Boise, Idaho, USA
| | - Calvin L Jones
- Mechanical & Biomedical Engineering, Boise State University, Boise, Idaho, USA
| | - Brian T Penney
- Mechanical & Biomedical Engineering, Boise State University, Boise, Idaho, USA
| | | |
Collapse
|
5
|
Hameed HA, Paturej J, Erbaş A. Phase behavior and dissociation kinetics of lamins in a polymer model of progeria. J Chem Phys 2025; 162:185101. [PMID: 40337942 DOI: 10.1063/5.0265578] [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: 02/14/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
One of the key structural proteins in the eukaryotic cell nucleus is lamin. Lamins can assemble into a two-dimensional protein meshwork at the nuclear periphery, known as the nuclear lamina, which provides rigidity and shape to the nucleus. Mutations in lamin proteins that alter the structure of the nuclear lamina underlie laminopathic diseases, including Hutchinson-Gilford Progeria Syndrome (HGPS). Experiments have shown that, compared to healthy cells, lamin supramolecular structures (e.g., protofilaments) assemble into a thicker lamina in HGPS, where they form highly stable nematic microdomains at the nuclear periphery, reminiscent of liquid crystals. This significantly alters the morphological and mechanical properties of the nucleus. In this study, we investigate the aggregation of lamin fibrous structures and their dissociation kinetics from the nuclear periphery by modeling them as coarse-grained, rod-like polymer chains confined within a rigid spherical shell. Our model reproduces the formation of multidirectional nematic domains at the nuclear surface and the reduced lamin dissociation observed in HGPS nuclei by adjusting lamin concentration, lamin-lamin (head-tail), and lamin-shell association strengths. While nematic phase formation requires relatively strong lamin-shell affinity under any non-vanishing inter-lamin attraction, the thickness of the lamina layer is primarily controlled by the head-tail association strength in the model. Furthermore, the unbinding kinetics of lamin chains from the lamina exhibit a concentration-dependent facilitated dissociation, suppressed by strong intra-lamin interactions, reminiscent of diseased nuclei. Overall, our calculations reveal the physical mechanisms by which mutations affecting native lamin interactions and concentration could lead to an abnormal nuclear lamina in laminopathic diseases.
Collapse
Affiliation(s)
- Hadiya Abdul Hameed
- UNAM-National Nanotechnology Research Center and Institute of Materials Science and Nanotechnology, Bilkent University, Ankara 06800, Turkiye
| | - Jarosław Paturej
- Institute of Physics, University of Silesia at Katowice, Chorzów 41-500, Poland
| | - Aykut Erbaş
- UNAM-National Nanotechnology Research Center and Institute of Materials Science and Nanotechnology, Bilkent University, Ankara 06800, Turkiye
- Institute of Physics, University of Silesia at Katowice, Chorzów 41-500, Poland
| |
Collapse
|
6
|
Dang Y, Lattner J, Lahola-Chomiak AA, Afonso DA, Ulbricht E, Taubenberger A, Rulands S, Tabler JM. Self-propagating wave drives morphogenesis of skull bones in vivo. Nat Commun 2025; 16:4330. [PMID: 40346043 PMCID: PMC12064835 DOI: 10.1038/s41467-025-59164-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/13/2025] [Indexed: 05/11/2025] Open
Abstract
Cellular motion is a key feature of tissue morphogenesis and is often driven by migration. However, migration need not explain cell motion in contexts where there is little free space or no obvious substrate, such as those found during organogenesis of mesenchymal organs including the embryonic skull. Through ex vivo imaging, biophysical modeling, and perturbation experiments, we find that mechanical feedback between cell fate and stiffness drives bone expansion and controls bone size in vivo. This mechanical feedback system is sufficient to propagate a wave of differentiation that establishes a collagen gradient which we find sufficient to describe patterns of osteoblast motion. Our work provides a mechanism for coordinated motion that may not rely upon cell migration but on emergent properties of the mesenchymal collective. Identification of such alternative mechanisms of mechanochemical coupling between differentiation and morphogenesis will help in understanding how directed cellular motility arises in complex environments with inhomogeneous material properties.
Collapse
Affiliation(s)
- Yiteng Dang
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
- Center for Systems Biology, Dresden, Germany
| | - Johanna Lattner
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Diana Alves Afonso
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | | | | | - Steffen Rulands
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
- Center for Systems Biology, Dresden, Germany
- Arnold-Sommerfeld-Center for Theoretical Physics, Ludwig-Maximilians-Universität München, München, Germany
| | - Jacqueline M Tabler
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
7
|
Fraczek PM, Duran P, Yang BA, Ferre V, Alawieh L, Castor-Macias JA, Wong VT, Guzman SD, Piotto C, Itsani K, Larouche JA, Aguilar CA. Vitamin A retinoic acid contributes to muscle stem cell and mitochondrial function loss in old age. JCI Insight 2025; 10:e183706. [PMID: 40131371 DOI: 10.1172/jci.insight.183706] [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] [Indexed: 03/27/2025] Open
Abstract
Adult stem cells decline in number and function in old age, and identifying factors that can delay or revert age-associated adult stem cell dysfunction are vital for maintaining a healthy lifespan. Here we show that vitamin A, a micronutrient that is derived from diet and metabolized into retinoic acid, acts as an antioxidant and transcriptional regulator in muscle stem cells. We first show that obstruction of dietary vitamin A in young animals drives mitochondrial and cell cycle dysfunction in muscle stem cells that mimics old age. Next, we pharmacologically targeted retinoic acid signaling in myoblasts and aged muscle stem cells ex vivo and in vivo and observed reductions in oxidative damage, enhanced mitochondrial function, and improved maintenance of quiescence through fatty acid oxidation. We next detected that the receptor for vitamin A-derived retinol, stimulated by retinoic acid 6 or Stra6, was diminished with muscle stem cell activation and in old age. To understand the relevance of Stra6 loss, we knocked down Stra6 and observed an accumulation of mitochondrial reactive oxygen species, as well as changes in mitochondrial morphology and respiration. These results demonstrate that vitamin A regulates mitochondria and metabolism in muscle stem cells and highlight a unique mechanism connecting stem cell function with vitamin intake.
Collapse
Affiliation(s)
- Paula M Fraczek
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | - Pamela Duran
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | - Benjamin A Yang
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | - Valeria Ferre
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | - Leanne Alawieh
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | | | - Vivian T Wong
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | - Steve D Guzman
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | - Celeste Piotto
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | | | | | - Carlos A Aguilar
- Department of Biomedical Engineering
- Biointerfaces Institute, and
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
8
|
Henretta S, Lammerding J. Nuclear envelope proteins, mechanotransduction, and their contribution to breast cancer progression. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:14. [PMID: 40337116 PMCID: PMC12052594 DOI: 10.1038/s44341-025-00018-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 03/18/2025] [Indexed: 05/09/2025]
Abstract
Breast cancer cells frequently exhibit changes in the expression of nuclear envelope (NE) proteins such as lamins and emerin that determine the physical properties of the nucleus and contribute to cellular mechanotransduction. This review explores the emerging interplay between NE proteins, the physical challenges incurred during metastatic progression, and mechanotransduction. Improved insights into the underlying mechanisms may ultimately lead to better prognostic tools and treatment strategies for metastatic breast cancer.
Collapse
Affiliation(s)
- Sarah Henretta
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY USA
| | - Jan Lammerding
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY USA
| |
Collapse
|
9
|
Rouhi L. Cardiac phenotypes in LMNA mutations. Curr Opin Cardiol 2025; 40:131-138. [PMID: 39998502 PMCID: PMC11968229 DOI: 10.1097/hco.0000000000001209] [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] [Indexed: 02/26/2025]
Abstract
PURPOSE OF REVIEW This review highlights the diverse cardiac manifestations of LMNA mutations, focusing on their underlying molecular mechanisms and clinical implications. As LMNA mutations are implicated in cardiomyopathies, such as dilated cardiomyopathy (DCM), arrhythmogenic cardiomyopathy (ARVC), and conduction system diseases, understanding these phenotypes is critical for advancing diagnosis and management strategies. RECENT FINDINGS Recent studies reveal that LMNA mutations disrupt nuclear envelope stability, activating the DNA damage response (DDR) and compromising chromatin organization and mechanotransduction. Mouse models have elucidated pathways linking LMNA dysfunction to fibrosis, arrhythmias, and myocardial remodeling. Emerging evidence demonstrates that fibroblasts play a crucial role in cardiac phenotypes. Advances in genetic screening have also underscored the importance of early identification and risk stratification, particularly for arrhythmias and sudden cardiac death. SUMMARY The diverse spectrum of LMNA-related cardiac phenotypes, from isolated conduction defects to severe DCM and ARVC, underscores the necessity of personalized care strategies. Bridging insights from molecular studies and clinical research paves the way for targeted therapies to slow disease progression and improve patient outcomes. Future efforts should prioritize translational research on molecular mechanisms with potential in mouse models, alongside a deeper exploration of genotype-phenotype correlations, to refine and implement effective therapeutic interventions.
Collapse
Affiliation(s)
- Leila Rouhi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| |
Collapse
|
10
|
Karlinski Zur M, Bhattacharya B, Solomonov I, Ben Dror S, Savidor A, Levin Y, Prior A, Sapir T, Harris T, Olender T, Schmidt R, Schwarz JM, Sagi I, Buxboim A, Reiner O. Altered extracellular matrix structure and elevated stiffness in a brain organoid model for disease. Nat Commun 2025; 16:4094. [PMID: 40312467 PMCID: PMC12045990 DOI: 10.1038/s41467-025-59252-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/15/2025] [Indexed: 05/03/2025] Open
Abstract
The viscoelastic properties of tissues influence their morphology and cellular behavior, yet little is known about changes in these properties during brain malformations. Lissencephaly, a severe cortical malformation caused by LIS1 mutations, results in a smooth cortex. Here, we show that human-derived brain organoids with LIS1 mutation exhibit increased stiffness compared to controls at multiple developmental stages. This stiffening correlates with abnormal extracellular matrix (ECM) expression and organization, as well as elevated water content, measured by diffusion-weighted MRI. Short-term MMP9 treatment reduces both stiffness and water diffusion levels to control values. Additionally, a computational microstructure mechanical model predicts mechanical changes based on ECM organization. These findings suggest that LIS1 plays a critical role in ECM regulation during brain development and that its mutation leads to significant viscoelastic alterations.
Collapse
Grants
- AARG-NTF-21-849529 Alzheimer's Association
- We express our gratitude for the help of Dr. Arpan Parichha and Alfredo Isaac Ponce Arias. Orly Reiner is an incumbent of the Berstein-Mason professorial chair of Neurochemistry and the Head of the M. Judith Ruth Institute for Preclinical Brain Research. Our research has been supported by a research grant from Ethel Lena Levy, the Selsky Memory Research Project, the Gladys Monroy and Larry Marks Center for Brain Disorders, the Advantage Trust, the Nella and Leon Benoziyo Center for Neurological Diseases, the David and Fela Shapell Family Center for Genetic Disorders Research, the Abish-Frenkel RNA center, the Brenden- Mann Women's Innovation Impact Fund, The Irving B. Harris Fund for New Directions in Brain Research, the Irving Bieber, M.D. and Toby Bieber, M.D. Memorial Research Fund, The Leff Family, Barbara & Roberto Kaminitz, Sergio & Sônia Lozinsky, Debbie Koren, Jack and Lenore Lowenthal, and the Dears Foundation. A research grant from the Estates of Ethel H. Smith, Gerald Alexander, Mr. and Mrs. George Zbeda, David A. Fishstrom, Norman Fidelman, Hermine Miller, Olga Klein Astrachan, Hermine Miller, and The Maurice and Vivienne Wohl Biology Endowment, Supported by a research grant from Emily Merjan, the ISF grant (545/21), and the United States-Israel Binational Science Foundation (BSF; Grant No. 2023009).
Collapse
Affiliation(s)
- Maayan Karlinski Zur
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute, Rehovot, Israel
| | - Bidisha Bhattacharya
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute, Rehovot, Israel
| | - Inna Solomonov
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sivan Ben Dror
- The Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus, Jerusalem, Israel
| | - Alon Savidor
- The De Botton Protein Profiling Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Yishai Levin
- The De Botton Protein Profiling Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Amir Prior
- The De Botton Protein Profiling Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Sapir
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute, Rehovot, Israel
| | - Talia Harris
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Tsviya Olender
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Rita Schmidt
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
| | - J M Schwarz
- Physics Department, Syracuse University, Syracuse, NY, USA
| | - Irit Sagi
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Amnon Buxboim
- The Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus, Jerusalem, Israel.
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, The Edmond J. Safra Campus, Jerusalem, Israel.
- The Alexender Grass Center for Bioengineering, The Hebrew University of Jerusalem, The Edmond J. Safra Campus, Jerusalem, Israel.
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
- Department of Molecular Neuroscience, Weizmann Institute, Rehovot, Israel.
| |
Collapse
|
11
|
Wu Y, Song Y, Soto J, Hoffman T, Lin X, Zhang A, Chen S, Massad RN, Han X, Qi D, Yeh KW, Fang Z, Eoh J, Gu L, Rowat AC, Gu Z, Li S. Viscoelastic extracellular matrix enhances epigenetic remodeling and cellular plasticity. Nat Commun 2025; 16:4054. [PMID: 40307238 PMCID: PMC12043949 DOI: 10.1038/s41467-025-59190-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
Extracellular matrices of living tissues exhibit viscoelastic properties, yet how these properties regulate chromatin and the epigenome remains unclear. Here, we show that viscoelastic substrates induce changes in nuclear architecture and epigenome, with more pronounced effects on softer surfaces. Fibroblasts on viscoelastic substrates display larger nuclei, lower chromatin compaction, and differential expression of distinct sets of genes related to the cytoskeleton and nuclear function, compared to those on elastic surfaces. Slow-relaxing viscoelastic substrates reduce lamin A/C expression and enhance nuclear remodeling. These structural changes are accompanied by a global increase in euchromatin marks and local increase in chromatin accessibility at cis-regulatory elements associated with neuronal and pluripotent genes. Consequently, viscoelastic substrates improve the reprogramming efficiency from fibroblasts into neurons and induced pluripotent stem cells. Collectively, our findings unravel the roles of matrix viscoelasticity in epigenetic regulation and cell reprogramming, with implications for designing smart materials for cell fate engineering.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Yang Song
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, 610041, Chengdu, China
| | - Jennifer Soto
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Tyler Hoffman
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Xiao Lin
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Aaron Zhang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Siyu Chen
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Ramzi N Massad
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Xiao Han
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Dongping Qi
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Kun-Wei Yeh
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Zhiwei Fang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Joon Eoh
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Luo Gu
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Amy C Rowat
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Zhen Gu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, 310027, Hangzhou, China
| | - Song Li
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Broad Stem Cell Research Center, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
12
|
Hitpass Romero K, Stevenson TJ, Smyth LCD, Watkin B, McCullough SJC, Vinnell L, Smith AM, Schweder P, Correia JA, Kipnis J, Dragunow M, Rustenhoven J. Age-related meningeal extracellular matrix remodeling compromises CNS lymphatic function. J Neuroinflammation 2025; 22:109. [PMID: 40247257 PMCID: PMC12007191 DOI: 10.1186/s12974-025-03436-0] [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: 03/09/2025] [Accepted: 04/01/2025] [Indexed: 04/19/2025] Open
Abstract
Efficient clearance of central nervous system (CNS) waste proteins and appropriate immune surveillance is essential for brain health. These processes are facilitated by lymphatic networks present in the meninges that drain cerebrospinal fluid (CSF). Age-related impairments to meningeal lymphatic drainage contribute to CNS waste accumulation and immune dysfunction, yet the underlying mechanisms remain poorly understood. Here, we identify extracellular matrix (ECM) remodeling in the aged dura as a key driver of CSF clearance deficits, demonstrating that peri-lymphatic collagen accumulation disrupts lymphatic function. Exploring immune-derived factors contributing to this ECM remodeling, we identify transforming growth factor beta 1 (TGFβ1) as a major regulator using primary human dural fibroblasts. Using a novel mouse model with constitutively active TGFβ receptor 1 (TGFβR1) signaling in dural fibroblasts, we show that excessive peri-lymphatic collagen deposition impairs meningeal lymphatic drainage and alters meningeal immunity. Mechanistically, we reveal that ECM-associated matrix stiffness disrupts lymphatic junction integrity and impairs lymphangiogenesis in human lymphatic endothelial cells. These findings establish dural immune cell and fibroblast-mediated ECM remodeling as a critical regulator of CSF clearance and highlight it as a potential therapeutic target for restoring brain waste clearance in aging.
Collapse
Affiliation(s)
- Kate Hitpass Romero
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Taylor J Stevenson
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Leon C D Smyth
- Brain Immunology and Glia Center, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Immunology Graduate Program, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Ben Watkin
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Samuel J C McCullough
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Luca Vinnell
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Amy M Smith
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Patrick Schweder
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand
- Auckland City Hospital, Auckland, 1023, New Zealand
| | - Jason A Correia
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand
- Auckland City Hospital, Auckland, 1023, New Zealand
| | - Jonathan Kipnis
- Brain Immunology and Glia Center, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Immunology Graduate Program, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Justin Rustenhoven
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, New Zealand.
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
13
|
Saito A, Nagayama K, Okada H, Onodera S, Aida N, Nakamura T, Sawada T, Hojo H, Kato S, Azuma T. Downregulation of Nesprin1 by Runx2 deficiency is critical for the development of skeletal laminopathy-like pathology. Proc Natl Acad Sci U S A 2025; 122:e2320138122. [PMID: 40208950 PMCID: PMC12012476 DOI: 10.1073/pnas.2320138122] [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: 11/28/2023] [Accepted: 01/31/2025] [Indexed: 04/12/2025] Open
Abstract
Runx2 is a master regulator of bone formation, and its dysfunction causes cleidocranial dysplasia (CCD) in humans. When iPS cells were generated from patients with CCD and Runx2-deficient iPS cells were generated using gene-editing techniques, abnormal laminopathy-like nuclei were observed. Runx2-deficient cells showed reduced Lamin A/C expression, but not protein levels. However, in Runx2-deficient cells, both the gene expression and protein levels of Nesprin1 were reduced, perinuclear actin fibers were sparser, and nuclear stiffness was reduced. Forced expression of Lamin A/C increased nuclear stiffness but did not improve nuclear morphology. In contrast, the induction of Nesprin1 expression alone normalized nuclear stiffness and restored nuclear morphology and perinuclear actin distribution. In Runx2-null cells, mechanical stress-induced phosphorylation of emerin was not observed. In contrast, forced expression of Nesprin1 in Runx2-null cells resulted in phosphorylation of emerin, indicating the restoration of intracellular tension. These observations were confirmed by atomic force microscopy. Therefore, the intracellular tension was inferred to pull the nuclear membrane into its normal shape. CUT&RUN assay and single RNA-seq analysis showed that an aberrant nuclear membrane caused loss of nuclear lamina gene regulation machinery, making the progression of normal osteogenic differentiation impossible; however, supplementation with Nesprin1 restored gene regulation mechanisms and promoted preosteoblast formation with normal nuclear morphology. Nesprin1 expression induced by Runx2 is essential for epigenetic regulation of the nuclear lamina. We propose CCD as a type of laminopathy involving defective expression of Nesprin1 regulated by Runx2.
Collapse
Affiliation(s)
- Akiko Saito
- Department of Biochemistry, Tokyo Dental College, Tokyo101-0061, Japan
- Oral Health Science Center, Tokyo Dental College, Tokyo101-0061, Japan
| | - Kazuaki Nagayama
- Department of Mechanical Systems Engineering, Ibaraki University, Ibaraki316-8511, Japan
| | - Hiroyuki Okada
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
| | - Shoko Onodera
- Department of Biochemistry, Tokyo Dental College, Tokyo101-0061, Japan
- Oral Health Science Center, Tokyo Dental College, Tokyo101-0061, Japan
| | - Natsuko Aida
- Department of Biochemistry, Tokyo Dental College, Tokyo101-0061, Japan
- Oral Health Science Center, Tokyo Dental College, Tokyo101-0061, Japan
| | - Takashi Nakamura
- Department of Biochemistry, Tokyo Dental College, Tokyo101-0061, Japan
- Oral Health Science Center, Tokyo Dental College, Tokyo101-0061, Japan
| | - Takashi Sawada
- Department of Histology and Developmental Biology, Tokyo Dental College, Tokyo101-0061, Japan
| | - Hironori Hojo
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
| | - Shigeaki Kato
- Department of Pharmacology, Iryo Sosei University, Fukushima970-8551, Japan
- Research Institute of Innovative Medicine, Tokiwa Foundation, Fukushima973-8403, Japan
| | - Toshifumi Azuma
- Department of Biochemistry, Tokyo Dental College, Tokyo101-0061, Japan
- Oral Health Science Center, Tokyo Dental College, Tokyo101-0061, Japan
- Obitsusankei Hospital, Saitama350-0021, Japan
| |
Collapse
|
14
|
Ba J, Zheng C, Lai Y, He X, Pan Y, Zhao Y, Xie H, Wu B, Deng X, Wang N. High matrix stiffness promotes senescence of type II alveolar epithelial cells by lysosomal degradation of lamin A/C in pulmonary fibrosis. Respir Res 2025; 26:128. [PMID: 40205454 PMCID: PMC11984030 DOI: 10.1186/s12931-025-03201-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Cellular senescence is one of the key steps in the progression of pulmonary fibrosis, and the senescence of type II alveolar epithelial cells (AEC IIs) may potentially accelerate the progression of pulmonary fibrosis. However, the molecular mechanisms underlying cellular senescence in pulmonary fibrosis remain unclear. METHODS The researchers first conducted in vitro experiments to investigate whether AEC IIs cultured on high matrix stiffness would lead to cellular senescence. Next, samples from mouse pulmonary fibrosis models and clinical idiopathic pulmonary fibrosis (IPF) patients were tested to observe extracellular matrix deposition, lamin A/C levels, and cellular senescence status in lung tissue. Construct lamin A/C knockdown and overexpression systems separately in AEC IIs, and observe whether changes in lamin A/C levels lead to cellular senescence. Further explore the degradation mechanism of lamin A/C using protein degradation inhibitors. RESULTS In vitro experiments have found that high matrix stiffness promotes senescence of AEC IIs. In a mouse model of pulmonary fibrosis, AEC IIs were found to exhibit significant cellular senescence on day 21. In clinical IPF samples, it was found that senescent cells expressed low levels of lamin A/C. In the lamin A/C SiRNA knockdown system, it was further confirmed that AEC IIs with low levels of lamin A/C are more prone to cellular senescence. Under high matrix stiffness, lamin A/C in AEC IIs is degraded through the autophagy lysosome pathway. The use of chloroquine can effectively alleviate cellular senescence. CONCLUSIONS High matrix stiffness degrades lamin A/C in pulmonary fibrosis through lysosomal degradation pathways, promoting AEC II senescence. Inhibition the degradation of lamin A/C could alleviate AEC II senescence.
Collapse
Affiliation(s)
- Junhui Ba
- Department of Medical Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Changyu Zheng
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yimei Lai
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Xin He
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yuxi Pan
- Department of Oncology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yanqiu Zhao
- Shenzhen Samii Medical Center, Shenzhen, Guangdong Province, China
| | - Huihui Xie
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Benquan Wu
- Department of Medical Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China.
| | - Xiao Deng
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China.
| | - Nan Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China.
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong Province, China.
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China.
| |
Collapse
|
15
|
Vilardo L, Cifola I, Nardella M, Pelucchi P, Ciotti MT, Bianchi A, Rinaldi A, Arisi I, Brandi R, d'Onofrio M, Galvanetto N, Gatti G, Catalano M, Lanzuolo C, Guglielmi L, D'Agnano I. Lamin A/C regulates cerebellar granule cell maturation. Cell Biol Toxicol 2025; 41:66. [PMID: 40186700 PMCID: PMC11972193 DOI: 10.1007/s10565-025-10011-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
Lamin A/C is a nuclear type V intermediate filament protein part of the meshwork structure underlying the inner nuclear membrane (nuclear lamina), which plays numerous roles, including maintenance of nuclear shape, heterochromatin organization, and transcriptional regulation. Our group has demonstrated the role of Lamin A/C in different pathophysiological conditions. Here, we investigated for the first time how Lamin A/C affects neuronal maturation in rat cerebellar granule cells (GCs). Primary rat cerebellar GCs where we silenced the Lmna gene constituted our key model; this provided a rather homogeneous cellular system showing a neuronal population in vitro. We then validated our findings in another in vivo murine model with knock-out of the Lmna gene and in an in vitro human neuronal model with silencing of the LMNA gene. We observed across three different models that Lamin A/C down-regulation affects neurons maturation by protecting the cells from glutamate-evoked excitotoxicity and correlates with an inhibition of calcium influxes and a down-regulation of pro-inflammatory cytokine pathways. Consistent with previous findings from our group, this study corroborates that Lamin A/C plays a key role in neural development and opens new significant implications for a better comprehension of the mechanisms involved in neurodegenerative diseases, where changes in the nuclear envelope are linked to neuroinflammatory processes and damage.
Collapse
Affiliation(s)
- Laura Vilardo
- CNR, Institute for Biomedical Technologies (ITB), Segrate, MI, Italy
| | - Ingrid Cifola
- CNR, Institute for Biomedical Technologies (ITB), Segrate, MI, Italy
| | - Marta Nardella
- Institute of Biochemistry and Cell Biology (IBBC), CNR, Monterotondo Scalo, RM, Italy
| | - Paride Pelucchi
- CNR, Institute for Biomedical Technologies (ITB), Segrate, MI, Italy
| | - Maria Teresa Ciotti
- Institute of Biochemistry and Cell Biology (IBBC), CNR, Monterotondo Scalo, RM, Italy
| | - Andrea Bianchi
- Istituto Nazionale Genetica Molecolare (INGM), Milan, Italy
| | - Arianna Rinaldi
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Ivan Arisi
- European Brain Research Institute (EBRI) "Rita Levi Montalcini", Rome, Italy
| | - Rossella Brandi
- European Brain Research Institute (EBRI) "Rita Levi Montalcini", Rome, Italy
| | - Mara d'Onofrio
- European Brain Research Institute (EBRI) "Rita Levi Montalcini", Rome, Italy
| | | | - Giuliana Gatti
- Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | - Loredana Guglielmi
- Faculty of Health and Medical Sciences, School of Biosciences, University of Surrey, Guildford, UK.
| | - Igea D'Agnano
- CNR, Institute for Biomedical Technologies (ITB), Segrate, MI, Italy.
| |
Collapse
|
16
|
Courbot O, Elosegui-Artola A. The role of extracellular matrix viscoelasticity in development and disease. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:10. [PMID: 40191103 PMCID: PMC11968406 DOI: 10.1038/s44341-025-00014-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 02/14/2025] [Indexed: 04/09/2025]
Abstract
For several decades, research has studied the influence of the extracellular matrix (ECM) mechanical properties in cell response, primarily emphasising its elasticity as the main determinant of cell and tissue behaviour. However, the ECM is not purely elastic; it is viscoelastic. ECM viscoelasticity has now emerged as a major regulator of collective cell dynamics. This review highlights recent findings on the role of ECM viscoelasticity in development and pathology.
Collapse
Affiliation(s)
- Olivia Courbot
- Cell and Tissue Mechanobiology Laboratory, The Francis Crick Institute, London, UK
- Department of Physics, King’s College London, London, UK
| | - Alberto Elosegui-Artola
- Cell and Tissue Mechanobiology Laboratory, The Francis Crick Institute, London, UK
- Department of Physics, King’s College London, London, UK
| |
Collapse
|
17
|
Bellitto D, Bozzo M, Ravera S, Bertola N, Rosamilia F, Milia J, Barboro P, Vargas GC, Di Lisa D, Pastorino L, Lantieri F, Castagnola P, Iervasi E, Ponassi M, Profumo A, Tkachenko K, Rosano C, Candiani S, Bachetti T. A multi-omics approach reveals impaired lipid metabolism and oxidative stress in a zebrafish model of Alexander disease. Redox Biol 2025; 81:103544. [PMID: 40023981 PMCID: PMC11915002 DOI: 10.1016/j.redox.2025.103544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 03/04/2025] Open
Abstract
Alexander disease (AxD) is a rare leukodystrophy caused by heterozygous mutations in the GFAP gene. To date, several in vitro and in vivo models have been generated in an attempt to unravel the main mechanisms underlying this complex disease. However, none of these models is suitable for investigating the global dysregulation caused by AxD. To address this shortcoming, we have generated a stable transgenic zebrafish line (zAxD) carrying the human GFAP p.R239C mutation, which is associated with severe phenotypes of AxD type I patients. We then performed transcriptomics and proteomics analyses on the whole larvae of our zAxD model, confirming the involvement of several pathways such as the immune system response and inflammation, oxidative stress, extracellular matrix, lipoxidation and lipid metabolism, which were previously reported in more limited omic studies. Interestingly, new pathways emerged as well, including tyrosine and butanoate metabolic processes. Biochemical assays confirmed alterations in cell respiration and lipid metabolism as well as elevated oxidative stress. These findings confirm the reliability of the zAxD model to apply a whole-organism approach to investigate the molecular basis of the disease.
Collapse
Affiliation(s)
- Deianira Bellitto
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova, Genova, Italy
| | - Matteo Bozzo
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova, Genova, Italy
| | - Silvia Ravera
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Nadia Bertola
- IRCCS Ospedale Policlinico San Martino, Genova, Unità Patologia Clinica, Italy
| | - Francesca Rosamilia
- Bioinformatica Clinica, Direzione Scientifica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Jessica Milia
- Centro di Ricerca, Sviluppo e Studi Superiori in Sardegna (CRS4), Pula, Italy
| | - Paola Barboro
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Donatella Di Lisa
- Dipartimento di Informatica, Bioingegneria, Robotica e Ingegneria dei Sistemi, Università di Genova, Genova, Italy
| | - Laura Pastorino
- Dipartimento di Informatica, Bioingegneria, Robotica e Ingegneria dei Sistemi, Università di Genova, Genova, Italy
| | - Francesca Lantieri
- Dipartimento di Scienze della Salute, Università di Genova, Genova, Italy
| | - Patrizio Castagnola
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova, Genova, Italy
| | - Erika Iervasi
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Marco Ponassi
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Aldo Profumo
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | | | - Simona Candiani
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | | |
Collapse
|
18
|
Wang X, Lang Z, Yan Z, Xu J, Zhang J, Jiao L, Zhang H. Dilated cardiomyopathy: from genes and molecules to potential treatments. Mol Cell Biochem 2025:10.1007/s11010-025-05269-0. [PMID: 40155570 DOI: 10.1007/s11010-025-05269-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Dilated cardiomyopathy is a myocardial condition marked by the enlargement of the heart's ventricular chambers and the gradual decline in systolic function, frequently resulting in congestive heart failure. Dilated cardiomyopathy has obvious familial characteristics, and mutations in related pathogenic genes can account for about 50% of patients with dilated cardiomyopathy. The most common genes related to dilated cardiomyopathy include TTN, LMNA, MYH7, etc. With more and more research on these genes, it will undoubtedly provide more potential targets and therapeutic pathways for the treatment of dilated cardiomyopathy. In addition, myocardial inflammation, myocardial metabolism abnormalities and cardiomyocyte apoptosis all have an important impact on the pathogenesis of dilated cardiomyopathy. Approximately half of sudden deaths among children and adolescents, along with the majority of patients undergoing heart transplantation, stem from cardiomyopathy. Therefore, precise and prompt clinical diagnosis holds paramount importance. Currently, diagnosis primarily hinges on the patient's medical background and imaging tests, with the significance of genetic testing steadily gaining prominence. The primary treatment for dilated cardiomyopathy remains heart transplantation. However, the scarcity of donors and the risk of severe immune rejection underscore the pressing need for novel therapies. Presently, research is actively exploring preclinical treatments like stem cell therapy as potential solutions.
Collapse
Affiliation(s)
- Xiumei Wang
- Department of Anesthesiology and Operating Theater, The First Hospital of Lanzhou University, Lanzhou, 730000, The People's Republic of China
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Zekun Lang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Zeyi Yan
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Jing Xu
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Jinyuan Zhang
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Lianhang Jiao
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Haijun Zhang
- Department of Anesthesiology and Operating Theater, The First Hospital of Lanzhou University, Lanzhou, 730000, The People's Republic of China.
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China.
| |
Collapse
|
19
|
Goelzer M, Howard S, Zavala AG, Conway D, Rubin J, Uzer G. Depletion of SUN1/2 induces heterochromatin accrual in mesenchymal stem cells during adipogenesis. Commun Biol 2025; 8:428. [PMID: 40082539 PMCID: PMC11906923 DOI: 10.1038/s42003-025-07832-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 02/24/2025] [Indexed: 03/16/2025] Open
Abstract
Critical to the mechano-regulation of mesenchymal stem cells (MSC), Linker of the Nucleoskeleton and Cytoskeleton (LINC) complex transduces cytoskeletal forces to the nuclei. The LINC complex contains outer nuclear membrane Nesprin proteins that associate with the cytoskeleton and their inner nuclear membrane couplers, SUN proteins. Here we tested the hypothesis that severing of the LINC complex-mediated cytoskeletal connections may have different effects on chromatin organization and MSC differentiation than those due to ablation of SUN proteins. In cells cultured under adipogenic conditions, interrupting LINC complex function through dominant-negative KASH domain expression (dnKASH) increased adipogesis while heterochromatin H3K27 and H3K9 methylation was unaltered. In contrast, SUN1/2 depletion inhibited adipogenic gene expression and fat droplet formation; as well the anti-adipogenic effect of SUN1/2 depletion was accompanied by increased H3K9me3, which was enriched on Adipoq, silencing this fat locus. We conclude that releasing the nucleus from cytoskeletal constraints via dnKASH accelerates adipogenesis while depletion of SUN1/2 increases heterochromatin accrual on adipogenic genes in a fashion independent of LINC complex function. Therefore, while these two approaches both disable LINC complex functions, their divergent effects on the epigenetic landscape indicate they cannot be used interchangeably to study mechanical regulation of cell differentiation.
Collapse
Affiliation(s)
- Matthew Goelzer
- Boise State University, Boise, ID, USA
- Oral Roberts University, Tulsa, OK, USA
| | | | | | - Daniel Conway
- The Ohio State University University, Columbus, OH, USA
| | - Janet Rubin
- University of North Carolina at Chapel Hill, Chapel Hill, USA
| | | |
Collapse
|
20
|
Momotyuk E, Ebrahim N, Shakirova K, Dashinimaev E. Role of the cytoskeleton in cellular reprogramming: effects of biophysical and biochemical factors. Front Mol Biosci 2025; 12:1538806. [PMID: 40123979 PMCID: PMC11926148 DOI: 10.3389/fmolb.2025.1538806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
The cytoskeleton plays a crucial role in regulating cellular behavior, acting as both a structural framework and a mediator of mechanical and biochemical signals that influence cell fate. In the context of cellular reprogramming, modifications to the cytoskeleton can have profound effects on lineage commitment and differentiation efficiency. This review explores the impact of mechanical forces such as substrate stiffness, topography, extracellular fluid viscosity, and cell seeding density on cytoskeletal organization and mechanotransduction pathways, including Rho/ROCK and YAP/TAZ signaling. Additionally, we examine the influence of biochemical agents that modulate cytoskeletal dynamics, such as actin and microtubule polymerization inhibitors, and their effects on stem cell differentiation. By understanding how cytoskeletal remodeling governs cellular identity, this review highlights potential strategies for improving reprogramming efficiency and directing cell fate by manipulating mechanical and biochemical cues.
Collapse
Affiliation(s)
| | | | | | - Erdem Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
21
|
Persano F, Parodi A, Pallaeva T, Kolesova E, Zamyatnin AA, Pokrovsky VS, De Matteis V, Leporatti S, Cascione M. Atomic Force Microscopy: A Versatile Tool in Cancer Research. Cancers (Basel) 2025; 17:858. [PMID: 40075706 PMCID: PMC11899184 DOI: 10.3390/cancers17050858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
The implementation of novel analytic methodologies in cancer and biomedical research has enabled the quantification of parameters that were previously disregarded only a few decades ago. A notable example of this paradigm shift is the widespread integration of atomic force microscopy (AFM) into biomedical laboratories, significantly advancing our understanding of cancer cell biology and treatment response. AFM allows for the meticulous monitoring of different parameters at the molecular and nanoscale levels, encompassing critical aspects such as cell morphology, roughness, adhesion, stiffness, and elasticity. These parameters can be systematically investigated in correlation with specific cell treatment, providing important insights into morpho-mechanical properties during normal and treated conditions. The resolution of this system holds the potential for its systematic adoption in clinics; its application could produce useful diagnostic information regarding the aggressiveness of cancer and the efficacy of treatment. This review endeavors to analyze the current literature, underscoring the pivotal role of AFM in biomedical research, especially in cancer cases, while also contemplating its prospective application in a clinical context.
Collapse
Affiliation(s)
- Francesca Persano
- Mathematics and Physics Department “Ennio De Giorgi”, University of Salento, Via Arnesano, 73100 Lecce, Italy; (F.P.); (V.D.M.)
- CNR Nanotec-Istituto di Nanotecnologia, Via Monteroni, 73100 Lecce, Italy
| | - Alessandro Parodi
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (T.P.); (E.K.); (V.S.P.)
| | - Tatiana Pallaeva
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (T.P.); (E.K.); (V.S.P.)
- Federal Scientific Research Center Crystallography and Photonics, Russian Academy of Sciences, 119333 Moscow, Russia
| | - Ekaterina Kolesova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (T.P.); (E.K.); (V.S.P.)
| | - Andrey A. Zamyatnin
- Department of Biological Chemistry, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Vadim S. Pokrovsky
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (T.P.); (E.K.); (V.S.P.)
- N.N. Blokhin Medical Research Center of Oncology, 115478 Moscow, Russia
- Patrice Lumumba People’s Friendship University, 117198 Moscow, Russia
| | - Valeria De Matteis
- Mathematics and Physics Department “Ennio De Giorgi”, University of Salento, Via Arnesano, 73100 Lecce, Italy; (F.P.); (V.D.M.)
- Institute for Microelectronics and Microsystems (IMM), National Research Council (CNR), Via Monteroni, 73100 Lecce, Italy
| | - Stefano Leporatti
- CNR Nanotec-Istituto di Nanotecnologia, Via Monteroni, 73100 Lecce, Italy
| | - Mariafrancesca Cascione
- Mathematics and Physics Department “Ennio De Giorgi”, University of Salento, Via Arnesano, 73100 Lecce, Italy; (F.P.); (V.D.M.)
- Institute for Microelectronics and Microsystems (IMM), National Research Council (CNR), Via Monteroni, 73100 Lecce, Italy
| |
Collapse
|
22
|
Jin L, Qin Y, Zhao Y, Zhou X, Zeng Y. Endothelial cytoskeleton in mechanotransduction and vascular diseases. J Biomech 2025; 182:112579. [PMID: 39938443 DOI: 10.1016/j.jbiomech.2025.112579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
The cytoskeleton is an important structural component that regulates various aspects of cell morphology, movement, and intracellular signaling. It plays a pivotal role in the cellular response to biomechanical stimuli, particularly in endothelial cells, which are critical for vascular homeostasis and the pathogenesis of cardiovascular diseases. Mechanical forces, such as shear and tension, activate intracellular signaling cascades that regulate transcription, translation, and cellular behaviors. Despite extensive research into cytoskeletal functions, the precise mechanisms by which the cytoskeleton transduces mechanical signals remain incompletely understood. This review focuses on the role of cytoskeletal components in membrane, cytoplasm, and nucleus in mechanotransduction, with an emphasis on their structure, mechanical and biological behaviors, dynamic interactions, and response to mechanical forces. The collaboration between membrane cytoskeleton, cytoplasmic cytoskeleton, and nucleoskeleton is indispensable for endothelial cells to respond to mechanical stimuli. Understanding their mechanoresponsive mechanisms is essential for advancing therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Linlu Jin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Yixue Qin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Yunran Zhao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Xintong Zhou
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China.
| |
Collapse
|
23
|
Karling T, Weavers H. Immune cells adapt to confined environments in vivo to optimise nuclear plasticity for migration. EMBO Rep 2025; 26:1238-1268. [PMID: 39915297 PMCID: PMC11894099 DOI: 10.1038/s44319-025-00381-0] [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: 04/30/2024] [Revised: 01/05/2025] [Accepted: 01/17/2025] [Indexed: 03/12/2025] Open
Abstract
Cells navigating in complex 3D microenvironments frequently encounter narrow spaces that physically challenge migration. While in vitro studies identified nuclear stiffness as a key rate-limiting factor governing the movement of many cell types through artificial constraints, how cells migrating in vivo respond dynamically to confinement imposed by local tissue architecture, and whether these encounters trigger molecular adaptations, is unclear. Here, we establish an innovative in vivo model for mechanistic analysis of nuclear plasticity as Drosophila immune cells transition into increasingly confined microenvironments. Integrating live in vivo imaging with molecular genetic analyses, we demonstrate how rapid molecular adaptation upon environmental confinement (including fine-tuning of the nuclear lamina) primes leukocytes for enhanced nuclear deformation while curbing damage (including rupture and micronucleation), ultimately accelerating movement through complex tissues. We find nuclear dynamics in vivo are further impacted by large organelles (phagosomes) and the plasticity of neighbouring cells, which themselves deform during leukocyte passage. The biomechanics of cell migration in vivo are thus shaped both by factors intrinsic to individual immune cells and the malleability of the surrounding microenvironment.
Collapse
Affiliation(s)
- Tua Karling
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Helen Weavers
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
24
|
Morival J, Hazelwood A, Lammerding J. Feeling the force from within - new tools and insights into nuclear mechanotransduction. J Cell Sci 2025; 138:JCS263615. [PMID: 40059756 PMCID: PMC11959624 DOI: 10.1242/jcs.263615] [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: 04/03/2025] Open
Abstract
The ability of cells to sense and respond to mechanical signals is essential for many biological processes that form the basis of cell identity, tissue development and maintenance. This process, known as mechanotransduction, involves crucial feedback between mechanical force and biochemical signals, including epigenomic modifications that establish transcriptional programs. These programs, in turn, reinforce the mechanical properties of the cell and its ability to withstand mechanical perturbation. The nucleus has long been hypothesized to play a key role in mechanotransduction due to its direct exposure to forces transmitted through the cytoskeleton, its role in receiving cytoplasmic signals and its central function in gene regulation. However, parsing out the specific contributions of the nucleus from those of the cell surface and cytoplasm in mechanotransduction remains a substantial challenge. In this Review, we examine the latest evidence on how the nucleus regulates mechanotransduction, both via the nuclear envelope (NE) and through epigenetic and transcriptional machinery elements within the nuclear interior. We also explore the role of nuclear mechanotransduction in establishing a mechanical memory, characterized by a mechanical, epigenetic and transcriptomic cell state that persists after mechanical stimuli cease. Finally, we discuss current challenges in the field of nuclear mechanotransduction and present technological advances that are poised to overcome them.
Collapse
Affiliation(s)
- Julien Morival
- Weill Institute for Cell and Molecular Biology, Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Anna Hazelwood
- Weill Institute for Cell and Molecular Biology, Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology, Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
25
|
Ramirez A, Sriram V, Abbouchi Y, Patolia R, Passaro E, Kaluzienski M, Maisel K. Inflammation modulates lymph node biomechanics in a sex-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639945. [PMID: 40060646 PMCID: PMC11888317 DOI: 10.1101/2025.02.24.639945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
Abstract
Lymph nodes are highly specialized immune organs that orchestrate the adaptive immune response. In the lymph nodes, naïve B and T lymphocytes encounter cognate antigens, sparking their activation and response to foreign substances. Lymph nodes grow in response to an immune challenge, at least in part to accommodate increased numbers of infiltrating and proliferating B and T lymphocytes. This behavior is supported by a robust three-dimensional network of extracellular matrix (ECM) fibers and fibroblastic reticular cells (FRCs). ECM fibers and FRCs work synergistically to alternate stretching and contractile forces between them allowing the lymph node to maintain structural integrity during rapid tissue reconstruction. These changes ultimately alter the material properties of the lymph node, which can impact cell migration, proliferation, and differentiation. Recent work has investigated the physiological implications of the changing lymph node microenvironment; however, the biophysical properties of the lymph nodes during these changes remain largely unexplored. Here, we use multiple particle tracking microrheology (MPT), a minimally invasive nanoparticle-based technique to investigate the biophysical properties (elastic/loss moduli, microviscosity, pore size) of lymph nodes post inflammatory stimulus. Our results highlight mechanical changes both during the initial phases of the acute inflammatory response and upon resolution of inflammation, a topic that is relatively understudied. We show that B and T cell rich areas restructure independently, with T cell zones remodeling significantly and exhibiting nearly a 3-fold higher elastic modulus. Additionally, for the first time, we show that biological sex modulates lymph node biomechanics in acute inflammation: Lymph nodes from female mice showed a ~20-fold increase in elastic and loss moduli at peak inflammation, while lymph nodes from male mice had a ~5-fold decrease in both moduli. Additionally, lymph nodes from female mice appeared to permanently remodel during the resolution of acute inflammation resulting in the maintenance of an overall higher elastic and loss modulus, while lymph nodes from male mice returned to the biomechanics of untreated lymph nodes. We also found that at least some of the changes in biomechanical properties were correlated with changes in ECM materials in the lymph nodes, suggesting a structure-function relationship. Overall, our studies provide key insights into how biomechanical properties in lymph nodes are altered during inflammation, a previously unstudied area, and lay the foundation for structure-function relationships involved in immune response. Additionally, we demonstrate a robust technique for the analysis of the lymph node interstitial tissue properties and how they vary with inflammatory stimuli.
Collapse
Affiliation(s)
- Ann Ramirez
- Fischell Department of Bioengineering, University of Maryland, College Park
| | - Vedanth Sriram
- Fischell Department of Bioengineering, University of Maryland, College Park
- Biophysics Program, University of Maryland, College Park
| | - Yassmin Abbouchi
- Fischell Department of Bioengineering, University of Maryland, College Park
| | - Reina Patolia
- Fischell Department of Bioengineering, University of Maryland, College Park
| | - Emily Passaro
- Fischell Department of Bioengineering, University of Maryland, College Park
| | | | - Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, College Park
- Biophysics Program, University of Maryland, College Park
| |
Collapse
|
26
|
Verdugo-Avello F, Wychowaniec JK, Villacis-Aguirre CA, D'Este M, Toledo JR. Bone microphysiological models for biomedical research. LAB ON A CHIP 2025; 25:806-836. [PMID: 39906932 DOI: 10.1039/d4lc00762j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Bone related disorders are highly prevalent, and many of these pathologies still do not have curative and definitive treatment methods. This is due to a complex interplay of multiple factors, such as the crosstalk between different tissues and cellular components, all of which are affected by microenvironmental factors. Moreover, these bone pathologies are specific, and current treatment results vary from patient to patient owing to their intrinsic biological variability. Current approaches in drug development to deliver new drug candidates against common bone disorders, such as standard two-dimensional (2D) cell culture and animal-based studies, are now being replaced by more relevant diseases modelling, such as three-dimension (3D) cell culture and primary cells under human-focused microphysiological systems (MPS) that can resemble human physiology by mimicking 3D tissue organization and cell microenvironmental cues. In this review, various technological advancements for in vitro bone modeling are discussed, highlighting the progress in biomaterials used as extracellular matrices, stem cell biology, and primary cell culture techniques. With emphasis on examples of modeling healthy and disease-associated bone tissues, this tutorial review aims to survey current approaches of up-to-date bone-on-chips through MPS technology, with special emphasis on the scaffold and chip capabilities for mimicking the bone extracellular matrix as this is the key environment generated for cell crosstalk and interaction. The relevant bone models are studied with critical analysis of the methods employed, aiming to serve as a tool for designing new and translational approaches. Additionally, the features reported in these state-of-the-art studies will be useful for modeling bone pathophysiology, guiding future improvements in personalized bone models that can accelerate drug discovery and clinical translation.
Collapse
Affiliation(s)
- Francisco Verdugo-Avello
- Biotechnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| | | | - Carlos A Villacis-Aguirre
- Biotechnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| | - Matteo D'Este
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
| | - Jorge R Toledo
- Biotechnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| |
Collapse
|
27
|
Waldherr A, Fogtman A. Radiation symptoms resemble laminopathies and the physical underlying cause may sit at the lamin A C-terminus. Mol Med 2025; 31:69. [PMID: 39979866 PMCID: PMC11844092 DOI: 10.1186/s10020-025-01081-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/13/2025] [Indexed: 02/22/2025] Open
Abstract
Ionizing radiation causes three divergent effects in the human body: On one side, tissue death (= deterministic effects) sets on, on the other side, mutations and cancer growth (= stochastic effects) can occur. In recent years, the additional phenomenon of accelerated aging has come to light. In the following, we argue that these seemingly contradictory radiation responses namely: (i) increased cancer growth, (ii) ablation of cancer tissue or (iii) deterministic senescence, share an underlying cause from damage at the lamin A C-terminus. In other words, besides the typically described genomic radiation impact, we propose an additional destabilization pathway via oxidation at the nuclear envelope. We propose five concrete hypotheses that draw a direct mechanistic model from radiation damage and cellular oxidative stress, to micronuclei and clinical symptoms. In conjunction with lamin B compensation, we might be able to explain why deterministic or stochastic responses dominate. If our model holds true, a novel target for radiotherapeutics and radiooncology arises, and a rationale to closer connect laminopathy and radioprotection research.
Collapse
Affiliation(s)
- Alexandra Waldherr
- Max-Planck Institute for Biology, Max-Planck-Ring 5, 72076, Tübingen, Germany.
- European Space Agency, Space Medicine Team, EAC European Astronaut Center, EAC Linder Höhe, 51147, Troisdorf, Germany.
| | - Anna Fogtman
- European Space Agency, Space Medicine Team, EAC European Astronaut Center, EAC Linder Höhe, 51147, Troisdorf, Germany
| |
Collapse
|
28
|
Sinha S, Fleck M, Ayushman M, Tong X, Mikos G, Jones S, Soto L, Yang F. Matrix Stiffness Regulates GBM Migration and Chemoradiotherapy Responses via Chromatin Condensation in 3D Viscoelastic Matrices. ACS APPLIED MATERIALS & INTERFACES 2025; 17:10342-10359. [PMID: 39912753 DOI: 10.1021/acsami.4c16993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Glioblastoma multiforme (GBM) progression is associated with changes in matrix stiffness, and different regions of the tumor niche exhibit distinct stiffnesses. Using elastic hydrogels, previous work has demonstrated that matrix stiffness modulates GBM behavior and drug responses. However, brain tissue is viscoelastic, and how stiffness impacts the GBM invasive phenotype and response to therapy within a viscoelastic niche remains largely unclear. Here, we report a three-dimensional (3D) viscoelastic GBM hydrogel system that models the stiffness heterogeneity present within the tumor niche. We find that GBM cells exhibit enhanced migratory ability, proliferation, and resistance to radiation in soft matrices, mimicking the tumor core and perifocal margins. Conversely, GBM cells remain confined and demonstrate increased resistance to chemotherapy in stiff matrices mimicking edematous tumor regions. We identify that stiffness-induced changes in the GBM phenotype are regulated by nuclear mechanosensing and chromatin condensation. Pharmacologically decondensing the chromatin significantly impedes GBM migration and overcomes stiffness-induced chemoresistance and radioresistance. Our findings highlight that stiffness regulates aggressive GBM behavior in viscoelastic matrices through mechanotransduction processes. Finally, we reveal the critical role of chromatin condensation in mediating GBM migration and therapy resistance, offering a potential new therapeutic target to improve GBM treatment outcomes.
Collapse
Affiliation(s)
- Sauradeep Sinha
- Department of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Mark Fleck
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Manish Ayushman
- Department of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Xinming Tong
- Department of Orthopaedic Surgery, Stanford University, Stanford, California 94305, United States
| | - Georgios Mikos
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Sarah Jones
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Luis Soto
- Department of Radiation Oncology, Stanford University, Stanford, California 94305, United States
| | - Fan Yang
- Department of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department of Orthopaedic Surgery, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
29
|
Campo F, Neroni A, Pignatelli C, Pellegrini S, Marzinotto I, Valla L, Manenti F, Policardi M, Lampasona V, Piemonti L, Citro A. Bioengineering of a human iPSC-derived vascularized endocrine pancreas for type 1 diabetes. Cell Rep Med 2025; 6:101938. [PMID: 39922198 PMCID: PMC11866511 DOI: 10.1016/j.xcrm.2025.101938] [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: 03/15/2024] [Revised: 11/18/2024] [Accepted: 01/13/2025] [Indexed: 02/10/2025]
Abstract
Intrahepatic islet transplantation in patients with type 1 diabetes is limited by donor availability and lack of engraftment. Alternative β cell sources and transplantation sites are needed. We demonstrate the feasibility to repurpose a decellularized lung as an endocrine pancreas for β cell replacement. We bioengineer an induced pluripotent stem cell (iPSC)-based version, fabricating a human iPSC-based vascularized endocrine pancreas (iVEP) using iPSC-derived β cells (iPSC-derived islets [SC-islets]) and endothelial cells (iECs). SC-islets and iECs are aggregated into vascularized iβ spheroids (ViβeSs), and over 7 days of culture, spheroids integrate into the bioengineered vasculature, generating a functional, perfusable human endocrine organ. In vitro, the vascularized extracellular matrix (ECM) sustained SC-islet engraftment and survival with a significantly preserved β cell mass and a physiologic insulin release. In vivo, iVEP restores normoglycemia in diabetic NSG mice. We report a human iVEP providing a controlled in vitro insulin-secreting phenotype and in vivo function.
Collapse
Affiliation(s)
- Francesco Campo
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Alessia Neroni
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Cataldo Pignatelli
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Pellegrini
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Marzinotto
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Libera Valla
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
| | - Fabio Manenti
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Policardi
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vito Lampasona
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
30
|
Urbanska M, Ge Y, Winzi M, Abuhattum S, Ali SS, Herbig M, Kräter M, Toepfner N, Durgan J, Florey O, Dori M, Calegari F, Lolo FN, del Pozo MÁ, Taubenberger A, Cannistraci CV, Guck J. De novo identification of universal cell mechanics gene signatures. eLife 2025; 12:RP87930. [PMID: 39960760 PMCID: PMC11832173 DOI: 10.7554/elife.87930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025] Open
Abstract
Cell mechanical properties determine many physiological functions, such as cell fate specification, migration, or circulation through vasculature. Identifying factors that govern the mechanical properties is therefore a subject of great interest. Here, we present a mechanomics approach for establishing links between single-cell mechanical phenotype changes and the genes involved in driving them. We combine mechanical characterization of cells across a variety of mouse and human systems with machine learning-based discriminative network analysis of associated transcriptomic profiles to infer a conserved network module of five genes with putative roles in cell mechanics regulation. We validate in silico that the identified gene markers are universal, trustworthy, and specific to the mechanical phenotype across the studied mouse and human systems, and demonstrate experimentally that a selected target, CAV1, changes the mechanical phenotype of cells accordingly when silenced or overexpressed. Our data-driven approach paves the way toward engineering cell mechanical properties on demand to explore their impact on physiological and pathological cell functions.
Collapse
Affiliation(s)
- Marta Urbanska
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und MedizinErlangenGermany
| | - Yan Ge
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Maria Winzi
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Shada Abuhattum
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und MedizinErlangenGermany
| | - Syed Shafat Ali
- Center for Complex Network Intelligence, Tsinghua Laboratory of Brain and Intelligence, Department of Computer Science and School of Biomedical Engineering, Tsinghua UniversityBeijingChina
- Department of Computer Science and Department of Economics, Jamia Millia IslamiaNew DelhiIndia
| | - Maik Herbig
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und MedizinErlangenGermany
- Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Martin Kräter
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und MedizinErlangenGermany
| | - Nicole Toepfner
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Joanne Durgan
- Signalling Programme, The Babraham InstituteCambridgeUnited Kingdom
| | - Oliver Florey
- Signalling Programme, The Babraham InstituteCambridgeUnited Kingdom
| | - Martina Dori
- Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Federico Calegari
- Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Fidel-Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Miguel Ángel del Pozo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Anna Taubenberger
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Carlo Vittorio Cannistraci
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Center for Complex Network Intelligence, Tsinghua Laboratory of Brain and Intelligence, Department of Computer Science and School of Biomedical Engineering, Tsinghua UniversityBeijingChina
- Center for Systems Biology DresdenDresdenGermany
- Cluster of Excellence Physics of Life, Technische Universität DresdenDresdenGermany
| | - Jochen Guck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und MedizinErlangenGermany
| |
Collapse
|
31
|
Wu X, Fei W, Shen T, Ye L, Li C, Chu S, Liu M, Cheng X, Qin J. Unveiling the potential of biomechanics in pioneering innovative strategies for cancer therapy. Theranostics 2025; 15:2903-2932. [PMID: 40083943 PMCID: PMC11898300 DOI: 10.7150/thno.108605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/02/2025] [Indexed: 03/16/2025] Open
Abstract
Mechanical force transmission is pivotal in tumor biology, profoundly affecting cancer cell behaviors such as proliferation, metastasis, and resistance to therapy. To explore novel biomechanical-based therapeutic strategies for cancer treatment, this paper deciphers the advances in biomechanical measurement approaches and the impact of biomechanical signals on fundamental oncological processes such as tumor microenvironment remodeling, angiogenesis, metastasis, and drug resistance. Then, the mechanisms of biomechanical signal transduction of tumor cells are demonstrated to identify novel targets for tumor therapy. Additionally, this study proposes a novel tumor treatment strategy, the biomechanical regulation tumor nanotherapeutics, including smart biomaterials designed to disturb mechanical signaling pathways and innovative nanodrugs that interfere transduction of biomechanical signals to improve tumor therapeutic outcomes. These methods mark a departure from conventional pharmacological therapies to novel strategies that utilize mechanical forces to impede tumor progression and enhance tumor responsiveness to treatment. In general, this review highlights the critical role of biomechanical signals in cancer biology from a holistic perspective and underscores the potential of biomechanical interventions as a transformative class of therapeutics. By integrating mechanobiology into the development of cancer treatments, this paper paves the way for more precise and effective strategies that leverage the inherent physical properties of the tumor microenvironment.
Collapse
Affiliation(s)
- Xiaodong Wu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Weidong Fei
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Tao Shen
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Lei Ye
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Chaoqun Li
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Siran Chu
- Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Mingqi Liu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xiaodong Cheng
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, 310006, China
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Hangzhou, 310006, China
| | - Jiale Qin
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, 310006, China
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Hangzhou, 310006, China
| |
Collapse
|
32
|
Shen K, Lv Z, Yang Y, Wang H, Liu J, Chen Q, Liu Z, Zhang M, Liu J, Cheng Y. A Wet-Adhesion and Swelling-Resistant Hydrogel for Fast Hemostasis, Accelerated Tissue Injury Healing and Bioelectronics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414092. [PMID: 39713944 DOI: 10.1002/adma.202414092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/01/2024] [Indexed: 12/24/2024]
Abstract
Hydrogel bioadhesives with adequate wet adhesion and swelling resistance are urgently needed in clinic. However, the presence of blood or body fluid usually weakens the interfacial bonding strength, and even leads to adhesion failure. Herein, profiting from the unique coupling structure of carboxylic and phenyl groups in one component (N-acryloyl phenylalanine) for interfacial drainage and matrix toughening as well as various electrostatic interactions mediated by zwitterions, a novel hydrogel adhesive (PAAS) is developed with superior tissue adhesion properties and matrix swelling resistance in challenging wet conditions (adhesion strength of 85 kPa, interfacial toughness of 450 J m-2, burst pressure of 514 mmHg, and swelling ratio of <4%). The PAAS hydrogel can not only realize fast hemostasis of liver, heart, artery rupture, and sealing of pulmonary air-leakage but also accelerate the recovery of stomach and liver defects in rat, rabbit, and pig models. Moreover, PAAS hydrogel can precisely and durably monitor various physiological activities (pulse, electrocardiogram, and electromyogram) even under humid environments (immersion in water for 3 days), and can be employed for the evaluation of in vivo sealing efficiency for artery rupture. The work provides a promising hydrogel adhesive for clinical hemostasis, tissue injury repair, and bioelectronics.
Collapse
Affiliation(s)
- Kaixiang Shen
- Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, School of Chemistry, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhuting Lv
- Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, School of Chemistry, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuxuan Yang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Haoyue Wang
- Institute of High Voltage Physics and Engineering, School of Electrical Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jiancheng Liu
- Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, School of Chemistry, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Qifei Chen
- Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, School of Chemistry, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zheng Liu
- Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, School of Chemistry, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Mengyuan Zhang
- Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, School of Chemistry, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jiaying Liu
- Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, School of Chemistry, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yilong Cheng
- Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, School of Chemistry, Xi'an Jiaotong University, Xi'an, 710049, China
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
33
|
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 2025; 24:312-322. [PMID: 39487316 DOI: 10.1038/s41563-024-02038-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
|
34
|
Paganelli F, Poli A, Truocchio S, Martelli AM, Palumbo C, Lattanzi G, Chiarini F. At the nucleus of cancer: how the nuclear envelope controls tumor progression. MedComm (Beijing) 2025; 6:e70073. [PMID: 39866838 PMCID: PMC11758262 DOI: 10.1002/mco2.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 01/28/2025] Open
Abstract
Historically considered downstream effects of tumorigenesis-arising from changes in DNA content or chromatin organization-nuclear alterations have long been seen as mere prognostic markers within a genome-centric model of cancer. However, recent findings have placed the nuclear envelope (NE) at the forefront of tumor progression, highlighting its active role in mediating cellular responses to mechanical forces. Despite significant progress, the precise interplay between NE components and cancer progression remains under debate. In this review, we provide a comprehensive and up-to-date overview of how changes in NE composition affect nuclear mechanics and facilitate malignant transformation, grounded in the latest molecular and functional studies. We also review recent research that uses advanced technologies, including artificial intelligence, to predict malignancy risk and treatment outcomes by analyzing nuclear morphology. Finally, we discuss how progress in understanding nuclear mechanics has paved the way for mechanotherapy-a promising cancer treatment approach that exploits the mechanical differences between cancerous and healthy cells. Shifting the perspective on NE alterations from mere diagnostic markers to potential therapeutic targets, this review calls for further investigation into the evolving role of the NE in cancer, highlighting the potential for innovative strategies to transform conventional cancer therapies.
Collapse
Affiliation(s)
- Francesca Paganelli
- Department of Biomedical and Neuromotor SciencesAlma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Alessandro Poli
- IFOM ETS ‐ The AIRC Institute of Molecular OncologyMilanItaly
| | - Serena Truocchio
- Department of Biomedical and Neuromotor SciencesAlma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Alberto M. Martelli
- Department of Biomedical and Neuromotor SciencesAlma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Carla Palumbo
- Department of BiomedicalMetabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza”Unit of BolognaBolognaItaly
- IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Francesca Chiarini
- Department of BiomedicalMetabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| |
Collapse
|
35
|
Zhang X, Huang N, Mu Y, Chen H, Zhu M, Zhang S, Liu P, Zhang H, Deng H, Feng K, Shang Q, Liu X, Zhang C, Shi M, Yang L, Sun J, Kong G, Geng J, Lu S, Li Z. Mechanical Force-Induced cGAS Activation in Carcinoma Cells Facilitates Splenocytes into Liver to Drive Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2401127. [PMID: 39737867 PMCID: PMC11848607 DOI: 10.1002/advs.202401127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 11/27/2024] [Indexed: 01/01/2025]
Abstract
Liver metastasis is the main cause of cancer-related mortality. During the metastasis process, circulating carcinoma cells hardly pass through narrow capillaries, leading to nuclear deformation. However, the effects of nuclear deformation and its underlying mechanisms on metastasis need further study. Here, it is shown that mechanical force-induced nuclear deformation exacerbates liver metastasis by activating the cGAS-STING pathway, which promotes splenocyte infiltration in the liver. Mechanical force results in nuclear deformation and rupture of the nuclear envelope with inevitable DNA leakage. Cytoplasmic DNA triggers the activation of cGAS-STING pathway, enhancing the production of IL6, TNFα, and CCL2. Additionally, splenocyte recruitment by the proinflammatory cytokines support carcinoma cell survival and colonization in the liver. Importantly, both intervening activity of cGAS and blocking of splenocyte migration to the liver efficiently ameliorate liver metastasis. Overall, these findings reveal a mechanism by which mechanical force-induced nuclear deformation exacerbates liver metastasis by regulating splenocyte infiltration into the liver and support targeting cGAS and blocking splenocyte recruitment as candidate therapeutic approaches for liver metastasis.
Collapse
|
36
|
Zheng F, Wu T, Wang F, Tang H, Cui X, Liu D, Chen P, Fu J, Li C, Jiang J. Effect of low-intensity pulsed ultrasound on the mineralization of force-treated cementoblasts and orthodontically induced inflammatory root resorption via the Lamin A/C-Yes associated protein axis. J Periodontal Res 2025; 60:189-199. [PMID: 39095980 DOI: 10.1111/jre.13330] [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: 04/08/2024] [Revised: 07/07/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
AIMS Orthodontic treatment commonly results in orthodontically induced inflammatory root resorption (OIIRR). This condition arises from excessive orthodontic force, which triggerslocal inflammatory responses and impedes cementoblasts' mineralization capacity. Low-intensity pulsed ultrasound (LIPUS) shows potential in reducing OIIRR. However, the precise mechanisms through which LIPUS reduces OIIRR remain unclear. This study aimed to explore the effects and mechanisms of LIPUS on the mineralization of force-treated cementoblasts and its impact on OIIRR. METHODS We established a rat OIIRR model and locally administered LIPUS stimulation for 7 and 14 days. We analyzed root resorption volume, osteoclast differentiation, and the expression of osteocalcin and yes-associated protein 1 (YAP1) using micro-computed tomography (micro-CT), hematoxylin and eosin, tartrate-resistant acid phosphatase, immunofluorescence and immunohistochemistry staining. In vitro, we applied compressive force and LIPUS to the immortalized mouse cementoblasts (OCCM30). We assessed mineralization using alkaline phosphatase (ALP) staining, alizarin red staining, real-time quantitative polymerase chain reaction, Western blotting and immunofluorescence staining. RESULTS In rats, LIPUS reduced OIIRR, as evidenced by micro-CT analysis and histological staining. In vitro, LIPUS enhanced mineralization of force-treated OCCM30 cells, as indicated by ALP and alizarin red staining, upregulated mRNA expression of mineralization-related genes, and increased protein expression of mineralization markers. Mechanistically, LIPUS activated YAP1 signaling via the cytoskeleton-Lamin A/C pathway, supported by immunofluorescence and Western blot analysis. CONCLUSION This study demonstrates that LIPUS promotes mineralization in force-treated cementoblasts and reduces OIIRR by activating YAP1 through the cytoskeletal-Lamin A/C signaling pathway. These findings provide fresh insights into how LIPUS benefits orthodontic treatment and suggest potential strategies for preventing and treating OIIRR.
Collapse
Affiliation(s)
- Fu Zheng
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Tong Wu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Feifei Wang
- Center of Digital Dentistry, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Hongyi Tang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Xinyu Cui
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Duo Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Peng Chen
- Department of Orthodontics, School of Stomatology Affiliated to Medical College, Zhejiang University, Hangzhou, China
| | - Jiangfeng Fu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Cuiying Li
- Central Laboratory, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Jiuhui Jiang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
37
|
Bosch‐Calvet M, Pérez‐Venteo A, Cebria‐Xart A, Garcia‐Cajide M, Mauvezin C. Nuclear stiffness through lamin A/C overexpression differentially modulates chromosomal instability biomarkers. Biol Cell 2025; 117:e12001. [PMID: 40012191 PMCID: PMC11865694 DOI: 10.1111/boc.12001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/22/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND INFORMATION Mitosis is crucial for the faithful transmission of genetic material, and disruptions can result in chromosomal instability (CIN), a hallmark of cancer. CIN is a known driver of tumor heterogeneity and anti-cancer drug resistance, thus highlighting the need to assess CIN levels in cancer cells to design effective targeted therapy. While micronuclei are widely recognized as CIN markers, we have recently identified the toroidal nucleus, a novel ring-shaped nuclear phenotype arising as well from chromosome mis-segregation. RESULTS Here, we examined whether increasing nuclear envelope stiffness through lamin A/C overexpression could affect the formation of toroidal nuclei and micronuclei. Interestingly, lamin A/C overexpression led to an increase in toroidal nuclei while reducing micronuclei prevalence. We demonstrated that chromatin compaction and nuclear stiffness drive the formation of toroidal nuclei. Furthermore, inhibition of autophagy and lysosomal function elevated the frequency of toroidal nuclei without affecting the number of micronuclei in the whole cell population. We demonstrated that this divergence between the two CIN biomarkers is independent of defects in lamin A processing. CONCLUSIONS AND SIGNIFICANCE These findings uncover a complex interplay between nuclear architecture and levels of CIN, advancing our understanding of the mechanisms supporting genomic stability and further contributing to cancer biology.
Collapse
Affiliation(s)
- Mireia Bosch‐Calvet
- Departament de BiomedicinaFacultat de Medicina i Ciències de la SalutUniversitat de BarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Alejandro Pérez‐Venteo
- Departament de BiomedicinaFacultat de Medicina i Ciències de la SalutUniversitat de BarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Alex Cebria‐Xart
- Institut de Recerca Sant Joan de Déu (IRSJD)BarcelonaSpain
- Cancer Science Programme, Laboratory of Pediatric Cancer EpigeneticsInstitute for Research in Biomedicine (IRB Barcelona)BarcelonaSpain
| | - Marta Garcia‐Cajide
- Departament de BiomedicinaFacultat de Medicina i Ciències de la SalutUniversitat de BarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Caroline Mauvezin
- Departament de BiomedicinaFacultat de Medicina i Ciències de la SalutUniversitat de BarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| |
Collapse
|
38
|
Anlaş AA, Sprenger MT, Wang M, Ontko N, Phan S, Discher DE. Matrix stiffness induces heritable changes in chromosome numbers, consistent with solid tumor heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635370. [PMID: 39975263 PMCID: PMC11838315 DOI: 10.1101/2025.01.28.635370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Solid tumors often have an abundance of collagen-I that stiffens the tissue, and they are invariably driven by mutations that include chromosome losses and gains. These observations are linked here by showing that 3D matrix stiffness induces heritable changes to a cell ' s DNA. We use live-cell chromosome reporters (ChReporters) and hydrogels of tunable stiffness to show mitotic compression, micronuclei counts, ChReporter losses and heterogeneity all increase as functions of stiffness. Increased mistakes occur despite suppressed cell division in stiff matrix and minimal size variation between spheroids. Colonies of ChReporter-negative cells within cancer spheroids align with Luria-Delbruck ' s seminal theory for heritable mutations, which predicts inter-spheroid variances that exceed Poisson statistics. Suppression of the contractility motor Myosin-II also increases chromosome loss in 3D but not 2D and does not affect spheroid growth - thus clarifying Myosin-II ' s putative role as a tumor suppressor. Consistent with experiments, pan-cancer analyses of clinical data associates chromosome losses and gains with collagen-I levels and genetic variation. Stiff extracellular matrix thus drives mechano-evolution of solid tumors as a Darwin-Lamarck process with heterogeneity that complicates therapy.
Collapse
|
39
|
Gharacheh H, Abaci A, Alkhoury K, Choudhury E, Liaw CY, Chester SA, Guvendiren M. Comparative evaluation of melt- vs. solution-printed poly(ε-caprolactone)/hydroxyapatite scaffolds for bone tissue engineering applications. SOFT MATTER 2025; 21:844-854. [PMID: 39611880 DOI: 10.1039/d4sm01197j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Material extrusion-based three-dimensional (3D) printing is a widely used manufacturing technology for fabricating scaffolds and devices in bone tissue engineering (BTE). This technique involves two fundamentally different extrusion approaches: solution-based and melt-based printing. In solution-based printing, a polymer solution is extruded and solidifies via solvent evaporation, whereas in melt-based printing, the polymer is melted at elevated temperatures and solidifies as it cools post-extrusion. Solution-based printing can also be enhanced to generate micro/nano-scale porosity through phase separation by printing the solution into a nonsolvent bath. The choice of the printing method directly affects scaffold properties and the biological response of stem cells. In this study, we selected polycaprolactone (PCL), a biodegradable polymer frequently used in BTE, blended with hydroxyapatite (HA) nanoparticles, a bioceramic known for promoting bone formation, to investigate the effects of the printing approach on scaffold properties and performance in vitro using human mesenchymal stem cells (hMSCs). Our results showed that while both printing methods produced scaffolds with similar strut and overall scaffold dimensions, solvent-based printing resulted in porous struts, higher surface roughness, lower stiffness, and increased crystallinity compared to melt-based printing. Although stem cell viability and proliferation were not significantly influenced by the printing approach, melt-printed scaffolds promoted a more spread morphology and exhibited pronounced vinculin staining. Furthermore, composite scaffolds outperformed their neat counterparts, with melt-printed composite scaffolds significantly enhancing bone formation. This study highlights the critical role of the printing process in determining scaffold properties and performance, providing valuable insights for optimizing scaffold design in BTE.
Collapse
Affiliation(s)
- Hadis Gharacheh
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| | - Alperen Abaci
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| | - Keven Alkhoury
- Department of Mechanical & Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Ediha Choudhury
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Chya-Yan Liaw
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| | - Shawn A Chester
- Department of Mechanical & Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Murat Guvendiren
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| |
Collapse
|
40
|
Luo Y, Chen J, Gu M, Luo Y. Optimizing gelation time for cell shape control through active learning. SOFT MATTER 2025; 21:970-981. [PMID: 39807042 DOI: 10.1039/d4sm01130a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Hydrogels are popular platforms for cell encapsulation in biomedicine and tissue engineering due to their soft, porous structures, high water content, and excellent tunability. Recent studies highlight that the timing of network formation can be just as important as mechanical properties in influencing cell morphologies. Conventionally, time-dependent properties can be achieved through multi-step processes. In contrast, one-pot synthesis can improve both the efficiency and uniformity of cell encapsulation. Reaction kinetics are sensitive to temperatures and pH conditions, thus, monitoring gelation time across different conditions is essential for formulation. In this work, we choose tetra-poly(ethylene glycol) (TPEG) macromers as a model system to examine the relationship between the rate of polymer network formation and cell morphology. Previous studies of this system focused on reactions at neutral pH and room temperature, leaving much of the formulation space underexplored. We use Gaussian process regression (GPR) to minimize response surface errors by strategically selecting additional investigation points based on prior knowledge. Then we extend the knowledge from pre-trained data at neutral pH to a new surface at physiological pH. We find that the gelation time surface can effectively predict the aspect ratio of the encapsulated cells. Additionally, through focal adhesion kinase inhibition, we show that cell shape is influenced by the properties of the forming network in the initial hours as cells develop connections with the matrix. We demonstrate the utility of a high-throughput microrheology approach in enhancing fabrications of synthetic extracellular matrix and cell assemblies.
Collapse
Affiliation(s)
- Yuxin Luo
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06510, USA.
| | - Juan Chen
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06510, USA.
| | - Mengyang Gu
- Department of Statistics and Applied Probability, University of California, Santa Barbara, Santa Barbara, CA 93117, USA
| | - Yimin Luo
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06510, USA.
| |
Collapse
|
41
|
Gräf R, Batsios P, Grafe M, Meyer I, Mitic K. Nuclear Envelope Dynamics in Dictyostelium Amoebae. Cells 2025; 14:186. [PMID: 39936978 PMCID: PMC11816917 DOI: 10.3390/cells14030186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/13/2025] Open
Abstract
In the last decades, the study of many nuclear envelope components in Dictyostelium amoebae has revealed conserved mechanisms of nuclear envelope dynamics that root back unexpectedly deep into the eukaryotic tree of life. In this review, we describe the state of the art in nuclear envelope research in this organism starting from early work on nuclear pore complexes to characterization of the first true lamin in a non-metazoan organism and its associated nuclear envelope transmembrane proteins, such as the HeH-family protein Src1 and the LINC complex protein Sun1. We also describe the dynamic processes during semi-closed mitosis, including centrosome insertion into the nuclear envelope, and processes involved in the restoration of nuclear envelope permeability around mitotic exit and compare them to the situation in cells with open or fully closed mitosis.
Collapse
Affiliation(s)
- Ralph Gräf
- Department of Cell Biology, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam-Golm, Germany; (M.G.); (I.M.); (K.M.)
| | - Petros Batsios
- Sigma-Aldrich Chemie GmbH, Eschenstraße 5, 82024 Taufkirchen, Germany;
| | - Marianne Grafe
- Department of Cell Biology, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam-Golm, Germany; (M.G.); (I.M.); (K.M.)
| | - Irene Meyer
- Department of Cell Biology, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam-Golm, Germany; (M.G.); (I.M.); (K.M.)
| | - Kristina Mitic
- Department of Cell Biology, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam-Golm, Germany; (M.G.); (I.M.); (K.M.)
| |
Collapse
|
42
|
Katoh K. Integrin and Its Associated Proteins as a Mediator for Mechano-Signal Transduction. Biomolecules 2025; 15:166. [PMID: 40001469 PMCID: PMC11853369 DOI: 10.3390/biom15020166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/11/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Mechano-signal transduction is a process in which cells perceive extracellular mechanical signals, convert them into intracellular biochemical signals, and produce a response. Integrins are cell surface receptors that sense the extracellular mechanical cues and bind to the extracellular matrix (ECM). This binding induces integrin clustering and activation. Cytoplasmic tails of activated integrins interact and induce cytoskeleton tensions via several adaptor proteins. Integrins monitor extracellular stiffness via cytoskeleton tensions and modulate ECM stiffness via downstream signaling pathways regulating the expression of genes of ECM components. Integrin-mediated mechano-transduction is very crucial for the cell as it regulates the cell physiology both in normal and diseased conditions according to extracellular mechanical cues. It regulates cell proliferation, survival, and migration. Abnormal mechanical cues such as extreme and prolonged mechanical stress result in pathological conditions including fibrosis, cancers, skin, and autoimmune disorders. This paper aims to explore the role of integrins and their associated proteins in mechano-signal transduction. It highlights the integrins and their associated proteins as targets for therapy development. Furthermore, it also presents the challenges to the targeted drug development, which can be drug resistance and cytotoxicity. It is concluded in this paper that research on integrin-mediated mechano-signal transduction and its relationship with cell physiology and pathologies will be an important step towards the development of effective therapies.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology, Tsukuba 305-8521, Japan
| |
Collapse
|
43
|
Li K, Jan YN. Experimental tools and emerging principles of organellar mechanotransduction. Trends Cell Biol 2025:S0962-8924(24)00279-4. [PMID: 39828483 DOI: 10.1016/j.tcb.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
Mechanotransduction is the process by which cells detect mechanical forces and convert them into biochemical or electrical signals. This process occurs across various cellular compartments, including the plasma membrane, cytoskeleton, and intracellular organelles. While research has focused mainly on force sensing at the plasma membrane, the mechanisms and significance of intracellular mechanotransduction are just beginning to be understood. This review summarizes current techniques for studying organellar mechanobiology, and highlights advances in our understanding of the mechanosensitive events occurring in organelles such as the endoplasmic reticulum (ER), Golgi apparatus, and endolysosomes. Additionally, some open questions and promising directions are identified for future research.
Collapse
Affiliation(s)
- Kai Li
- Institute for Medical Physiology, Chinese Institutes for Medical Research, Beijing, 100069, China; School of Basic Medicine, Capital Medical University, Beijing, 100069, China
| | - Yuh Nung Jan
- Department of Physiology, University of California at San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
44
|
Wang Z, Wu J, Lv Z, Liang P, Li Q, Li Y, Guo Y. LMNA-related cardiomyopathy: From molecular pathology to cardiac gene therapy. J Adv Res 2025:S2090-1232(25)00001-3. [PMID: 39827909 DOI: 10.1016/j.jare.2025.01.001] [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: 11/19/2024] [Revised: 12/29/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND The genetic variants of LMNA cause an array of diseases that often affect the heart. LMNA-related cardiomyopathy exhibits high-penetrance and early-onset phenotypes that lead to late-stage heart failure or lethal arrhythmia. As a subtype of dilated cardiomyopathy and arrhythmogenic cardiomyopathy, LMNA-related cardiac dysfunction is resistant to existing cardiac therapeutic strategies, leaving a major unmet clinical need in cardiomyopathy management. AIM OF REVIEW Here we comprehensively summarize current knowledge about the genetic basis, disease models and pathological mechanisms of LMNA-related cardiomyopathy. Recent translational studies were highlighted to indicate new therapeutic modalities such as gene supplementation, gene silencing and genome editing therapy, which offer potential opportunities to overcome the difficulties in the development of specific drugs for this disease. KEY SCIENTIFIC CONCEPTS OF REVIEW LMNA-related cardiomyopathy involves many diverse disease mechanisms that preclude small-molecule drugs that target only a small fraction of the mechanisms. Agreeing to this notion, the first-in-human clinical trial for this disease recently reported futility. By contrast, gene therapy offers the new hope to directly intervene LMNA variants and demonstrates a tremendous potential for breakthrough therapy for this disease. Concepts in this review are also applicable to studies of other genetic diseases that lack effective therapeutics.
Collapse
Affiliation(s)
- Ze Wang
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Jiahao Wu
- Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengyuan Lv
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Ping Liang
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China.
| | - Qirui Li
- Department of Cardiology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| | - Yifei Li
- Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Yuxuan Guo
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| |
Collapse
|
45
|
Li H, Li D, Wang X, Zeng Z, Pahlavan S, Zhang W, Wang X, Wang K. Progress in Biomaterials-Enhanced Vascularization by Modulating Physical Properties. ACS Biomater Sci Eng 2025; 11:33-54. [PMID: 39615049 DOI: 10.1021/acsbiomaterials.4c01106] [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] [Indexed: 01/06/2025]
Abstract
Sufficient vascular system and adequate blood perfusion is crucial for ensuring nutrient and oxygen supply within biomaterials. Actively exploring the optimal physical properties of biomaterials in various application scenarios has provided clues for enhancing vascularization within materials, leading to improved outcomes in tissue engineering and clinical translation. Here we focus on reviewing the physical properties of biomaterials, including pore structure, surface topography, and stiffness, and their effects on promoting vascularization. This angiogenic capability has the potential to provide better standardized research models and personalized treatment strategies for bone regeneration, wound healing, islet transplantation and cardiac repair.
Collapse
Affiliation(s)
- Hao Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Dayan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xue Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Ziyuan Zeng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Wei Zhang
- TianXinFu (Beijing) Medical Appliance Co., Ltd., Beijing 102200, China
| | - Xi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| |
Collapse
|
46
|
Wang Z, Wang W, Luo Q, Song G. High matrix stiffness accelerates migration of hepatocellular carcinoma cells through the integrin β1-Plectin-F-actin axis. BMC Biol 2025; 23:8. [PMID: 39789506 PMCID: PMC11721467 DOI: 10.1186/s12915-025-02113-1] [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: 09/07/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Abundant research indicates that increased extracellular matrix (ECM) stiffness significantly enhances the malignant characteristics of hepatocellular carcinoma (HCC) cells. Plectin, an essential cytoskeletal linker protein, has recently emerged as a promoter of cancer progression, particularly in the context of cancer cell invasion and metastasis. However, the responsiveness of plectin to changes in ECM stiffness and its impact on HCC progression remain unclear. In this study, we aimed to investigate whether plectin responds to variations in ECM stiffness and to explore its involved molecular mechanisms in regulating HCC cell migration. RESULTS Our results showed that, when compared with control group (7 kPa), high ECM stiffness (53 kPa) boosts HCC cell migration by upregulating plectin and integrin β1 expression and increasing F-actin polymerization. Knockdown of integrin β1 negated the high stiffness-upregulated plectin expression. Furthermore, reducing either plectin or integrin β1 levels, or using latrunculin A, effectively prevented the high ECM stiffness-induced F-actin polymerization and HCC cell migration. CONCLUSIONS These findings demonstrate that integrin β1-plectin-F-actin axis is necessary for high matrix stiffness-driven migration of HCC cells, and provide evidence for the critical role of plectin in mechanotransduction in HCC cells.
Collapse
Affiliation(s)
- Zhihui Wang
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing, 400030, China
| | - Wenbin Wang
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing, 400030, China
| | - Qing Luo
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing, 400030, China
| | - Guanbin Song
- College of Bioengineering, Chongqing University, Chongqing, 400030, China.
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
47
|
Maeda H, Sasaki H. Blastocoel expansion and AMOT degradation cooperatively promote YAP nuclear localization during epiblast formation. Dev Biol 2025; 517:234-247. [PMID: 39486633 DOI: 10.1016/j.ydbio.2024.10.007] [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: 07/04/2024] [Revised: 09/30/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
The epiblast is a pluripotent cell population formed in the late blastocyst stage of preimplantation embryos. During the process of epiblast formation from the inner cell mass (ICM) of the early blastocyst, activation of the Hippo pathway transcription factor TEAD by the nuclear translocation of the coactivator protein YAP is required for the robust expression of pluripotency factors. However, the mechanisms that alter YAP localization during epiblast formation remain unknown. Here, we reveal two such mechanisms. Expansion of the blastocoel promotes nuclear YAP localization by increasing cytoplasmic F-actin and reducing YAP phosphorylation. Additionally, cell differentiation regulates YAP. Expression of the junctional Hippo component, AMOT, gradually decreases during epiblast formation through a tankyrase-mediated degradation. SOX2 expression in the ICM is necessary for the reduction of AMOT and YAP phosphorylation. These two mechanisms function in parallel. Thus, the blastocoel-F-actin and SOX2-AMOT axes cooperatively suppress YAP phosphorylation and promote YAP nuclear localization during epiblast formation. The cooperation of these two distinct mechanisms likely contributes to the robustness of epiblast cell differentiation.
Collapse
Affiliation(s)
- Hinako Maeda
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroshi Sasaki
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
48
|
Na J, Tai C, Wang Z, Yang Z, Chen X, Zhang J, Zheng L, Fan Y. Stiff extracellular matrix drives the differentiation of mesenchymal stem cells toward osteogenesis by the multiscale 3D genome reorganization. Biomaterials 2025; 312:122715. [PMID: 39094522 DOI: 10.1016/j.biomaterials.2024.122715] [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: 03/22/2024] [Revised: 07/17/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Abstract
Extracellular matrix (ECM) stiffness is a major driver of stem cell fate. However, the involvement of the three-dimensional (3D) genomic reorganization in response to ECM stiffness remains unclear. Here, we generated comprehensive 3D chromatin landscapes of mesenchymal stem cells (MSCs) exposed to various ECM stiffness. We found that there were more long-range chromatin interactions, but less compartment A in MSCs cultured on stiff ECM than those cultured on soft ECM. However, the switch from compartment B in MSCs cultured on soft ECM to compartment A in MSCs cultured on stiff ECM included genes encoding proteins primarily enriched in cytoskeleton organization. At the topologically associating domains (TADs) level, stiff ECM tends to have merged TADs on soft ECM. These merged TADs on stiff ECM include upregulated genes encoding proteins enriched in osteogenesis, such as SP1, ETS1, and DCHS1, which were validated by quantitative real-time polymerase chain reaction and found to be consistent with the increase of alkaline phosphatase staining. Knockdown of SP1 or ETS1 led to the downregulation of osteogenic marker genes, including COL1A1, RUNX2, ALP, and OCN in MSCs cultured on stiff ECM. Our study provides an important insight into the stiff ECM-mediated promotion of MSC differentiation towards osteogenesis, emphasizing the influence of mechanical cues on the reorganization of 3D genome architecture and stem cell fate.
Collapse
Affiliation(s)
- Jing Na
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Chengzheng Tai
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Ziyi Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Zhijie Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Xinyuan Chen
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Jing Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, 100083, China.
| | - Lisha Zheng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China.
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China.
| |
Collapse
|
49
|
Wang B, Luo Q, Medalia O. Lamins and chromatin join forces. Adv Biol Regul 2025; 95:101059. [PMID: 39547851 DOI: 10.1016/j.jbior.2024.101059] [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: 09/25/2024] [Revised: 10/28/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
The intricate interplay between lamins and chromatin underpins the structural integrity and functional organization of the eukaryotic nucleus. Lamins, type V intermediate filament proteins, form a robust meshwork beneath the inner nuclear membrane that is crucial for sustaining nuclear architecture through interactions with lamin-associated domains (LADs). LADs are predominantly heterochromatic regions in which compacted chromatin is enriched at the nuclear periphery, interacting with lamins and lamin-associated proteins. Disruptions of these interactions are implicated in a spectrum of diseases, including laminopathies, cancer, and age-related pathologies, highlighting the importance of lamin-LAD interactions. Thus, a detailed understanding of lamin-chromatin interactions may provide new insights into chromatin organization and shed light on the mechanism behind certain disease states. Here, we discuss the current state of knowledge of lamin-chromatin interactions from a biochemical and structural point of view.
Collapse
Affiliation(s)
- Baihui Wang
- Department of Biochemistry, University of Zurich, Winterthur 190, 8057, Zurich, Switzerland.
| | - Qiang Luo
- Department of Biochemistry, University of Zurich, Winterthur 190, 8057, Zurich, Switzerland.
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Winterthur 190, 8057, Zurich, Switzerland.
| |
Collapse
|
50
|
Dong R, Kang M, Qu Y, Hou T, Zhao J, Cheng X. Incorporating Hydrogel (with Low Polymeric Content) into 3D-Printed PLGA Scaffolds for Local and Sustained Release of BMP2 in Repairing Large Segmental Bone Defects. Adv Healthc Mater 2025; 14:e2403613. [PMID: 39491519 DOI: 10.1002/adhm.202403613] [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: 09/21/2024] [Indexed: 11/05/2024]
Abstract
Treating large bone defects remains a considerable challenge for clinicians: bone repair requires scaffolds with mechanical properties and bioactivities. Herein, based on crosslinking o-phthalaldehyde (OPA) with amine groups, 4-arm polyethylene glycol (4armPEG)-OPA/Gelatin hydrogel loaded with bone morphogenetic protein 2 (BMP2) is prepared and a three dimensional (3D)-printed poly (lactic-co-glycolic acid) (PLGA) porous scaffold is filled with the hydrogel solution. The composite scaffold, with a compression modulus of 0.68 ± 0.097 GPa similar to the cancellous bone, has a porosity of 56.67 ± 4.72% and a pore size of about 380 µm, promoting bone growth. The hydrogel forms a porous network at low concentrations, aiding protein release and cell migration. The hydrogel degrades in approximately three weeks, and the scaffold takes five months, matching bone repair timelines. BMP2 release experiment shows a sustained BMP2 release with a 72.4 ± 0.53% release ratio. The ALP activity test and alizarin red staining shows effective osteogenic promotion, while RT-PCR confirms BMP2@Gel enhanced COL-1 and OPN expression. Animal experiments further validate the composite scaffold's bone repair efficacy. This study demonstrates the effectiveness of the hydrogel in releasing BMP2 and the mechanical support of the 3D-printed PLGA porous scaffold, providing a new treatment for bone defects.
Collapse
Affiliation(s)
- Rongpeng Dong
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin, 130014, China
| | - Mingyang Kang
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin, 130014, China
| | - Yang Qu
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin, 130014, China
| | - Tingting Hou
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin, 130014, China
| | - Jianwu Zhao
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin, 130014, China
| | - Xueliang Cheng
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin, 130014, China
| |
Collapse
|