1
|
Li W, Xu B, Huang Y, Wang X, Yu D. Rodent models in sensorineural hearing loss research: A comprehensive review. Life Sci 2024; 358:123156. [PMID: 39442868 DOI: 10.1016/j.lfs.2024.123156] [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: 07/31/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Sensorineural hearing loss (SNHL) constitutes a major global health challenge, affecting millions of individuals and substantially impairing social integration and quality of life. The complexity of the auditory system and the multifaceted nature of SNHL necessitate advanced methodologies to understand its etiology, progression, and potential therapeutic interventions. This review provides a comprehensive overview of the current animal models used in SNHL research, focusing on their selection based on specific characteristics and their contributions to elucidating pathophysiological mechanisms and evaluating novel treatment strategies. It discusses the most commonly used rodent models in hearing research, including mice, rats, guinea pigs, Mongolian gerbils, and chinchillas. Through a comparative analysis, this review underscores the importance of selecting models that align with specific research objectives in SNHL studies, discussing the advantages and limitations of each model. By advocating for a multidisciplinary approach that leverages the strengths of various animal models with technological advancements, this review aims to facilitate significant advancements in the prevention, diagnosis, and treatment of sensorineural hearing loss.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200100, PR China
| | - Baoying Xu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Yuqi Huang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Xueling Wang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200100, PR China
| | - Dehong Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China.
| |
Collapse
|
2
|
Wu W, Zhao Z, Wang Y, Zhu G, Tan K, Liu M, Li L. Biomechanical Effects of Mechanical Stress on Cells Involved in Fracture Healing. Orthop Surg 2024; 16:811-820. [PMID: 38439564 PMCID: PMC10984830 DOI: 10.1111/os.14026] [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: 10/20/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/06/2024] Open
Abstract
Fracture healing is a complex staged repair process in which the mechanical environment plays a key role. Bone tissue is very sensitive to mechanical stress stimuli, and the literature suggests that appropriate stress can promote fracture healing by altering cellular function. However, fracture healing is a coupled process involving multiple cell types that balance and limit each other to ensure proper fracture healing. The main cells that function during different stages of fracture healing are different, and the types and molecular mechanisms of stress required are also different. Most previous studies have used a single mechanical stimulus on individual mechanosensitive cells, and there is no relatively uniform standard for the size and frequency of the mechanical stress. Analyzing the mechanisms underlying the effects of mechanical stimulation on the metabolic regulation of signaling pathways in cells such as in bone marrow mesenchymal stem cells (BMSCs), osteoblasts, chondrocytes, and osteoclasts is currently a challenging research hotspot. Grasping how stress affects the function of different cells at the molecular biology level can contribute to the refined management of fracture healing. Therefore, in this review, we summarize the relevant literature and describe the effects of mechanical stress on cells associated with fracture healing, and their possible signaling pathways, for the treatment of fractures and the further development of regenerative medicine.
Collapse
Affiliation(s)
- Weiyong Wu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihui Zhao
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Yongqing Wang
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Gengbao Zhu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Kemeng Tan
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Meiyue Liu
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Lili Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| |
Collapse
|
3
|
Kureel SK, Blair B, Sheetz MP. Recent Advancement in Elimination Strategies and Potential Rejuvenation Targets of Senescence. Adv Biol (Weinh) 2024; 8:e2300461. [PMID: 37857532 DOI: 10.1002/adbi.202300461] [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: 08/29/2023] [Indexed: 10/21/2023]
Abstract
Cellular senescence is a state of exiting the cell cycle, resisting apoptosis, and changing phenotype. Senescent cells (SCs) can be identified by large, distorted morphology and irreversible inability to replicate. In early development, senescence has beneficial roles like tissue patterning and wound healing, where SCs are cleared by the immune system. However, there is a steep rise in SC number as organisms age. The issue with SC accumulation stems from the loss of cellular function, alterations of the microenvironment, and secretions of pro-inflammatory molecules, consisting of cytokines, chemokines, matrix metalloproteinases (MMPs), interleukins, and extracellular matrix (ECM)-associated molecules. This secreted cocktail is referred to as the senescence-associated secretory phenotype (SASP), a hallmark of cellular senescence. The SASP promotes inflammation and displays a bystander effect where paracrine signaling turns proliferating cells into senescent states. To alleviate age-associated diseases, researchers have developed novel methods and techniques to selectively eliminate SCs in aged individuals. Although studies demonstrated that selectively killing SCs improves age-related disorders, there are drawbacks to SC removal. Considering favorable aspects of senescence in the body, this paper reviews recent advancements in elimination strategies and potential rejuvenation targets of senescence to bring researchers in the field up to date.
Collapse
Affiliation(s)
- Sanjay Kumar Kureel
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Brandon Blair
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Michael P Sheetz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
4
|
Alijagic A, Kotlyar O, Larsson M, Salihovic S, Hedbrant A, Eriksson U, Karlsson P, Persson A, Scherbak N, Färnlund K, Engwall M, Särndahl E. Immunotoxic, genotoxic, and endocrine disrupting impacts of polyamide microplastic particles and chemicals. ENVIRONMENT INTERNATIONAL 2024; 183:108412. [PMID: 38183898 DOI: 10.1016/j.envint.2023.108412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/06/2023] [Accepted: 12/28/2023] [Indexed: 01/08/2024]
Abstract
Due to their exceptional properties and cost effectiveness, polyamides or nylons have emerged as widely used materials, revolutionizing diverse industries, including industrial 3D printing or additive manufacturing (AM). Powder-based AM technologies employ tonnes of polyamide microplastics to produce complex components every year. However, the lack of comprehensive toxicity assessment of particulate polyamides and polyamide-associated chemicals, especially in the light of the global microplastics crisis, calls for urgent action. This study investigated the physicochemical properties of polyamide-12 microplastics used in AM, and assessed a number of toxicity endpoints focusing on inflammation, immunometabolism, genotoxicity, aryl hydrocarbon receptor (AhR) activation, endocrine disruption, and cell morphology. Specifically, microplastics examination by means of field emission scanning electron microscopy revealed that work flow reuse of material created a fraction of smaller particles with an average size of 1-5 µm, a size range readily available for uptake by human cells. Moreover, chemical analysis by means of gas chromatography high-resolution mass spectrometry detected several polyamide-associated chemicals including starting material, plasticizer, thermal stabilizer/antioxidant, and migrating slip additive. Even if polyamide particles and chemicals did not induce an acute inflammatory response, repeated and prolonged exposure of human primary macrophages disclosed a steady increase in the levels of proinflammatory chemokine Interleukin-8 (IL-8/CXCL-8). Moreover, targeted metabolomics disclosed that polyamide particles modulated the kynurenine pathway and some of its key metabolites. The p53-responsive luciferase reporter gene assay showed that particles per se were able to activate p53, being indicative of a genotoxic stress. Polyamide-associated chemicals triggered moderate activation of AhR and elicited anti-androgenic activity. Finally, a high-throughput and non-targeted morphological profiling by Cell Painting assay outlined major sites of bioactivity of polyamide-associated chemicals and indicated putative mechanisms of toxicity in the cells. These findings reveal that the increasing use of polyamide microplastics may pose a potential health risk for the exposed individuals, and it merits more attention.
Collapse
Affiliation(s)
- Andi Alijagic
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro SE-701 82, Sweden; Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro SE-701 82, Sweden; School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro SE-701 82, Sweden.
| | - Oleksandr Kotlyar
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro SE-701 82, Sweden; Centre for Applied Autonomous Sensor Systems (AASS), Mobile Robotics and Olfaction Lab (MRO), Örebro University, SE-701 82 Örebro, Sweden
| | - Maria Larsson
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro SE-701 82, Sweden
| | - Samira Salihovic
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro SE-701 82, Sweden; School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro SE-701 82, Sweden
| | - Alexander Hedbrant
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro SE-701 82, Sweden; School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro SE-701 82, Sweden
| | - Ulrika Eriksson
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro SE-701 82, Sweden
| | - Patrik Karlsson
- Department of Mechanical Engineering, Örebro University, Örebro SE-701 82, Sweden
| | - Alexander Persson
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro SE-701 82, Sweden; School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro SE-701 82, Sweden
| | - Nikolai Scherbak
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro SE-701 82, Sweden
| | | | - Magnus Engwall
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro SE-701 82, Sweden
| | - Eva Särndahl
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro SE-701 82, Sweden; School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro SE-701 82, Sweden
| |
Collapse
|
5
|
Eftekhari BS, Song D, Janmey PA. Electrical Stimulation of Human Mesenchymal Stem Cells on Conductive Substrates Promotes Neural Priming. Macromol Biosci 2023; 23:e2300149. [PMID: 37571815 PMCID: PMC10880582 DOI: 10.1002/mabi.202300149] [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: 04/06/2023] [Revised: 06/29/2023] [Indexed: 08/13/2023]
Abstract
Electrical stimulation (ES) within a conductive scaffold is potentially beneficial in encouraging the differentiation of stem cells toward a neuronal phenotype. To improve stem cell-based regenerative therapies, it is essential to use electroconductive scaffolds with appropriate stiffnesses to regulate the amount and location of ES delivery. Herein, biodegradable electroconductive substrates with different stiffnesses are fabricated from chitosan-grafted-polyaniline (CS-g-PANI) copolymers. Human mesenchymal stem cells (hMSCs) cultured on soft conductive scaffolds show a morphological change with significant filopodial elongation after electrically stimulated culture along with upregulation of neuronal markers and downregulation of glial markers. Compared to stiff conductive scaffolds and non-conductive CS scaffolds, soft conductive CS-g-PANI scaffolds promote increased expression of microtubule-associated protein 2 (MAP2) and neurofilament heavy chain (NF-H) after application of ES. At the same time, there is a decrease in the expression of the glial markers glial fibrillary acidic protein (GFAP) and vimentin after ES. Furthermore, the elevation of intracellular calcium [Ca2+ ] during spontaneous, cell-generated Ca2+ transients further suggests that electric field stimulation of hMSCs cultured on conductive substrates can promote a neural-like phenotype. The findings suggest that the combination of the soft conductive CS-g-PANI substrate and ES is a promising new tool for enhancing neuronal tissue engineering outcomes.
Collapse
Affiliation(s)
| | - Dawei Song
- Department of Physiology and Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A. Janmey
- Department of Bioengineering, University of Pennsylvania, Philadelphia, USA
- Department of Physiology and Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
6
|
Pekeč T, Venkatachalapathy S, Shim AR, Paysan D, Grzmil M, Schibli R, Béhé M, Shivashankar GV. Detecting radio- and chemoresistant cells in 3D cancer co-cultures using chromatin biomarkers. Sci Rep 2023; 13:20662. [PMID: 38001169 PMCID: PMC10673941 DOI: 10.1038/s41598-023-47287-2] [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: 07/31/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
The heterogenous treatment response of tumor cells limits the effectiveness of cancer therapy. While this heterogeneity has been linked to cell-to-cell variability within the complex tumor microenvironment, a quantitative biomarker that identifies and characterizes treatment-resistant cell populations is still missing. Herein, we use chromatin organization as a cost-efficient readout of the cells' states to identify subpopulations that exhibit distinct responses to radiotherapy. To this end, we developed a 3D co-culture model of cancer spheroids and patient-derived fibroblasts treated with radiotherapy. Using the model we identified treatment-resistant cells that bypassed DNA damage checkpoints and exhibited an aggressive growth phenotype. Importantly, these cells featured more condensed chromatin which primed them for treatment evasion, as inhibiting chromatin condensation and DNA damage repair mechanisms improved the efficacy of not only radio- but also chemotherapy. Collectively, our work shows the potential of using chromatin organization to cost-effectively study the heterogeneous treatment susceptibility of cells and guide therapeutic design.
Collapse
Affiliation(s)
- Tina Pekeč
- Laboratory for Nanoscale Biology, Paul Scherrer Institute, Villigen, Switzerland
| | | | - Anne R Shim
- Laboratory for Nanoscale Biology, Paul Scherrer Institute, Villigen, Switzerland
| | - Daniel Paysan
- Laboratory for Nanoscale Biology, Paul Scherrer Institute, Villigen, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Michal Grzmil
- Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, Villigen, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, Villigen, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Martin Béhé
- Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, Villigen, Switzerland
| | - G V Shivashankar
- Laboratory for Nanoscale Biology, Paul Scherrer Institute, Villigen, Switzerland.
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
7
|
Nakamura H, Rho E, Lee CT, Itoh K, Deng D, Watanabe S, Razavi S, Matsubayashi HT, Zhu C, Jung E, Rangamani P, Watanabe S, Inoue T. ActuAtor, a Listeria-inspired molecular tool for physical manipulation of intracellular organizations through de novo actin polymerization. Cell Rep 2023; 42:113089. [PMID: 37734382 PMCID: PMC10872831 DOI: 10.1016/j.celrep.2023.113089] [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: 06/21/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023] Open
Abstract
Form and function are often interdependent throughout biology. Inside cells, mitochondria have particularly attracted attention since both their morphology and functionality are altered under pathophysiological conditions. However, directly assessing their causal relationship has been beyond reach due to the limitations of manipulating mitochondrial morphology in a physiologically relevant manner. By engineering a bacterial actin regulator, ActA, we developed tools termed "ActuAtor" that inducibly trigger actin polymerization at arbitrary subcellular locations. The ActuAtor-mediated actin polymerization drives striking deformation and/or movement of target organelles, including mitochondria, Golgi apparatus, and nucleus. Notably, ActuAtor operation also disperses non-membrane-bound entities such as stress granules. We then implemented ActuAtor in functional assays, uncovering the physically fragmented mitochondria being slightly more susceptible to degradation, while none of the organelle functions tested are morphology dependent. The modular and genetically encoded features of ActuAtor should enable its application in studies of the form-function interplay in various intracellular contexts.
Collapse
Affiliation(s)
- Hideki Nakamura
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kyoto University Hakubi Center for Advanced Research, Yoshida-honmachi, Sakyo-ku, Kyoto 606-8501, Japan; Kyoto University Graduate School of Engineering, Department of Synthetic Chemistry and Biological Chemistry, Katsura Int'tech Center, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto 615-8530, Japan.
| | - Elmer Rho
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christopher T Lee
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Kie Itoh
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daqi Deng
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Satoshi Watanabe
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shiva Razavi
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hideaki T Matsubayashi
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Cuncheng Zhu
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Eleanor Jung
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Shigeki Watanabe
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Takanari Inoue
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
8
|
Petit C, Bonnet C, Safieddine S. Deafness: from genetic architecture to gene therapy. Nat Rev Genet 2023; 24:665-686. [PMID: 37173518 DOI: 10.1038/s41576-023-00597-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2023] [Indexed: 05/15/2023]
Abstract
Progress in deciphering the genetic architecture of human sensorineural hearing impairment (SNHI) or loss, and multidisciplinary studies of mouse models, have led to the elucidation of the molecular mechanisms underlying auditory system function, primarily in the cochlea, the mammalian hearing organ. These studies have provided unparalleled insights into the pathophysiological processes involved in SNHI, paving the way for the development of inner-ear gene therapy based on gene replacement, gene augmentation or gene editing. The application of these approaches in preclinical studies over the past decade has highlighted key translational opportunities and challenges for achieving effective, safe and sustained inner-ear gene therapy to prevent or cure monogenic forms of SNHI and associated balance disorders.
Collapse
Affiliation(s)
- Christine Petit
- Institut Pasteur, Université Paris Cité, Inserm, Institut de l'Audition, F-75012, Paris, France.
- Collège de France, F-75005, Paris, France.
| | - Crystel Bonnet
- Institut Pasteur, Université Paris Cité, Inserm, Institut de l'Audition, F-75012, Paris, France
| | - Saaïd Safieddine
- Institut Pasteur, Université Paris Cité, Inserm, Institut de l'Audition, F-75012, Paris, France
- Centre National de la Recherche Scientifique, F-75016, Paris, France
| |
Collapse
|
9
|
Zhang H, Zhu H, Feng J, Zhang Z, Zhang S, Wang Z, Sun L, Zhang W, Gao B, Zhang Y, Lin M. Reprogramming of Activated Pancreatic Stellate Cells via Mechanical Modulation of Transmembrane Force-sensitive N-cadherin Receptor. J Mol Biol 2023; 435:167819. [PMID: 36089055 DOI: 10.1016/j.jmb.2022.167819] [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: 07/16/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 02/04/2023]
Abstract
Cancer has been the leading cause of death due mainly to tumor metastasis. The tumor microenvironment plays a key role in tumor metastasis. As the main stromal cells in tumor microenvironment originated from activated fibroblast, cancer-associated fibroblasts (CAFs) play a major role in promoting tumor metastasis. A promising therapeutic avenue is reprogramming of CAFs into tumor-restraining quiescence state. In this study, we observed that CAF-like active pancreatic stellate cells (PSCs) interact with each other via N-cadherin, a force-sensitive transmembrane receptor. Since N-cadherin ligation mediated mechanotransduction has been reported to restrict integrin mediated signalling, we thus hypothesized that the reprogramming of activated PSCs by mechanical modulation of N-cadherin ligation might be possible. To test this hypothesis, we grafted N-cadherin ligand (HAVDI peptide) onto soft polyethylene glycol hydrogel substrate prior to cell adhesion to mimic cell-cell interaction via N-cadherin ligation. We found that the activated PSCs could be reprogrammed to their original quiescent state when transferred onto the substrate with immobilized HAVDI peptide. These results reveal a key role of mechanosensing by intercellular transmembrane receptor in reprogramming of activated PSCs, and provide a potential way for designing novel therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Huan Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Hongyuan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Jinteng Feng
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Zheng Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Simei Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Lin Sun
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Wencheng Zhang
- Department of Endocrinology, Second Affiliated Hospital of Air Force Military Medical University, Xi'an 710038, PR China
| | - Bin Gao
- Department of Endocrinology, Second Affiliated Hospital of Air Force Military Medical University, Xi'an 710038, PR China
| | - Ying Zhang
- Xijing 986 Hospital Department, Fourth Military Medical University, Xi'an 710054, PR China
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
10
|
Golloshi R, Playter C, Freeman TF, Das P, Raines TI, Garretson JH, Thurston D, McCord RP. Constricted migration is associated with stable 3D genome structure differences in cancer cells. EMBO Rep 2022; 23:e52149. [PMID: 35969179 PMCID: PMC9535800 DOI: 10.15252/embr.202052149] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 07/01/2022] [Accepted: 07/12/2022] [Indexed: 01/14/2023] Open
Abstract
To spread from a localized tumor, metastatic cancer cells must squeeze through constrictions that cause major nuclear deformations. Since chromosome structure affects nucleus stiffness, gene regulation, and DNA repair, here, we investigate the relationship between 3D genome structure and constricted migration in cancer cells. Using melanoma (A375) cells, we identify phenotypic differences in cells that have undergone multiple rounds of constricted migration. These cells display a stably higher migration efficiency, elongated morphology, and differences in the distribution of Lamin A/C and heterochromatin. Hi-C experiments reveal differences in chromosome spatial compartmentalization specific to cells that have passed through constrictions and related alterations in expression of genes associated with migration and metastasis. Certain features of the 3D genome structure changes, such as a loss of B compartment interaction strength, are consistently observed after constricted migration in clonal populations of A375 cells and in MDA-MB-231 breast cancer cells. Our observations suggest that consistent types of chromosome structure changes are induced or selected by passage through constrictions and that these may epigenetically encode stable differences in gene expression and cellular migration phenotype.
Collapse
Affiliation(s)
- Rosela Golloshi
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| | - Christopher Playter
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| | - Trevor F Freeman
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| | - Priyojit Das
- UT‐ORNL Graduate School of Genome Science and TechnologyUniversity of TennesseeKnoxvilleTNUSA
| | - Thomas Isaac Raines
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| | - Joshua H Garretson
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| | - Delaney Thurston
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| | - Rachel Patton McCord
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| |
Collapse
|
11
|
Das R, Sakaue T, Shivashankar GV, Prost J, Hiraiwa T. How enzymatic activity is involved in chromatin organization. eLife 2022; 11:79901. [PMID: 36472500 PMCID: PMC9810329 DOI: 10.7554/elife.79901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 12/06/2022] [Indexed: 12/12/2022] Open
Abstract
Spatial organization of chromatin plays a critical role in genome regulation. Previously, various types of affinity mediators and enzymes have been attributed to regulate spatial organization of chromatin from a thermodynamics perspective. However, at the mechanistic level, enzymes act in their unique ways and perturb the chromatin. Here, we construct a polymer physics model following the mechanistic scheme of Topoisomerase-II, an enzyme resolving topological constraints of chromatin, and investigate how it affects interphase chromatin organization. Our computer simulations demonstrate Topoisomerase-II's ability to phase separate chromatin into eu- and heterochromatic regions with a characteristic wall-like organization of the euchromatic regions. We realized that the ability of the euchromatic regions to cross each other due to enzymatic activity of Topoisomerase-II induces this phase separation. This realization is based on the physical fact that partial absence of self-avoiding interaction can induce phase separation of a system into its self-avoiding and non-self-avoiding parts, which we reveal using a mean-field argument. Furthermore, motivated from recent experimental observations, we extend our model to a bidisperse setting and show that the characteristic features of the enzymatic activity-driven phase separation survive there. The existence of these robust characteristic features, even under the non-localized action of the enzyme, highlights the critical role of enzymatic activity in chromatin organization.
Collapse
Affiliation(s)
- Rakesh Das
- Mechanobiology Institute, National University of SingaporeSingaporeSingapore
| | - Takahiro Sakaue
- Department of Physics and Mathematics, Aoyama Gakuin UniversityKanagawaJapan
| | - GV Shivashankar
- ETH ZurichZurichSwitzerland,Paul Scherrer InstituteVilligenSwitzerland
| | - Jacques Prost
- Mechanobiology Institute, National University of SingaporeSingaporeSingapore,Laboratoire Physico Chimie Curie, Institut Curie, Paris Science et Lettres Research UniversityParisFrance
| | - Tetsuya Hiraiwa
- Mechanobiology Institute, National University of SingaporeSingaporeSingapore
| |
Collapse
|
12
|
Gao Q, Kim BS, Gao G. Advanced Strategies for 3D Bioprinting of Tissue and Organ Analogs Using Alginate Hydrogel Bioinks. Mar Drugs 2021; 19:708. [PMID: 34940707 PMCID: PMC8708555 DOI: 10.3390/md19120708] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/12/2021] [Accepted: 12/12/2021] [Indexed: 12/15/2022] Open
Abstract
Alginate is a natural polysaccharide that typically originates from various species of algae. Due to its low cost, good biocompatibility, and rapid ionic gelation, the alginate hydrogel has become a good option of bioink source for 3D bioprinting. However, the lack of cell adhesive moieties, erratic biodegradability, and poor printability are the critical limitations of alginate hydrogel bioink. This review discusses the pivotal properties of alginate hydrogel as a bioink for 3D bioprinting technologies. Afterward, a variety of advanced material formulations and biofabrication strategies that have recently been developed to overcome the drawbacks of alginate hydrogel bioink will be focused on. In addition, the applications of these advanced solutions for 3D bioprinting of tissue/organ mimicries such as regenerative implants and in vitro tissue models using alginate-based bioink will be systematically summarized.
Collapse
Affiliation(s)
- Qiqi Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, No. 5, South Street, Zhongguancun, Haidian District, Beijing 100081, China;
| | - Byoung-Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 626841, Kyungnam, Korea;
| | - Ge Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, No. 5, South Street, Zhongguancun, Haidian District, Beijing 100081, China;
- Department of Medical Technology, Beijing Institute of Technology, No. 5, South Street, Zhongguancun, Haidian District, Beijing 100081, China
| |
Collapse
|
13
|
Nader GPDF, Williart A, Piel M. Nuclear deformations, from signaling to perturbation and damage. Curr Opin Cell Biol 2021; 72:137-145. [PMID: 34461580 DOI: 10.1016/j.ceb.2021.07.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/13/2021] [Accepted: 07/28/2021] [Indexed: 01/07/2023]
Abstract
During cell growth and motility in crowded tissues or interstitial spaces, cells must integrate multiple physical and biochemical environmental inputs. After a number of recent studies, the view of the nucleus as a passive object that cells have to drag along has become obsolete, placing the nucleus as a central player in sensing some of these inputs. In the present review, we will focus on changes in nuclear shape caused by external and internal forces. Depending on their magnitude, nuclear deformations can generate signaling events that modulate cell behavior and fate, or be a source of perturbations or even damage, having detrimental effects on cellular functions. On very large deformations, nuclear envelope rupture events become frequent, leading to uncontrolled nucleocytoplasmic mixing and DNA damage. We will also discuss the consequences of repeated compromised nuclear integrity, which can trigger DNA surveillance mechanisms, with critical consequences to cell fate and tissue homeostasis.
Collapse
Affiliation(s)
| | - Alice Williart
- Institut Curie and Institut Pierre Gilles de Gennes, PSL Research University, CNRS, UMR 144, Paris, France
| | - Matthieu Piel
- Institut Curie and Institut Pierre Gilles de Gennes, PSL Research University, CNRS, UMR 144, Paris, France.
| |
Collapse
|
14
|
Lang F, Contreras-Gerenas MF, Gelléri M, Neumann J, Kröger O, Sadlo F, Berniak K, Marx A, Cremer C, Wagenknecht HA, Allgayer H. Tackling Tumour Cell Heterogeneity at the Super-Resolution Level in Human Colorectal Cancer Tissue. Cancers (Basel) 2021; 13:cancers13153692. [PMID: 34359592 PMCID: PMC8345115 DOI: 10.3390/cancers13153692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Tumour cell heterogeneity is the most fundamental problem in cancer diagnosis and therapy. Micro-diagnostic technologies able to differentiate the heterogeneous molecular, especially metastatic, potential of single cells or cell clones already within early primary tumours of carcinoma patients would be of utmost importance. Single molecule localisation microscopy (SMLM) has recently allowed the imaging of subcellular features at the nanoscale. However, the technology has mostly been limited to cultured cell lines only. We introduce a first-in-field approach for quantitative SMLM-analysis of chromatin nanostructure in individual cells in resected, routine-pathology colorectal carcinoma patient tissue sections, illustrating, as a first example, changes in nuclear chromatin nanostructure and microRNA intracellular distribution within carcinoma cells as opposed to normal cells, chromatin accessibility and microRNAs having been shown to be critical in gene regulation and metastasis. We believe this technology to have an enormous potential for future differential diagnosis between individual cells in the tissue context. Abstract Tumour cell heterogeneity, and its early individual diagnosis, is one of the most fundamental problems in cancer diagnosis and therapy. Single molecule localisation microscopy (SMLM) resolves subcellular features but has been limited to cultured cell lines only. Since nuclear chromatin architecture and microRNAs are critical in metastasis, we introduce a first-in-field approach for quantitative SMLM-analysis of chromatin nanostructure in individual cells in resected, routine-pathology colorectal carcinoma (CRC) patient tissue sections. Chromatin density profiles proved to differ for cells in normal and carcinoma colorectal tissues. In tumour sections, nuclear size and chromatin compaction percentages were significantly different in carcinoma versus normal epithelial and other cells of colorectal tissue. SMLM analysis in nuclei from normal colorectal tissue revealed abrupt changes in chromatin density profiles at the nanoscale, features not detected by conventional widefield microscopy. SMLM for microRNAs relevant for metastasis was achieved in colorectal cancer tissue at the nuclear level. Super-resolution microscopy with quantitative image evaluation algorithms provide powerful tools to analyse chromatin nanostructure and microRNAs of individual cells from normal and tumour tissue at the nanoscale. Our new perspectives improve the differential diagnosis of normal and (metastatically relevant) tumour cells at the single-cell level within the heterogeneity of primary tumours of patients.
Collapse
Affiliation(s)
- Fabian Lang
- Institute of Organic Chemistry, Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, Campus Süd, 76131 Karlsruhe, Germany; (F.L.); (H.-A.W.)
| | - María F. Contreras-Gerenas
- Department of Experimental Surgery—Cancer Metastasis, Mannheim Medical Faculty, Ruprecht-Karls University of Heidelberg, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany;
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany; (M.G.); (J.N.); (C.C.)
| | - Márton Gelléri
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany; (M.G.); (J.N.); (C.C.)
| | - Jan Neumann
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany; (M.G.); (J.N.); (C.C.)
| | - Ole Kröger
- Interdisciplinary Centre for Scientific Computing (IWR), University Heidelberg, Mathematikon B, Im Neuenheimer Feld 205, 69120 Heidelberg, Germany; (O.K.); (F.S.)
| | - Filip Sadlo
- Interdisciplinary Centre for Scientific Computing (IWR), University Heidelberg, Mathematikon B, Im Neuenheimer Feld 205, 69120 Heidelberg, Germany; (O.K.); (F.S.)
| | - Krzysztof Berniak
- Department of Cell Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7 Street, 30-387 Krakow, Poland;
| | - Alexander Marx
- Institute of Pathology, Mannheim Medical Faculty, Ruprecht-Karls University of Heidelberg, Theodor-Kutzer-Ufer 1, 68167 Mannheim, Germany;
| | - Christoph Cremer
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany; (M.G.); (J.N.); (C.C.)
- Interdisciplinary Centre for Scientific Computing (IWR), University Heidelberg, Mathematikon B, Im Neuenheimer Feld 205, 69120 Heidelberg, Germany; (O.K.); (F.S.)
- Institute of Pharmacy & Molecular Biotechnology, Ruprecht-Karls University of Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Hans-Achim Wagenknecht
- Institute of Organic Chemistry, Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, Campus Süd, 76131 Karlsruhe, Germany; (F.L.); (H.-A.W.)
| | - Heike Allgayer
- Department of Experimental Surgery—Cancer Metastasis, Mannheim Medical Faculty, Ruprecht-Karls University of Heidelberg, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany;
- Correspondence: ; Tel.: +49-(0)621-383-71630/-1406/-71635; Fax: +49-(0)621-383-71631
| |
Collapse
|
15
|
Li Y, Tang W, Guo M. The Cell as Matter: Connecting Molecular Biology to Cellular Functions. MATTER 2021; 4:1863-1891. [PMID: 35495565 PMCID: PMC9053450 DOI: 10.1016/j.matt.2021.03.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Viewing cell as matter to understand the intracellular biomolecular processes and multicellular tissue behavior represents an emerging research area at the interface of physics and biology. Cellular material displays various physical and mechanical properties, which can strongly affect both intracellular and multicellular biological events. This review provides a summary of how cells, as matter, connect molecular biology to cellular and multicellular scale functions. As an impact in molecular biology, we review recent progresses in utilizing cellular material properties to direct cell fate decisions in the communities of immune cells, neurons, stem cells, and cancer cells. Finally, we provide an outlook on how to integrate cellular material properties in developing biophysical methods for engineered living systems, regenerative medicine, and disease treatments.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wenhui Tang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
16
|
Li L, Wang H, Xiong C, Luo D, Chen H, Liu Y. Quantify the combined effects of temperature and force on the stability of DNA hairpin. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2021; 33:185102. [PMID: 33711825 DOI: 10.1088/1361-648x/abee38] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/12/2021] [Indexed: 06/12/2023]
Abstract
OxDNA, as a successful coarse-grain model, has been applied to reproduce the thermodynamic and mechanical properties of both single- and double-stranded DNA. In current simulation, oxDNA is extended to explore the combined effects of temperature and force on the stability of DNA hairpin and its free energy landscape. Simulations were carried out at different forces and temperatures, at each temperature, a 18-base-pair DNA hairpin dynamically transited between folded state and unfolded state, and the separation between two states is consistent with the full contour length of single-stranded DNA in the unfolded state. Two methods were used to identify the critical force of DNA hairpin at each temperature and the critical forces obtained from two methods were consistent with each other and gradually decreased with the increasing temperature from 300 K to 326 K. The critical force at 300 K is reasonably consistent with the single molecule result of DNA hairpin with the same stem length. The two-state free energy landscape can be elucidated from the probability distribution of DNA hairpin extension and its dependence on the force and temperature is totally different. The increasing temperature not only reduces the free energy barrier, but also alters the position of transition point along the extension coordinate, resulting in the reduction of folding distance and the extension of unfolding distance, but their sum is not obviously dependent on the temperature. Generally, an assumption that the location of transition state in two-state energy landscape is independent of the stretching force is used to analyze the data of the single molecule experiment, but current simulation results indicate that effects of stretching forces on the location of transition state in two-state energy landscape are dependent on temperature. At relatively high temperature, stretching force can also change the location of transition state in the free energy landscape.
Collapse
Affiliation(s)
- Lin Li
- College of Physics, Guizhou University, Guiyang 550025, People's Republic of China
| | - Hongchang Wang
- School of Physics and Electronic Science, Guizhou Normal University, Guiyang 550025, People's Republic of China
| | - Caiyun Xiong
- College of Physics, Guizhou University, Guiyang 550025, People's Republic of China
| | - Di Luo
- College of Physics, Guizhou University, Guiyang 550025, People's Republic of China
| | - Hu Chen
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Department of Physics, Xiamen University, Xiamen, People's Republic of China
| | - Yanhui Liu
- College of Physics, Guizhou University, Guiyang 550025, People's Republic of China
| |
Collapse
|
17
|
Trinh DC, Alonso-Serra J, Asaoka M, Colin L, Cortes M, Malivert A, Takatani S, Zhao F, Traas J, Trehin C, Hamant O. How Mechanical Forces Shape Plant Organs. Curr Biol 2021; 31:R143-R159. [PMID: 33561417 DOI: 10.1016/j.cub.2020.12.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Plants produce organs of various shapes and sizes. While much has been learned about genetic regulation of organogenesis, the integration of mechanics in the process is also gaining attention. Here, we consider the role of forces as instructive signals in organ morphogenesis. Turgor pressure is the primary cause of mechanical signals in developing organs. Because plant cells are glued to each other, mechanical signals act, in essence, at multiple scales, through cell wall contiguity and water flux. In turn, cells use such signals to resist mechanical stress, for instance, by reinforcing their cell walls. We show that the three elemental shapes behind plant organs - spheres, cylinders and lamina - can be actively maintained by such a mechanical feedback. Combinations of this 3-letter alphabet can generate more complex shapes. Furthermore, mechanical conflicts emerge at the boundary between domains exhibiting different growth rates or directions. These secondary mechanical signals contribute to three other organ shape features - folds, shape reproducibility and growth arrest. The further integration of mechanical signals with the molecular network offers many fruitful prospects for the scientific community, including the role of proprioception in organ shape robustness or the definition of cell and organ identities as a result of an interplay between biochemical and mechanical signals.
Collapse
Affiliation(s)
- Duy-Chi Trinh
- Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCBL, INRAE, CNRS, 46 Allée d'Italie, 69364 Lyon Cedex 07, France; Department of Pharmacological, Medical and Agronomical Biotechnology, University of Science and Technology of Hanoi, Cau Giay District, Hanoi, Vietnam
| | - Juan Alonso-Serra
- Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCBL, INRAE, CNRS, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Mariko Asaoka
- Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCBL, INRAE, CNRS, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Leia Colin
- Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCBL, INRAE, CNRS, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Matthieu Cortes
- Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCBL, INRAE, CNRS, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Alice Malivert
- Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCBL, INRAE, CNRS, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Shogo Takatani
- Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCBL, INRAE, CNRS, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Feng Zhao
- Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCBL, INRAE, CNRS, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Jan Traas
- Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCBL, INRAE, CNRS, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Christophe Trehin
- Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCBL, INRAE, CNRS, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Olivier Hamant
- Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCBL, INRAE, CNRS, 46 Allée d'Italie, 69364 Lyon Cedex 07, France.
| |
Collapse
|
18
|
Goswami R, Asnacios A, Hamant O, Chabouté ME. Is the plant nucleus a mechanical rheostat? CURRENT OPINION IN PLANT BIOLOGY 2020; 57:155-163. [PMID: 33128898 DOI: 10.1016/j.pbi.2020.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 08/29/2020] [Accepted: 09/11/2020] [Indexed: 06/11/2023]
Abstract
Beyond its biochemical nature, the nucleus is also a physical object. There is accumulating evidence that its mechanics plays a key role in gene expression, cytoskeleton organization, and more generally in cell and developmental biology. Building on data mainly obtained from the animal literature, we show how nuclear mechanics may orchestrate development and gene expression. In other words, the nucleus may play the additional role of a mechanical rheostat. Although data from plant systems are still scarce, we pinpoint recent advances and highlight some differences with animal systems. Building on this survey, we propose a list of prospects for future research in plant nuclear mechanotransduction and development.
Collapse
Affiliation(s)
- Rituparna Goswami
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, 67084 Strasbourg, France; Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, UCB Lyon 1, ENS de Lyon, INRA, CNRS, 69364 Lyon, France
| | - Atef Asnacios
- Laboratoire Matières et Systèmes Complexes, Université de Paris, CNRS, Université Paris-Diderot, 75013 Paris, France
| | - Olivier Hamant
- Laboratoire de Reproduction et Développement des Plantes, Université de Lyon, UCB Lyon 1, ENS de Lyon, INRA, CNRS, 69364 Lyon, France.
| | - Marie-Edith Chabouté
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, 67084 Strasbourg, France.
| |
Collapse
|
19
|
Effects of extracellular matrix viscoelasticity on cellular behaviour. Nature 2020; 584:535-546. [PMID: 32848221 DOI: 10.1038/s41586-020-2612-2] [Citation(s) in RCA: 1043] [Impact Index Per Article: 208.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/17/2020] [Indexed: 11/08/2022]
Abstract
Substantial research over the past two decades has established that extracellular matrix (ECM) elasticity, or stiffness, affects fundamental cellular processes, including spreading, growth, proliferation, migration, differentiation and organoid formation. Linearly elastic polyacrylamide hydrogels and polydimethylsiloxane (PDMS) elastomers coated with ECM proteins are widely used to assess the role of stiffness, and results from such experiments are often assumed to reproduce the effect of the mechanical environment experienced by cells in vivo. However, tissues and ECMs are not linearly elastic materials-they exhibit far more complex mechanical behaviours, including viscoelasticity (a time-dependent response to loading or deformation), as well as mechanical plasticity and nonlinear elasticity. Here we review the complex mechanical behaviours of tissues and ECMs, discuss the effect of ECM viscoelasticity on cells, and describe the potential use of viscoelastic biomaterials in regenerative medicine. Recent work has revealed that matrix viscoelasticity regulates these same fundamental cell processes, and can promote behaviours that are not observed with elastic hydrogels in both two- and three-dimensional culture microenvironments. These findings have provided insights into cell-matrix interactions and how these interactions differentially modulate mechano-sensitive molecular pathways in cells. Moreover, these results suggest design guidelines for the next generation of biomaterials, with the goal of matching tissue and ECM mechanics for in vitro tissue models and applications in regenerative medicine.
Collapse
|
20
|
Domingues C, Geraldo AM, Anjo SI, Matos A, Almeida C, Caramelo I, Lopes-da-Silva JA, Paiva A, Carvalho J, Pires das Neves R, Manadas B, Grãos M. Cofilin-1 Is a Mechanosensitive Regulator of Transcription. Front Cell Dev Biol 2020; 8:678. [PMID: 32903827 PMCID: PMC7438942 DOI: 10.3389/fcell.2020.00678] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
The mechanical properties of the extracellular environment are interrogated by cells and integrated through mechanotransduction. Many cellular processes depend on actomyosin-dependent contractility, which is influenced by the microenvironment’s stiffness. Here, we explored the influence of substrate stiffness on the proteome of proliferating undifferentiated human umbilical cord-matrix mesenchymal stem/stromal cells. The relative abundance of several proteins changed significantly by expanding cells on soft (∼3 kPa) or stiff substrates (GPa). Many such proteins are associated with the regulation of the actin cytoskeleton, a major player of mechanotransduction and cell physiology in response to mechanical cues. Specifically, Cofilin-1 levels were elevated in cells cultured on soft comparing with stiff substrates. Furthermore, Cofilin-1 was de-phosphorylated (active) and present in the nuclei of cells kept on soft substrates, in contrast with phosphorylated (inactive) and widespread distribution in cells on stiff. Soft substrates promoted Cofilin-1-dependent increased RNA transcription and faster RNA polymerase II-mediated transcription elongation. Cofilin-1 is part of a novel mechanism linking mechanotransduction and transcription.
Collapse
Affiliation(s)
- Catarina Domingues
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - A Margarida Geraldo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Sandra Isabel Anjo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - André Matos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Polytechnic Institute of Coimbra, Coimbra College of Agriculture, Coimbra, Portugal
| | - Cláudio Almeida
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Polytechnic Institute of Coimbra, Coimbra College of Agriculture, Coimbra, Portugal
| | - Inês Caramelo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | | | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, Coimbra, Portugal
| | - João Carvalho
- Centro de Física da Universidade de Coimbra (CFisUC), Department of Physics, University of Coimbra, Coimbra, Portugal
| | - Ricardo Pires das Neves
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - Bruno Manadas
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Mário Grãos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal.,Biocant, Technology Transfer Association, Cantanhede, Portugal
| |
Collapse
|
21
|
Abstract
The mechanical reprogramming of fibroblasts, followed by their redifferentiation into rejuvenated fibroblasts in an optimized 3D collagen matrix, made these cells more contractile and more efficient at synthesizing matrix components including laminin, fibronectin, and collagen-IV. Moreover, the rejuvenated fibroblasts obtained through this approach exhibited a decrease in DNA damage. The rejuvenated fibroblasts derived from this method precisely align into tissue architectures, suggesting its potential application as clinical implants in tissue engineering and regenerative medicine. Over the course of the aging process, fibroblasts lose contractility, leading to reduced connective-tissue stiffness. A promising therapeutic avenue for functional rejuvenation of connective tissue is reprogrammed fibroblast replacement, although major hurdles still remain. Toward this, we recently demonstrated that the laterally confined growth of fibroblasts on micropatterned substrates induces stem-cell-like spheroids. In this study, we embedded these partially reprogrammed spheroids in collagen-I matrices of varying densities, mimicking different three-dimensional (3D) tissue constraints. In response to such matrix constraints, these spheroids regained their fibroblastic properties and sprouted to form 3D connective-tissue networks. Interestingly, we found that these differentiated fibroblasts exhibit reduced DNA damage, enhanced cytoskeletal gene expression, and actomyosin contractility. In addition, the rejuvenated fibroblasts show increased matrix protein (fibronectin and laminin) deposition and collagen remodeling compared to the parental fibroblast tissue network. Furthermore, we show that the partially reprogrammed cells have comparatively open chromatin compaction states and may be more poised to redifferentiate into contractile fibroblasts in 3D-collagen matrix. Collectively, our results highlight efficient fibroblast rejuvenation through laterally confined reprogramming, which has important implications in regenerative medicine.
Collapse
|
22
|
Chromatin Viscoelasticity Measured by Local Dynamic Analysis. Biophys J 2020; 118:2258-2267. [PMID: 32320676 PMCID: PMC7203068 DOI: 10.1016/j.bpj.2020.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 12/25/2022] Open
Abstract
The nucleus in eukaryotic cells is a crowded environment that consists of genetic code along the DNA, together with a condensed solution of proteins, RNA, and other molecules. It is subjected to highly dynamic processes, including cell division, transcription, and DNA repair. In addition, the genome in the nucleus is subjected to external forces applied by the cytoplasmic skeleton and neighboring cells, as well as to internal nuclear forces. These forces oppose the need to maintain the genome order, which may be compensated by the internal nuclear viscoelastic properties that can restrain these forces. The structural and mechanical properties of chromatin inside the nucleus are still not fully clear; however, their importance for the proper functioning of the cells is unquestionable. Different approaches have been developed for this aim, ranging from directly measuring the dynamic and elastic properties of chromatin to studying the interactions of chromatin with the surrounding envelope and nuclear bodies. Although the elasticity of naked DNA in vitro is well characterized, the elasticity of chromatin in live cells is more complex and is still not fully understood. Here, we studied the elastic properties of chromatin by dynamic measurements in live cells, followed by viscoelastic modeling. We measured the trajectories of single chromatin loci, centromeres, and telomeres in live cells and analyzed their dynamics using the Langevin formalism. We assumed that the overall effect of the chromatin network forces can be modeled for each locus by a local harmonic potential and calculated the effective force constant. In addition, we assumed that this harmonic force results from the chromatin network formed by the internal polymer structure together with cross-links formed by the protein complex. We show that lamin A has the greatest effect on chromatin viscoelasticity and that its removal leads to a significant reduction in the local harmonic force.
Collapse
|
23
|
Percipalle P, Vartiainen M. Cytoskeletal proteins in the cell nucleus: a special nuclear actin perspective. Mol Biol Cell 2020; 30:1781-1785. [PMID: 31306096 PMCID: PMC6727747 DOI: 10.1091/mbc.e18-10-0645] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The emerging role of cytoskeletal proteins in the cell nucleus has become a new frontier in cell biology. Actin and actin-binding proteins regulate chromatin and gene expression, but importantly they are beginning to be essential players in genome organization. These actin-based functions contribute to genome stability and integrity while affecting DNA replication and global transcription patterns. This is likely to occur through interactions of actin with nuclear components including nuclear lamina and subnuclear organelles. An exciting future challenge is to understand how these actin-based genome-wide mechanisms may regulate development and differentiation by interfering with the mechanical properties of the cell nucleus and how regulated actin polymerization plays a role in maintaining nuclear architecture. With a special focus on actin, here we summarize how cytoskeletal proteins operate in the nucleus and how they may be important to consolidate nuclear architecture for sustained gene expression or silencing.
Collapse
Affiliation(s)
- Piergiorgio Percipalle
- Biology Program, Science Division, New York University Abu Dhabi, 12988 Abu Dhabi, United Arab Emirates.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Maria Vartiainen
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
24
|
Targeting cell plasticity for regeneration: From in vitro to in vivo reprogramming. Adv Drug Deliv Rev 2020; 161-162:124-144. [PMID: 32822682 DOI: 10.1016/j.addr.2020.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022]
Abstract
The discovery of induced pluripotent stem cells (iPSCs), reprogrammed to pluripotency from somatic cells, has transformed the landscape of regenerative medicine, disease modelling and drug discovery pipelines. Since the first generation of iPSCs in 2006, there has been enormous effort to develop new methods that increase reprogramming efficiency, and obviate the need for viral vectors. In parallel to this, the promise of in vivo reprogramming to convert cells into a desired cell type to repair damage in the body, constitutes a new paradigm in approaches for tissue regeneration. This review article explores the current state of reprogramming techniques for iPSC generation with a specific focus on alternative methods that use biophysical and biochemical stimuli to reduce or eliminate exogenous factors, thereby overcoming the epigenetic barrier towards vector-free approaches with improved clinical viability. We then focus on application of iPSC for therapeutic approaches, by giving an overview of ongoing clinical trials using iPSCs for a variety of health conditions and discuss future scope for using materials and reagents to reprogram cells in the body.
Collapse
|
25
|
Zhao X, Guo S, Lu C, Chen J, Le S, Fu H, Yan J. Single-molecule manipulation quantification of site-specific DNA binding. Curr Opin Chem Biol 2019; 53:106-117. [DOI: 10.1016/j.cbpa.2019.08.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/24/2019] [Accepted: 08/24/2019] [Indexed: 10/25/2022]
|
26
|
d'Angelo M, Benedetti E, Tupone MG, Catanesi M, Castelli V, Antonosante A, Cimini A. The Role of Stiffness in Cell Reprogramming: A Potential Role for Biomaterials in Inducing Tissue Regeneration. Cells 2019; 8:E1036. [PMID: 31491966 PMCID: PMC6770247 DOI: 10.3390/cells8091036] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/30/2019] [Accepted: 09/04/2019] [Indexed: 01/12/2023] Open
Abstract
The mechanotransduction is the process by which cells sense mechanical stimuli such as elasticity, viscosity, and nanotopography of extracellular matrix and translate them into biochemical signals. The mechanotransduction regulates several aspects of the cell behavior, including migration, proliferation, and differentiation in a time-dependent manner. Several reports have indicated that cell behavior and fate are not transmitted by a single signal, but rather by an intricate network of many signals operating on different length and timescales that determine cell fate. Since cell biology and biomaterial technology are fundamentals in cell-based regenerative therapies, comprehending the interaction between cells and biomaterials may allow the design of new biomaterials for clinical therapeutic applications in tissue regeneration. In this work, we present the most relevant mechanism by which the biomechanical properties of extracellular matrix (ECM) influence cell reprogramming, with particular attention on the new technologies and materials engineering, in which are taken into account not only the biochemical and biophysical signals patterns but also the factor time.
Collapse
Affiliation(s)
- Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Maria Grazia Tupone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Mariano Catanesi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
| |
Collapse
|
27
|
Mohammed D, Versaevel M, Bruyère C, Alaimo L, Luciano M, Vercruysse E, Procès A, Gabriele S. Innovative Tools for Mechanobiology: Unraveling Outside-In and Inside-Out Mechanotransduction. Front Bioeng Biotechnol 2019; 7:162. [PMID: 31380357 PMCID: PMC6646473 DOI: 10.3389/fbioe.2019.00162] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/20/2019] [Indexed: 12/26/2022] Open
Abstract
Cells and tissues can sense and react to the modifications of the physico-chemical properties of the extracellular environment (ECM) through integrin-based adhesion sites and adapt their physiological response in a process called mechanotransduction. Due to their critical localization at the cell-ECM interface, transmembrane integrins are mediators of bidirectional signaling, playing a key role in “outside-in” and “inside-out” signal transduction. After presenting the basic conceptual fundamentals related to cell mechanobiology, we review the current state-of-the-art technologies that facilitate the understanding of mechanotransduction signaling pathways. Finally, we highlight innovative technological developments that can help to advance our understanding of the mechanisms underlying nuclear mechanotransduction.
Collapse
Affiliation(s)
- Danahe Mohammed
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Marie Versaevel
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Céline Bruyère
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Laura Alaimo
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Marine Luciano
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Eléonore Vercruysse
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Anthony Procès
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium.,Department of Neurosciences, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Sylvain Gabriele
- Mechanobiology and Soft Matter Group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| |
Collapse
|