1
|
Zhang R, Sougawa N, Mao D, Inoue H, Goda S. Signaling pathways of pro-IL-1β production induced by mechanical stress in gingival epithelial cells. J Oral Biosci 2025:100626. [PMID: 39921162 DOI: 10.1016/j.job.2025.100626] [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: 11/13/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
OBJECTIVES Mechanical stress on the teeth and alveolar bone caused by bruxism, orthodontics, and implants affects the periodontal tissues, causing gingival recession and alveolar bone resorption, and entire body, including the heart and vascular system. Although the same forces exerted on the alveolar bone and teeth are exerted on gingival epithelial cells, little is known about the effects of mechanical stress on these cells. This study investigated the effects of mechanical stress on gingival epithelial cells. METHODS Ca9-22 cells (human gingival epithelial cells) were used. They were seeded onto the silicone and stretched cyclically. Mechanical stress-stimulated Ca9-22 cells were evaluated for pro-inflammatory interleukin (pro-IL)-1β production using Western blotting and analyzed to assess the phosphorylation level of intracellular signaling molecules. RESULTS Mechanical stress induced pro-IL-1β upregulation in Ca9-22 cells, which was significantly inhibited by ruthenium red. Ruthenium red significantly inhibited mechanical stress-induced phosphorylation of focal adhesion kinase (FAK), P130Cas, and extracellular signal-regulated kinase 1 and 2 (ERK1/2) induced by mechanical stress. Additionally, Y15 significantly inhibited the upregulation of pro-IL-1β expression and phosphorylation of FAK, P130Cas, and ERK1/2 stimulated by mechanical stress. CONCLUSIONS In Ca9-22 cells, mechanical stress may increase pro-IL-1β production via mechanosensitive ion channels and FAK. These findings revealed the mechanisms of inflammation in mechanically-stressed Ca9-22 cells and may aid in the development of therapeutic approaches to prevent bone resorption.
Collapse
Affiliation(s)
- Ruixuan Zhang
- Graduate School of Dentistry, Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Nagako Sougawa
- Department of Physiology, Osaka Dental University, Osaka, Japan.
| | - Dan Mao
- Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Hiroshi Inoue
- Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Seiji Goda
- Department of Physiology, Osaka Dental University, Osaka, Japan
| |
Collapse
|
2
|
Lei M, Wang W, Zhang H, Gong J, Cai H, Wang Z, Zhu L, Yang X, Wang S, Ma C. Piezo1 Regulates Stiffness-Dependent DRG Axon Regeneration via Modifying Cytoskeletal Dynamics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405705. [PMID: 39514408 DOI: 10.1002/advs.202405705] [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: 05/24/2024] [Revised: 08/23/2024] [Indexed: 11/16/2024]
Abstract
Despite medical interventions, the regenerative capacity of the peripheral nervous system is limited. Dorsal root ganglion (DRG) neurons possess the capacity to detect mechanical signals from their microenvironment, but the impact and mechanism by which these signals regulate axon regrowth and even regeneration in DRG neurons remain unclear. In this study, DRG neurons from newborn rats are cultured on substrates with varying degrees of stiffness in vitro to investigate the role of mechanical signals in axon regrowth. The findings reveal that substrate stiffness plays a crucial role in regulating axon regrowth, with an optimal stiffness required for this process. In addition, the data demonstrate that Piezo1, a mechanosensitive cation channel, detects substrate stiffness at the growth cone and regulates axon regrowth through activating downstream Ca2+-CaMKII-FAK-actin cascade signaling pathway. Interestingly, knocking down Piezo1 in adult rat DRG neurons leads to enhanced axon regeneration and accelerated recovery of sensory function after sciatic nerve injury. Overall, these findings contribute to the understanding of the role of mechanical signals in axon regeneration and highlight microenvironmental stiffness as a promising therapeutic target for repairing nerve injuries.
Collapse
Affiliation(s)
- Mengshi Lei
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Weiyou Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Hong Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jihong Gong
- Key Laboratory of Cognitive Science, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central Minzu University, Wuhan, 430079, China
| | - Hanmian Cai
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhili Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Le Zhu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaofei Yang
- Key Laboratory of Cognitive Science, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central Minzu University, Wuhan, 430079, China
| | - Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hubei Key Laboratory of Brain-inspired Intelligent Systems, Wuhan, 430074, China
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hubei Key Laboratory of Brain-inspired Intelligent Systems, Wuhan, 430074, China
| |
Collapse
|
3
|
Luu N, Liao J, Fang Y, Chen W. Advances in ligand-based surface engineering strategies for fine-tuning T cell mechanotransduction toward efficient immunotherapy. Biophys J 2024:S0006-3495(24)02240-9. [PMID: 39600091 DOI: 10.1016/j.bpj.2024.11.1512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/16/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
T cell-based immunotherapy has recently emerged as a promising strategy to treat cancer, requiring the activation of antigen-directed cytotoxicity to eliminate cancer cells. Mechanical signaling, although often overshadowed by its biochemical counterpart, plays a crucial role in T cell anticancer responses, from activation to cytolytic killing. Rapid advancements in the fields of chemistry, biomaterials, and micro/nanoengineering offer an interdisciplinary approach to incorporating mechano- and immunomodulatory ligands, including but not limited to synthetic peptides, small molecules, cytokines, and artificial antigens, onto the biomaterial-based platforms to modulate mechanotransducive processes in T cells. The surface engineering of these immunomodulatory ligands with optimization of ligand density, geometrical arrangement, and mobility has been proven to better mimic the natural ligation between immunoreceptors and ligands to directly enhance or inhibit mechanotransduction pathways in T cells, through triggering upstream mechanosensitive channels, adhesion molecules, cytoskeletal components, or downstream mechanoimmunological regulators. Despite its tremendous potential, current research on this new biomaterial surface engineering approach for mechanomodulatory T cell activation and effector functions remains in a nascent stage. This review highlights the recent progress in this new direction, focusing on achievements in mechanomodulatory ligand-based surface engineering strategies and underlying principles, and outlooks the further research in the rapidly evolving field of T cell mechanotransduction engineering for efficient immunotherapy.
Collapse
Affiliation(s)
- Ngoc Luu
- Department of Biomedical Engineering, New York University, Brooklyn, New York
| | - Junru Liao
- Department of Biomedical Engineering, New York University, Brooklyn, New York
| | - Yifei Fang
- Department of Biomedical Engineering, New York University, Brooklyn, New York
| | - Weiqiang Chen
- Department of Biomedical Engineering, New York University, Brooklyn, New York; Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, New York; Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York.
| |
Collapse
|
4
|
Lan Y, Lu J, Zhang S, Jie C, Chen C, Xiao C, Qin C, Cheng D. Piezo1-Mediated Mechanotransduction Contributes to Disturbed Flow-Induced Atherosclerotic Endothelial Inflammation. J Am Heart Assoc 2024; 13:e035558. [PMID: 39450718 DOI: 10.1161/jaha.123.035558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/12/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Disturbed flow generates oscillatory shear stress (OSS), which in turn leads to endothelial inflammation and atherosclerosis. Piezo1, a biomechanical force sensor, plays a crucial role in the cardiovascular system. However, the specific role of Piezo1 in atherosclerosis remains to be fully elucidated. METHODS AND RESULTS We detected the expression of Piezo1 in atherosclerotic mice and endothelial cells from regions with disturbed blood flow. The pharmacological inhibitor Piezo1 inhibitor (GsMTx4) was used to evaluate the impact of Piezo1 on plaque progression and endothelial inflammation. We examined Piezo1's direct response to OSS in vitro and its effects on endothelial inflammation. Furthermore, mechanistic studies were conducted to explore the potential molecular cascade through which Piezo1 mediates endothelial inflammation in response to OSS. Our findings revealed the upregulation of Piezo1 in apoE-/- (apolipoprotein E) atherosclerotic mice, which is associated with disturbed flow. Treatment with GsMTx4 not only delayed plaque progression but also mitigated endothelial inflammation in both chronic and disturbed flow-induced atherosclerosis. Piezo1 was shown to facilitate calcium ions (Ca2+) influx in response to OSS, thereby activating endothelial inflammation. This inflammatory response was attenuated in the absence of Piezo1. Additionally, we identified that under OSS, Piezo1 activates the Ca2+/CaM/CaMKII (calmodulin/calmodulin-dependent protein kinases Ⅱ) pathways, which subsequently stimulate downstream kinases FAK (focal adhesion kinase) and Src. This leads to the activation of the OSS-sensitive YAP (yes-associated protein), ultimately triggering endothelial inflammation. CONCLUSIONS Our study highlights the key role of Piezo1 in atherosclerotic endothelial inflammation, proposing the Piezo1-Ca2+/CaM/CaMKII-FAK/Src-YAP axis as a previously unknown endothelial mechanotransduction pathway. Piezo1 is expected to become a potential therapeutic target for atherosclerosis and cardiovascular diseases.
Collapse
Affiliation(s)
- Yining Lan
- Department of Neurology The First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
| | - Jing Lu
- Department of Neurology The First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
| | - Shaohan Zhang
- The Second Affiliated Hospital of Qiqihar Medical College Qiqihar Heilongjiang China
| | - Chunxiao Jie
- Department of Neurology The First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
| | - Chunyong Chen
- Department of Neurology The First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
| | - Chao Xiao
- Department of Neurology The First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
- Department of Neurology Liuzhou People's Hospital Liuzhou Guangxi China
| | - Chao Qin
- Department of Neurology The First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
| | - Daobin Cheng
- Department of Neurology The First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
| |
Collapse
|
5
|
Zeng Y, Shen J, Zhou X, Ouyang Z, Zhong J, Qin Y, Jin L, He X, Li L, Xie J, Liu X. Osteogenic differentiation of bone mesenchymal stem cells on linearly aligned triangular micropatterns. J Mater Chem B 2024; 12:8420-8430. [PMID: 39093007 DOI: 10.1039/d4tb01218f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Mesenchymal stem cells (MSCs) hold promise for regenerative medicine, particularly for bone tissue engineering. However, directing MSC differentiation towards specific lineages, such as osteogenic, while minimizing undesired phenotypes remains a challenge. Here, we investigate the influence of micropatterns on the behavior and lineage commitment of rat bone marrow-derived MSCs (rBMSCs), focusing on osteogenic differentiation. Linearly aligned triangular micropatterns (TPs) and circular micropatterns (CPs) coated with fibronectin were fabricated to study their effects on rBMSC morphology and differentiation and the underlying mechanobiological mechanisms. TPs, especially TP15 (15 μm), induced the cell elongation and thinning, while CPs also promoted the cell stretching, as evidenced by the decreased circularity and increased aspect ratio. TP15 significantly promoted osteogenic differentiation, with increased expression of osteogenic genes (Runx2, Spp1, Alpl, Bglap, Col1a1) and decreased expression of adipogenic genes (Pparg, Cebpa, Fabp4). Conversely, CPs inhibited both osteogenic and adipogenic differentiation. Mechanistically, TP15 increased Piezo1 activity, cytoskeletal remodeling including the aggregates of F-actin and myosin filaments at the cell periphery, YAP1 nuclear translocation, and integrin upregulation. Piezo1 inhibition suppressed the osteogenic genes expression, myosin remodeling, and YAP1 nuclear translocation, indicating Piezo1-mediated the mechanotransduction in rBMSCs on TPs. TP15 also induced osteogenic differentiation of BMSCs from aging rats, with upregulated Piezo1 and nuclear translocation of YAP1. Therefore, triangular micropatterns, particularly TP15, promote osteogenesis and inhibit adipogenesis of rBMSCs through Piezo1-mediated myosin and YAP1 pathways. Our study provides novel insights into the mechanobiological mechanisms governing MSC behaviors on micropatterns, offering new strategies for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Junyi Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Xintong Zhou
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Zhi Ouyang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Jian Zhong
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Yixue Qin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Linlu Jin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Xueling He
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Liang Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| |
Collapse
|
6
|
Karkempetzaki AI, Ravid K. Piezo1 and Its Function in Different Blood Cell Lineages. Cells 2024; 13:482. [PMID: 38534326 PMCID: PMC10969519 DOI: 10.3390/cells13060482] [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: 02/07/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Mechanosensation is a fundamental function through which cells sense mechanical stimuli by initiating intracellular ion currents. Ion channels play a pivotal role in this process by orchestrating a cascade of events leading to the activation of downstream signaling pathways in response to particular stimuli. Piezo1 is a cation channel that reacts with Ca2+ influx in response to pressure sensation evoked by tension on the cell lipid membrane, originating from cell-cell, cell-matrix, or hydrostatic pressure forces, such as laminar flow and shear stress. The application of such forces takes place in normal physiological processes of the cell, but also in the context of different diseases, where microenvironment stiffness or excessive/irregular hydrostatic pressure dysregulates the normal expression and/or activation of Piezo1. Since Piezo1 is expressed in several blood cell lineages and mutations of the channel have been associated with blood cell disorders, studies have focused on its role in the development and function of blood cells. Here, we review the function of Piezo1 in different blood cell lineages and related diseases, with a focus on megakaryocytes and platelets.
Collapse
Affiliation(s)
- Anastasia Iris Karkempetzaki
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA;
- Whitaker Cardiovascular Institute, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
- School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Katya Ravid
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA;
- Whitaker Cardiovascular Institute, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| |
Collapse
|
7
|
Luu N, Bajpai A, Li R, Park S, Noor M, Ma X, Chen W. Aging-associated decline in vascular smooth muscle cell mechanosensation is mediated by Piezo1 channel. Aging Cell 2024; 23:e14036. [PMID: 37941511 PMCID: PMC10861209 DOI: 10.1111/acel.14036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 11/10/2023] Open
Abstract
Aging of the vasculature is associated with detrimental changes in vascular smooth muscle cell (VSMC) mechanosensitivity to extrinsic forces in their surrounding microenvironment. However, how chronological aging alters VSMCs' ability to sense and adapt to mechanical perturbations remains unexplored. Here, we show defective VSMC mechanosensation in aging measured with ultrasound tweezers-based micromechanical system, force instantaneous frequency spectrum, and transcriptome analyses. The study reveals that aged VSMCs adapt to a relatively inert mechanobiological state with altered actin cytoskeletal integrity, resulting in an impairment in their mechanosensitivity and dynamic mechanoresponse to mechanical perturbations. The aging-associated decline in mechanosensation behaviors is mediated by hyperactivity of Piezo1-dependent calcium signaling. Inhibition of Piezo1 alleviates vascular aging and partially restores the loss in dynamic contractile properties in aged cells. Altogether, our study reveals the signaling pathway underlying aging-associated aberrant mechanosensation in VSMC and identifies Piezo1 as a potential therapeutic mechanobiological target to alleviate vascular aging.
Collapse
Affiliation(s)
- Ngoc Luu
- Department of Biomedical EngineeringNew York UniversityBrooklynNew YorkUSA
| | - Apratim Bajpai
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNew YorkUSA
| | - Rui Li
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNew YorkUSA
| | - Seojin Park
- Department of Biomedical EngineeringNew York UniversityBrooklynNew YorkUSA
| | - Mahad Noor
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNew YorkUSA
| | - Xiao Ma
- Department of Biomedical EngineeringNew York UniversityBrooklynNew YorkUSA
| | - Weiqiang Chen
- Department of Biomedical EngineeringNew York UniversityBrooklynNew YorkUSA
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNew YorkUSA
- Laura and Isaac Perlmutter Cancer CenterNYU Langone HealthNew YorkUSA
| |
Collapse
|
8
|
Xia K, Chen X, Wang W, Liu Q, Zhao M, Ma J, Jia H. Roles of mechanosensitive ion channels in immune cells. Heliyon 2024; 10:e23318. [PMID: 38148826 PMCID: PMC10750075 DOI: 10.1016/j.heliyon.2023.e23318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
Mechanosensitive ion channels are a class of membrane-integrated proteins that convert externalmechanical forces, including stretching, pressure, gravity, and osmotic pressure changes, some of which can be caused by pathogen invasion, into electrical and chemical signals transmitted to the cytoplasm. In recent years, with the identification of many of these channels, their roles in the initiation and progression of many diseases have been gradually revealed. Multiple studies have shown that mechanosensitive ion channels regulate the proliferation, activation, and inflammatory responses of immune cells by being expressed on the surface of immune cells and further responding to mechanical forces. Nonetheless, further clarification is required regarding the signaling pathways of immune-cell pattern-recognition receptors and on the impact of microenvironmental changes and mechanical forces on immune cells. This review summarizes the roles of mechanosensitive ion channels in immune cells.
Collapse
Affiliation(s)
- Kexin Xia
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Xiaolin Chen
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Wenyan Wang
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Qianwen Liu
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Mai Zhao
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Road 100, Shanghai, 200080, China
| | - Jiacheng Ma
- The Department of Information Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, 999077, China
| | - Hao Jia
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry & Molecular Cellular Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
9
|
Wang Z, Chen X, Chen N, Yan H, Wu K, Li J, Ru Q, Deng R, Liu X, Kang R. Mechanical Factors Regulate Annulus Fibrosus (AF) Injury Repair and Remodeling: A Review. ACS Biomater Sci Eng 2024; 10:219-233. [PMID: 38149967 DOI: 10.1021/acsbiomaterials.3c01091] [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: 12/28/2023]
Abstract
Low back pain is a common chronic disease that can severely affect the patient's work and daily life. The breakdown of spinal mechanical homeostasis caused by intervertebral disc (IVD) degeneration is a leading cause of low back pain. Annulus fibrosus (AF), as the outer layer structure of the IVD, is often the first affected part. AF injury caused by consistent stress overload will further accelerate IVD degeneration. Therefore, regulating AF injury repair and remodeling should be the primary goal of the IVD repair strategy. Mechanical stimulation has been shown to promote AF regeneration and repair, but most studies only focus on the effect of single stress on AF, and lack realistic models and methods that can mimic the actual mechanical environment of AF. In this article, we review the effects of different types of stress stimulation on AF injury repair and remodeling, suggest possible beneficial load combinations, and explore the underlying molecular mechanisms. It will provide the theoretical basis for designing better tissue engineering therapy using mechanical factors to regulate AF injury repair and remodeling in the future.
Collapse
Affiliation(s)
- Zihan Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210028, P.R. China
| | - Xin Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210028, P.R. China
| | - Nan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210028, P.R. China
| | - Hongjie Yan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210028, P.R. China
| | - Ke Wu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210028, P.R. China
| | - Jitao Li
- School of Physics and Telecommunications Engineering, Zhoukou Normal University, Zhoukou, Henan Province 466001, P.R. China
| | - Qingyuan Ru
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210028, P.R. China
| | - Rongrong Deng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210028, P.R. China
| | - Xin Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210028, P.R. China
| | - Ran Kang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210028, P.R. China
- Department of Orthopedics, Nanjing Lishui Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu Province 210028, P.R. China
| |
Collapse
|
10
|
Cheng D, Wang J, Yao M, Cox CD. Joining forces: crosstalk between mechanosensitive PIEZO1 ion channels and integrin-mediated focal adhesions. Biochem Soc Trans 2023; 51:1897-1906. [PMID: 37772664 DOI: 10.1042/bst20230042] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 09/30/2023]
Abstract
Both integrin-mediated focal adhesions (FAs) and mechanosensitive ion channels such as PIEZO1 are critical in mechanotransduction processes that influence cell differentiation, development, and cancer. Ample evidence now exists for regulatory crosstalk between FAs and PIEZO1 channels with the molecular mechanisms underlying this process remaining unclear. However, an emerging picture is developing based on spatial crosstalk between FAs and PIEZO1 revealing a synergistic model involving the cytoskeleton, extracellular matrix (ECM) and calcium-dependent signaling. Already cell type, cell contractility, integrin subtypes and ECM composition have been shown to regulate this crosstalk, implying a highly fine-tuned relationship between these two major mechanosensing systems. In this review, we summarize the latest advances in this area, highlight the physiological implications of this crosstalk and identify gaps in our knowledge that will improve our understanding of cellular mechanosensing.
Collapse
Affiliation(s)
- Delfine Cheng
- The Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Junfan Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mingxi Yao
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Charles D Cox
- The Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Kensington, NSW 2052, Australia
| |
Collapse
|
11
|
Liu C, Gao X, Lou J, Li H, Chen Y, Chen M, Zhang Y, Hu Z, Chang X, Luo M, Zhai Y, Li C. Aberrant mechanical loading induces annulus fibrosus cells apoptosis in intervertebral disc degeneration via mechanosensitive ion channel Piezo1. Arthritis Res Ther 2023; 25:117. [PMID: 37420255 PMCID: PMC10327399 DOI: 10.1186/s13075-023-03093-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 06/16/2023] [Indexed: 07/09/2023] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is closely associated with the structural damage in the annulus fibrosus (AF). Aberrant mechanical loading is an important inducement of annulus fibrosus cells (AFCs) apoptosis, which contributes to the AF structural damage and aggravates IVDD, but the underlying mechanism is still unclear. This study aims to investigate the mechanism of a mechanosensitive ion channel protein Piezo1 in aberrant mechanical loading-induced AFCs apoptosis and IVDD. METHODS Rats were subjected to lumbar instability surgery to induce the unbalanced dynamic and static forces to establish the lumbar instability model. MRI and histological staining were used to evaluate the IVDD degree. A cyclic mechanical stretch (CMS)-stimulated AFCs apoptosis model was established by a Flexcell system in vitro. Tunel staining, mitochondrial membrane potential (MMP) detection, and flow cytometry were used to evaluate the apoptosis level. The activation of Piezo1 was detected using western blot and calcium fluorescent probes. Chemical activator Yoda1, chemical inhibitor GSMTx4, and a lentiviral shRNA-Piezo1 system (Lv-Piezo1) were utilized to regulate the function of Piezo1. High-throughput RNA sequencing (RNA-seq) was used to explore the mechanism of Piezo1-induced AFCs apoptosis. The Calpain activity and the activation of Calpain2/Bax/Caspase3 axis were evaluated by the Calpain activity kit and western blot with the siRNA-mediated Calapin1 or Calpain2 knockdown. Intradiscal administration of Lv-Piezo1 was utilized to evaluate the therapeutic effect of Piezo1 silencing in IVDD rats. RESULTS Lumbar instability surgery promoted the expression of Piezo1 in AFCs and stimulated IVDD in rats 4 weeks after surgery. CMS elicited distinct apoptosis of AFCs, with enhanced Piezo1 activation. Yoda1 further promoted CMS-induced apoptosis of AFCs, while GSMTx4 and Lv-Piezo1 exhibited opposite effects. RNA-seq showed that knocking down Piezo1 inhibited the calcium signaling pathway. CMS enhanced Calpain activity and elevated the expression of BAX and cleaved-Caspase3. Calpain2, but not Calpain1 knockdown, inhibited the expression of BAX and cleaved-Caspase3 and alleviated AFCs apoptosis. Lv-Piezo1 significantly alleviated the progress of IVDD in rats after lumbar instability surgery. CONCLUSIONS Aberrant mechanical loading induces AFCs apoptosis to promote IVDD by activating Piezo1 and downstream Calpain2/BAX/Caspase3 pathway. Piezo1 is expected to be a potential therapeutic target in treating IVDD.
Collapse
Affiliation(s)
- Chenhao Liu
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
- Department of Orthopedics, Qinghai Provincial People's Hospital, Xining, 810007, Qinghai, China
| | - Xiaoxin Gao
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
| | - Jinhui Lou
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
| | - Haiyin Li
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
| | - Yuxuan Chen
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
- Center of Traumatic Orthopedics, People's Liberation Army 990 Hospital, Xinyang, 464000, Henan, China
| | - Molong Chen
- Department of Orthopedics/Sports Medicine Center, The First Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
| | - Yuyao Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
| | - Zhilei Hu
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
| | - Xian Chang
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
| | - Menglin Luo
- Clinical Laboratory, Qinghai Provincial People's Hospital, Xining, 810007, Qinghai, China
| | - Yu Zhai
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China.
| | - Changqing Li
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China.
| |
Collapse
|
12
|
Liu L, Qu Y, Cheng L, Yoon CW, He P, Monther A, Guo T, Chittle S, Wang Y. Engineering chimeric antigen receptor T cells for solid tumour therapy. Clin Transl Med 2022; 12:e1141. [PMID: 36495108 PMCID: PMC9736813 DOI: 10.1002/ctm2.1141] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/22/2022] [Accepted: 11/26/2022] [Indexed: 12/13/2022] Open
Abstract
Cell-based immunotherapy, for example, chimeric antigen receptor T (CAR-T) cell immunotherapy, has revolutionized cancer treatment, particularly for blood cancers. However, factors such as insufficient T cell tracking, tumour heterogeneity, inhibitory tumour microenvironment (TME) and T cell exhaustion limit the broad application of CAR-based immunotherapy for solid tumours. In particular, the TME is a complex and evolving entity, which is composed of cells of different types (e.g., cancer cells, immune cells and stromal cells), vasculature, soluble factors and extracellular matrix (ECM), with each component playing a critical role in CAR-T immunotherapy. Thus, developing approaches to mitigate the inhibitory TME factors is critical for future success in applying CAR-T cells for solid tumour treatment. Accordingly, understanding the bilateral interaction of CAR-T cells with the TME is in pressing need to pave the way for more efficient therapeutics. In the following review, we will discuss TME-associated aspects with an emphasis on T cell trafficking, ECM barriers, abnormal vasculature, solid tumour heterogenicity and immune suppressive microenvironment. We will then summarize current engineering strategies to overcome the challenges posed by the TME-associated factors. Lastly, the future directions for engineering efficient CAR-T cells for solid tumour therapy will be discussed.
Collapse
Affiliation(s)
- Longwei Liu
- Department of BioengineeringInstitute of Engineering in MedicineUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Yunjia Qu
- Department of BioengineeringInstitute of Engineering in MedicineUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Leonardo Cheng
- Department of BioengineeringInstitute of Engineering in MedicineUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Chi Woo Yoon
- Department of BioengineeringInstitute of Engineering in MedicineUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Peixiang He
- Department of BioengineeringInstitute of Engineering in MedicineUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Abdula Monther
- Department of BioengineeringInstitute of Engineering in MedicineUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Tianze Guo
- Department of BioengineeringInstitute of Engineering in MedicineUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Sarah Chittle
- Department of BioengineeringInstitute of Engineering in MedicineUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Yingxiao Wang
- Department of BioengineeringInstitute of Engineering in MedicineUniversity of CaliforniaLa JollaCaliforniaUSA
| |
Collapse
|