1
|
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
|
2
|
Bai J, Yan M, Xu Y, Wang Y, Yao Y, Jin P, Zhang Y, Qu Y, Niu L, Li H. YAP enhances mitochondrial OXPHOS in tumor-infiltrating Treg through upregulating Lars2 on stiff matrix. J Immunother Cancer 2024; 12:e010463. [PMID: 39551603 PMCID: PMC11574482 DOI: 10.1136/jitc-2024-010463] [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] [Accepted: 10/27/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Tumor-infiltrating regulatory T cells (TI-Tregs) are well-adapted to thrive in the challenging tumor microenvironment (TME) by undergoing metabolic reprogramming, notably shifting from glycolysis to mitochondrial oxidative phosphorylation (OXPHOS) for energy production. The extracellular matrix is an important component of the TME, contributing to the regulation of both tumor and immune cell metabolism patterns by activating mechanosensors such as YAP. Whether YAP plays a part in regulating TI-Treg mitochondrial function and the underlying mechanisms are yet to be elucidated. METHODS To gain insights into the effect of matrix stiffness on YAP activation in Tregs, alterations in stiffness were performed both in vitro and in vivo. YAP conditional knockout mice were used to determine the role of YAP in TI-Tregs. RNA-seq, quantitative PCR, flow cytometry, lentivirus infection and mitochondrial function assay were employed to uncover the mechanism of YAP modulating mitochondrial function in TI-Tregs. A YAP inhibitor and a low leucine diet were applied to tumor-bearing mice to seek the potential antitumor strategy. RESULTS In this study, we found that YAP, as a mechanotransducer, was activated by matrix stiffness in TI-Tregs. A deficiency in YAP significantly hindered the immunosuppressive capability of TI-Tregs by disrupting mitochondrial function. Mechanically, YAP enhanced mitochondrial OXPHOS by upregulating the transcription of Lars2 (Leucyl-tRNA synthetase 2, mitochondrial), which was essential for mitochondrial protein translation in TI-Tregs. Since Lars2 relied much on its substrate amino acid, leucine, the combination of a low leucine diet and YAP inhibitor synergistically induced mitochondrial dysfunction in TI-Tregs, ultimately restraining tumor growth. CONCLUSIONS This finding uncovered a new understanding of how YAP shapes mitochondrial function in TI-Tregs in response to mechanical signals within the TME, making the combined strategy of traditional medicine and diet adjustment a promising approach for tumor therapy.
Collapse
Affiliation(s)
- Jingchao Bai
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Meinan Yan
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Yihan Xu
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Youhui Wang
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Yuan Yao
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Peng Jin
- Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuhan Zhang
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Yang Qu
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Liling Niu
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Hui Li
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| |
Collapse
|
3
|
Santiago-Carvalho I, Ishikawa M, Borges da Silva H. Channel plan: control of adaptive immune responses by pannexins. Trends Immunol 2024; 45:892-902. [PMID: 39393945 PMCID: PMC11560585 DOI: 10.1016/j.it.2024.09.009] [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: 07/16/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024]
Abstract
The development of mammalian adaptive (i.e., B and T cell-mediated) immune responses is tightly controlled at transcriptional, epigenetic, and metabolic levels. Signals derived from the extracellular milieu are crucial regulators of adaptive immunity. Beyond the traditionally studied cytokines and chemokines, many other extracellular metabolites can bind to specialized receptors and regulate T and B cell immune responses. These molecules often accumulate extracellularly through active export by plasma membrane transporters. For example, mammalian immune and non-immune cells express pannexin (PANX)1-3 channels on the plasma membrane, which release many distinct small molecules, notably intracellular ATP. Here, we review novel findings defining PANXs as crucial regulators of T and B cell immune responses in disease contexts such as cancer or viral infections.
Collapse
Affiliation(s)
| | - Masaki Ishikawa
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | | |
Collapse
|
4
|
Xing H, Liu H, Chang Z, Zhang J. Research progress on the immunological functions of Piezo1 a receptor molecule that responds to mechanical force. Int Immunopharmacol 2024; 139:112684. [PMID: 39008939 DOI: 10.1016/j.intimp.2024.112684] [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: 04/17/2024] [Revised: 06/30/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
The human immune system is capable of defending against, monitoring, and self-stabilizing various immune cells. Differentiation, proliferation, and development of these cells are regulated by biochemical signals. Moreover, biophysical signals, such as mechanical forces, have been found to affect immune cell function, thus introducing a new area of immunological research. Piezo1, a mechanically sensitive ion channel, was awarded the Nobel Prize for Physiology and Medicine in 2021. This channel is present on the surface of many cells, and when stimulated by mechanical force, it controls calcium (Ca2+) inside the cells, leading to changes in downstream signals and thus regulating cell functions. Piezo1 is also expressed in various innate and adaptive immune cells and plays a major role in the immune function. In this review, we will explore the physiological functions and regulatory mechanisms of Piezo1 and its impact on innate and adaptive immunity. This may offer new insights into diagnostics and therapeutics for the prevention and treatment of diseases and surgical infections.
Collapse
Affiliation(s)
- Hao Xing
- Department of Orthopaedics, The 960th Hospital of PLA, Jinan 250031, China
| | - Huan Liu
- Department of Orthopaedics, The 960th Hospital of PLA, Jinan 250031, China; The Second Medical University of Shandong, Weifang, Shandong 261000, China
| | - Zhengqi Chang
- Department of Orthopaedics, The 960th Hospital of PLA, Jinan 250031, China.
| | - Ji Zhang
- Department of Immunology, Basic Medical College, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
5
|
Pang R, Sun W, Yang Y, Wen D, Lin F, Wang D, Li K, Zhang N, Liang J, Xiong C, Liu Y. PIEZO1 mechanically regulates the antitumour cytotoxicity of T lymphocytes. Nat Biomed Eng 2024; 8:1162-1176. [PMID: 38514773 DOI: 10.1038/s41551-024-01188-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 02/05/2024] [Indexed: 03/23/2024]
Abstract
The killing function of cytotoxic T cells can be enhanced biochemically. Here we show that blocking the mechanical sensor PIEZO1 in T cells strengthens their traction forces and augments their cytotoxicity against tumour cells. By leveraging cytotoxic T cells collected from tumour models in mice and from patients with cancers, we show that PIEZO1 upregulates the transcriptional factor GRHL3, which in turn induces the expression of the E3 ubiquitin ligase RNF114. RNF114 binds to filamentous actin, causing its downregulation and rearrangement, which depresses traction forces in the T cells. In mice with tumours, the injection of cytotoxic T cells collected from the animals and treated with a PIEZO1 antagonist promoted their infiltration into the tumour and attenuated tumour growth. As an immunomechanical regulator, PIEZO1 could be targeted to enhance the outcomes of cancer immunotherapies.
Collapse
Affiliation(s)
- Ruiyang Pang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Weihao Sun
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Yingyun Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Dahan Wen
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Feng Lin
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Dingding Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Center for Bioinformatics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Kailong Li
- Department of Biochemistry and Biophysics, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Centre, Beijing, China
| | - Ning Zhang
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Institute of Blood Transfusion, Peking Union Medical College & Chinese Academy of Medical Sciences, Chengdu, China
| | - Junbo Liang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Center for Bioinformatics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.
| | - Chunyang Xiong
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China.
| | - Yuying Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.
- Clinical Immunology Centre, CAMS, Beijing, China.
| |
Collapse
|
6
|
Zhou R, Fu W, Vasylyev D, Waxman SG, Liu CJ. Ion channels in osteoarthritis: emerging roles and potential targets. Nat Rev Rheumatol 2024; 20:545-564. [PMID: 39122910 DOI: 10.1038/s41584-024-01146-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
Osteoarthritis (OA) is a highly prevalent joint disease that causes substantial disability, yet effective approaches to disease prevention or to the delay of OA progression are lacking. Emerging evidence has pinpointed ion channels as pivotal mediators in OA pathogenesis and as promising targets for disease-modifying treatments. Preclinical studies have assessed the potential of a variety of ion channel modulators to modify disease pathways involved in cartilage degeneration, synovial inflammation, bone hyperplasia and pain, and to provide symptomatic relief in models of OA. Some of these modulators are currently being evaluated in clinical trials. This review explores the structures and functions of ion channels, including transient receptor potential channels, Piezo channels, voltage-gated sodium channels, voltage-dependent calcium channels, potassium channels, acid-sensing ion channels, chloride channels and the ATP-dependent P2XR channels in the osteoarthritic joint. The discussion spans channel-targeting drug discovery and potential clinical applications, emphasizing opportunities for further research, and underscoring the growing clinical impact of ion channel biology in OA.
Collapse
Affiliation(s)
- Renpeng Zhou
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Wenyu Fu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Dmytro Vasylyev
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
7
|
Thien ND, Hai-Nam N, Anh DT, Baecker D. Piezo1 and its inhibitors: Overview and perspectives. Eur J Med Chem 2024; 273:116502. [PMID: 38761789 DOI: 10.1016/j.ejmech.2024.116502] [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: 04/05/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
The cation channel Piezo1, a crucial mechanotransducer found in various organs and tissues, has gained considerable attention as a therapeutic target in recent years. Following this trend, several Piezo1 inhibitors have been discovered and studied for potential pharmacological properties. This review provides an overview of the structural and functional importance of Piezo1, as well as discussing the biological activities of Piezo1 inhibitors based on their mechanism of action. The compounds addressed include the toxin GsMTx4, Aβ peptides, certain fatty acids, ruthenium red and gadolinium, Dooku1, as well as the natural products tubeimoside I, salvianolic acid B, jatrorrhzine, and escin. The findings revealed that misexpression of Piezo1 can be associated with a number of chronic diseases, including hypertension, cancer, and hemolytic anemia. Consequently, inhibiting Piezo1 and the subsequent calcium influx can have beneficial effects on various pathological processes, as shown by many in vitro and in vivo studies. However, the development of Piezo1 inhibitors is still in its beginnings, with many opportunities and challenges remaining to be explored.
Collapse
Affiliation(s)
- Nguyen Duc Thien
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 100000, Viet Nam
| | - Nguyen Hai-Nam
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 100000, Viet Nam
| | - Duong Tien Anh
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 100000, Viet Nam.
| | - Daniel Baecker
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, Berlin, 14195, Germany.
| |
Collapse
|
8
|
Ifejeokwu OV, Do A, El Khatib SM, Ho NH, Zavala A, Othy S, Acharya MM. Immune Checkpoint Inhibition-related Neuroinflammation Disrupts Cognitive Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601087. [PMID: 39005282 PMCID: PMC11244914 DOI: 10.1101/2024.07.01.601087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Combinatorial blockade of Cytotoxic T-lymphocyte associated protein 4 (CTLA-4) and Programmed Cell Death Protein 1 (PD-1) significantly improve the progression-free survival of individuals with metastatic cancers, including melanoma. In addition to unleashing anti-tumor immunity, combination immune checkpoint inhibition (ICI) disrupts immune-regulatory networks critical for maintaining homeostasis in various tissues, including the central nervous system (CNS). Although ICI- and cancer-related cognitive impairments (CRCI) in survivors are increasingly becoming evident, our understanding of ICI-induced immune-related adverse effects (IREA) in the CNS remains incomplete. Here, our murine melanoma model reveals that combination ICI impairs hippocampal-dependent learning and memory, as well as memory consolidation processes. Mechanistically, combination ICI disrupted synaptic integrity, and neuronal plasticity, reduced myelin, and further predisposed CNS for exaggerated experimental autoimmune encephalomyelitis. Combination ICI substantially altered both lymphoid and myeloid cells in the CNS. Neurogenesis was unaffected, however, microglial activation persisted for two-months post- ICI, concurrently with cognitive deficits, which parallels clinical observations in survivors. Overall, our results demonstrate that blockade of CTLA-4 and PD-1 alters neuro-immune homeostasis and activates microglia, promoting long-term neurodegeneration and driving cognitive impairments. Therefore, limiting microglial activation is a potential avenue to mitigate CNS IRAE while maintaining the therapeutic benefits of rapidly evolving ICIs and their combinations. SIGNIFICANCE Despite the superior therapeutic efficacy of immune checkpoint inhibition (ICI) for cancers, its undesired effects on brain function are not fully understood. Here, we demonstrate that combination ICI elevates neuroinflammation, activates microglia, leading to detrimental neurodegenerative and neurocognitive sequelae.
Collapse
|
9
|
Sarna NS, Desai SH, Kaufman BG, Curry NM, Hanna AM, King MR. Enhanced and sustained T cell activation in response to fluid shear stress. iScience 2024; 27:109999. [PMID: 38883838 PMCID: PMC11177201 DOI: 10.1016/j.isci.2024.109999] [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: 12/08/2023] [Revised: 04/08/2024] [Accepted: 05/14/2024] [Indexed: 06/18/2024] Open
Abstract
The efficacy of T cell therapies in treating solid tumors is limited by poor in vivo persistence, proliferation, and cytotoxicity, which can be attributed to limited and variable ex vivo activation. Herein, we present a 10-day kinetic profile of T cells subjected to fluid shear stress (FSS) ex vivo, with and without stimulation utilizing bead-conjugated anti-CD3/CD28 antibodies. We demonstrate that mechanical stimulation via FSS combined with bead-bound anti-CD3/CD28 antibodies yields a synergistic effect, resulting in amplified and sustained downstream signaling (NF-κB, c-Fos, and NFAT), expression of activation markers (CD69 and CD25), proliferation and production of pro-inflammatory cytokines (IFN-γ, TNF-α, and IL-2). This study represents the first characterization of the dynamic response of primary T cells to FSS. Collectively, our findings underscore the critical role of mechanosensitive ion channel-mediated mechanobiological signaling in T cell activation and fitness, enabling the development of strategies to address the current challenges associated with poor immunotherapy outcomes.
Collapse
Affiliation(s)
- Nicole S Sarna
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37235, United States
| | - Shanay H Desai
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37235, United States
- Department of Neuroscience, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37235, United States
| | - Benjamin G Kaufman
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37235, United States
| | - Natalie M Curry
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37235, United States
| | - Anne M Hanna
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37235, United States
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37235, United States
| |
Collapse
|
10
|
Zhang J, Li J, Hou Y, Lin Y, Zhao H, Shi Y, Chen K, Nian C, Tang J, Pan L, Xing Y, Gao H, Yang B, Song Z, Cheng Y, Liu Y, Sun M, Linghu Y, Li J, Huang H, Lai Z, Zhou Z, Li Z, Sun X, Chen Q, Su D, Li W, Peng Z, Liu P, Chen W, Huang H, Chen Y, Xiao B, Ye L, Chen L, Zhou D. Osr2 functions as a biomechanical checkpoint to aggravate CD8 + T cell exhaustion in tumor. Cell 2024; 187:3409-3426.e24. [PMID: 38744281 DOI: 10.1016/j.cell.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/04/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Alterations in extracellular matrix (ECM) architecture and stiffness represent hallmarks of cancer. Whether the biomechanical property of ECM impacts the functionality of tumor-reactive CD8+ T cells remains largely unknown. Here, we reveal that the transcription factor (TF) Osr2 integrates biomechanical signaling and facilitates the terminal exhaustion of tumor-reactive CD8+ T cells. Osr2 expression is selectively induced in the terminally exhausted tumor-specific CD8+ T cell subset by coupled T cell receptor (TCR) signaling and biomechanical stress mediated by the Piezo1/calcium/CREB axis. Consistently, depletion of Osr2 alleviates the exhaustion of tumor-specific CD8+ T cells or CAR-T cells, whereas forced Osr2 expression aggravates their exhaustion in solid tumor models. Mechanistically, Osr2 recruits HDAC3 to rewire the epigenetic program for suppressing cytotoxic gene expression and promoting CD8+ T cell exhaustion. Thus, our results unravel Osr2 functions as a biomechanical checkpoint to exacerbate CD8+ T cell exhaustion and could be targeted to potentiate cancer immunotherapy.
Collapse
Affiliation(s)
- Jinjia Zhang
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Junhong Li
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yongqiang Hou
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yao Lin
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China; Changping Laboratory, 102206 Beijing, China
| | - Hao Zhao
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yiran Shi
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Kaiyun Chen
- Fujian State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Cheng Nian
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jiayu Tang
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Lei Pan
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Yunzhi Xing
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Huan Gao
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Bingying Yang
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Zengfang Song
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yao Cheng
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yue Liu
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Min Sun
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yueyue Linghu
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jiaxin Li
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Haitao Huang
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhangjian Lai
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhien Zhou
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Zifeng Li
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiufeng Sun
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Qinghua Chen
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Dongxue Su
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wengang Li
- Department of Hepatobiliary and Pancreatic & Organ Transplantation Surgery, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhihai Peng
- Department of Hepatobiliary and Pancreatic & Organ Transplantation Surgery, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Pingguo Liu
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Department of Hepatobiliary Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China
| | - Wei Chen
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hongling Huang
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yixin Chen
- Fujian State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Bailong Xiao
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Frontier Research Center for Biological Structure, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China; Changping Laboratory, 102206 Beijing, China.
| | - Lanfen Chen
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China.
| | - Dawang Zhou
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
11
|
Song T, Hui W, Huang M, Guo Y, Yu M, Yang X, Liu Y, Chen X. Dynamic Changes in Ion Channels during Myocardial Infarction and Therapeutic Challenges. Int J Mol Sci 2024; 25:6467. [PMID: 38928173 PMCID: PMC11203447 DOI: 10.3390/ijms25126467] [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: 04/20/2024] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
In different areas of the heart, action potential waveforms differ due to differences in the expressions of sodium, calcium, and potassium channels. One of the characteristics of myocardial infarction (MI) is an imbalance in oxygen supply and demand, leading to ion imbalance. After MI, the regulation and expression levels of K+, Ca2+, and Na+ ion channels in cardiomyocytes are altered, which affects the regularity of cardiac rhythm and leads to myocardial injury. Myocardial fibroblasts are the main effector cells in the process of MI repair. The ion channels of myocardial fibroblasts play an important role in the process of MI. At the same time, a large number of ion channels are expressed in immune cells, which play an important role by regulating the in- and outflow of ions to complete intracellular signal transduction. Ion channels are widely distributed in a variety of cells and are attractive targets for drug development. This article reviews the changes in different ion channels after MI and the therapeutic drugs for these channels. We analyze the complex molecular mechanisms behind myocardial ion channel regulation and the challenges in ion channel drug therapy.
Collapse
Affiliation(s)
- Tongtong Song
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
- Department of Anatomy, College of Basic Medical Sciences, Jilin University, Changchun 130012, China
| | - Wenting Hui
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| | - Min Huang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| | - Yan Guo
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| | - Meiyi Yu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| | - Xiaoyu Yang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| | - Yanqing Liu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| | - Xia Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| |
Collapse
|
12
|
Hurrell BP, Shen S, Li X, Sakano Y, Kazemi MH, Quach C, Shafiei-Jahani P, Sakano K, Ghiasi H, Akbari O. Piezo1 channels restrain ILC2s and regulate the development of airway hyperreactivity. J Exp Med 2024; 221:e20231835. [PMID: 38530239 PMCID: PMC10965393 DOI: 10.1084/jem.20231835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/16/2024] [Accepted: 02/22/2024] [Indexed: 03/27/2024] Open
Abstract
Mechanosensitive ion channels sense force and pressure in immune cells to drive the inflammatory response in highly mechanical organs. Here, we report that Piezo1 channels repress group 2 innate lymphoid cell (ILC2)-driven type 2 inflammation in the lungs. Piezo1 is induced on lung ILC2s upon activation, as genetic ablation of Piezo1 in ILC2s increases their function and exacerbates the development of airway hyperreactivity (AHR). Conversely, Piezo1 agonist Yoda1 reduces ILC2-driven lung inflammation. Mechanistically, Yoda1 inhibits ILC2 cytokine secretion and proliferation in a KLF2-dependent manner, as we found that Piezo1 engagement reduces ILC2 oxidative metabolism. Consequently, in vivo Yoda1 treatment reduces the development of AHR in experimental models of ILC2-driven allergic asthma. Human-circulating ILC2s express and induce Piezo1 upon activation, as Yoda1 treatment of humanized mice reduces human ILC2-driven AHR. Our studies define Piezo1 as a critical regulator of ILC2s, and we propose the potential of Piezo1 activation as a novel therapeutic approach for the treatment of ILC2-driven allergic asthma.
Collapse
Affiliation(s)
- Benjamin P. Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stephen Shen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xin Li
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yoshihiro Sakano
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mohammad Hossein Kazemi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christine Quach
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kei Sakano
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Homayon Ghiasi
- Department of Surgery, Center for Neurobiology and Vaccine Development, Ophthalmology Research, Cedars-Sinai Burns and Allen Research Institute, Los Angeles, CA, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
13
|
Hou YJ, Yang XX, He L, Meng HX. Pathological mechanisms of cold and mechanical stress in modulating cancer progression. Hum Cell 2024; 37:593-606. [PMID: 38538930 DOI: 10.1007/s13577-024-01049-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/22/2024] [Indexed: 04/15/2024]
Abstract
Environmental temperature and cellular mechanical force are the inherent factors that participate in various biological processes and regulate cancer progress, which have been hot topics worldwide. They occupy a dominant part in the cancer tissues through different approaches. However, extensive investigation regarding pathological mechanisms in the carcinogenic field. After research, we found cold stress via two means to manipulate tumors: neuroscience and mechanically sensitive ion channels (MICHs) such as TRP families to regulate the physiological and pathological activities. Excessive cold stimulation mediated neuroscience acting on every cancer stage through the hypothalamus-pituitary-adrenocorticoid (HPA) to reach the target organs. Comparatively speaking, mechanical force via Piezo of MICHs controls cancer development. The progression of cancer depends on the internal activation of proto-oncogenes and the external tumorigenic factors; the above two means eventually lead to genetic disorders at the molecular level. This review summarizes the interaction of bidirectional communication between them and the tumor. It covers the main processes from cytoplasm to nucleus related to metastasis cascade and tumor immune escape.
Collapse
Affiliation(s)
- Yun-Jing Hou
- Harbin Medical University, Harbin, China
- Department of Precision Medicine Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin-Xin Yang
- Harbin Medical University, Harbin, China
- Department of Precision Medicine Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lin He
- Department of Stomatology, Heilongjiang Provincial Hospital, Harbin, China
| | - Hong-Xue Meng
- Harbin Medical University, Harbin, China.
- Department of Pathology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, China.
| |
Collapse
|
14
|
Yang B, Ma D, Zhu X, Wu Z, An Q, Zhao J, Gao X, Zhang L. Roles of TRP and PIEZO receptors in autoimmune diseases. Expert Rev Mol Med 2024; 26:e10. [PMID: 38659380 PMCID: PMC11140548 DOI: 10.1017/erm.2023.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/15/2023] [Accepted: 08/21/2023] [Indexed: 04/26/2024]
Abstract
Autoimmune diseases are pathological autoimmune reactions in the body caused by various factors, which can lead to tissue damage and organ dysfunction. They can be divided into organ-specific and systemic autoimmune diseases. These diseases usually involve various body systems, including the blood, muscles, bones, joints and soft tissues. The transient receptor potential (TRP) and PIEZO receptors, which resulted in David Julius and Ardem Patapoutian winning the Nobel Prize in Physiology or Medicine in 2021, attracted people's attention. Most current studies on TRP and PIEZO receptors in autoimmune diseases have been carried out on animal model, only few clinical studies have been conducted. Therefore, this study aimed to review existing studies on TRP and PIEZO to understand the roles of these receptors in autoimmune diseases, which may help elucidate novel treatment strategies.
Collapse
Affiliation(s)
- Baoqi Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Dan Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xueqing Zhu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Zewen Wu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Qi An
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Jingwen Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xinnan Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| |
Collapse
|
15
|
Zhang Y, Zou W, Dou W, Luo H, Ouyang X. Pleiotropic physiological functions of Piezo1 in human body and its effect on malignant behavior of tumors. Front Physiol 2024; 15:1377329. [PMID: 38690080 PMCID: PMC11058998 DOI: 10.3389/fphys.2024.1377329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Mechanosensitive ion channel protein 1 (Piezo1) is a large homotrimeric membrane protein. Piezo1 has various effects and plays an important and irreplaceable role in the maintenance of human life activities and homeostasis of the internal environment. In addition, recent studies have shown that Piezo1 plays a vital role in tumorigenesis, progression, malignancy and clinical prognosis. Piezo1 is involved in regulating the malignant behaviors of a variety of tumors, including cellular metabolic reprogramming, unlimited proliferation, inhibition of apoptosis, maintenance of stemness, angiogenesis, invasion and metastasis. Moreover, Piezo1 regulates tumor progression by affecting the recruitment, activation, and differentiation of multiple immune cells. Therefore, Piezo1 has excellent potential as an anti-tumor target. The article reviews the diverse physiological functions of Piezo1 in the human body and its major cellular pathways during disease development, and describes in detail the specific mechanisms by which Piezo1 affects the malignant behavior of tumors and its recent progress as a new target for tumor therapy, providing new perspectives for exploring more potential effects on physiological functions and its application in tumor therapy.
Collapse
Affiliation(s)
- Yihan Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
- The Second Clinical Medicine School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wen Zou
- The Second Clinical Medicine School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wenlei Dou
- The Second Clinical Medicine School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hongliang Luo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xi Ouyang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
- The Second Clinical Medicine School, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
16
|
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
|
17
|
Yanamandra AK, Zhang J, Montalvo G, Zhou X, Biedenweg D, Zhao R, Sharma S, Hoth M, Lautenschläger F, Otto O, Del Campo A, Qu B. PIEZO1-mediated mechanosensing governs NK-cell killing efficiency and infiltration in three-dimensional matrices. Eur J Immunol 2024; 54:e2350693. [PMID: 38279603 DOI: 10.1002/eji.202350693] [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: 07/31/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/28/2024]
Abstract
Natural killer (NK) cells play a vital role in eliminating tumorigenic cells. Efficient locating and killing of target cells in complex three-dimensional (3D) environments are critical for their functions under physiological conditions. However, the role of mechanosensing in regulating NK-cell killing efficiency in physiologically relevant scenarios is poorly understood. Here, we report that the responsiveness of NK cells is regulated by tumor cell stiffness. NK-cell killing efficiency in 3D is impaired against softened tumor cells, whereas it is enhanced against stiffened tumor cells. Notably, the durations required for NK-cell killing and detachment are significantly shortened for stiffened tumor cells. Furthermore, we have identified PIEZO1 as the predominantly expressed mechanosensitive ion channel among the examined candidates in NK cells. Perturbation of PIEZO1 abolishes stiffness-dependent NK-cell responsiveness, significantly impairs the killing efficiency of NK cells in 3D, and substantially reduces NK-cell infiltration into 3D collagen matrices. Conversely, PIEZO1 activation enhances NK killing efficiency as well as infiltration. In conclusion, our findings demonstrate that PIEZO1-mediated mechanosensing is crucial for NK killing functions, highlighting the role of mechanosensing in NK-cell killing efficiency under 3D physiological conditions and the influence of environmental physical cues on NK-cell functions.
Collapse
Affiliation(s)
- Archana K Yanamandra
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
- INM - Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Jingnan Zhang
- INM - Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Galia Montalvo
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
- Department of Experimental Physics, Saarland University, Saarbrücken, Germany
- Center for Biophysics, Saarland University, Saarbrücken, Germany
| | - Xiangda Zhou
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Doreen Biedenweg
- Institute of Physics, University of Greifswald, Greifswald, Germany
| | - Renping Zhao
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Shulagna Sharma
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Markus Hoth
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Franziska Lautenschläger
- Department of Experimental Physics, Saarland University, Saarbrücken, Germany
- Center for Biophysics, Saarland University, Saarbrücken, Germany
| | - Oliver Otto
- Institute of Physics, University of Greifswald, Greifswald, Germany
| | - Aránzazu Del Campo
- INM - Leibniz Institute for New Materials, Saarbrücken, Germany
- Chemistry Department, Saarland University, Saarbrücken, Germany
| | - Bin Qu
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
18
|
Coste B, Delmas P. PIEZO Ion Channels in Cardiovascular Functions and Diseases. Circ Res 2024; 134:572-591. [PMID: 38422173 DOI: 10.1161/circresaha.123.322798] [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] [Indexed: 03/02/2024]
Abstract
The cardiovascular system provides blood supply throughout the body and as such is perpetually applying mechanical forces to cells and tissues. Thus, this system is primed with mechanosensory structures that respond and adapt to changes in mechanical stimuli. Since their discovery in 2010, PIEZO ion channels have dominated the field of mechanobiology. These have been proposed as the long-sought-after mechanosensitive excitatory channels involved in touch and proprioception in mammals. However, more and more pieces of evidence point to the importance of PIEZO channels in cardiovascular activities and disease development. PIEZO channel-related cardiac functions include transducing hemodynamic forces in endothelial and vascular cells, red blood cell homeostasis, platelet aggregation, and arterial blood pressure regulation, among others. PIEZO channels contribute to pathological conditions including cardiac hypertrophy and pulmonary hypertension and congenital syndromes such as generalized lymphatic dysplasia and xerocytosis. In this review, we highlight recent advances in understanding the role of PIEZO channels in cardiovascular functions and diseases. Achievements in this quickly expanding field should open a new road for efficient control of PIEZO-related diseases in cardiovascular functions.
Collapse
Affiliation(s)
- Bertrand Coste
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| | - Patrick Delmas
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| |
Collapse
|
19
|
Jin T, Fei M, Luo S, Wang H. Piezo1 as a potential player in intracranial hemorrhage: from perspectives on biomechanics and hematoma metabolism. J Biomed Res 2024; 38:1-12. [PMID: 38808569 PMCID: PMC11461532 DOI: 10.7555/jbr.37.20230241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 05/30/2024] Open
Abstract
Intracranial hemorrhage (ICH) causes numerous neurological deficits and deaths worldwide each year, leaving a significant health burden on the public. The pathophysiology of ICH is complicated, and involves both primary and secondary injury. Hematoma, as the prime pathology of ICH, undergoes metabolism and triggers biochemical and biomechanical alterations in the brain, leading to secondary injury. Past endeavors mainly aimed at biochemical-initiated mechanisms for causing secondary injury have made limited progress in recent years, although ICH itself is also highly biomechanics-related. The discovery of the mechanical-activated cation channel Piezo1 provides a new avenue to further explore underlying mechanisms of secondary injury. The current article reviews the structure and gating mechanisms of Piezo1, its roles in the physiology/pathophysiology of neurons, astrocytes, microglia, and bone-marrow-derived macrophages, and especially its roles in erythrocytic turnover and iron metabolism, revealing a potential interplay between the biomechanics and biochemistry of hematoma in ICH. Collectively, these advances provide deeper insights into the secondary injury of ICH and lay the foundations for future research.
Collapse
Affiliation(s)
- Tianle Jin
- Department of Neurosurgery, Nanjing BenQ Medical Center, the Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, Jiangsu 210019, China
| | - Maoxing Fei
- Department of Neurosurgery, Nanjing Jinling Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210002, China
| | - Shiqiao Luo
- Department of Neurosurgery, the Affiliated Jinling Hospital of Nanjing Medical University, Nanjing, Jiangsu 210002, China
| | - Handong Wang
- Department of Neurosurgery, Nanjing BenQ Medical Center, the Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, Jiangsu 210019, China
| |
Collapse
|
20
|
Liu CSC, Mandal T, Biswas P, Hoque MA, Bandopadhyay P, Sinha BP, Sarif J, D'Rozario R, Sinha DK, Sinha B, Ganguly D. Piezo1 mechanosensing regulates integrin-dependent chemotactic migration in human T cells. eLife 2024; 12:RP91903. [PMID: 38393325 PMCID: PMC10942591 DOI: 10.7554/elife.91903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024] Open
Abstract
T cells are crucial for efficient antigen-specific immune responses and thus their migration within the body, to inflamed tissues from circulating blood or to secondary lymphoid organs, plays a very critical role. T cell extravasation in inflamed tissues depends on chemotactic cues and interaction between endothelial adhesion molecules and cellular integrins. A migrating T cell is expected to sense diverse external and membrane-intrinsic mechano-physical cues, but molecular mechanisms of such mechanosensing in cell migration are not established. We explored if the professional mechanosensor Piezo1 plays any role during integrin-dependent chemotaxis of human T cells. We found that deficiency of Piezo1 in human T cells interfered with integrin-dependent cellular motility on ICAM-1-coated surface. Piezo1 recruitment at the leading edge of moving T cells is dependent on and follows focal adhesion formation at the leading edge and local increase in membrane tension upon chemokine receptor activation. Piezo1 recruitment and activation, followed by calcium influx and calpain activation, in turn, are crucial for the integrin LFA1 (CD11a/CD18) recruitment at the leading edge of the chemotactic human T cells. Thus, we find that Piezo1 activation in response to local mechanical cues constitutes a membrane-intrinsic component of the 'outside-in' signaling in human T cells, migrating in response to chemokines, that mediates integrin recruitment to the leading edge.
Collapse
Affiliation(s)
- Chinky Shiu Chen Liu
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical BiologyKolkataIndia
| | - Tithi Mandal
- Department of Biological Sciences, Indian Institute of Science Education and ResearchKolkataIndia
| | - Parijat Biswas
- Department of Biological Sciences, Indian Association for Cultivation of ScienceKolkataIndia
| | - Md Asmaul Hoque
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical BiologyKolkataIndia
- Academy of Scientific and Innovative ResearchGhaziabadIndia
| | - Purbita Bandopadhyay
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical BiologyKolkataIndia
- Academy of Scientific and Innovative ResearchGhaziabadIndia
| | - Bishnu Prasad Sinha
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical BiologyKolkataIndia
- Academy of Scientific and Innovative ResearchGhaziabadIndia
| | - Jafar Sarif
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical BiologyKolkataIndia
- Academy of Scientific and Innovative ResearchGhaziabadIndia
| | - Ranit D'Rozario
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical BiologyKolkataIndia
- Academy of Scientific and Innovative ResearchGhaziabadIndia
| | - Deepak Kumar Sinha
- Department of Biological Sciences, Indian Association for Cultivation of ScienceKolkataIndia
| | - Bidisha Sinha
- Department of Biological Sciences, Indian Institute of Science Education and ResearchKolkataIndia
| | - Dipyaman Ganguly
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical BiologyKolkataIndia
| |
Collapse
|
21
|
He H, Zhou J, Xu X, Zhou P, Zhong H, Liu M. Piezo channels in the intestinal tract. Front Physiol 2024; 15:1356317. [PMID: 38379701 PMCID: PMC10877011 DOI: 10.3389/fphys.2024.1356317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/24/2024] [Indexed: 02/22/2024] Open
Abstract
The intestine is the largest mechanosensitive organ in the human body whose epithelial cells, smooth muscle cells, neurons and enteroendocrine cells must sense and respond to various mechanical stimuli such as motility, distension, stretch and shear to regulate physiological processes including digestion, absorption, secretion, motility and immunity. Piezo channels are a newly discovered class of mechanosensitive ion channels consisting of two subtypes, Piezo1 and Piezo2. Piezo channels are widely expressed in the intestine and are involved in physiological and pathological processes. The present review summarizes the current research progress on the expression, function and regulation of Piezo channels in the intestine, with the aim of providing a reference for the future development of therapeutic strategies targeting Piezo channels.
Collapse
Affiliation(s)
- Haolong He
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jingying Zhou
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xuan Xu
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Pinxi Zhou
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Huan Zhong
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Acupuncture and Moxibustion Bioinformatics, Education Department of Hunan Province, Changsha, Hunan, China
| | - Mi Liu
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Acupuncture and Moxibustion Bioinformatics, Education Department of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
22
|
Janssen E, van den Dries K, Ventre M, Cambi A. Mechanobiology of myeloid cells. Curr Opin Cell Biol 2024; 86:102311. [PMID: 38176349 DOI: 10.1016/j.ceb.2023.102311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/06/2024]
Abstract
Tissue-resident myeloid cells sense and transduce mechanical signals such as stiffness, stretch and compression. In the past two years, our understanding of the mechanosensitive signalling pathways in myeloid cells has significantly expanded. Moreover, it is increasingly clear which mechanical signals induce myeloid cells towards a pro- or anti-inflammatory phenotype. This is especially relevant in the context of altered matrix mechanics in immune-related pathologies or in the response to implanted biomaterials. A detailed understanding of myeloid cell mechanosensing may eventually lead to more effective cell-based immunotherapies for cancer, the development of mechanically inspired therapies to target fibrosis, and the engineering of safer implants. This review covers these recent advances in the emerging field of mechanoimmunology of myeloid cells.
Collapse
Affiliation(s)
- Eline Janssen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, the Netherlands
| | - Koen van den Dries
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, the Netherlands
| | - Maurizio Ventre
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, P.le Tecchio 80, 80125, Naples, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Fondazione Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Naples, Italy; Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Italy
| | - Alessandra Cambi
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, the Netherlands.
| |
Collapse
|
23
|
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
|
24
|
abiff M, Alshebremi M, Bonner M, Myers JT, Kim BG, Tomchuck SL, Santin A, Kingsley D, Choi SH, Huang AY. Piezo1 facilitates optimal T cell activation during tumor challenge. Oncoimmunology 2023; 12:2281179. [PMID: 38126029 PMCID: PMC10732680 DOI: 10.1080/2162402x.2023.2281179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 11/03/2023] [Indexed: 12/23/2023] Open
Abstract
Functional effector T cells in the tumor microenvironment (TME) are critical for successful anti-tumor responses. T cell anti-tumor function is dependent on their ability to differentiate from a naïve state, infiltrate into the tumor site, and exert cytotoxic functions. The factors dictating whether a particular T cell can successfully undergo these processes during tumor challenge are not yet completely understood. Piezo1 is a mechanosensitive cation channel with high expression on both CD4+ and CD8+ T cells. Previous studies have demonstrated that Piezo1 optimizes T cell activation and restrains the CD4+ regulatory T cell (Treg) pool in vitro and under inflammatory conditions in vivo. However, little is known about the role Piezo1 plays on CD4+ and CD8+ T cells in cancer. We hypothesized that disruption of Piezo1 on T cells impairs anti-tumor immunity in vivo by hindering inflammatory T cell responses. We challenged mice with T cell Piezo1 deletion (P1KO) with tumor models dependent on T cells for immune rejection. P1KO mice had the more aggressive tumors, higher tumor growth rates and were unresponsive to immune-mediated therapeutic interventions. We observed a decreased CD4:CD8 ratio in both the secondary lymphoid organs and TME of P1KO mice that correlated inversely with tumor size. Poor CD4+ helper T cell responses underpinned the immunodeficient phenotype of P1KO mice. Wild type CD8+ T cells are sub-optimally activated in vivo with P1KO CD4+ T cells, taking on a CD25loPD-1hi phenotype. Together, our results suggest that Piezo1 optimizes T cell activation in the context of a tumor response.
Collapse
Affiliation(s)
- muta abiff
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Mohammad Alshebremi
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Melissa Bonner
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jay T. Myers
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Byung-Gyu Kim
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Suzanne L. Tomchuck
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alicia Santin
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Daniel Kingsley
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sung Hee Choi
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alex Y. Huang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Center for Pediatric Immunotherapy, Angie Fowler AYA Cancer Institute, UH Rainbow Babies & Children’s Hospital, Cleveland, OH, USA
| |
Collapse
|
25
|
Zhu Y, Garcia-Sanchez J, Dalal R, Sun Y, Kapiloff MS, Goldberg JL, Liu WW. Differential expression of PIEZO1 and PIEZO2 mechanosensitive channels in ocular tissues implicates diverse functional roles. Exp Eye Res 2023; 236:109675. [PMID: 37820892 PMCID: PMC10843266 DOI: 10.1016/j.exer.2023.109675] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/08/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
PIEZO1 and PIEZO2 are mechanosensitive ion channels that regulate many important physiological processes including vascular blood flow, touch, and proprioception. As the eye is subject to mechanical stress and is highly perfused, these channels may play important roles in ocular function and intraocular pressure regulation. PIEZO channel expression in the eye has not been well defined, in part due to difficulties in validating available antibodies against PIEZO1 and PIEZO2 in ocular tissues. It is also unclear if PIEZO1 and PIEZO2 are differentially expressed. To address these questions, we used single-molecule fluorescence in situ hybridization (smFISH) together with transgenic reporter mice expressing PIEZO fusion proteins under the control of their endogenous promoters to compare the expression and localization of PIEZO1 and PIEZO2 in mouse ocular tissues relevant to glaucoma. We detected both PIEZO1 and PIEZO2 expression in the trabecular meshwork, ciliary body, and in the ganglion cell layer (GCL) of the retina. Piezo1 mRNA was more abundantly expressed than Piezo2 mRNA in these ocular tissues. Piezo1 but not Piezo2 mRNA was detected in the inner nuclear layer and outer nuclear layer of the retina. Our results suggest that PIEZO1 and PIEZO2 are differentially expressed and may have distinct roles as mechanosensors in glaucoma-relevant ocular tissues.
Collapse
Affiliation(s)
- Ying Zhu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Julian Garcia-Sanchez
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Roopa Dalal
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Yang Sun
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Michael S Kapiloff
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wendy W Liu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
26
|
Zhu H, He W, Ye P, Chen J, Wu X, Mu X, Wu Y, Pang H, Han F, Nie X. Piezo1 in skin wound healing and related diseases: Mechanotransduction and therapeutic implications. Int Immunopharmacol 2023; 123:110779. [PMID: 37582313 DOI: 10.1016/j.intimp.2023.110779] [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: 05/25/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/17/2023]
Abstract
Skin wound healing is a multifaceted and intricate process involving inflammation, tissue proliferation, and scar formation, all of which are accompanied by the continuous application of mechanical forces. Mechanotransduction is the mechanism by which the skin receives and reacts to physical signals from the internal and external environment, converting them into intracellular biochemical signals. This intricate process relies on specialized proteins known as mechanotransducers, with Piezo1 being a critical mechanosensitive ion channel that plays a central role in this process. This article provides an overview of the structural characteristics of Piezo1 and summarizes its effects on corresponding cells or tissues at different stages of skin trauma, including how it regulates skin sensation and skin-related diseases. The aim is to reveal the potential diagnostic and therapeutic value of Piezo1 in skin trauma and skin-related diseases. Piezo1 has been reported to be a vital mediator of mechanosensation and transduction in various organs and tissues. Given its high expression in the skin, Piezo1, as a significant cell membrane ion channel, is essential in activating intracellular signaling cascades that trigger several cellular physiological functions, including cell migration and muscle contraction. These functions contribute to the regulation and improvement of wound healing.
Collapse
Affiliation(s)
- Huan Zhu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Wenjie He
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Penghui Ye
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Jitao Chen
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Xingqian Wu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Xingrui Mu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Youzhi Wu
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland (UQ), Brisbane, QLD 4072, Australia.
| | - Huiwen Pang
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland (UQ), Brisbane, QLD 4072, Australia.
| | - Felicity Han
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland (UQ), Brisbane, QLD 4072, Australia.
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; School Medical Office, Zunyi Medical University, Zunyi 563006, China; Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland (UQ), Brisbane, QLD 4072, Australia; School of Biomedical Sciences, Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia.
| |
Collapse
|
27
|
Miyamoto S, Kondo T, Maruyama K. Senso-immunology: the past, present, and future. J Biochem 2023; 174:305-315. [PMID: 37461198 DOI: 10.1093/jb/mvad052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/13/2023] [Indexed: 09/29/2023] Open
Abstract
Pain and mechanical stimulation are thought to be alarm systems that alert the brain to physical abnormalities. When we experience unpleasant feelings in infected or traumatized tissues, our awareness is directed to the afflicted region, prompting activities such as resting or licking the tissue. Despite extensive research into the molecular biology of nociceptors, it was unclear whether their role was limited to the generation and transmission of unpleasant feelings or whether they actively modulate the pathogenesis of infected or traumatized tissues. Recently, it has become clear how the sensory and immune systems interact with one another and share similar receptors and ligands to modify the pathogenesis of various diseases. In this paper, we summarize the mechanisms of crosstalk between the sensory and immune systems and the impact of this new interdisciplinary field, which should be dubbed 'senso-immunology,' on medical science.
Collapse
Affiliation(s)
- Satoshi Miyamoto
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 3N7, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takeshi Kondo
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido 060-8636, Japan
| | - Kenta Maruyama
- National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8787, Japan
| |
Collapse
|
28
|
Kwak K, Sohn H, George R, Torgbor C, Manzella-Lapeira J, Brzostowski J, Pierce SK. B cell responses to membrane-presented antigens require the function of the mechanosensitive cation channel Piezo1. Sci Signal 2023; 16:eabq5096. [PMID: 37751477 PMCID: PMC10691204 DOI: 10.1126/scisignal.abq5096] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/05/2023] [Indexed: 09/28/2023]
Abstract
The demand for a vaccine for coronavirus disease 2019 (COVID-19) highlighted gaps in our understanding of the requirements for B cell responses to antigens, particularly to membrane-presented antigens, as occurs in vivo. We found that human B cell responses to membrane-presented antigens required the function of Piezo1, a plasma membrane mechanosensitive cation channel. Simply making contact with a glass probe induced calcium (Ca2+) fluxes in B cells that were blocked by the Piezo1 inhibitor GsMTx4. When placed on glass surfaces, the plasma membrane tension of B cells increased, which stimulated Ca2+ influx and spreading of B cells over the glass surface, which was blocked by the Piezo1 inhibitor OB-1. B cell responses to membrane-presented antigens but not to soluble antigens were inhibited both by Piezo1 inhibitors and by siRNA-mediated knockdown of Piezo1. Thus, the activation of Piezo1 defines an essential event in B cell activation to membrane-presented antigens that may be exploited to improve the efficacy of vaccines.
Collapse
Affiliation(s)
- Kihyuck Kwak
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Haewon Sohn
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Rachel George
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Charles Torgbor
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Javier Manzella-Lapeira
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Joseph Brzostowski
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Susan K. Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
29
|
Liu C, Xia Y, Fu S, Meng F, Feng B, Xu L, Li L, Zuo X. Inhibition of Piezo1 Ameliorates Intestinal Inflammation and Limits the Activation of Group 3 Innate Lymphoid Cells in Experimental Colitis. J Innate Immun 2023; 15:709-723. [PMID: 37725937 PMCID: PMC10601687 DOI: 10.1159/000533525] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 08/04/2023] [Indexed: 09/21/2023] Open
Abstract
Piezo1, the mechanosensory ion channel, has attracted increasing attention for its essential roles in various inflammatory responses and immune-related diseases. Although most of the key immune cells in inflammatory bowel disease (IBD) have been reported to be regulated by Piezo1, the specific role of Piezo1 in colitis has yet to be intensively studied. The present study investigated the impact of pharmacological inhibition of Piezo1 on dextran sulfate sodium (DSS)-induced colitis and explored the role of Piezo1 in intestinal immune cells in the context of colitis. We observed upregulated expression of Piezo1 in the colon tissue of mice with DSS-induced colitis. Pharmacological inhibition of Piezo1 by GsMTx4 diminished the severity of colitis. Piezo1 inhibition downregulated the expression of pro-inflammatory mediators Il1b, Il6, and Ptgs2 in colonic tissue and suppressed the production of IL-6 from macrophages and dendritic cells without altering the balance of T helper (Th) cells. In particular, Piezo1 did not affect cell viability but regulated cell proliferation and production of IL-17A in group 3 innate lymphoid cells (ILC3s), which is dependent on the PI3K-Akt-mTOR signaling pathway. Our findings uncover Piezo1 as an effective regulator of gut inflammation. Targeting Piezo1 could be a promising strategy to modulate intestinal immunity in IBD.
Collapse
Affiliation(s)
- Chang Liu
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China,
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan, China,
| | - Yanan Xia
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Shichen Fu
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Fanyi Meng
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Bingcheng Feng
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Leiqi Xu
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Lixiang Li
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital, Shandong University, Jinan, China
| | - Xiuli Zuo
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital, Shandong University, Jinan, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
30
|
Otero-Sobrino Á, Blanco-Carlón P, Navarro-Aguadero MÁ, Gallardo M, Martínez-López J, Velasco-Estévez M. Mechanosensitive Ion Channels: Their Physiological Importance and Potential Key Role in Cancer. Int J Mol Sci 2023; 24:13710. [PMID: 37762011 PMCID: PMC10530364 DOI: 10.3390/ijms241813710] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Mechanosensitive ion channels comprise a broad group of proteins that sense mechanical extracellular and intracellular changes, translating them into cation influx to adapt and respond to these physical cues. All cells in the organism are mechanosensitive, and these physical cues have proven to have an important role in regulating proliferation, cell fate and differentiation, migration and cellular stress, among other processes. Indeed, the mechanical properties of the extracellular matrix in cancer change drastically due to high cell proliferation and modification of extracellular protein secretion, suggesting an important contribution to tumor cell regulation. In this review, we describe the physiological significance of mechanosensitive ion channels, emphasizing their role in cancer and immunity, and providing compelling proof of the importance of continuing to explore their potential as new therapeutic targets in cancer research.
Collapse
Affiliation(s)
- Álvaro Otero-Sobrino
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Pablo Blanco-Carlón
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Miguel Ángel Navarro-Aguadero
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Miguel Gallardo
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Joaquín Martínez-López
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| | - María Velasco-Estévez
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| |
Collapse
|
31
|
Evtugina NG, Peshkova AD, Khabirova AI, Andrianova IA, Abdullayeva S, Ayombil F, Shepeliuk T, Grishchuk EL, Ataullakhanov FI, Litvinov RI, Weisel JW. Activation of Piezo1 channels in compressed red blood cells augments platelet-driven contraction of blood clots. J Thromb Haemost 2023; 21:2418-2429. [PMID: 37268065 PMCID: PMC10949619 DOI: 10.1016/j.jtha.2023.05.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND Piezo1 is a mechanosensitive cationic channel that boosts intracellular [Ca2+]i. Compression of red blood cells (RBCs) during platelet-driven contraction of blood clots may cause the activation of Piezo1. OBJECTIVES To establish relationships between Piezo1 activity and blood clot contraction. METHODS Effects of a Piezo1 agonist, Yoda1, and antagonist, GsMTx-4, on clot contraction in vitro were studied in human blood containing physiological [Ca2+]. Clot contraction was induced by exogenous thrombin. Activation of Piezo1 was assessed by Ca2+ influx in RBCs and with other functional and morphologic features. RESULTS Piezo1 channels in compressed RBCs are activated naturally during blood clot contraction and induce an upsurge in the intracellular [Ca2+]i, followed by phosphatidylserine exposure. Adding the Piezo1 agonist Yoda1 to whole blood increased the extent of clot contraction due to Ca2+-dependent volumetric shrinkage of RBCs and increased platelet contractility due to their hyperactivation by the enhanced generation of endogenous thrombin on activated RBCs. Addition of rivaroxaban, the inhibitor of thrombin formation, or elimination of Ca2+ from the extracellular space abrogated the stimulating effect of Yoda1 on clot contraction. The Piezo1 antagonist, GsMTx-4, caused a decrease in the extent of clot contraction relative to the control both in whole blood and in platelet-rich plasma. Activated Piezo1 in compressed and deformed RBCs amplified the platelet contractility as a positive feedback mechanism during clot contraction. CONCLUSION The results obtained demonstrate that the Piezo1 channel expressed on RBCs comprises a mechanochemical modulator of blood clotting that may be considered a potential therapeutic target to correct hemostatic disorders.
Collapse
Affiliation(s)
- Natalia G Evtugina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | - Alina D Peshkova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation; Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Alina I Khabirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | - Izabella A Andrianova
- Department of Internal Medicine, Division of Hematology and Program in Molecular Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Shahnoza Abdullayeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | - Francis Ayombil
- Division of Hematology and the Raymond G. Perelman Center for Cellular and Molecular Therapeutics, the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Taisia Shepeliuk
- Department of Physiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ekaterina L Grishchuk
- Department of Physiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Fazoil I Ataullakhanov
- Department of Physiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
32
|
Zheng Q, Liu H, Yu W, Dong Y, Zhou L, Deng W, Hua F. Mechanical properties of the brain: Focus on the essential role of Piezo1-mediated mechanotransduction in the CNS. Brain Behav 2023; 13:e3136. [PMID: 37366640 PMCID: PMC10498085 DOI: 10.1002/brb3.3136] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/24/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND The brain is a highly mechanosensitive organ, and changes in the mechanical properties of brain tissue influence many physiological and pathological processes. Piezo type mechanosensitive ion channel component 1 (Piezo1), a protein found in metazoans, is highly expressed in the brain and involved in sensing changes of the mechanical microenvironment. Numerous studies have shown that Piezo1-mediated mechanotransduction is closely related to glial cell activation and neuronal function. However, the precise role of Piezo1 in the brain requires further elucidation. OBJECTIVE This review first discusses the roles of Piezo1-mediated mechanotransduction in regulating the functions of a variety of brain cells, and then briefly assesses the impact of Piezo1-mediated mechanotransduction on the progression of brain dysfunctional disorders. CONCLUSIONS Mechanical signaling contributes significantly to brain function. Piezo1-mediated mechanotransduction regulates processes such as neuronal differentiation, cell migration, axon guidance, neural regeneration, and oligodendrocyte axon myelination. Additionally, Piezo1-mediated mechanotransduction plays significant roles in normal aging and brain injury, as well as the development of various brain diseases, including demyelinating diseases, Alzheimer's disease, and brain tumors. Investigating the pathophysiological mechanisms through which Piezo1-mediated mechanotransduction affects brain function will give us a novel entry point for the diagnosis and treatment of numerous brain diseases.
Collapse
Affiliation(s)
- Qingcui Zheng
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Jiangxi Province Key Laboratory of Molecular MedicineThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Hailin Liu
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Jiangxi Province Key Laboratory of Molecular MedicineThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Wen Yu
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Yao Dong
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Jiangxi Province Key Laboratory of Molecular MedicineThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Lanqian Zhou
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Jiangxi Province Key Laboratory of Molecular MedicineThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Wenze Deng
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Fuzhou Hua
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| |
Collapse
|
33
|
Yuan W, Zhang X, Fan X. The Role of the Piezo1 Mechanosensitive Channel in Heart Failure. Curr Issues Mol Biol 2023; 45:5830-5848. [PMID: 37504285 PMCID: PMC10378680 DOI: 10.3390/cimb45070369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/02/2023] [Accepted: 07/08/2023] [Indexed: 07/29/2023] Open
Abstract
Mechanotransduction (MT) is inseparable from the pathobiology of heart failure (HF). However, the effects of mechanical forces on HF remain unclear. This review briefly describes how Piezo1 functions in HF-affected cells, including endothelial cells (ECs), cardiac fibroblasts (CFs), cardiomyocytes (CMs), and immune cells. Piezo1 is a mechanosensitive ion channel that has been extensively studied in recent years. Piezo1 responds to different mechanical forces and converts them into intracellular signals. The pathways that modulate the Piezo1 switch have also been briefly described. Experimental drugs that specifically activate Piezo1-like proteins, such as Yoda1, Jedi1, and Jedi2, are available for clinical studies to treat Piezo1-related diseases. The only mechanosensitive ion-channel-specific inhibitor available is GsMTx4, which can turn off Piezo1 by modulating the local membrane tension. Ultrasound waves can modulate Piezo1 switching in vitro with the assistance of microbubbles. This review provides new possible targets for heart failure therapy by exploring the cellular functions of Piezo1 that are involved in the progression of the disease. Modulation of Piezo1 activity may, therefore, effectively delay the progression of heart failure.
Collapse
Affiliation(s)
- Weihua Yuan
- National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, 3333 Binsheng Rd, Hangzhou 310052, China
| | - Xicheng Zhang
- National Clinical Research Center for Child Health, Department of Cardiac Surgery, Children's Hospital, Zhejiang University School of Medicine, 3333 Binsheng Rd, Hangzhou 310052, China
| | - Xiangming Fan
- National Clinical Research Center for Child Health, Department of Cardiac Surgery, Children's Hospital, Zhejiang University School of Medicine, 3333 Binsheng Rd, Hangzhou 310052, China
| |
Collapse
|
34
|
Zhao T, Chu Z, Chu CH, Dong S, Li G, Wu J, Tang C. Macrophages induce gingival destruction via Piezo1-mediated MMPs-degrading collagens in periodontitis. Front Immunol 2023; 14:1194662. [PMID: 37261355 PMCID: PMC10228731 DOI: 10.3389/fimmu.2023.1194662] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/03/2023] [Indexed: 06/02/2023] Open
Abstract
Macrophages are an integral part of the innate immune response in periodontal tissue and play a crucial role in the progression of periodontitis. Here we reported that macrophages also provoke periodontitis-induced gingival destruction through Piezol-mediated collagen degradation. We discovered that the PIEZO1 expression was markedly elevated in patients with periodontitis through transcriptomic profiling. Moreover, Piezo1 promoted macrophage polarization toward the M1 type in response to lipopolysaccharide (LPS) and induced production of proinflammatory cytokines, which in turn stimulated production of matrix metalloproteinases (MMPs) leading to collagen degradation. Our study suggests that Piezol might be a potential therapeutic target for treating periodontitis-induced gingival destruction.
Collapse
Affiliation(s)
- Tong Zhao
- Department of Dental Implantology and Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Zhuangzhuang Chu
- Department of Dental Implantology and Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Catherine Huihan Chu
- Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
- Department of Dental Orthodontic, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuo Dong
- Department of Dental Implantology and Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Guoqing Li
- Department of Dental Implantology and Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Jin Wu
- Department of Dental Implantology and Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Chunbo Tang
- Department of Dental Implantology and Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| |
Collapse
|
35
|
Sonkodi B, Csorba A, Marsovszky L, Balog A, Kopper B, Nagy ZZ, Resch MD. Evidence of Disruption in Neural Regeneration in Dry Eye Secondary to Rheumatoid Arthritis. Int J Mol Sci 2023; 24:ijms24087514. [PMID: 37108693 PMCID: PMC10140938 DOI: 10.3390/ijms24087514] [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: 04/07/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
The purpose of our study was to analyze abnormal neural regeneration activity in the cornea through means of confocal microscopy in rheumatoid arthritis patients with concomitant dry eye disease. We examined 40 rheumatoid arthritis patients with variable severity and 44 volunteer age- and gender-matched healthy control subjects. We found that all examined parameters were significantly lower (p < 0.05) in rheumatoid arthritis patients as opposed to the control samples: namely, the number of fibers, the total length of the nerves, the number of branch points on the main fibers and the total nerve-fiber area. We examined further variables, such as age, sex and the duration of rheumatoid arthritis. Interestingly, we could not find a correlation between the above variables and abnormal neural structural changes in the cornea. We interpreted these findings via implementing our hypotheses. Correspondingly, one neuroimmunological link between dry eye and rheumatoid arthritis could be through the chronic Piezo2 channelopathy-induced K2P-TASK1 signaling axis. This could accelerate neuroimmune-induced sensitization on the spinal level in this autoimmune disease, with Langerhans-cell activation in the cornea and theorized downregulated Piezo1 channels in these cells. Even more importantly, suggested principal primary-damage-associated corneal keratocyte activation could be accompanied by upregulation of Piezo1. Both activation processes on the periphery would skew the plasticity of the Th17/Treg ratio, resulting in Th17/Treg imbalance in dry eye, secondary to rheumatoid arthritis. Hence, chronic somatosensory-terminal Piezo2 channelopathy-induced impaired Piezo2-Piezo1 crosstalk could result in a mixed picture of disrupted functional regeneration but upregulated morphological regeneration activity of these somatosensory axons in the cornea, providing the demonstrated abnormal neural corneal morphology.
Collapse
Affiliation(s)
- Balázs Sonkodi
- Department of Health Sciences and Sport Medicine, Hungarian University of Sports Science, 1123 Budapest, Hungary
| | - Anita Csorba
- Department of Ophthalmology, Semmelweis University, 1085 Budapest, Hungary
| | - László Marsovszky
- Department of Ophthalmology, Semmelweis University, 1085 Budapest, Hungary
| | - Attila Balog
- Department of Rheumatology and Immunology, Faculty of Medicine, Albert Szent-Györgyi Health Center, University of Szeged, 6725 Szeged, Hungary
| | - Bence Kopper
- Faculty of Kinesiology, Hungarian University of Sports Science, 1123 Budapest, Hungary
| | - Zoltán Zsolt Nagy
- Department of Ophthalmology, Semmelweis University, 1085 Budapest, Hungary
| | - Miklós D Resch
- Department of Ophthalmology, Semmelweis University, 1085 Budapest, Hungary
| |
Collapse
|
36
|
Chen S, Li Z, Chen D, Cui H, Wang J, Li Z, Li X, Zheng Z, Zhan Z, Liu H. Piezo1-mediated mechanotransduction promotes entheseal pathological new bone formation in ankylosing spondylitis. Ann Rheum Dis 2023; 82:533-545. [PMID: 36543525 DOI: 10.1136/ard-2022-223428] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The aim of this study was to identify the role of Piezo1-mediated mechanotransduction in entheseal pathological new bone formation and to explore the underlying molecular mechanism. METHODS Spinal ligament tissues were collected from 14 patients with ankylosing spondylitis (AS) and 14 non-AS controls and bulk RNA sequencing was conducted. Collagen antibody-induced arthritis models were established to observe pathological new bone formation. Pharmacological inhibition and genetic ablation of Piezo1 was performed in animal models to identify the essential role of Piezo1. Entheseal osteo-chondral lineage cells were collected and in vitro cell culture system was established to study the role and underlying mechanism of Piezo1 in regulation of chondrogenesis, osteogenesis and its own expression. RESULTS Piezo1 was aberrantly upregulated in ligaments and entheseal tissues from patients with AS and animal models. Pharmaceutical and genetic inhibition of Piezo1 attenuated while activation of Piezo1 promoted pathological new bone formation. Mechanistically, activation of CaMKII (Calcium/calmodulin dependent protein kinase II) signalling was found essential for Piezo1-mediated mechanotransduction. In addition, Piezo1 was upregulated by AS-associated inflammatory cytokines. CONCLUSION Piezo1-mediated mechanotransduction promotes entheseal pathological new bone formation through CaMKII signalling in AS.
Collapse
Affiliation(s)
- Siwen Chen
- Department of Spine Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, Guangdong, China
| | - Zihao Li
- Department of Spine Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, Guangdong, China
| | - Dongying Chen
- Deparment of Rheumatology and Immunology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Haowen Cui
- Department of Spine Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, Guangdong, China
| | - Jianru Wang
- Department of Spine Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, Guangdong, China
| | - Zemin Li
- Department of Spine Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, Guangdong, China
| | - Xiang Li
- Department of Spine Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, Guangdong, China
| | - Zhaomin Zheng
- Department of Spine Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, Guangdong, China
| | - Zhongping Zhan
- Deparment of Rheumatology and Immunology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Hui Liu
- Department of Spine Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, Guangdong, China
| |
Collapse
|
37
|
Abstract
Immune responses are governed by signals from the tissue microenvironment, and in addition to biochemical signals, mechanical cues and forces arising from the tissue, its extracellular matrix and its constituent cells shape immune cell function. Indeed, changes in biophysical properties of tissue alter the mechanical signals experienced by cells in many disease conditions, in inflammatory states and in the context of ageing. These mechanical cues are converted into biochemical signals through the process of mechanotransduction, and multiple pathways of mechanotransduction have been identified in immune cells. Such pathways impact important cellular functions including cell activation, cytokine production, metabolism, proliferation and trafficking. Changes in tissue mechanics may also represent a new form of 'danger signal' that alerts the innate and adaptive immune systems to the possibility of injury or infection. Tissue mechanics can change temporally during an infection or inflammatory response, offering a novel layer of dynamic immune regulation. Here, we review the emerging field of mechanoimmunology, focusing on how mechanical cues at the scale of the tissue environment regulate immune cell behaviours to initiate, propagate and resolve the immune response.
Collapse
|
38
|
Immunoregulatory Role of the Mechanosensitive Ion Channel Piezo1 in Inflammation and Cancer. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010213. [PMID: 36615408 PMCID: PMC9822220 DOI: 10.3390/molecules28010213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/22/2022] [Accepted: 12/25/2022] [Indexed: 12/28/2022]
Abstract
Piezo1 was originally identified as a mechanically activated, nonselective cation ion channel, with significant permeability to calcium ions, is evolutionally conserved, and is involved in the proliferation and development of various types of cells, in the context of various types of mechanical or innate stimuli. Recently, our study and work by others have reported that Piezo1 from all kinds of immune cells is involved in regulating many diseases, including infectious inflammation and cancer. This review summarizes the recent progress made in understanding the immunoregulatory role and mechanisms of the mechanical receptor Piezo1 in inflammation and cancer and provides new insight into the biological significance of Piezo1 in regulating immunity and tumors.
Collapse
|
39
|
Yoon CW, Pan Y, Wang Y. The application of mechanobiotechnology for immuno-engineering and cancer immunotherapy. Front Cell Dev Biol 2022; 10:1064484. [PMID: 36483679 PMCID: PMC9725026 DOI: 10.3389/fcell.2022.1064484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/08/2022] [Indexed: 11/24/2022] Open
Abstract
Immune-engineering is a rapidly emerging field in the past few years, as immunotherapy evolved from a paradigm-shifting therapeutic approach for cancer treatment to promising immuno-oncology models in clinical trials and commercial products. Linking the field of biomedical engineering with immunology, immuno-engineering applies engineering principles and utilizes synthetic biology tools to study and control the immune system for diseases treatments and interventions. Over the past decades, there has been a deeper understanding that mechanical forces play crucial roles in regulating immune cells at different stages from antigen recognition to actual killing, which suggests potential opportunities to design and tailor mechanobiology tools to novel immunotherapy. In this review, we first provide a brief introduction to recent technological and scientific advances in mechanobiology for immune cells. Different strategies for immuno-engineering are then discussed and evaluated. Furthermore, we describe the opportunities and challenges of applying mechanobiology and related technologies to study and engineer immune cells and ultimately modulate their function for immunotherapy. In summary, the synergetic integration of cutting-edge mechanical biology techniques into immune-engineering strategies can provide a powerful platform and allow new directions for the field of immunotherapy.
Collapse
Affiliation(s)
- Chi Woo Yoon
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Yijia Pan
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
40
|
Liu H, Zhao H, Sun Y. Tumor microenvironment and cellular senescence: Understanding therapeutic resistance and harnessing strategies. Semin Cancer Biol 2022; 86:769-781. [PMID: 34799201 DOI: 10.1016/j.semcancer.2021.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/24/2021] [Accepted: 11/08/2021] [Indexed: 01/27/2023]
Abstract
The tumor microenvironment (TME) is a major contributor to cancer malignancy including development of therapeutic resistance, a process mediated in part through intercellular crosstalk. Besides diverse soluble factors responsible for pro-survival pathway activation, immune evasion and extracellular matrix (ECM) remodeling further promote cancer resistance. Importantly, therapy-induced senescence (TIS) of cells in the TME is frequently observed in anticancer regimens, an off-target effect that can generate profound impacts on disease progression. By conferring the resistance and fueling the repopulation of remaining cancerous cells, TIS is responsible for tumor relapse and distant metastasis in posttreatment stage. This pathological trajectory can be substantially driven by the pro-inflammatory feature of senescent cells, termed as the senescence-associated secretory phenotype (SASP). Targeting strategies to selectively and efficiently remove senescent cells before they exert non-autonomous but largely deleterious effects, are emerging as an effective solution to prevent drug resistance acquired from a treatment-remodeled TME. In this review, we summarize the TME composition and key activities that affect tissue homeostasis and support treatment resistance. Promising opportunities that allow TME-manipulation and senescent cell-targeting (senotherapy) are discussed, with translational pipelines to overcome therapeutic barriers in clinical oncology projected.
Collapse
Affiliation(s)
- Hanxin Liu
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Huifang Zhao
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Yu Sun
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, 264003, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China; Department of Medicine and VAPSHCS, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
41
|
Shahidullah M, Rosales JL, Delamere N. Activation of Piezo1 Increases Na,K-ATPase-Mediated Ion Transport in Mouse Lens. Int J Mol Sci 2022; 23:12870. [PMID: 36361659 PMCID: PMC9656371 DOI: 10.3390/ijms232112870] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 10/31/2023] Open
Abstract
Lens ion homeostasis depends on Na,K-ATPase and NKCC1. TRPV4 and TRPV1 channels, which are mechanosensitive, play important roles in mechanisms that regulate the activity of these transporters. Here, we examined another mechanosensitive channel, piezo1, which is also expressed in the lens. The purpose of the study was to examine piezo1 function. Recognizing that activation of TRPV4 and TRPV1 causes changes in lens ion transport mechanisms, we carried out studies to determine whether piezo1 activation changes either Na,K-ATPase-mediated or NKCC1-mediated ion transport. We also examined channel function of piezo1 by measuring calcium entry. Rb uptake was measured as an index of inwardly directed potassium transport by intact mouse lenses. Intracellular calcium concentration was measured in Fura-2 loaded cells by a ratiometric imaging technique. Piezo1 immunolocalization was most evident in the lens epithelium. Potassium (Rb) uptake was increased in intact lenses as well as in cultured lens epithelium exposed to Yoda1, a piezo1 agonist. The majority of Rb uptake is Na,K-ATPase-dependent, although there also is a significant NKCC-dependent component. In the presence of ouabain, an Na,K-ATPase inhibitor, Yoda1 did not increase Rb uptake. In contrast, Yoda1 increased Rb uptake to a similar degree in the presence or absence of 1 µM bumetanide, an NKCC inhibitor. The Rb uptake response to Yoda1 was inhibited by the selective piezo1 antagonist GsMTx4, and also by the nonselective antagonists ruthenium red and gadolinium. In parallel studies, Yoda1 was observed to increase cytoplasmic calcium concentration in cells loaded with Fura-2. The calcium response to Yoda1 was abolished by gadolinium or ruthenium red. The calcium and Rb uptake responses to Yoda1 were absent in calcium-free bathing solution, consistent with calcium entry when piezo1 is activated. Taken together, these findings point to stimulation of Na,K-ATPase, but not NKCC, when piezo1 is activated. Na,K-ATPase is the principal mechanism responsible for ion and water homeostasis in the lens. The functional role of lens piezo1 is a topic for further study.
Collapse
Affiliation(s)
- Mohammad Shahidullah
- Department of Physiology, University of Arizona, 1501 N Campbell Avenue, Tucson, AZ 85724, USA
- Department of Ophthalmology and Vision Science, University of Arizona, 1501 N Campbell Avenue, Tucson, AZ 85724, USA
| | - Joaquin Lopez Rosales
- Department of Physiology, University of Arizona, 1501 N Campbell Avenue, Tucson, AZ 85724, USA
| | - Nicholas Delamere
- Department of Physiology, University of Arizona, 1501 N Campbell Avenue, Tucson, AZ 85724, USA
- Department of Ophthalmology and Vision Science, University of Arizona, 1501 N Campbell Avenue, Tucson, AZ 85724, USA
| |
Collapse
|
42
|
Yang K, He X, Wu Z, Yin Y, Pan H, Zhao X, Sun T. The emerging roles of piezo1 channels in animal models of multiple sclerosis. Front Immunol 2022; 13:976522. [PMID: 36177027 PMCID: PMC9513475 DOI: 10.3389/fimmu.2022.976522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/24/2022] [Indexed: 11/23/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory, demyelinating, and neurodegenerative disease in the central nervous system (CNS). Its pathogenesis is quite complex: Accumulated evidence suggests that biochemical signals as well as mechanical stimuli play important roles in MS. In both patients and animal models of MS, brain viscoelasticity is reduced during disease progression. Piezo mechanosensitive channels are recently discovered, and their three-dimensional structure has been solved. Both the membrane dome mechanism and the membrane footprint hypothesis have been proposed to explain their mechanosensitivity. While membrane-mediated forces alone appear to be sufficient to induce Piezo gating, tethers attached to the membrane or to the channel itself also seem to play a role. Current research indicates that Piezo1 channels play a key role in multiple aspects of MS pathogenesis. Activation of Piezo1 channels in axon negatively regulates CNS myelination. in addition, the inhibition of Piezo1 in CD4+ T cells and/or T regulatory cells (Treg) attenuates experimental autoimmune encephalitis (EAE) symptoms. Although more work has to be done to clarify the roles of Piezo1 channels in MS, they might be a promising future drug target for MS treatment.
Collapse
Affiliation(s)
- Kai Yang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
- *Correspondence: Kai Yang, ; Taolei Sun,
| | - Xueai He
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Zhengqi Wu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Yimeng Yin
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Hanyu Pan
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Xinyue Zhao
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
- *Correspondence: Kai Yang, ; Taolei Sun,
| |
Collapse
|
43
|
Huang J, Zhang K, Du R, Liu W, Zhang H, Tian T, Wang Y, Wang G, Yin T. The Janus-faced role of Piezo1 in cardiovascular health under mechanical stimulation. Genes Dis 2022. [PMID: 37492728 PMCID: PMC10363580 DOI: 10.1016/j.gendis.2022.08.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
In recent years, cardiovascular health problems are becoming more and more serious. At the same time, mechanical stimulation closely relates to cardiovascular health. In this context, Piezo1, which is very sensitive to mechanical stimulation, has attracted our attention. Here, we review the critical significance of Piezo1 in mechanical stimulation of endothelial cells, NO production, lipid metabolism, DNA damage protection, the development of new blood vessels and maturation, narrowing of blood vessels, blood pressure regulation, vascular permeability, insulin sensitivity, and maintenance of red blood cell function. Besides, Piezo1 may participate in the occurrence and development of atherosclerosis, diabetes, hypertension, and other cardiovascular diseases. It is worth noting that Piezo1 has dual effects on maintaining cardiovascular health. On the one hand, the function of Piezo1 is necessary to maintain cardiovascular health; on the other hand, under some extreme mechanical stimulation, the overexpression of Piezo1 may bring adverse factors such as inflammation. Therefore, this review discusses the Janus-faced role of Piezo1 in maintaining cardiovascular health and puts forward new ideas to provide references for gene therapy or nanoagents targeting Piezo1.
Collapse
|
44
|
Wang Y, Yang H, Jia A, Wang Y, Yang Q, Dong Y, Hou Y, Cao Y, Dong L, Bi Y, Liu G. Dendritic cell Piezo1 stimulated by mechanical stiffness or inflammatory signals directs the differentiation of T H1 and T reg cells in cancer. eLife 2022; 11:79957. [PMID: 35993548 PMCID: PMC9451538 DOI: 10.7554/elife.79957] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/20/2022] [Indexed: 11/13/2022] Open
Abstract
Dendritic cells (DCs) play an important role in anti-tumor immunity by inducing T cell differentiation. Herein, we found that the DC mechanical sensor Piezo1 stimulated by mechanical stiffness or inflammatory signals directs the reciprocal differentiation of TH1 and regulatory T (Treg) cells in cancer. Genetic deletion of Piezo1 in DCs inhibited the generation of TH1 cells while driving the development of Treg cells in promoting cancer growth in mice. Mechanistically, Piezo1-deficient DCs regulated the secretion of the polarizing cytokines TGFβ1 and IL-12, leading to increased TGFβR2-p-Smad3 activity and decreased IL-12Rβ2-p-STAT4 activity while inducing the reciprocal differentiation of Treg and TH1 cells. In addition, Piezo1 integrated the SIRT1-hypoxia-inducible factor-1 alpha (HIF1α)-dependent metabolic pathway and calcium-calcineurin-NFAT signaling pathway to orchestrate reciprocal TH1 and Treg lineage commitment through DC-derived IL-12 and TGFβ1. Our studies provide critical insight for understanding the role of the DC-based mechanical regulation of immunopathology in directing T cell lineage commitment in tumor microenvironments.
Collapse
Affiliation(s)
| | - Hui Yang
- Fudan University, Shanghai, China
| | - Anna Jia
- Beijing Normal University, Beijing, China
| | - Yufei Wang
- Beijing Normal University, Beijing, China
| | - Qiuli Yang
- Beijing Normal University, Beijing, China
| | | | - Yueru Hou
- Beijing Normal University, Beijing, China
| | - Yejin Cao
- Beijing Normal University, Beijing, China
| | - Lin Dong
- Beijing Normal University, Beijing, China
| | - Yujing Bi
- Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | | |
Collapse
|
45
|
Cahalan MD. Voltage-gated Ca 2+ channel proteins do not function as ion channels in T cells. Sci Signal 2022; 15:eabq5594. [PMID: 35700266 DOI: 10.1126/scisignal.abq5594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Calcium (Ca2+) signaling has long been known to be crucial for T cell activation. Erdogmus et al. tested the function of voltage-gated Ca2+ channel (CaV) proteins and discovered a nonchannel function mediated by an accessory subunit but found no evidence for CaV channel activity in T cells.
Collapse
Affiliation(s)
- Michael D Cahalan
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA.,Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
46
|
Li X, Hu J, Zhao X, Li J, Chen Y. Piezo channels in the urinary system. Exp Mol Med 2022; 54:697-710. [PMID: 35701561 PMCID: PMC9256749 DOI: 10.1038/s12276-022-00777-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/25/2022] [Accepted: 02/16/2022] [Indexed: 12/24/2022] Open
Abstract
The Piezo channel family, including Piezo1 and Piezo2, includes essential mechanosensitive transduction molecules in mammals. Functioning in the conversion of mechanical signals to biological signals to regulate a plethora of physiological processes, Piezo channels, which have a unique homotrimeric three-blade propeller-shaped structure, utilize a cap-motion and plug-and-latch mechanism to gate their ion-conducting pathways. Piezo channels have a wide range of biological roles in various human systems, both in vitro and in vivo. Currently, there is a lack of comprehensive understanding of their antagonists and agonists, and therefore further investigation is needed. Remarkably, increasingly compelling evidence demonstrates that Piezo channel function in the urinary system is important. This review article systematically summarizes the existing evidence of the importance of Piezo channels, including protein structure, mechanogating mechanisms, and pharmacological characteristics, with a particular focus on their physiological and pathophysiological roles in the urinary system. Collectively, this review aims to provide a direction for future clinical applications in urinary system diseases.
Collapse
Affiliation(s)
- Xu Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Junwei Hu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Xuedan Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Juanjuan Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yuelai Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
47
|
Velasco-Estevez M, Koch N, Klejbor I, Caratis F, Rutkowska A. Mechanoreceptor Piezo1 Is Downregulated in Multiple Sclerosis Brain and Is Involved in the Maturation and Migration of Oligodendrocytes in vitro. Front Cell Neurosci 2022; 16:914985. [PMID: 35722613 PMCID: PMC9204635 DOI: 10.3389/fncel.2022.914985] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
Mechanical properties of the brain such as intracranial pressure or stiffness of the matrix play an important role in the brain’s normal physiology and pathophysiology. The physical properties are sensed by the cells through mechanoreceptors and translated into ion currents which activate multiple biochemical cascades allowing the cells to adapt and respond to changes in their microenvironment. Piezo1 is one of the first identified mechanoreceptors. It modulates various central nervous system functions such as axonal growth or activation of astrocytes. Piezo1 signaling was also shown to play a role in the pathophysiology of Alzheimer’s disease. Here, we explore the expression of the mechanoreceptor Piezo1 in human MO3.13 oligodendrocytes and human MS/non-MS patients’ brains and investigate its putative effects on oligodendrocyte proliferation, maturation, and migration. We found that Piezo1 is expressed in human oligodendrocytes and oligodendrocyte progenitor cells in the human brain and that its inhibition with GsMTx4 leads to an increment in proliferation and migration of MO3.13 oligodendrocytes. Activation of Piezo1 with Yoda-1 induced opposite effects. Further, we observed that expression of Piezo1 decreased with MO3.13 maturation in vitro. Differences in expression were also observed between healthy and multiple sclerosis brains. Remarkably, the data showed significantly lower expression of Piezo1 in the white matter in multiple sclerosis brains compared to its expression in the white matter in healthy controls. There were no differences in Piezo1 expression between the white matter plaque and healthy-appearing white matter in the multiple sclerosis brain. Taken together, we here show that Piezo1-induced signaling can be used to modulate oligodendrocyte function and that it may be an important player in the pathophysiology of multiple sclerosis.
Collapse
Affiliation(s)
- Maria Velasco-Estevez
- H12O-CNIO Hematological Malignancies Group, Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Nina Koch
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Ilona Klejbor
- Department of Anatomy, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Fionä Caratis
- Department of Anatomy and Neurobiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Rutkowska
- Department of Anatomy and Neurobiology, Medical University of Gdańsk, Gdańsk, Poland
- *Correspondence: Aleksandra Rutkowska,
| |
Collapse
|
48
|
Tang H, Zeng R, He E, Zhang I, Ding C, Zhang A. Piezo-Type Mechanosensitive Ion Channel Component 1 (Piezo1): A Promising Therapeutic Target and Its Modulators. J Med Chem 2022; 65:6441-6453. [DOI: 10.1021/acs.jmedchem.2c00085] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Hairong Tang
- Pharm-X Center, Laboratory of Medicinal Chemical Biology & Frontiers on Drug Discovery (RLMCBFDD), School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ruoqing Zeng
- Pharm-X Center, Laboratory of Medicinal Chemical Biology & Frontiers on Drug Discovery (RLMCBFDD), School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ende He
- Pharm-X Center, Laboratory of Medicinal Chemical Biology & Frontiers on Drug Discovery (RLMCBFDD), School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | | | - Chunyong Ding
- Pharm-X Center, Laboratory of Medicinal Chemical Biology & Frontiers on Drug Discovery (RLMCBFDD), School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ao Zhang
- Pharm-X Center, Laboratory of Medicinal Chemical Biology & Frontiers on Drug Discovery (RLMCBFDD), School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Lingang National Laboratory, Shanghai 200210,China
| |
Collapse
|
49
|
Swain SM, Romac JMJ, Vigna SR, Liddle RA. Piezo1-mediated stellate cell activation causes pressure-induced pancreatic fibrosis in mice. JCI Insight 2022; 7:158288. [PMID: 35451372 PMCID: PMC9089793 DOI: 10.1172/jci.insight.158288] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/02/2022] [Indexed: 11/17/2022] Open
Abstract
Pancreatic fibrosis is a complication of chronic pancreatitis and is a prominent feature of pancreatic cancer. Pancreatic fibrosis is commonly observed in patients with prolonged pancreatic duct obstruction, which elevates intrapancreatic pressure. We show here that increased pancreatic duct pressure causes fibrosis and describes the mechanism by which pressure increases deposition of extracellular matrix proteins and fibrosis. We found that pancreatic stellate cells (PSCs), the source of the extracellular matrix proteins in fibrosis, express the mechanically activated ion channel Piezo1. By increasing intracellular calcium, mechanical stress or the Piezo1 agonist Yoda1-activated PSCs manifest by loss of perinuclear fat droplets and increased TGF-β1, fibronectin, and type I collagen expression. These effects were blocked by the Piezo1 inhibitor GsMTx4 and absent in PSCs from mice with conditional genetic deletion of Piezo1 in stellate cells, as was pancreatic duct ligation-induced fibrosis. Although TRPV4 has been proposed to have direct mechanosensing properties, we discovered that PSCs from Trpv4-KO mice were protected against Yoda1-triggered activation. Moreover, mice devoid of TRPV4 were protected from pancreatic duct ligation-induced fibrosis. Thus, high pressure within the pancreas stimulates Piezo1 channel opening, and subsequent activation of TRPV4 leads to stellate cell activation and pressure-induced chronic pancreatitis and fibrosis.
Collapse
Affiliation(s)
- Sandip M Swain
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Joelle M-J Romac
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Steven R Vigna
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Rodger A Liddle
- Department of Medicine, Duke University, Durham, North Carolina, USA.,Department of Veterans Affairs Healthcare System, Durham, North Carolina, USA
| |
Collapse
|
50
|
Targeting extracellular matrix stiffness and mechanotransducers to improve cancer therapy. J Hematol Oncol 2022; 15:34. [PMID: 35331296 PMCID: PMC8943941 DOI: 10.1186/s13045-022-01252-0] [Citation(s) in RCA: 171] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer microenvironment is critical for tumorigenesis and cancer progression. The extracellular matrix (ECM) interacts with tumor and stromal cells to promote cancer cells proliferation, migration, invasion, angiogenesis and immune evasion. Both ECM itself and ECM stiffening-induced mechanical stimuli may activate cell membrane receptors and mechanosensors such as integrin, Piezo1 and TRPV4, thereby modulating the malignant phenotype of tumor and stromal cells. A better understanding of how ECM stiffness regulates tumor progression will contribute to the development of new therapeutics. The rapidly expanding evidence in this research area suggests that the regulators and effectors of ECM stiffness represent potential therapeutic targets for cancer. This review summarizes recent work on the regulation of ECM stiffness in cancer, the effects of ECM stiffness on tumor progression, cancer immunity and drug resistance. We also discuss the potential targets that may be druggable to intervene ECM stiffness and tumor progression. Based on these advances, future efforts can be made to develop more effective and safe drugs to interrupt ECM stiffness-induced oncogenic signaling, cancer progression and drug resistance.
Collapse
|