1
|
Nagi JS, Doiron AL. 20 nm nanoparticles trigger calcium influx to endothelial cells via a TRPV4 channel. Biomater Sci 2025; 13:2728-2743. [PMID: 40192740 DOI: 10.1039/d4bm01691b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
While increased intracellular calcium (Ca2+) has been identified as a key effect of nanoparticles on endothelial cells, the mechanism has not been fully elucidated or examined under shear stress. Here, we show the effect of several types of 20 nm particles on Ca2+ in the presence of shear stress in human umbilical vein endothelial cells (HUVECs), human coronary artery endothelial cells (HCAECs), and human cardiac microvascular endothelial cells (HMVEC-Cs). Intracellular Ca2+ levels increased by nearly three-fold in these cell types upon exposure to 100 μg mL-1 20 nm Au particles, which was not seen in response to larger or smaller particles. An antagonist to the calcium channel - transient receptor potential vanilloid-type 4 (TRPV4) - drastically reduced the amount of calcium by 9.3-fold in HUVECs exposed to 0.6 Pa shear stress and 100 μg mL-1 20 nm gold particles, a trend upheld in both HCAECs and HMVEC-Cs. Cell alignment in the direction of fluid flow is a well-known phenomenon in endothelial cells, and interestingly, cells in the presence of 20 nm particles with fluid flow had a higher alignment index than cells in the fluid flow alone. When compared with previous works, these results indicated that 20 nm particles may be inducing endothelial permeability by activating the TRPV4 channel in vitro. The potential of nanoparticle delivery technologies hinges on an improved understanding of this effect toward improved delivery with limited toxicity.
Collapse
Affiliation(s)
- Jaspreet Singh Nagi
- Department of Electrical and Biomedical Engineering, University of Vermont, Burlington, VT 05405, USA.
| | - Amber L Doiron
- Department of Electrical and Biomedical Engineering, University of Vermont, Burlington, VT 05405, USA.
| |
Collapse
|
2
|
Mocciaro E, Kidd M, Johnson K, Bishop E, Johnson K, Zeng YP, Perrotta C, Micci MA. Mechanosensitive ion channel Piezo1 modulates the response of rat hippocampus neural stem cells to rapid stretch injury. PLoS One 2025; 20:e0323191. [PMID: 40359437 PMCID: PMC12074584 DOI: 10.1371/journal.pone.0323191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
Traumatic brain injury (TBI) is one of the primary causes of long-term brain disabilities among military personnel and civilians, regardless of gender. A plethora of secondary events are triggered by a primary brain insult, increasing the complexity of TBI. One of the most affected brain regions is the hippocampus, where neurogenesis occurs throughout life due to the presence of neural stem cells (NSC). Preclinical models have been extensively used to better understand TBI and develop effective treatments. Among these, rapid stretch injury has been used to mimic the effect of mechanical stress produced by a TBI on neurons and glia in vitro. In this study, we aimed to determine the impact of rapid stretch on the viability, proliferation, and differentiation of NSC isolated from rat hippocampus (Hipp-NSC) and to determine the role of the stretch-activated ion channel Piezo-1 in modulating their response to mechanical stress. We found that while rapid stretch (30 and 50 PSI) reduced Hipp-NSC viability (measured as a function of LDH release), it did not change their proliferation and differentiation potentials. Interestingly, rapid stretch in the presence of a selective Piezo-1 inhibitor, GsMTx4, or Piezo1 targeting siRNA, directed Hipp-NSC differentiation toward a neurogenic lineage. Additionally, we found that inhibiting Piezo1 with the addition of a rapid stretch injury increased the expression of miRNAs known to regulate neurogenesis. This work uses a novel approach for studying the effect of mechanical stress on NSC in vitro and points to the critical role the stretch-activated ion channel Piezo-1 has in modulating the impact of TBI on hippocampal neurogenesis.
Collapse
Affiliation(s)
- Emanuele Mocciaro
- Gene Expression Regulation Unit, San Raffaele Scientific Institute, Milan, Italy
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Madison Kidd
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kevin Johnson
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Elizabeth Bishop
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kathia Johnson
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Ya Ping Zeng
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Cristiana Perrotta
- Gene Expression Regulation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Maria-Adelaide Micci
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
3
|
Lee LW, Lee GH, Su IH, Lu CH, Lin KH, Wen FL, Tang MJ. Mechanobiological mechanism of cyclic stretch-induced cell columnarization. Cell Rep 2025; 44:115662. [PMID: 40338742 DOI: 10.1016/j.celrep.2025.115662] [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: 08/23/2024] [Revised: 01/24/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025] Open
Abstract
In vivo, epithelial cells maintain structural integrity under dynamic mechanical perturbations. To study this, we treated various epithelial cell lines with long-term cyclic stretch (CS). Surprisingly, cells transitioned from cuboidal to columnar shape (columnarization) in MDCK cells, while others only elongated. This change correlated with actin accumulation at the top and stress fiber realignment at the bottom. Blocking mechanical stimulation via FAK inhibition or reducing vinculin partially prevented columnarization; however, disrupting tight junctions or cellular contractility substantially blocked it. The MK4 cells, derived from MDCK cells with weaker cell-cell junctions, showed less columnarization under CS, whereas overexpressing Caveolin-1 (Cav1) in MK4 cells enhanced junctions and promoted columnar formation. Atomic force microscopy studies revealed increased apical junctional stiffness in both CS-treated MDCK and Cav1-overexpressing MK4 cells. This, combined with a mathematical model, elucidated the physical characteristics and changes in cell tension post-stretch, revealing the mechanobiological foundation of epithelial cell columnarization.
Collapse
Affiliation(s)
- Lun-Wei Lee
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan 70101, Taiwan
| | - Gang-Hui Lee
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan 70101, Taiwan
| | - I-Hsiu Su
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan 70101, Taiwan
| | - Chia-Hsuan Lu
- Department of Computer Science, University of Oxford, Wolfson Building, Parks Road, Oxford OXI 3QD, UK
| | - Keng-Hui Lin
- Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
| | - Fu-Lai Wen
- Department of Physics and Center for Complex Systems, National Central University, Taoyuan 32001, Taiwan; Physics Division, National Center for Theoretical Sciences, Taipei 10617, Taiwan.
| | - Ming-Jer Tang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
4
|
Mhalhel K, Cavallaro M, Pansera L, Franco GA, Montalbano G, Laurà R, Abbate F, Germanà A, Levanti M, Aragona M. Ion-Channel Proteins in the Prepubertal Bitch Reproductive System: The Immunolocalization of ASIC2, ASIC4, and PIEZO2. Int J Mol Sci 2025; 26:4388. [PMID: 40362625 PMCID: PMC12072602 DOI: 10.3390/ijms26094388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 04/21/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Ion channels play a crucial role in various physiological processes, yet their functions in the reproductive system remain underexplored. This study investigates the expression and the localization of ASIC2, ASIC4, and PIEZO2 ion channels in the reproductive tracts of prepubertal bitches. Western blotting on samples from eight prepubertal bitches confirmed the presence of these ion channels in ovarian, uterine, and uterine tubes tissues, and validated antibody specificity. Immunohistochemistry revealed that all primordial follicles expressed these ion channels, while only some developing follicles showed immunolabeling. These findings suggest ion channels' potential involvement in oocyte differentiation and maturation. The localization of these channels in uterine tubes, uterine lining, and glandular epithelium suggests a role in tissue maintenance, oocyte transport, and embryo implantation. Additionally, their expression in the tunica media of reproductive vasculature points to a potential role in vascular regulation. Future studies are needed to elucidate the specific mechanisms underlying the role of these channels in reproductive physiology.
Collapse
Affiliation(s)
- Kamel Mhalhel
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, Polo Universitario dell’ Annunziata, 98168 Messina, Italy; (K.M.); (M.C.); (L.P.); (R.L.); (F.A.); (A.G.); (M.L.)
| | - Mauro Cavallaro
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, Polo Universitario dell’ Annunziata, 98168 Messina, Italy; (K.M.); (M.C.); (L.P.); (R.L.); (F.A.); (A.G.); (M.L.)
| | - Lidia Pansera
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, Polo Universitario dell’ Annunziata, 98168 Messina, Italy; (K.M.); (M.C.); (L.P.); (R.L.); (F.A.); (A.G.); (M.L.)
| | - Gianluca Antonio Franco
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell’ Annunziata, 98168 Messina, Italy;
| | - Giuseppe Montalbano
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, Polo Universitario dell’ Annunziata, 98168 Messina, Italy; (K.M.); (M.C.); (L.P.); (R.L.); (F.A.); (A.G.); (M.L.)
| | - Rosaria Laurà
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, Polo Universitario dell’ Annunziata, 98168 Messina, Italy; (K.M.); (M.C.); (L.P.); (R.L.); (F.A.); (A.G.); (M.L.)
| | - Francesco Abbate
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, Polo Universitario dell’ Annunziata, 98168 Messina, Italy; (K.M.); (M.C.); (L.P.); (R.L.); (F.A.); (A.G.); (M.L.)
| | - Antonino Germanà
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, Polo Universitario dell’ Annunziata, 98168 Messina, Italy; (K.M.); (M.C.); (L.P.); (R.L.); (F.A.); (A.G.); (M.L.)
| | - Maria Levanti
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, Polo Universitario dell’ Annunziata, 98168 Messina, Italy; (K.M.); (M.C.); (L.P.); (R.L.); (F.A.); (A.G.); (M.L.)
| | - Marialuisa Aragona
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, Polo Universitario dell’ Annunziata, 98168 Messina, Italy; (K.M.); (M.C.); (L.P.); (R.L.); (F.A.); (A.G.); (M.L.)
| |
Collapse
|
5
|
Knoepp F, Abid S, Houssaini A, Lipskaia L, Gökyildirim MY, Born E, Marcos E, Arhatte M, Glogowska E, Vienney N, Günther A, Kraut S, Breitenborn-Mueller I, Quanz K, Fenner-Nau D, Derumeaux G, Weissmann N, Honoré E, Adnot S. Piezo1 in PASMCs: Critical for Hypoxia-Induced Pulmonary Hypertension Development. Circ Res 2025; 136:1031-1048. [PMID: 40181773 PMCID: PMC12036789 DOI: 10.1161/circresaha.124.325475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/25/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a life-threatening and progressive yet incurable disease. The hallmarks of PH comprise (1) sustained contraction and (2) excessive proliferation of pulmonary arterial smooth muscle cells (PASMCs). A major stimulus to which PASMCs are exposed during PH development is altered mechanical stress, originating from increased blood pressure, changes in blood flow velocity, and a progressive stiffening of pulmonary arteries. Mechanosensitive ion channels, including Piezo1 (Piezo-type mechanosensitive ion channel component-1), perceive such mechanical stimuli and translate them into a variety of cellular responses, including contractility or proliferation. Thus, the objective of the present study was to elucidate the specific role of Piezo1 in PASMCs for PH development and progression. METHODS The cell-type specific function of Piezo1 in PH was assessed in (1) PASMCs and lung tissues from patients with PH and (2) 2 mouse strains characterized by smooth muscle cell-specific, conditional Piezo1 knockout. Taking advantage of these strains, the smooth muscle cell-specific role of Piezo1 in PH development and progression was assessed in isolated, perfused, and ventilated mouse lungs, wire myography, and proliferation assays. Finally, in vivo function of smooth muscle cell-specific Piezo1 knockout was evaluated upon induction of chronic hypoxia-induced PH in these mice with insights into pulmonary vascular cell senescence. RESULTS Compared with healthy controls, PASMCs from patients with PH featured an elevated Piezo1 expression and increased proliferative phenotype. Smooth muscle cell-specific Piezo1 deletion, as confirmed via quantitative real-time polymerase chain reaction and patch clamp recordings, prevented the hypoxia-induced increase in PASMC proliferation in mice. Moreover, Piezo1 knockout reduced hypoxic pulmonary vasoconstriction in isolated, perfused, and ventilated mouse lungs, endothelial-denuded pulmonary arteries, and hemodynamic measurements in vivo. Consequently, Piezo1-deficient mice were considerably protected against chronic hypoxia-induced PH development with ameliorated right heart hypertrophy and improved hemodynamic function. In addition, distal pulmonary capillaries were preserved in the Piezo1-knockout mice, associated with a lower number of senescent endothelial cells. CONCLUSIONS This study provides evidence that Piezo1 expressed in PASMCs is critically involved in the pathogenesis of PH by controlling pulmonary vascular tone, arterial remodeling, and associated lung capillary rarefaction due to endothelial cell senescence.
Collapse
MESH Headings
- Animals
- Ion Channels/genetics
- Ion Channels/metabolism
- Ion Channels/deficiency
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/genetics
- Hypoxia/complications
- Hypoxia/metabolism
- Mice, Knockout
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice
- Cell Proliferation
- Male
- Cells, Cultured
- Mice, Inbred C57BL
- Female
- Vascular Remodeling
- Disease Models, Animal
Collapse
Affiliation(s)
- Fenja Knoepp
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Shariq Abid
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
- Medical Research Center, Liaquat University of Medical and Health Sciences, Pakistan (S. Abid)
| | - Amal Houssaini
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
- Institute for Lung Health, Justus Liebig University, Giessen, Germany (A.H., M.Y.G., S. Adnot)
| | - Larissa Lipskaia
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
| | - Mira Yasemin Gökyildirim
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
- Institute for Lung Health, Justus Liebig University, Giessen, Germany (A.H., M.Y.G., S. Adnot)
| | - Emmanuelle Born
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
| | - Elisabeth Marcos
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
| | - Malika Arhatte
- Centre National de la Recherche Scientifique, Institut national de la santé et de la recherche médicale, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France (M.A., E.G., E.H.)
| | - Edyta Glogowska
- Centre National de la Recherche Scientifique, Institut national de la santé et de la recherche médicale, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France (M.A., E.G., E.H.)
| | - Nora Vienney
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
| | - Andreas Günther
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Simone Kraut
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Ingrid Breitenborn-Mueller
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Karin Quanz
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Dagmar Fenner-Nau
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Geneviève Derumeaux
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Eric Honoré
- Centre National de la Recherche Scientifique, Institut national de la santé et de la recherche médicale, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France (M.A., E.G., E.H.)
| | - Serge Adnot
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
- Institute for Lung Health, Justus Liebig University, Giessen, Germany (A.H., M.Y.G., S. Adnot)
| |
Collapse
|
6
|
Budde I, Schlichting A, Ing D, Schimmelpfennig S, Kuntze A, Fels B, Romac JMJ, Swain SM, Liddle RA, Stevens A, Schwab A, Pethő Z. Piezo1-induced durotaxis of pancreatic stellate cells depends on TRPC1 and TRPV4 channels. J Cell Sci 2025; 138:jcs263846. [PMID: 40019468 DOI: 10.1242/jcs.263846] [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: 04/26/2024] [Accepted: 02/23/2025] [Indexed: 03/01/2025] Open
Abstract
Pancreatic stellate cells (PSCs) are primarily responsible for producing the stiff tumor tissue in pancreatic ductal adenocarcinoma (PDAC). Thereby, PSCs generate a stiffness gradient between the healthy pancreas and the tumor. This gradient induces durotaxis, a form of directional cell migration driven by differential stiffness. However, the molecular sensors behind durotaxis are still unclear. To investigate the role of mechanosensitive ion channels in PSC durotaxis, we established a two-dimensional stiffness gradient mimicking PDAC. Using pharmacological and genetic methods, we investigated the contribution of the ion channels Piezo1, TRPC1 and TRPV4 in PSC durotaxis. We found that PSC migration towards a stiffer substrate is diminished by altering Piezo1 activity. Moreover, disrupting TRPC1 along with TRPV4 abolishes PSC durotaxis even when Piezo1 is functional. Our results demonstrate that optimal PSC durotaxis requires an intermediary level of ion channel activity, which we simulated via a numerically discretized mathematical model. These findings suggest that mechanosensitive Piezo1 channels detect the differential stiffness microenvironment. The resulting intracellular signals are amplified by TRPV4 and TRPC1 channels to guide efficient PSC durotaxis.
Collapse
Affiliation(s)
- Ilka Budde
- Institute of Physiology II , University of Münster, Robert-Koch Str. 27B, 48149 Münster, Germany
| | - André Schlichting
- Institute for Analysis and Numerics , University of Münster, Einsteinstr. 62, 48149 Münster, Germany
- Institute of Applied Analysis , University of Ulm, Helmholtzstraße 18, 89081 Ulm, Germany
| | - David Ing
- Institute of Physiology II , University of Münster, Robert-Koch Str. 27B, 48149 Münster, Germany
| | - Sandra Schimmelpfennig
- Institute of Physiology II , University of Münster, Robert-Koch Str. 27B, 48149 Münster, Germany
| | - Anna Kuntze
- Institute of Physiology II , University of Münster, Robert-Koch Str. 27B, 48149 Münster, Germany
- Institute of Applied Analysis , University of Ulm, Helmholtzstraße 18, 89081 Ulm, Germany
| | - Benedikt Fels
- Institute of Physiology II , University of Münster, Robert-Koch Str. 27B, 48149 Münster, Germany
- Gerhard-Domagk-Institute of Pathology , University of Münster, 48149 Münster, Germany
| | - Joelle M-J Romac
- Institute of Physiology, University of Lübeck, 23562 Lübeck, Germany
| | - Sandip M Swain
- Institute of Physiology, University of Lübeck, 23562 Lübeck, Germany
| | - Rodger A Liddle
- Institute of Physiology, University of Lübeck, 23562 Lübeck, Germany
| | - Angela Stevens
- Institute for Analysis and Numerics , University of Münster, Einsteinstr. 62, 48149 Münster, Germany
| | - Albrecht Schwab
- Institute of Physiology II , University of Münster, Robert-Koch Str. 27B, 48149 Münster, Germany
| | - Zoltán Pethő
- Institute of Physiology II , University of Münster, Robert-Koch Str. 27B, 48149 Münster, Germany
| |
Collapse
|
7
|
Bontempo A, Heidari A, Pastore MR, Madonia R, Sadik A, Schweizer M, Cayabyab M. Yoda1, a Piezo1 agonist, induced latent HIV reactivation associated with upregulation of CD3/TCR complex and HLA genes. RESEARCH SQUARE 2025:rs.3.rs-6208371. [PMID: 40297703 PMCID: PMC12036472 DOI: 10.21203/rs.3.rs-6208371/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
There is currently no cure for HIV because of the presence of latent viral reservoirs in people with HIV (PWH) on antiretroviral therapy (ART). Latency-reversing agents (LRAs) that can effectively reactivate and destroy latent HIV are being developed as a possible cure for HIV. Here, we identify Yoda1, a Piezo1 agonist, as a novel LRA. Yoda1 reactivated latent HIV in vitro ACH2 cells and ex vivo PBMCs from an HIV patient on ART. Yoda1 induced infectious virus production and HIV gene expression via Piezo1 activation and calcium signaling. Transcriptomic and proteomic analyses revealed a unique latent HIV reactivation pathway involving T cell activation, upregulation of TCR/CD3 and HLA genes, as well as modulation of host and viral transcription and translation that favors viral gene expression. These findings suggest further testing and development of Yoda1 as an effective LRA to reactivate latent HIV and destroy latent reservoirs for the cure of HIV.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mark Cayabyab
- Nova Southeastern University College of Dental Medicine
| |
Collapse
|
8
|
Fabiano AR, Newman MW, Dombroski JA, Rowland SJ, Knoblauch SV, Kusunose J, Gibson‐Corley KN, Kaufman BG, Ren L, Caskey CF, King MR. Applying Ultrasound to Mechanically and Noninvasively Sensitize Prostate Tumors to TRAIL-Mediated Apoptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412995. [PMID: 39976192 PMCID: PMC12005757 DOI: 10.1002/advs.202412995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/26/2025] [Indexed: 02/21/2025]
Abstract
Non-surgical and safe prostate cancer (PCa) therapies are in demand. Soluble tumor necrosis factor (TNF-α) related apoptosis inducing ligand (TRAIL), a cancer-specific drug, shows preclinical efficacy but has a short circulation half-life. This research has shown that physiological fluid shear stress activates mechanosensitive ion channels (MSCs), such as Piezo1, enhancing TRAIL-mediated apoptosis in cancer cells. Herein, noninvasive, focal ultrasound (FUS) is implemented to augment the pro-apoptotic effects of TRAIL. Using thermally safe FUS parameters, it is observed that TRAIL sensitivity increases with higher FUS pressure in PCa cells, mediated by Piezo1. This is confirmed by examining the effects of calcium chelation, MSC inhibitors, and PIEZO knockdown. In vivo, a multi-dose study with 10 min FUS exposure shows that 0 and 4-h intervals between TRAIL and FUS significantly reduce tumor burden, with an increase in apoptosis evident by enhanced cleaved-caspase 3 expression. This mechanotherapy offers a clinically translatable approach by utilizing widely available FUS technology, applicable to treat additional cancer types.
Collapse
Affiliation(s)
- Abigail R. Fabiano
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
- Department of BioengineeringRice UniversityHoustonTX77005USA
| | - Malachy W. Newman
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Jenna A. Dombroski
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Schyler J. Rowland
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | | | - Jiro Kusunose
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTN37235USA
| | - Katherine N. Gibson‐Corley
- Department of PathologyMicrobiology and ImmunologyDivision of Comparative MedicineVanderbilt University Medical CenterNashvilleTN37235USA
| | | | - Liqin Ren
- Department of BioengineeringRice UniversityHoustonTX77005USA
| | - Charles F. Caskey
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTN37235USA
- Department of Radiology and Radiological SciencesVanderbilt UniversityNashvilleTN37235USA
| | - Michael R. King
- Department of BioengineeringRice UniversityHoustonTX77005USA
| |
Collapse
|
9
|
Ghnamah Y, Palmer CD, Livnat-Levanon N, Grupper M, Rosenzweig AC, Lewinson O. Prokaryotic mechanosensitive channels mediate copper influx. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.644891. [PMID: 40196475 PMCID: PMC11974724 DOI: 10.1101/2025.03.24.644891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Copper is an essential micronutrient in all kingdoms of life, requiring a meticulous balance between acquisition and toxic overload. While copper import in eukaryotes has been investigated extensively, few prokaryotic copper importers have been identified, leading to the notion that cytoplasmic copper uptake is unnecessary in prokaryotes. Here we report that mechanosensitive channels are key players in prokaryotic copper import. Deletion of the gene encoding the E. coli small mechanosensitive channel, Ec MscS, leads to significantly reduced copper influx. Conversely, overexpression of Ec MscS leads to increased copper influx, elevated intracellular copper content, and renders cells hypersensitive to copper. Furthermore, specific channel blockers and competing permeating ions inhibit Ec MscS copper conductance, lowering intracellular copper accumulation and alleviating copper hypersensitivity. These findings extend beyond E. coli , as other prokaryotic small mechanosensitive channels of bacterial and archaeal origin also facilitate copper influx. Taken together, these results uncover a previously unknown moonlighting function for mechanosensitive channels as a pathway for prokaryotic copper uptake.
Collapse
|
10
|
Romero LO, Bade M, Elsherif L, Williams JD, Kong X, Adebiyi A, Ataga KI, Ma S, Cordero-Morales J, Vásquez V. Enhanced PIEZO1 Function Contributes to the Pathogenesis of Sickle Cell Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.643952. [PMID: 40166330 PMCID: PMC11957036 DOI: 10.1101/2025.03.19.643952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Sickle cell disease (SCD), an inherited blood disorder caused by a mutation in the β-globin gene, is characterized by sickle erythrocytes that are prone to hemolysis, causing anemia and vaso-occlusion crises. In sickle erythrocytes, hemoglobin aggregation is followed by altered cation permeability and subsequent dehydration. Interventions to restore cation permeability can decrease hemolysis and ameliorate the symptoms associated with SCD. PIEZO1 is a non-selective mechanosensitive cation channel that regulates erythrocyte volume. Gain-of-function (GOF) mutations in PIEZO1 cause hemolytic anemia by increasing cation permeability, leading to erythrocyte dehydration in humans and mice. Although PIEZO1 plays a key role in erythrocyte homeostasis, its role in SCD remains unknown. Here, we demonstrate that the function of the PIEZO1 channel is upregulated in sickle erythrocytes of humans and mice, and this enhancement can be restored through a dietary intervention. We found that PIEZO1 function in sickle erythrocytes resembles that of the GOF mutation causing hemolytic anemia. A diet enriched in the ω -3 fatty acid eicosapentaenoic (EPA) acid decreases PIEZO1 function in sickle erythrocytes and hemolysis in a mouse model of SCD. Furthermore, EPA decreases hemolysis and reduces inflammatory markers. We propose that PIEZO1 contributes to the increase in nonselective cationic conductance (i.e., Psickle), which leads to dehydration downstream of hemoglobin polymerization. Our results suggest that reducing PIEZO1 function is a promising therapeutic approach to reestablishing normal cation permeability in SCD.
Collapse
|
11
|
Angeli S, Neophytou C, Kalli M, Stylianopoulos T, Mpekris F. The mechanopathology of the tumor microenvironment: detection techniques, molecular mechanisms and therapeutic opportunities. Front Cell Dev Biol 2025; 13:1564626. [PMID: 40171226 PMCID: PMC11958720 DOI: 10.3389/fcell.2025.1564626] [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: 01/22/2025] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
The mechanical properties of the tumor microenvironment (TME) undergo significant changes during tumor growth, primarily driven by alterations in extracellular (ECM) stiffness and tumor viscoelasticity. These mechanical changes not only promote tumor progression but also hinder therapeutic efficacy by impairing drug delivery and activating mechanotransduction pathways that regulate crucial cellular processes such as migration, proliferation, and resistance to therapy. In this review, we examine the mechanisms through which tumor cells sense and transmit mechanical signals to maintain homeostasis in the biomechanically altered TME. We explore current computational modelling strategies for mechanotransduction pathways, highlighting the need for developing models that incorporate additional components of the mechanosignaling machinery. Furthermore, we review available methods for measuring the mechanical properties of tumors in clinical settings and strategies aiming at restoring the TME and blocking deregulated mechanotransduction pathways. Finally, we propose that proper characterization and a deeper understanding of the mechanical landscape of the TME, both at the tissue and cellular levels, are essential for developing therapeutic strategies that account for the influence of mechanical forces on treatment efficacy.
Collapse
Affiliation(s)
| | | | | | | | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
12
|
Torgbor C, Sohn H, Dizon BLP, Mutic EC, George R, Kwak K, Akkaya M, Ulker EB, Traver M, Brzostowski J, Galloway DA, Thompson CD, Çuburu N, Schiller JT, Pierce SK. In the activation of HPV-specific human B cells HPV-VLP vaccines mimic membrane-associated antigens. Proc Natl Acad Sci U S A 2025; 122:e2414514122. [PMID: 40030014 PMCID: PMC11912367 DOI: 10.1073/pnas.2414514122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 01/10/2025] [Indexed: 03/19/2025] Open
Abstract
B cell responses to membrane-presented antigens appear to be strongly favored over soluble antigens in vivo suggesting that vaccines that mimic membrane-presented antigens may be highly efficacious. We recently demonstrated that human B cell responses to membrane-associated but not to soluble antigens in vitro depended on the expression and activity of the plasma membrane mechanosensitive ion channel, Piezo1. Here, we provide evidence that the efficacy of the current human papillomavirus virus-like particle (HPV VLP) vaccines may be due in part to their inherent ability to mimic Piezo1-dependent membrane presentation of antigens to B cells. We compared HPV-specific human B cell responses to HPV VLPs versus soluble HPV pentameric capsomeres and showed that although both induced calcium responses, only HPV VLP-induced responses were blocked by Piezo1 inhibitors. The kinetics of internalization of HPV-VLP and capsomeres into HPV-specific B cells were similar and neither required Piezo1 function as shown by small interfering RNA (siRNA)-mediated knockdown of Piezo. However, trafficking of HPV-VLPs into intracellular major histocompatibility complex (MHC) class II, lysosomal associated membrane protein 1 (LAMP1)+ antigen-processing compartments was Piezo1-dependent, whereas trafficking of capsomeres was not. In addition, a time course of intracellular trafficking suggested that colocalization of HPV-VLP with MHC classII was more stable over time as compared to capsomeres. Taken together these findings suggest that the ability of HPV-VLP vaccines to mimic Piezo1-dependent membrane antigen presentation may be exploited in the design of highly effective human vaccines.
Collapse
Affiliation(s)
- Charles Torgbor
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852
| | - Haewon Sohn
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852
| | - Brian L P Dizon
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852
- Rheumatology Fellowship and Training Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20814
| | - Evan C Mutic
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852
| | - Rachel George
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852
| | - Kihyuck Kwak
- 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
| | - Munir Akkaya
- Division of Rheumatology and Immunology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
- Pelotonia Institute for Immuno-Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Esin Bayrali Ulker
- Division of Rheumatology and Immunology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
- Pelotonia Institute for Immuno-Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Maria Traver
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852
| | - Joseph Brzostowski
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852
| | - Denise A Galloway
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Cynthia D Thompson
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20814
| | - Nicolas Çuburu
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20814
| | - John T Schiller
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20814
| | - Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852
| |
Collapse
|
13
|
Khairullina ZM, Vasileva VY, Chubinskiy-Nadezhdin VI. Piezo1 Ion Channels Regulate the Formation and Spreading of Human Endometrial Mesenchymal Stem Cell Spheroids. Int J Mol Sci 2025; 26:2474. [PMID: 40141118 PMCID: PMC11942067 DOI: 10.3390/ijms26062474] [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: 01/29/2025] [Revised: 03/03/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
Mesenchymal stem cells obtained from desquamated endometrium (eMSCs) are considered as reliable and promising objects for stem cell-based therapy. eMSCs aggregated into three-dimensional (3D) spheroids demonstrate greater efficiency compared to monolayer 2D eMSCs. However, molecular processes and specific mechanisms regulating the effectiveness of spheroids remain unknown. Regulation of a number of physiological reactions in MSCs is associated with the functioning of Ca2+-permeable mechanosensitive Piezo1 channels. In our previous study, we showed that selective Piezo1 activation by its selective agonist Yoda1 controls the migratory activity of 2D eMSCs. Here, we aimed to determine the effect of Yoda1 on eMSC spheroid formation and spreading. PIEZO1 mRNA expression was lower in spheroids compared to 2D culture. Spheroids formed with Yoda1 or spread in the presence of Yoda1 demonstrated lower spreading rates compared to control (Yoda1-free) spheroids. The spreading rates of control spheroids depended on the substrate stiffness, whereas spheroids formed with Yoda1 had similar spreading rates regardless of the surface properties. Our results demonstrate several Piezo1-dependent reactions of eMSC spheroids that could be modulated by selective Piezo1 activation.
Collapse
|
14
|
Purali N. Mechanosensitive Ion Channels: The Unending Riddle of Mechanotransduction. Bioelectricity 2025; 7:58-70. [PMID: 40342940 PMCID: PMC12054614 DOI: 10.1089/bioe.2024.0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025] Open
Abstract
Sensation begins at the periphery, where distinct transducer proteins, activated by specific physical stimuli, initiate biological events to convert the stimulus into electrical activity. These evoked pulse trains encode various properties of the stimulus and travel to higher centers, enabling perception of the physical environment. Transduction is an essential process in all of the five senses described by Aristotle. A substantial amount of information is already available on how G-protein coupled receptor proteins transduce exposure to light, odors, and tastants. Functional studies have revealed the presence of mechanosensitive (MS) ion channels, which act as force transducers, in a wide range of organisms from archaea to mammals. However, the molecular basis of mechanosensitivity is incompletely understood. Recently, the structure of a few MS channels and the molecular mechanisms linking mechanical force to channel gating have been partially revealed. This article reviews recent developments focusing on the molecular basis of mechanosensitivity and emerging methods to investigate MS channels.
Collapse
Affiliation(s)
- Nuhan Purali
- Faculty of Medicine, Department of Biophysics, Hacettepe University, Ankara, Turkey
| |
Collapse
|
15
|
Fu S, Pan X, Lu M, Dong J, Yan Z. Human TMC1 and TMC2 are mechanically gated ion channels. Neuron 2025; 113:411-425.e4. [PMID: 39674179 DOI: 10.1016/j.neuron.2024.11.009] [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: 04/06/2024] [Revised: 09/23/2024] [Accepted: 11/14/2024] [Indexed: 12/16/2024]
Abstract
Mammalian transmembrane channel-like proteins 1 and 2 (TMC1 and TMC2) have emerged as very promising candidate mechanotransduction channels in hair cells. However, controversy persists because the heterogeneously expressed TMC1/2 in cultured cells lack evidence of mechanical gating, primarily due to their absence from the plasma membrane. By employing domain swapping with OSCA1.1 and subsequent point mutations, we successfully identified membrane-localized mouse TMC1/2 mutants, demonstrating that they are mechanically gated in heterologous cells. Further, whole-genome CRISPRi screening enabled wild-type human TMC1/2 localization in the plasma membrane, where they responded robustly to poking stimuli. In addition, wild-type human TMC1/2 showed stretch-activated currents and clear single-channel current activities. Deafness-related TMC1 mutations altered the reversal potential of TMC1, indicating that TMC1/2 are pore-forming mechanotransduction channels. In summary, our study provides evidence that human TMC1/2 are pore-forming, mechanically activated ion channels, supporting their roles as mechanotransduction channels in hair cells.
Collapse
Affiliation(s)
- Songdi Fu
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Xueqi Pan
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China
| | - Mingshun Lu
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jianying Dong
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China
| | - Zhiqiang Yan
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China; Institute for Medical Physiology, Chinese Institutes for Medical Research, Beijing, China.
| |
Collapse
|
16
|
Ly AT, Freites JA, Bertaccini GA, Evans EL, Dickinson GD, Tobias DJ, Pathak MM. Single-particle tracking reveals heterogeneous PIEZO1 diffusion. Biophys J 2025:S0006-3495(25)00023-2. [PMID: 39844465 DOI: 10.1016/j.bpj.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/21/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025] Open
Abstract
The mechanically activated ion channel PIEZO1 is critical to numerous physiological processes, and is activated by diverse mechanical cues. The channel is gated by membrane tension and has been found to be mobile in the plasma membrane. We employed single-particle tracking (SPT) of endogenous, tdTomato-tagged PIEZO1 using total internal reflection fluorescence microscopy in live cells. Application of SPT unveiled a surprising heterogeneity of diffusing PIEZO1 subpopulations, which we labeled "mobile" and "immobile." We sorted these trajectories into the two aforementioned categories using trajectory spread. To evaluate the effects of the plasma membrane composition on PIEZO1 diffusion, we manipulated membrane composition by depleting or supplementing cholesterol, or by adding margaric acid to stiffen the membrane. To examine effects of channel activation on PIEZO1 mobility, we treated cells with Yoda1, a PIEZO1 agonist, and GsMTx-4, a channel inhibitor. We collected thousands of trajectories for each condition, and found that cholesterol removal and Yoda1 incubation increased the channel's propensity for mobility. Conversely, we found that GsMTx-4 incubation and cholesterol supplementation resulted in a lower chance of mobile trajectories, whereas margaric acid incubation did not have a significant effect on PIEZO1 mobility. The mobile trajectories were analyzed further by fitting the time-averaged mean-squared displacement as a function of lag time to a power law model, revealing that mobile PIEZO1 puncta exhibit anomalous subdiffusion. These studies illuminate the fundamental properties governing PIEZO1 diffusion in the plasma membrane and set the stage to determine how cellular processes and interactions may influence channel activity and mobility.
Collapse
Affiliation(s)
- Alan T Ly
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California
| | | | - Gabriella A Bertaccini
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California
| | - Elizabeth L Evans
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California
| | - George D Dickinson
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California
| | - Douglas J Tobias
- Department of Chemistry, UC Irvine, Irvine, California; Center for Complex Biological Systems, UC Irvine, Irvine, California.
| | - Medha M Pathak
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Department of Biomedical Engineering, UC Irvine, Irvine, California; Center for Complex Biological Systems, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California.
| |
Collapse
|
17
|
Moneme C, Olutoye OO, Sobstel MF, Zhang Y, Zhou X, Kaminer JL, Hsu BA, Shen C, Mandal A, Li H, Yu L, Balaji S, Keswani SG, Cheng LS. Activation of mechanoreceptor Piezo1 inhibits enteric neuronal growth and migration in vitro. Front Mol Neurosci 2024; 17:1474025. [PMID: 39759870 PMCID: PMC11695422 DOI: 10.3389/fnmol.2024.1474025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/30/2024] [Indexed: 01/07/2025] Open
Abstract
Introduction Dysfunction of the enteric nervous system (ENS) is linked to a myriad of gastrointestinal (GI) disorders. Piezo1 is a mechanosensitive ion channel found throughout the GI tract, but its role in the ENS is largely unknown. We hypothesize that Piezo1 plays an important role in the growth and development of the ENS. Methods Enteric neural crest-derived progenitor cells (ENPC) were isolated from adult mouse intestine and propagated in culture as neurospheres. ENPC-derived neurons were then subject to in vitro stretch in the presence or absence of Piezo1 antagonist (GsMTx4). Transcriptomes of stretched and unstretched ENPC-derived cells were compared using bulk RNA sequencing. Enteric neurons were also cultured under static conditions in the presence of Piezo1 agonist (Yoda1) or antagonist. Neuronal phenotype, migration, and recovery from injury were compared between groups. Results Though stretch did not cause upregulation of Piezo1 expression in enteric neurons, both stretch and Piezo1 activation produced similar alterations in neuronal morphology. Compared to control, neurite length was significantly shorter when stretched and in the presence of Piezo1 activation. Piezo1 inhibition prevented a significant reduction in neurite length in stretched neurons. Piezo1 inhibition also led to significantly increased neuronal migration, whereas Piezo1 activation resulted in significantly decreased neuronal migration and slower neuronal recovery from injury. Conclusion Mechanotransduction plays an important role in regulating normal GI function. Our results suggest that the Piezo1 mechanoreceptor may play an important role in the ENS as its activation leads to decreased neuronal growth and migration. Piezo1 could be an important target for diseases of ENS dysfunction and development.
Collapse
Affiliation(s)
- Chioma Moneme
- Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Oluyinka O. Olutoye
- Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Michał F. Sobstel
- Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Yuwen Zhang
- Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Xinyu Zhou
- Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Jacob L. Kaminer
- Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Britney A. Hsu
- Department of Pediatric Surgery, Texas Children's Hospital, Houston, TX, United States
| | - Chengli Shen
- Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Arabinda Mandal
- Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Hui Li
- Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatric Surgery, Texas Children's Hospital, Houston, TX, United States
| | - Ling Yu
- Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatric Surgery, Texas Children's Hospital, Houston, TX, United States
| | - Swathi Balaji
- Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatric Surgery, Texas Children's Hospital, Houston, TX, United States
| | - Sundeep G. Keswani
- Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatric Surgery, Texas Children's Hospital, Houston, TX, United States
| | - Lily S. Cheng
- Department of Surgery, University of Virginia, Charlottesville, VA, United States
- Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatric Surgery, Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
18
|
Stricker AM, Hutson MS, Page-McCaw A. Piezo-dependent surveillance of matrix stiffness generates transient cells that repair the basement membrane. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.573147. [PMID: 38187749 PMCID: PMC10769369 DOI: 10.1101/2023.12.22.573147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Basement membranes are extracellular matrix sheets separating tissue layers and providing mechanical support, and Collagen IV (Col4) is their most abundant protein. Although basement membranes are repaired after damage, little is known about repair, including whether and how damage is detected, what cells repair the damage, and how repair is controlled to avoid fibrosis. Using the intestinal basement membrane of adult Drosophila as a model, we show that after basement membrane damage, there is a sharp increase in enteroblasts transiently expressing Col4, or "matrix mender" cells. Enteroblast-derived Col4 is specifically required for matrix repair. The increase in matrix mender cells requires the mechanosensitive ion channel Piezo, expressed in intestinal stem cells. Matrix menders are induced by the loss of matrix stiffness, as specifically inhibiting Col4 crosslinking is sufficient for Piezo-dependent induction of matrix mender cells. Our data suggest that epithelial stem cells control basement membrane integrity by monitoring stiffness.
Collapse
Affiliation(s)
- Aubrie M. Stricker
- Department of Cell and Developmental Biology, Center for Matrix Biology, Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - M. Shane Hutson
- Department of Physics and Astronomy, Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology, Center for Matrix Biology, Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
19
|
Li Q, Li C, Li X, Liu X, Qian J, Li J, Li X, Zhang X. GsMTx4 Combined with Exercise Exerts Neuroprotective Effects by Regulating Neuronal Autophagy in Rats with Spinal Cord Injury. Neurochem Res 2024; 50:55. [PMID: 39666140 DOI: 10.1007/s11064-024-04304-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/05/2024] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
A sharp increase in intramedullary pressure after spinal cord injury (SCI) can aggravate secondary injury and lead to severe neurological deficits. Unfortunately, effective treatment options are currently lacking. The mechanosensitive ion channel Piezo1 plays an important role in the pathological process of SCI by transducing mechanical stress. The Piezo1 inhibitor GsMTx4 has been shown to have neuroprotective effects and may hold therapeutic potential for SCI. Given that single drug treatment strategy has limited effect on functional recovery after SCI, we explored the efficacy of combining GsMTx4 with exercise training in treating SCI in rats and investigated the underlying mechanisms. We used the T10 SCI rat model, administered GsMTx4 immediately after injury, and performed 4 weeks of body weight supported treadmill training starting (BWSTT) 2 weeks post injury. Subsequently, HE and LFB staining were used to observe the morphology of spinal cord tissue, WB was used to detect autophagy and apoptosis-related proteins, biochemical detection of calcium ion concentration and CTSD activity, IHC detection of LAMP1 expression, immunofluorescence labeling of NeuN and ChAT-positive motor neurons, as well as MBP and GFAP, and BBB scores were used to evaluate rat motor function. We found that the combined treatment of GsMTx4 drug and exercise training was more effective than single treatment alone. The combined treatment reduced calcium ion concentration, improved lysosomal function, enhanced autophagic flux, reduced cell apoptosis, and significantly improved the motor function of rats. This combined treatment regimen may pave the way for developing more comprehensive treatment strategies for SCI in the future.
Collapse
Affiliation(s)
- Qianxi Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Chenyu Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Xinyan Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Xinyu Liu
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Jinghua Qian
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Jianjun Li
- School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- China Rehabilitation Science Institute, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Xuemei Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Xin Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| |
Collapse
|
20
|
Teranishi A, Mori M, Ichiki R, Toda S, Shioi G, Okuda S. An actin bracket-induced elastoplastic transition determines epithelial folding irreversibility. Nat Commun 2024; 15:10476. [PMID: 39668169 PMCID: PMC11638340 DOI: 10.1038/s41467-024-54906-7] [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: 02/25/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024] Open
Abstract
During morphogenesis, epithelial sheets undergo sequential folding to form three-dimensional organ structures. The resulting folds are often irreversible, ensuring that morphogenesis progresses in one direction. However, the mechanism establishing folding irreversibility remains unclear. Here, we report a mechanical property of epithelia that determines folding irreversibility. Using a mechanical assay, we demonstrate that long-term, high-curvature folding induces plastic, irreversible deformations, while short-term or low-curvature folding results in an elastic, shape-restoring response. This elastic-plastic transition occurs in a switch-like manner, with critical thresholds in folding curvature and duration. The transition is induced by F-actin accumulating into a bracket-like structure across the fold, triggered by cells sensing deformations via mechanosensitive signaling pathways, including TRPC 3/6-mediated calcium influx and ligand-independent EGFR activation. These results demonstrate that cells control epithelial folding irreversibility by detecting folding characteristics and adaptively switching between elastic and plastic responses, providing mechanical insight into the directionality of morphogenesis.
Collapse
Affiliation(s)
- Aki Teranishi
- Division of Nano Life Science, Graduate School of Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Misato Mori
- Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Rihoko Ichiki
- Division of Nano Life Science, Graduate School of Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Satoshi Toda
- Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Go Shioi
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Satoru Okuda
- Nano Life Science Institute, Kanazawa University, Kanazawa, Japan.
- Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
21
|
Kinsella JA, Debant M, Parsonage G, Morley LC, Bajarwan M, Revill C, Foster R, Beech DJ. Pharmacology of PIEZO1 channels. Br J Pharmacol 2024; 181:4714-4732. [PMID: 39402010 DOI: 10.1111/bph.17351] [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: 02/06/2024] [Revised: 06/15/2024] [Accepted: 08/25/2024] [Indexed: 11/05/2024] Open
Abstract
PIEZO1 is a eukaryotic membrane protein that assembles as trimers to form calcium-permeable, non-selective cation channels with exquisite capabilities for mechanical force sensing and transduction of force into effect in diverse cell types that include blood cells, endothelial cells, epithelial cells, fibroblasts and stem cells and diverse systems that include bone, lymphatics and muscle. The channel has wide-ranging roles and is considered as a target for novel therapeutics in ailments spanning cancers and cardiovascular, dental, gastrointestinal, hepatobiliary, infectious, musculoskeletal, nervous system, ocular, pregnancy, renal, respiratory and urological disorders. The identification of PIEZO1 modulators is in its infancy but useful experimental tools emerged for activating, and to a lesser extent inhibiting, the channels. Elementary structure-activity relationships are known for the Yoda series of small molecule agonists, which show the potential for diverse physicochemical and pharmacological properties. Intriguing effects of Yoda1 include the stimulated removal of excess cerebrospinal fluid. Despite PIEZO1's broad expression, opportunities are suggested for selective positive or negative modulation without intolerable adverse effects. Here we provide a focused, non-systematic, narrative review of progress with this pharmacology and discuss potential future directions for research in the area.
Collapse
Affiliation(s)
- Jacob A Kinsella
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
- School of Chemistry, University of Leeds, Leeds, UK
| | - Marjolaine Debant
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Gregory Parsonage
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Lara C Morley
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Muath Bajarwan
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | | | | | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
22
|
Roeterink RMA, Casadevall I Solvas X, Collins DJ, Scott DJ. Force versus Response: Methods for Activating and Characterizing Mechanosensitive Ion Channels and GPCRs. Adv Healthc Mater 2024; 13:e2402167. [PMID: 39402780 DOI: 10.1002/adhm.202402167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/20/2024] [Indexed: 12/18/2024]
Abstract
Mechanotransduction is the process whereby cells convert mechanical signals into electrochemical responses, where mechanosensitive proteins mediate this interaction. To characterize these critical proteins, numerous techniques have been developed that apply forces and measure the subsequent cellular responses. While these approaches have given insight into specific aspects of many such proteins, subsequent validation and cross-comparison between techniques remain difficult given significant variations in reported activation thresholds and responses for the same protein across different studies. Accurately determining mechanosensitivity responses for various proteins, however, is essential for understanding mechanotransduction and potential physiological implications, including therapeutics. This critical review provides an assessment of current and emerging approaches used for mechanosensitive ion channel and G-Coupled Receptors (GPCRs) stimulation and measurement, with a specific focus on the ability to quantitatively measure mechanosensitive responses.
Collapse
Affiliation(s)
- Renate M A Roeterink
- Department of Biomedical Engineering, The University of Melbourne, VIC, Parkville, Victoria, 3010, Australia
- Department of Biosystems - MeBioS, KU Leuven, Willem de Croylaan 42, Leuven, 3001, Belgium
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
| | | | - David J Collins
- Department of Biomedical Engineering, The University of Melbourne, VIC, Parkville, Victoria, 3010, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Daniel J Scott
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
23
|
Tian C, Lyu T, Zhao X, Wang R, Wu Y, Yang D. Piezo1 channel: A global bibliometric analysis from 2010 to 2024. Channels (Austin) 2024; 18:2396354. [PMID: 39282983 PMCID: PMC11407379 DOI: 10.1080/19336950.2024.2396354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
In recent years, the Piezo1 channel has attracted great attention. Piezo1's research has made remarkable advance in many aspects. However, the overall trends and knowledge structures have not been systematically investigated from a worldwide viewpoint. Therefore, it is important to fill this knowledge gap and utilize a proper tool to show the research status, hotspots, and frontiers in the Piezo1 channel. In order to better investigate the hotspots and frontiers of the Piezo1 channel research, we retrieved relevant literature from Web of Science Core Collection (WoSCC) and applied CiteSpace to perform a bibliometric analysis. Our findings might serve as a reference for future research in this area.
Collapse
Affiliation(s)
- Chuanxi Tian
- Traditional Chinese Respiratory Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tianyi Lyu
- Department of Acupuncture, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Xirui Zhao
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ruoshui Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Wu
- Department of Acupuncture, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Daowen Yang
- Department of Traditional Chinese Medicine for Pulmonary Diseases, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
24
|
Baghdadi MB, Houtekamer RM, Perrin L, Rao-Bhatia A, Whelen M, Decker L, Bergert M, Pérez-Gonzàlez C, Bouras R, Gropplero G, Loe AKH, Afkhami-Poostchi A, Chen X, Huang X, Descroix S, Wrana JL, Diz-Muñoz A, Gloerich M, Ayyaz A, Matic Vignjevic D, Kim TH. PIEZO-dependent mechanosensing is essential for intestinal stem cell fate decision and maintenance. Science 2024; 386:eadj7615. [PMID: 39607940 DOI: 10.1126/science.adj7615] [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: 07/16/2023] [Revised: 07/27/2024] [Accepted: 10/07/2024] [Indexed: 11/30/2024]
Abstract
Stem cells perceive and respond to biochemical and physical signals to maintain homeostasis. Yet, it remains unclear how stem cells sense mechanical signals from their niche in vivo. In this work, we investigated the roles of PIEZO mechanosensitive channels in the intestinal stem cell (ISC) niche. We used mouse genetics and single-cell RNA sequencing analysis to assess the requirement for PIEZO channels in ISC maintenance. In vivo measurement of basement membrane stiffness showed that ISCs reside in a more rigid microenvironment at the bottom of the crypt. Three-dimensional and two-dimensional organoid systems combined with bioengineered substrates and a stretching device revealed that PIEZO channels sense extracellular mechanical stimuli to modulate ISC function. This study delineates the mechanistic cascade of PIEZO activation that coordinates ISC fate decision and maintenance.
Collapse
Affiliation(s)
- Meryem B Baghdadi
- Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ronja M Houtekamer
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Louisiane Perrin
- Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Abilasha Rao-Bhatia
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Myles Whelen
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Linda Decker
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Martin Bergert
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Réda Bouras
- Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Giacomo Gropplero
- Institut Curie, IPGG, PSL Research University, CNRS UMR 168, Paris, France
| | - Adrian K H Loe
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Amin Afkhami-Poostchi
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Xin Chen
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Xi Huang
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stephanie Descroix
- Institut Curie, IPGG, PSL Research University, CNRS UMR 168, Paris, France
| | - Jeffrey L Wrana
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Laboratory Medicine, St. Michael's Hospital, Toronto, ON, Canada
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Martijn Gloerich
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Arshad Ayyaz
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Tae-Hee Kim
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
25
|
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
|
26
|
Chen D, Tang Y, Lapinski PE, Wiggins D, Sevick EM, Davis MJ, King PD. EPHB4-RASA1 Inhibition of PIEZO1 Ras Activation Drives Lymphatic Valvulogenesis. Circ Res 2024; 135:1048-1066. [PMID: 39421925 PMCID: PMC11560524 DOI: 10.1161/circresaha.124.325383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND EPHB4 (ephrin receptor B4) and the RASA1 (p120 Ras GTPase-activating protein) are necessary for the development of lymphatic vessel (LV) valves. However, precisely how EPHB4 and RASA1 regulate LV valve development is unknown. In this study, we examine the mechanisms by which EPHB4 and RASA1 regulate the development of LV valves. METHODS We used LV-specific inducible EPHB4-deficient mice and EPHB4 knockin mice that express a form of EPHB4 that is unable to bind RASA1 yet retains protein tyrosine kinase activity (EPHB4 2YP) to study the role of EPHB4 and RASA1 in LV valve development in the embryo and LV valve maintenance in adults. We also used human dermal lymphatic endothelial cells in vitro to study the role of EPHB4 and RASA1 as regulators of LV valve specification induced by oscillatory shear stress, considered the trigger for LV valve specification in vivo. RESULTS LV valve specification, continued valve development postspecification, and LV valve maintenance were blocked upon induced loss of EPHB4 in LV. LV valve specification and maintenance were also impaired in EPHB4 2YP mice. Defects in LV valve development were reversed by inhibition of the Ras-MAPK (mitogen-activated protein kinase) signaling pathway. In human dermal lymphatic endothelial cells, loss of expression of EPHB4 or its ephrin b2 ligand, loss of expression of RASA1, and inhibition of physical interaction between EPHB4 and RASA1 resulted in dysregulated oscillatory shear stress-induced Ras-MAPK activation and impaired expression of LV specification markers that could be rescued by Ras-MAPK pathway inhibition. The same results were observed when human dermal lymphatic endothelial cells were stimulated with the Yoda1 agonist of the PIEZO1 oscillatory shear stress sensor. Although Yoda1 increased the number of LV valves when administered to wild-type embryos, it did not increase LV valve number when administered to EPHB4 2YP embryos. CONCLUSIONS EPHB4 is necessary for LV valve specification, continued valve development postspecification, and valve maintenance. LV valve specification requires physical interaction between EPHB4 and RASA1 to limit activation of the Ras-MAPK pathway in lymphatic endothelial cells. Specifically, EPHB4-RASA1 physical interaction is necessary to dampen Ras-MAPK activation induced through the PIEZO1 oscillatory shear stress sensor. These findings reveal the mechanism by which EPHB4 and RASA1 regulate the development of LV valves.
Collapse
Affiliation(s)
- Di Chen
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48103, USA
| | - Yipei Tang
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48103, USA
| | - Philip E. Lapinski
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48103, USA
| | - David Wiggins
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Eva M. Sevick
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Michael J. Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| | - Philip D. King
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48103, USA
| |
Collapse
|
27
|
Xiao B. Mechanisms of mechanotransduction and physiological roles of PIEZO channels. Nat Rev Mol Cell Biol 2024; 25:886-903. [PMID: 39251883 DOI: 10.1038/s41580-024-00773-5] [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: 08/02/2024] [Indexed: 09/11/2024]
Abstract
Mechanical force is an essential physical element that contributes to the formation and function of life. The discovery of the evolutionarily conserved PIEZO family, including PIEZO1 and PIEZO2 in mammals, as bona fide mechanically activated cation channels has transformed our understanding of how mechanical forces are sensed and transduced into biological activities. In this Review, I discuss recent structure-function studies that have illustrated how PIEZO1 and PIEZO2 adopt their unique structural design and curvature-based gating dynamics, enabling their function as dedicated mechanotransduction channels with high mechanosensitivity and selective cation conductivity. I also discuss our current understanding of the physiological and pathophysiological roles mediated by PIEZO channels, including PIEZO1-dependent regulation of development and functional homeostasis and PIEZO2-dominated mechanosensation of touch, tactile pain, proprioception and interoception of mechanical states of internal organs. Despite the remarkable progress in PIEZO research, this Review also highlights outstanding questions in the field.
Collapse
Affiliation(s)
- Bailong Xiao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
- Beijing Frontier Research Center of Biological Structure, Tsinghua University, Beijing, China.
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
| |
Collapse
|
28
|
Xu Y, Wang Y, Yang Y, Fang X, Wu L, Hu J, Li J, Mei S. Piezo1: the key regulators in central nervous system diseases. Front Cell Neurosci 2024; 18:1441806. [PMID: 39539343 PMCID: PMC11557416 DOI: 10.3389/fncel.2024.1441806] [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: 05/31/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
The occurrence and development of central nervous system (CNS) diseases is a multi-factor and multi-gene pathological process, and their diagnosis and treatment have always posed a serious challenge in the medical field. Therefore, exploring the relevant factors in the pathogenesis of CNS and improving the diagnosis and treatment rates has become an urgent problem. Piezo1 is a recently discovered mechanosensitive ion channel that opens in response to mechanical stimuli. A number of previous studies have shown that the Piezo channel family plays a crucial role in CNS physiology and pathology, especially in diseases related to CNS development and mechanical stimulation. This article comprehensively describes the biological properties of Piezo1, focuses on the potential association between Piezo1 and CNS disorders, and explores the pharmacological roles of Piezo1 agonists and inhibitors in treating CNS disorders.
Collapse
Affiliation(s)
- Yi Xu
- The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuheng Wang
- The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanling Yang
- The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaowei Fang
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lidong Wu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jialing Hu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jin Li
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shuchong Mei
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
29
|
Nagase T, Nagase M. Piezo ion channels: long-sought-after mechanosensors mediating hypertension and hypertensive nephropathy. Hypertens Res 2024; 47:2786-2799. [PMID: 39103520 DOI: 10.1038/s41440-024-01820-6] [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: 05/08/2024] [Revised: 07/03/2024] [Accepted: 07/07/2024] [Indexed: 08/07/2024]
Abstract
Recent advances in mechanobiology and the discovery of mechanosensitive ion channels have opened a new era of research on hypertension and related diseases. Piezo1 and Piezo2, first reported in 2010, are regarded as bona fide mechanochannels that mediate various biological and pathophysiological phenomena in multiple tissues and organs. For example, Piezo channels have pivotal roles in blood pressure control, triggering shear stress-induced nitric oxide synthesis and vasodilation, regulating baroreflex in the carotid sinus and aorta, and releasing renin from renal juxtaglomerular cells. Herein, we provide an overview of recent literature on the roles of Piezo channels in the pathogenesis of hypertension and related kidney damage, including our experimental data on the involvement of Piezo1 in podocyte injury and that of Piezo2 in renin expression and renal fibrosis in animal models of hypertensive nephropathy. The mechanosensitive ion channels Piezo1 and Piezo2 play various roles in the pathogenesis of systemic hypertension by acting on vascular endothelial cells, baroreceptors in the carotid artery and aorta, and the juxtaglomerular apparatus. Piezo channels also contribute to hypertensive nephropathy by acting on mesangial cells, podocytes, and perivascular mesenchymal cells.
Collapse
Affiliation(s)
- Takashi Nagase
- Kunitachi Aoyagien Tachikawa Geriatric Health Services Facility, Tokyo, Japan
| | - Miki Nagase
- Department of Anatomy, Kyorin University School of Medicine, Tokyo, Japan.
| |
Collapse
|
30
|
Moura TDBD, Nunes FB, Crestani BDV, Araujo TFC, Hanauer EL, Corleta HVE, Branchini G. Preeclampsia and transport of ions and small molecules: A literature review. Placenta 2024; 156:77-91. [PMID: 39293185 DOI: 10.1016/j.placenta.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/22/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Preeclampsia (PE) is a prevalent obstetric complication affecting approximately 3-5% of pregnancies worldwide and is a major cause of maternal and perinatal morbidity and mortality. Preeclampsia is considered a disease of the endothelial system that can progress to eclampsia, characterized by seizures. Early diagnosis and appropriate management are crucial to improving maternal and fetal outcomes, as preeclampsia can lead to severe complications such as placental abruption, fetal growth restriction, and stroke. The pathophysiology of PE is complex, involving a combination of genetic, acquired, and immunological factors. A central feature of the condition is inadequate placentation and impaired uteroplacental perfusion, leading to local hypoxia, endothelial dysfunction, vasoconstriction, and immunological dysregulation. Recent evidence suggests that dysregulation of ion transporters may play a significant role in the adaptation of uterine circulation during placentation. These transporters are essential for maintaining maternal-fetal homeostasis, influencing processes such as nutrient exchange, hormone synthesis, trophoblast cell migration, and the function of smooth muscle cells in blood vessels. In preeclampsia, adverse conditions like hypoxia and oxidative stress result in the downregulation of ion, solute, and water transporters, impairing their function. This review focuses on membrane transporters involved in PE, discussing functional alterations and their physiological implications. The goal of this investigation is to enhance understanding of how dysregulation of ion and small molecule transporters contributes to the development and progression of preeclampsia, underscoring the importance of exploring these signaling pathways for potential therapeutic interventions.
Collapse
Affiliation(s)
- Thaís Duarte Borges de Moura
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, RS, ZIP 90050170, Brazil
| | - Fernanda Bordignon Nunes
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, RS, ZIP 90050170, Brazil; Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), 6681 Ipiranga Av, Porto Alegre, RS, ZIP 90619-900, Brazil
| | - Bianca Dalla Vecchia Crestani
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, ZIP 90050170, Brazil
| | | | - Eduarda Luiza Hanauer
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, ZIP 90050170, Brazil
| | - Helena von Eye Corleta
- Departamento de Ginecologia e Obstetrícia, Faculdade de Medicina, Universidade Federal Do Rio Grande Do Sul (UFRGS), 2400 Ramiro Barcelos St, Porto Alegre, RS, ZIP 90035-003, Brazil
| | - Gisele Branchini
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, RS, ZIP 90050170, Brazil.
| |
Collapse
|
31
|
Garcia-Sanchez J, Lin D, Liu WW. Mechanosensitive ion channels in glaucoma pathophysiology. Vision Res 2024; 223:108473. [PMID: 39180975 PMCID: PMC11398070 DOI: 10.1016/j.visres.2024.108473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/27/2024]
Abstract
Force sensing is a fundamental ability that allows cells and organisms to interact with their physical environment. The eye is constantly subjected to mechanical forces such as blinking and eye movements. Furthermore, elevated intraocular pressure (IOP) can cause mechanical strain at the optic nerve head, resulting in retinal ganglion cell death (RGC) in glaucoma. How mechanical stimuli are sensed and affect cellular physiology in the eye is unclear. Recent studies have shown that mechanosensitive ion channels are expressed in many ocular tissues relevant to glaucoma and may influence IOP regulation and RGC survival. Furthermore, variants in mechanosensitive ion channel genes may be associated with risk for primary open angle glaucoma. These findings suggest that mechanosensitive channels may be important mechanosensors mediating cellular responses to pressure signals in the eye. In this review, we focus on mechanosensitive ion channels from three major channel families-PIEZO, two-pore potassium and transient receptor potential channels. We review the key properties of these channels, their effects on cell function and physiology, and discuss their possible roles in glaucoma pathophysiology.
Collapse
Affiliation(s)
- Julian Garcia-Sanchez
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Danting Lin
- 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
|
32
|
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
|
33
|
Xu Y, Wang Y, Mei S, Hu J, Wu L, Xu L, Bao L, Fang X. The mechanism and potential therapeutic target of piezo channels in pain. FRONTIERS IN PAIN RESEARCH 2024; 5:1452389. [PMID: 39398533 PMCID: PMC11466900 DOI: 10.3389/fpain.2024.1452389] [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: 06/20/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Pain is a common symptom of many clinical diseases; it adversely affects patients' physical and mental health, reduces their quality of life, and heavily burdens patients and society. Pain treatment is one of the most difficult problems today. There is an urgent need to explore the potential factors involved in the pathogenesis of pain to improve its diagnosis and treatment rate. Piezo1/2, a newly identified mechanosensitive ion channel opens in response to mechanical stimuli and plays a critical role in regulating pain-related diseases. Inhibition or downregulation of Piezo1/2 alleviates disease-induced pain. Therefore, in this study, we comprehensively discussed the biology of this gene, focusing on its potential relevance in pain-related diseases, and explored the pharmacological effects of drugs using this gene for the treatment of pain.
Collapse
Affiliation(s)
- Yi Xu
- Department of Emergency Medicine, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
- Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Yuheng Wang
- Department of Emergency Medicine, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
- Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Shuchong Mei
- Department of Emergency Medicine, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Jialing Hu
- Department of Emergency Medicine, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Lidong Wu
- Department of Emergency Medicine, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Luyang Xu
- Department of Emergency Medicine, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Lijie Bao
- Department of Emergency Medicine, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Xiaowei Fang
- Department of Emergency Medicine, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| |
Collapse
|
34
|
Amoakon JP, Lee J, Liyanage P, Arora K, Karlstaedt A, Mylavarapu G, Amin R, Naren AP. Defective CFTR modulates mechanosensitive channels TRPV4 and PIEZO1 and drives endothelial barrier failure. iScience 2024; 27:110703. [PMID: 39252977 PMCID: PMC11382128 DOI: 10.1016/j.isci.2024.110703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 06/25/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Cystic fibrosis (CF) is a genetic disease caused by a mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Despite reports of CFTR expression on endothelial cells, pulmonary vascular perturbations, and perfusion deficits in CF patients, the mechanism of pulmonary vascular disease in CF remains unclear. Here, our pilot study of 40 CF patients reveals a loss of small pulmonary blood vessels in patients with severe lung disease. Using a vessel-on-a-chip model, we establish a shear-stress-dependent mechanism of endothelial barrier failure in CF involving TRPV4, a mechanosensitive channel. Furthermore, we demonstrate that CFTR deficiency downregulates the function of PIEZO1, another mechanosensitive channel involved in angiogenesis and wound repair, and exacerbates loss of small pulmonary blood vessel. We also show that CFTR directly interacts with PIEZO1 and enhances its function. Our study identifies key cellular targets to mitigate loss of small pulmonary blood vessels in CF.
Collapse
Affiliation(s)
- Jean-Pierre Amoakon
- Department of Systems Biology and Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jesun Lee
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Pramodha Liyanage
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kavisha Arora
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Goutham Mylavarapu
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Raouf Amin
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anjaparavanda P Naren
- Department of Systems Biology and Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
35
|
Jin C, Su S, Yu S, Zhang Y, Chen K, Xiang M, Ma H. Essential Roles of PIEZO1 in Mammalian Cardiovascular System: From Development to Diseases. Cells 2024; 13:1422. [PMID: 39272994 PMCID: PMC11394449 DOI: 10.3390/cells13171422] [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: 07/15/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Mechanical force is the basis of cardiovascular development, homeostasis, and diseases. The perception and response of mechanical force by the cardiovascular system are crucial. However, the molecular mechanisms mediating mechanotransduction in the cardiovascular system are not yet understood. PIEZO1, a novel transmembrane mechanosensitive cation channel known for its regulation of touch sensation, has been found to be widely expressed in the mammalian cardiovascular system. In this review, we elucidate the role and mechanism of PIEZO1 as a mechanical sensor in cardiovascular development, homeostasis, and disease processes, including embryo survival, angiogenesis, cardiac development repair, vascular inflammation, lymphangiogenesis, blood pressure regulation, cardiac hypertrophy, cardiac fibrosis, ventricular remodeling, and heart failure. We further summarize chemical molecules targeting PIEZO1 for potential translational applications. Finally, we address the controversies surrounding emergent concepts and challenges in future applications.
Collapse
Affiliation(s)
- Chengjiang Jin
- Cardiovascular Key Laboratory of Zhejiang Province, National Key Laboratory of Vascular Implantable Devices, Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Sheng’an Su
- Cardiovascular Key Laboratory of Zhejiang Province, National Key Laboratory of Vascular Implantable Devices, Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Shuo Yu
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yue Zhang
- Cardiovascular Key Laboratory of Zhejiang Province, National Key Laboratory of Vascular Implantable Devices, Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Kaijie Chen
- Cardiovascular Key Laboratory of Zhejiang Province, National Key Laboratory of Vascular Implantable Devices, Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Meixiang Xiang
- Cardiovascular Key Laboratory of Zhejiang Province, National Key Laboratory of Vascular Implantable Devices, Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Hong Ma
- Cardiovascular Key Laboratory of Zhejiang Province, National Key Laboratory of Vascular Implantable Devices, Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
36
|
Pourteymour S, Fan J, Majhi RK, Guo S, Sun X, Huang Z, Liu Y, Winter H, Bäcklund A, Skenteris NT, Chernogubova E, Werngren O, Li Z, Skogsberg J, Li Y, Matic L, Hedin U, Maegdefessel L, Ehrenborg E, Tian Y, Jin H. PIEZO1 targeting in macrophages boosts phagocytic activity and foam cell apoptosis in atherosclerosis. Cell Mol Life Sci 2024; 81:331. [PMID: 39107572 PMCID: PMC11335255 DOI: 10.1007/s00018-024-05372-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024]
Abstract
The rising incidences of atherosclerosis have necessitated efforts to identify novel targets for therapeutic interventions. In the present study, we observed increased expression of the mechanosensitive calcium channel Piezo1 transcript in mouse and human atherosclerotic plaques, correlating with infiltration of PIEZO1-expressing macrophages. In vitro administration of Yoda1, a specific agonist for PIEZO1, led to increased foam cell apoptosis and enhanced phagocytosis by macrophages. Mechanistically, PIEZO1 activation resulted in intracellular F-actin rearrangement, elevated mitochondrial ROS levels and induction of mitochondrial fragmentation upon PIEZO1 activation, as well as increased expression of anti-inflammatory genes. In vivo, ApoE-/- mice treated with Yoda1 exhibited regression of atherosclerosis, enhanced stability of advanced lesions, reduced plaque size and necrotic core, increased collagen content, and reduced expression levels of inflammatory markers. Our findings propose PIEZO1 as a novel and potential therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Shirin Pourteymour
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden.
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Blindern, PO Box 1046, 0317, Oslo, Norway.
| | - Jingxue Fan
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, People's Republic of China
| | - Rakesh Kumar Majhi
- Department of Microbiology, Tumor and Cell Biology, Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden
| | - Shuyuan Guo
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, People's Republic of China
| | - Xin Sun
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People's Hospital, Shenzhen, People's Republic of China
| | - Zhen Huang
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, People's Republic of China
| | - Ying Liu
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, People's Republic of China
| | - Hanna Winter
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
| | - Alexandra Bäcklund
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Nikolaos-Taxiarchis Skenteris
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | - Olivera Werngren
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Zhaolong Li
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
| | - Josefin Skogsberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Yuhuang Li
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Lars Maegdefessel
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
| | - Ewa Ehrenborg
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Ye Tian
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, People's Republic of China.
| | - Hong Jin
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden.
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
37
|
Ubuzima P, Nshimiyimana E, Mukeshimana C, Mazimpaka P, Mugabo E, Mbyayingabo D, Mohamed AS, Habumugisha J. Exploring biological mechanisms in orthodontic tooth movement: Bridging the gap between basic research experiments and clinical applications - A comprehensive review. Ann Anat 2024; 255:152286. [PMID: 38810763 DOI: 10.1016/j.aanat.2024.152286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/21/2024] [Accepted: 05/21/2024] [Indexed: 05/31/2024]
Abstract
OBJECTIVES The molecular mechanisms behind orthodontic tooth movements (OTM) were investigated by clarifying the role of chemical messengers released by cells. METHODS Using the Cochrane library, Google scholar, and PubMed databases, a literature search was conducted, and studies published from 1984 to 2024 were considered. RESULTS Both bone growth and remodeling may occur when a tooth is subjected to mechanical stress. These chemicals have a significant effect on the stimulation and regulation of osteoblasts, osteoclasts, and osteocytes during alveolar bone remodeling. This regulation can take place in pathological conditions, such as periodontal diseases, or during OTM alone. This comprehensive review outlines key molecular mechanisms underlying OTM and explores various clinical assumptions associated with specific molecules and their functional domains during this process. Furthermore, clinical applications of certain molecules such as relaxin, prostaglandin E (PGE), and interleukin-1β (IL-1β) in accelerating OTM have been reported. Our findings underscore the existing gap between OTM clinical applications and basic research investigations. CONCLUSION A comprehensive understanding of orthodontic treatment is enriched by insights into biological systems. We reported the activation of osteoblasts, osteoclast precursor cells, osteoclasts, and osteocytes in response to mechanical stress, leading to targeted cellular and molecular interventions and facilitating rapid and regulated alveolar bone remodeling during tooth movement. Despite the shortcomings of clinical studies in accelerating OTM, this review highlights the crucial role of biological agents in this process and advocates for prioritizing high-quality human studies in future research to gain further insights from clinical trials.
Collapse
Affiliation(s)
- Pascal Ubuzima
- Department of Orthodontics, Affliated Hospital of Stomatology, Anhui Medical University Hefei, 69 Meishan Road, Hefei, Anhui, China; School of Dentistry, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Eugene Nshimiyimana
- Department of Orthodontics, Affliated Hospital of Stomatology, Anhui Medical University Hefei, 69 Meishan Road, Hefei, Anhui, China
| | - Christelle Mukeshimana
- Department of Orthodontics, Affliated Hospital of Stomatology, Anhui Medical University Hefei, 69 Meishan Road, Hefei, Anhui, China
| | - Patrick Mazimpaka
- School of Dentistry, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Eric Mugabo
- Department of Orthodontics, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, 72 Xiangya Road, Changsha, Hunan 410000, China
| | - Dieudonne Mbyayingabo
- Department of Orthodontics, Stomatological Hospital of Xi'an Jiaotong University, 98 XiWu Road, Xi'an, Shaanxi 710004, China
| | | | - Janvier Habumugisha
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1, Shikata-cho, Kitaku, Okayama 700-8525, Japan; Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| |
Collapse
|
38
|
Franco-Obregón A, Tai YK. Are Aminoglycoside Antibiotics TRPing Your Metabolic Switches? Cells 2024; 13:1273. [PMID: 39120305 PMCID: PMC11311832 DOI: 10.3390/cells13151273] [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: 07/03/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024] Open
Abstract
Transient receptor potential (TRP) channels are broadly implicated in the developmental programs of most tissues. Amongst these tissues, skeletal muscle and adipose are noteworthy for being essential in establishing systemic metabolic balance. TRP channels respond to environmental stimuli by supplying intracellular calcium that instigates enzymatic cascades of developmental consequence and often impinge on mitochondrial function and biogenesis. Critically, aminoglycoside antibiotics (AGAs) have been shown to block the capacity of TRP channels to conduct calcium entry into the cell in response to a wide range of developmental stimuli of a biophysical nature, including mechanical, electromagnetic, thermal, and chemical. Paradoxically, in vitro paradigms commonly used to understand organismal muscle and adipose development may have been led astray by the conventional use of streptomycin, an AGA, to help prevent bacterial contamination. Accordingly, streptomycin has been shown to disrupt both in vitro and in vivo myogenesis, as well as the phenotypic switch of white adipose into beige thermogenic status. In vivo, streptomycin has been shown to disrupt TRP-mediated calcium-dependent exercise adaptations of importance to systemic metabolism. Alternatively, streptomycin has also been used to curb detrimental levels of calcium leakage into dystrophic skeletal muscle through aberrantly gated TRPC1 channels that have been shown to be involved in the etiology of X-linked muscular dystrophies. TRP channels susceptible to AGA antagonism are critically involved in modulating the development of muscle and adipose tissues that, if administered to behaving animals, may translate to systemwide metabolic disruption. Regenerative medicine and clinical communities need to be made aware of this caveat of AGA usage and seek viable alternatives, to prevent contamination or infection in in vitro and in vivo paradigms, respectively.
Collapse
Affiliation(s)
- Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zürich, 8057 Zürich, Switzerland
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Yee Kit Tai
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| |
Collapse
|
39
|
Mim MS, Kumar N, Levis M, Unger MF, Miranda G, Gazzo D, Robinett T, Zartman JJ. Piezo regulates epithelial topology and promotes precision in organ size control. Cell Rep 2024; 43:114398. [PMID: 38935502 PMCID: PMC11606527 DOI: 10.1016/j.celrep.2024.114398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 05/09/2024] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
Mechanosensitive Piezo channels regulate cell division, cell extrusion, and cell death. However, systems-level functions of Piezo in regulating organogenesis remain poorly understood. Here, we demonstrate that Piezo controls epithelial cell topology to ensure precise organ growth by integrating live-imaging experiments with pharmacological and genetic perturbations and computational modeling. Notably, the knockout or knockdown of Piezo increases bilateral asymmetry in wing size. Piezo's multifaceted functions can be deconstructed as either autonomous or non-autonomous based on a comparison between tissue-compartment-level perturbations or between genetic perturbation populations at the whole-tissue level. A computational model that posits cell proliferation and apoptosis regulation through modulation of the cutoff tension required for Piezo channel activation explains key cell and tissue phenotypes arising from perturbations of Piezo expression levels. Our findings demonstrate that Piezo promotes robustness in regulating epithelial topology and is necessary for precise organ size control.
Collapse
Affiliation(s)
- Mayesha Sahir Mim
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Nilay Kumar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Megan Levis
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Maria F Unger
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Gabriel Miranda
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - David Gazzo
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Trent Robinett
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jeremiah J Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
40
|
Guo J, Li L, Chen F, Fu M, Cheng C, Wang M, Hu J, Pei L, Sun J. Forces Bless You: Mechanosensitive Piezo Channels in Gastrointestinal Physiology and Pathology. Biomolecules 2024; 14:804. [PMID: 39062518 PMCID: PMC11274378 DOI: 10.3390/biom14070804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
The gastrointestinal (GI) tract is an organ actively involved in mechanical processes, where it detects forces via a mechanosensation mechanism. Mechanosensation relies on specialized cells termed mechanoreceptors, which convert mechanical forces into electrochemical signals via mechanosensors. The mechanosensitive Piezo1 and Piezo2 are widely expressed in various mechanosensitive cells that respond to GI mechanical forces by altering transmembrane ionic currents, such as epithelial cells, enterochromaffin cells, and intrinsic and extrinsic enteric neurons. This review highlights recent research advances on mechanosensitive Piezo channels in GI physiology and pathology. Specifically, the latest insights on the role of Piezo channels in the intestinal barrier, GI motility, and intestinal mechanosensation are summarized. Additionally, an overview of Piezo channels in the pathogenesis of GI disorders, including irritable bowel syndrome, inflammatory bowel disease, and GI cancers, is provided. Overall, the presence of mechanosensitive Piezo channels offers a promising new perspective for the treatment of various GI disorders.
Collapse
Affiliation(s)
- Jing Guo
- Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, China; (J.G.); (C.C.); (M.W.); (J.H.)
| | - Li Li
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| | - Feiyi Chen
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| | - Minhan Fu
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| | - Cheng Cheng
- Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, China; (J.G.); (C.C.); (M.W.); (J.H.)
| | - Meizi Wang
- Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, China; (J.G.); (C.C.); (M.W.); (J.H.)
| | - Jun Hu
- Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, China; (J.G.); (C.C.); (M.W.); (J.H.)
| | - Lixia Pei
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| | - Jianhua Sun
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| |
Collapse
|
41
|
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
|
42
|
Kang T, Yang Z, Zhou M, Lan Y, Hong Y, Gong X, Wu Y, Li M, Chen X, Zhang W. The role of the Piezo1 channel in osteoblasts under cyclic stretching: A study on osteogenic and osteoclast factors. Arch Oral Biol 2024; 163:105963. [PMID: 38608563 DOI: 10.1016/j.archoralbio.2024.105963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/10/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024]
Abstract
OBJECTIVES Orthodontic tooth movement is a mechanobiological reaction induced by appropriate forces, including bone remodeling. The mechanosensitive Piezo channels have been shown to contribute to bone remodeling. However, information about the pathways through which Piezo channels affects osteoblasts remains limited. Thus, we aimed to investigate the influence of Piezo1 on the osteogenic and osteoclast factors in osteoblasts under mechanical load. MATERIALS AND METHODS Cyclic stretch (CS) experiments on MC3T3-E1 were conducted using a BioDynamic mechanical stretching device. The Piezo1 channel blocker GsMTx4 and the Piezo1 channel agonist Yoda1 were used 12 h before the application of CS. MC3T3-E1 cells were then subjected to 15% CS, and the expression of Piezo1, Piezo2, BMP-2, OCN, Runx2, RANKL, p-p65/p65, and ALP was measured using quantitative real-time polymerase chain reaction, western blot, alkaline phosphatase staining, and immunofluorescence staining. RESULTS CS of 15% induced the highest expression of Piezo channel and osteoblast factors. Yoda1 significantly increased the CS-upregulated expression of Piezo1 and ALP activity but not Piezo2 and RANKL. GsMTx4 downregulated the CS-upregulated expression of Piezo1, Piezo2, Runx2, OCN, p-65/65, and ALP activity but could not completely reduce CS-upregulated BMP-2. CONCLUSIONS The appropriate force is more suitable for promoting osteogenic differentiation in MC3T3-E1. The Piezo1 channel participates in osteogenic differentiation of osteoblasts through its influence on the expression of osteogenic factors like BMP-2, Runx2, and OCN and is involved in regulating osteoclasts by influencing phosphorylated p65. These results provide a foundation for further exploration of osteoblast function in orthodontic tooth movement.
Collapse
Affiliation(s)
- Ting Kang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Ziyuan Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Mengqi Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yanhua Lan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yaya Hong
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xinyi Gong
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yongjia Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Min Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuepeng Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China.
| | - Weifang Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
43
|
Byun KA, Lee JH, Lee SY, Oh S, Batsukh S, Cheon GW, Lee D, Hong JH, Son KH, Byun K. Piezo1 Activation Drives Enhanced Collagen Synthesis in Aged Animal Skin Induced by Poly L-Lactic Acid Fillers. Int J Mol Sci 2024; 25:7232. [PMID: 39000341 PMCID: PMC11242599 DOI: 10.3390/ijms25137232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Poly L-lactic acid (PLLA) fillers stimulate collagen synthesis by activating various immune cells and fibroblasts. Piezo1, an ion channel, responds to mechanical stimuli, including changes in extracellular matrix stiffness, by mediating Ca2+ influx. Given that elevated intracellular Ca2+ levels trigger signaling pathways associated with fibroblast proliferation, Piezo1 is a pivotal regulator of collagen synthesis and tissue fibrosis. The aim of the present study was to investigate the impact of PLLA on dermal collagen synthesis by activating Piezo1 in both an H2O2-induced cellular senescence model in vitro and aged animal skin in vivo. PLLA elevated intracellular Ca2+ levels in senescent fibroblasts, which was attenuated by the Piezo1 inhibitor GsMTx4. Furthermore, PLLA treatment increased the expression of phosphorylated ERK1/2 to total ERK1/2 (pERK1/2/ERK1/2) and phosphorylated AKT to total AKT (pAKT/AKT), indicating enhanced pathway activation. This was accompanied by upregulation of cell cycle-regulating proteins (CDK4 and cyclin D1), promoting the proliferation of senescent fibroblasts. Additionally, PLLA promoted the expression of phosphorylated mTOR/S6K1/4EBP1, TGF-β, and Collagen I/III in senescent fibroblasts, with GsMTx4 treatment mitigating these effects. In aged skin, PLLA treatment similarly upregulated the expression of pERK1/2/ERK1/2, pAKT/AKT, CDK4, cyclin D1, mTOR/S6K1/4EBP1, TGF-β, and Collagen I/III. In summary, our findings suggest Piezo1's involvement in PLLA-induced collagen synthesis, mediated by heightened activation of cell proliferation signaling pathways such as pERK1/2/ERK1/2, pAKT/AKT, and phosphorylated mTOR/S6K1/4EBP1, underscoring the therapeutic potential of PLLA in tissue regeneration.
Collapse
Affiliation(s)
- Kyung-A Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- LIBON Inc., Incheon 22006, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Je Hyuk Lee
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Doctorbom Clinic, Seoul 06614, Republic of Korea
| | - So Young Lee
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Sosorburam Batsukh
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Gwahn-woo Cheon
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Maylin Clinic, Pangyo 13529, Republic of Korea
| | - Dongun Lee
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea (J.H.H.)
| | - Jeong Hee Hong
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea (J.H.H.)
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kyunghee Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea (J.H.H.)
| |
Collapse
|
44
|
Wang HJ, Wang Y, Mirjavadi SS, Andersen T, Moldovan L, Vatankhah P, Russell B, Jin J, Zhou Z, Li Q, Cox CD, Su QP, Ju LA. Microscale geometrical modulation of PIEZO1 mediated mechanosensing through cytoskeletal redistribution. Nat Commun 2024; 15:5521. [PMID: 38951553 PMCID: PMC11217425 DOI: 10.1038/s41467-024-49833-6] [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: 07/30/2023] [Accepted: 06/20/2024] [Indexed: 07/03/2024] Open
Abstract
The microgeometry of the cellular microenvironment profoundly impacts cellular behaviors, yet the link between it and the ubiquitously expressed mechanosensitive ion channel PIEZO1 remains unclear. Herein, we describe a fluorescent micropipette aspiration assay that allows for simultaneous visualization of intracellular calcium dynamics and cytoskeletal architecture in real-time, under varied micropipette geometries. By integrating elastic shell finite element analysis with fluorescent lifetime imaging microscopy and employing PIEZO1-specific transgenic red blood cells and HEK cell lines, we demonstrate a direct correlation between the microscale geometry of aspiration and PIEZO1-mediated calcium signaling. We reveal that increased micropipette tip angles and physical constrictions lead to a significant reorganization of F-actin, accumulation at the aspirated cell neck, and subsequently amplify the tension stress at the dome of the cell to induce more PIEZO1's activity. Disruption of the F-actin network or inhibition of its mobility leads to a notable decline in PIEZO1 mediated calcium influx, underscoring its critical role in cellular mechanosensing amidst geometrical constraints.
Collapse
Affiliation(s)
- Haoqing Jerry Wang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW, 2042, Australia
| | - Yao Wang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Seyed Sajad Mirjavadi
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Tomas Andersen
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Laura Moldovan
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW, 2042, Australia
| | - Parham Vatankhah
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Blake Russell
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Jasmine Jin
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Zijing Zhou
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, NSW, 2010, Australia
| | - Qing Li
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Charles D Cox
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, NSW, 2010, Australia
- Faculty of Medicine, St. Vincent's Clinical School, University of New South Wale, Sydney, NSW, 2010, Australia
| | - Qian Peter Su
- Heart Research Institute, Camperdown, Newtown, NSW, 2042, Australia.
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
- Heart Research Institute, Camperdown, Newtown, NSW, 2042, Australia.
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
45
|
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
|
46
|
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
|
47
|
Ikiz ED, Hascup ER, Bae C, Hascup KN. Microglial Piezo1 mechanosensitive channel as a therapeutic target in Alzheimer's disease. Front Cell Neurosci 2024; 18:1423410. [PMID: 38957539 PMCID: PMC11217546 DOI: 10.3389/fncel.2024.1423410] [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: 04/25/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
Microglia are the resident macrophages of the central nervous system (CNS) that control brain development, maintain neural environments, respond to injuries, and regulate neuroinflammation. Despite their significant impact on various physiological and pathological processes across mammalian biology, there remains a notable gap in our understanding of how microglia perceive and transmit mechanical signals in both normal and diseased states. Recent studies have revealed that microglia possess the ability to detect changes in the mechanical properties of their environment, such as alterations in stiffness or pressure. These changes may occur during development, aging, or in pathological conditions such as trauma or neurodegenerative diseases. This review will discuss microglial Piezo1 mechanosensitive channels as potential therapeutic targets for Alzheimer's disease (AD). The structure, function, and modulation of Piezo1 will be discussed, as well as its role in facilitating microglial clearance of misfolded amyloid-β (Aβ) proteins implicated in the pathology of AD.
Collapse
Affiliation(s)
- Erol D. Ikiz
- Department of Chemistry, School of Integrated Sciences, Sustainability, and Public Health, College of Health, Science, and Technology, University of Illinois at Springfield, Springfield, IL, United States
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Erin R. Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, United States
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Chilman Bae
- School of Electrical, Computer, and Biomedical Engineering, Southern Illinois University at Carbondale, Carbondale, IL, United States
| | - Kevin N. Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, United States
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States
| |
Collapse
|
48
|
Morena F, Argentati C, Caponi S, Lüchtefeld I, Emiliani C, Vassalli M, Martino S. Piezo1 - Serine/threonine-protein phosphatase 2A - Cofilin1 biochemical mechanotransduction axis controls F-actin dynamics and cell migration. Heliyon 2024; 10:e32458. [PMID: 38933959 PMCID: PMC11201121 DOI: 10.1016/j.heliyon.2024.e32458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
This study sheds light on a ground-breaking biochemical mechanotransduction pathway and reveals how Piezo1 channels orchestrate cell migration. We observed an increased cell migration rate in HEK293T (HEK) cells treated with Yoda1, a Piezo1 agonist, or in HEK cells overexpressing Piezo1 (HEK + P). Conversely, a significant reduction in cell motility was observed in HEK cells treated with GsMTx4 (a channel inhibitor) or upon silencing Piezo1 (HEK-P). Our findings establish a direct correlation between alterations in cell motility, Piezo1 expression, abnormal F-actin microfilament dynamics, and the regulation of Cofilin1, a protein involved in severing F-actin microfilaments. Here, the conversion of inactive pCofilin1 to active Cofilin1, mediated by the serine/threonine-protein phosphatase 2A catalytic subunit C (PP2AC), resulted in increased severing of F-actin microfilaments and enhanced cell migration in HEK + P cells compared to HEK controls. However, this effect was negligible in HEK-P and HEK cells transfected with hsa-miR-133b, which post-transcriptionally inhibited PP2AC mRNA expression. In summary, our study suggests that Piezo1 regulates cell migration through a biochemical mechanotransduction pathway involving PP2AC-mediated Cofilin1 dephosphorylation, leading to changes in F-actin microfilament dynamics.
Collapse
Affiliation(s)
- Francesco Morena
- Department of Chemistry, Biology, and Biotechnologies, Via del Giochetto, University of Perugia, Perugia, Italy
| | - Chiara Argentati
- Department of Chemistry, Biology, and Biotechnologies, Via del Giochetto, University of Perugia, Perugia, Italy
| | - Silvia Caponi
- CNR, Istituto Officina dei Materiali-IOM c/o Dipartimento di Fisica e Geologia, University of Perugia, Perugia, Italy
| | - Ines Lüchtefeld
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Switzerland
| | - Carla Emiliani
- Department of Chemistry, Biology, and Biotechnologies, Via del Giochetto, University of Perugia, Perugia, Italy
| | | | - Sabata Martino
- Department of Chemistry, Biology, and Biotechnologies, Via del Giochetto, University of Perugia, Perugia, Italy
| |
Collapse
|
49
|
Lüchtefeld I, Pivkin IV, Gardini L, Zare-Eelanjegh E, Gäbelein C, Ihle SJ, Reichmuth AM, Capitanio M, Martinac B, Zambelli T, Vassalli M. Dissecting cell membrane tension dynamics and its effect on Piezo1-mediated cellular mechanosensitivity using force-controlled nanopipettes. Nat Methods 2024; 21:1063-1073. [PMID: 38802520 PMCID: PMC11166569 DOI: 10.1038/s41592-024-02277-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/10/2024] [Indexed: 05/29/2024]
Abstract
The dynamics of cellular membrane tension and its role in mechanosensing, which is the ability of cells to respond to physical stimuli, remain incompletely understood, mainly due to the lack of appropriate tools. Here, we report a force-controlled nanopipette-based method that combines fluidic force microscopy with fluorescence imaging for precise manipulation of the cellular membrane tension while monitoring the impact on single-cell mechanosensitivity. The force-controlled nanopipette enables control of the indentation force imposed on the cell cortex as well as of the aspiration pressure applied to the plasma membrane. We show that this setup can be used to concurrently monitor the activation of Piezo1 mechanosensitive ion channels via calcium imaging. Moreover, the spatiotemporal behavior of the tension propagation is assessed with the fluorescent membrane tension probe Flipper-TR, and further dissected using molecular dynamics modeling. Finally, we demonstrate that aspiration and indentation act independently on the cellular mechanobiological machinery, that indentation induces a local pre-tension in the membrane, and that membrane tension stays confined by links to the cytoskeleton.
Collapse
Affiliation(s)
- Ines Lüchtefeld
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland.
| | - Igor V Pivkin
- Institute of Computing, Università della Svizzera Italiana, Lugano, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Lucia Gardini
- National Institute of Optics, National Research Council, Florence, Italy
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Florence, Italy
| | | | | | - Stephan J Ihle
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland
| | - Andreas M Reichmuth
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland
| | - Marco Capitanio
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Florence, Italy
- Physics and Astronomy Department, University of Florence, Florence, Italy
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Tomaso Zambelli
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland.
| | - Massimo Vassalli
- James Watt School of Engineering, University of Glasgow, Glasgow, UK.
| |
Collapse
|
50
|
Fabiano AR, Robbins SC, Knoblauch SV, Rowland SJ, Dombroski JA, King MR. Multiplex, high-throughput method to study cancer and immune cell mechanotransduction. Commun Biol 2024; 7:674. [PMID: 38824207 PMCID: PMC11144229 DOI: 10.1038/s42003-024-06327-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/14/2024] [Indexed: 06/03/2024] Open
Abstract
Studying cellular mechanoresponses during cancer metastasis is limited by sample variation or complex protocols that current techniques require. Metastasis is governed by mechanotransduction, whereby cells translate external stimuli, such as circulatory fluid shear stress (FSS), into biochemical cues. We present high-throughput, semi-automated methods to expose cells to FSS using the VIAFLO96 multichannel pipetting device custom-fitted with 22 G needles, increasing the maximum FSS 94-fold from the unmodified tips. Specifically, we develop protocols to semi-automatically stain live samples and to fix, permeabilize, and intracellularly process cells for flow cytometry analysis. Our first model system confirmed that the pro-apoptotic effects of TRAIL therapeutics in prostate cancer cells can be enhanced via FSS-induced Piezo1 activation. Our second system implements this multiplex methodology to show that FSS exposure (290 dyn cm-2) increases activation of murine bone marrow-derived dendritic cells. These methodologies greatly improve the mechanobiology workflow, offering a high-throughput, multiplex approach.
Collapse
Affiliation(s)
- Abigail R Fabiano
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN, 37212, USA
| | - Spencer C Robbins
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN, 37212, USA
| | - Samantha V Knoblauch
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN, 37212, USA
| | - Schyler J Rowland
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN, 37212, USA
| | - Jenna A Dombroski
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN, 37212, USA
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN, 37212, USA.
| |
Collapse
|