1
|
Wang J, Fu C, Chang S, Stephens C, Li H, Wang D, Fu YC, Green KJ, Yan J, Yi R. PIEZO1-mediated calcium signaling reinforces mechanical properties of hair follicle stem cells to promote quiescence. SCIENCE ADVANCES 2025; 11:eadt2771. [PMID: 40435254 PMCID: PMC12118625 DOI: 10.1126/sciadv.adt2771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 04/23/2025] [Indexed: 06/01/2025]
Abstract
The mechanisms by which epithelial stem cells (SCs) sense mechanical cues within their niche and convert the information into biochemical signals to govern their function are not well understood. Here, we show that hair follicle SCs (HF-SCs) sense mechanical forces through cell adhesion and maintain quiescence in a PIEZO1-dependent mechanism. PIEZO1 interacts with E-cadherin in HF-SCs, and mechanical pulling of E-cadherin with a force of ~20 pN triggers PIEZO1-dependent, localized calcium flickers. Deletion of Piezo1 leads to reduced cumulative calcium influx and compromises quiescence. Single-cell genomic analyses identify a transcriptional network involving AP1 and NFATC1, which functions downstream of PIEZO1 and regulates the expression of extracellular matrix, cell adhesion, and actin cytoskeleton genes to reinforce the unique mechanical property of HF-SCs. These findings establish the force threshold necessary for PIEZO1 activation and reveal PIEZO1-dependent calcium influx as a key mechanism for sensing mechanical cues in the niche and regulating HF-SC activity.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Chaoyu Fu
- Department of Physics, National University of Singapore, Singapore 117551, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Sophie Chang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Christopher Stephens
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Haimin Li
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Dongmei Wang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Yuheng C. Fu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Kathleen J. Green
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Jie Yan
- Department of Physics, National University of Singapore, Singapore 117551, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Rui Yi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| |
Collapse
|
2
|
Santiago C, Siegrist J, Africawala N, Handler A, Tasnim A, Anjum R, Turecek J, Lehnert BP, Renauld S, Choi J, Nolan-Tamariz M, Iskols M, Magee AR, Paradis S, Sharma N, Ginty DD. Activity-dependent development of the body's touch receptors. Neuron 2025:S0896-6273(25)00298-3. [PMID: 40381613 DOI: 10.1016/j.neuron.2025.04.015] [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: 09/22/2023] [Revised: 02/12/2025] [Accepted: 04/16/2025] [Indexed: 05/20/2025]
Abstract
We report a role for activity in the development of the primary sensory neurons that detect touch. Genetic deletion of Piezo2, the principal mechanosensitive ion channel in somatosensory neurons, caused profound changes in the formation of mechanosensory end-organ structures. Peripheral-nervous-system-specific deletion of the voltage-gated sodium channel Nav1.6 (Scn8a), which resulted in altered electrophysiological responses to mechanical stimuli, also disrupted somatosensory neuron morphologies, supporting a role for neuronal activity in end-organ formation. Single-cell RNA sequencing of Piezo2 mutants revealed changes in gene expression in sensory neurons activated by light mechanical forces, whereas other neuronal classes were minimally affected, and genetic deletion of Piezo2-dependent genes partially reproduced the defects in mechanosensory neuron structures observed in Piezo2 mutants. These findings indicate that mechanically evoked neuronal activity acts early in life to shape the maturation of mechanosensory end-organs that underlie our sense of gentle touch.
Collapse
Affiliation(s)
- Celine Santiago
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Julianna Siegrist
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Nusrat Africawala
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Annie Handler
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Aniqa Tasnim
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA
| | - Josef Turecek
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Brendan P Lehnert
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Sophia Renauld
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jinheon Choi
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Nolan-Tamariz
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Alexandra R Magee
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA
| | - Nikhil Sharma
- Department of Biochemistry and Molecular Biophysics and Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Medical Center, New York, NY 10032, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
3
|
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
|
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
|
Ghosh S, Bishnoi B, Das S. Artery regeneration: Molecules, mechanisms and impact on organ function. Semin Cell Dev Biol 2025; 171:103611. [PMID: 40318557 DOI: 10.1016/j.semcdb.2025.103611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 05/07/2025]
Abstract
Replenishment of artery cells to repair or create new arteries is a promising strategy to re-vascularize ischemic tissue. However, limited understanding of cellular and molecular programs associated with artery (re-)growth impedes our efforts towards designing optimal therapeutic approaches. In this review, we summarize different cellular mechanisms that drive injury-induced artery regeneration in distinct organs and organisms. Artery formation during embryogenesis includes migration, self-amplification, and changes in cell fates. These processes are coordinated by multiple signaling pathways, like Vegf, Wnt, Notch, Cxcr4; many of which, also involved in injury-induced vascular responses. We also highlight how physiological and environmental factors determine the extent of arterial re-vascularization. Finally, we discuss different in vitro cellular reprogramming and tissue engineering approaches to promote artery regeneration, in vivo. This review provides the current understanding of endothelial cell fate reprogramming and explores avenues for regenerating arteries to restore organ function through efficient revascularization.
Collapse
Affiliation(s)
- Swarnadip Ghosh
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Bhavnesh Bishnoi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Soumyashree Das
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India.
| |
Collapse
|
6
|
Xie Z, Rose L, Feng J, Zhao Y, Lu Y, Kane H, Hibberd TJ, Hu X, Wang Z, Zang K, Yang X, Richardson Q, Othman R, Venezia O, Zhakyp A, Gao F, Abe N, Vigeland K, Wang H, Branch C, Duizer C, Deng L, Meng X, Zamidar L, Hauptschein M, Bergin R, Dong X, Chiu IM, Kim BS, Spencer NJ, Hu H, Jackson R. Enteric neuronal Piezo1 maintains mechanical and immunological homeostasis by sensing force. Cell 2025; 188:2417-2432.e19. [PMID: 40132579 PMCID: PMC12048284 DOI: 10.1016/j.cell.2025.02.031] [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: 05/06/2024] [Revised: 11/19/2024] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
The gastrointestinal (GI) tract experiences a myriad of mechanical forces while orchestrating digestion and barrier immunity. A central conductor of these processes, the enteric nervous system (ENS), detects luminal pressure to regulate peristalsis independently of extrinsic input from the central and peripheral nervous systems. However, how the ∼500 million enteric neurons that reside in the GI tract sense and respond to force remains unknown. Herein, we establish that the mechanosensor Piezo1 is functionally expressed in cholinergic enteric neurons. Optogenetic stimulation of Piezo1+ cholinergic enteric neurons drives colonic motility, while Piezo1 deficiency reduces cholinergic neuronal activity and slows peristalsis. Additionally, Piezo1 deficiency in cholinergic enteric neurons abolishes exercise-induced acceleration of GI motility. Finally, we uncover that enteric neuronal Piezo1 function is required for motility alterations in colitis and acts to prevent aberrant inflammation and tissue damage. This work uncovers how the ENS senses and responds to mechanical force.
Collapse
Affiliation(s)
- Zili Xie
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Lillian Rose
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Feng
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA; Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yonghui Zhao
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Yisi Lu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Harry Kane
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy J Hibberd
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Xueming Hu
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Zhen Wang
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Kaikai Zang
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Xingliang Yang
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | | | - Rahmeh Othman
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Olivia Venezia
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ademi Zhakyp
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Fang Gao
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Nobuya Abe
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Keren Vigeland
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Hongshen Wang
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Camren Branch
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Coco Duizer
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Liwen Deng
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Xia Meng
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Lydia Zamidar
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Max Hauptschein
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ronan Bergin
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Issac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Brian S Kim
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, New York, NY 10029, USA
| | - Nick J Spencer
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Hongzhen Hu
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, New York, NY 10029, USA; The Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Ruaidhrí Jackson
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Richa P, Häring M, Wang Q, Choudhury AR, Göpfert MC, Wolf F, Großhans J, Kong D. Synchronization in epithelial tissue morphogenesis. Curr Biol 2025:S0960-9822(25)00382-3. [PMID: 40239658 DOI: 10.1016/j.cub.2025.03.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/28/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025]
Abstract
Coordination of cell behavior is central to morphogenesis, when arrays of cells simultaneously undergo shape changes or dynamic rearrangements. In epithelia, cell shape changes invariably exert mechanical forces, which adjacent cells could sense to trigger an active response. However, molecular mechanisms for such mechano-transduction and especially their role for tissue-wide coordination in morphogenesis have remained ambiguous. Here, we investigate the function of Tmc, a key component of cellular mechano-transduction in vertebrate hearing, for coordination of cell dynamics in the epithelial amnioserosa of Drosophila embryos. We directly probed cell-cell mechano-transduction in vivo by opto-chemically inducing single-cell contractions and discovered a Tmc-dependent contraction response in neighboring cell groups. On the tissue scale, we uncover synchronization of neighboring cell area oscillations, which is impaired in Tmc mutants. A data-driven model of Tmc-dependent cell-cell interactions predicts that synchronization leads to an isotropic force map and effectively shields the tissue from external mechanical pulling. By microdissection, we detect equal junction tension along the axial and lateral axis in wild-type but increased lateral tension in Tmc mutants. Thus, Tmc transduces forces into an intracellular response that coordinates mechanical cell behavior in epithelial tissue.
Collapse
Affiliation(s)
- Prachi Richa
- Department of Biology, Philipps University, Hans-Meerwein-Straße 6, Marburg 35043, Germany; Göttingen Campus Institute for Dynamics of Biological Networks (CIDBN), Georg August University, Heinrich-Düker-Weg 12, Göttingen 37073, Germany
| | - Matthias Häring
- Göttingen Campus Institute for Dynamics of Biological Networks (CIDBN), Georg August University, Heinrich-Düker-Weg 12, Göttingen 37073, Germany; Institute for Nonlinear Dynamics, Georg August University, Friedrich-Hund-Pl. 1, Göttingen 37077, Germany; Max Planck Institute for Dynamics and Self-Organization, Am Faßberg 17, Göttingen 37077, Germany
| | - Qiyan Wang
- Department of Biology, Philipps University, Hans-Meerwein-Straße 6, Marburg 35043, Germany
| | - Ankit Roy Choudhury
- Department of Biology, Philipps University, Hans-Meerwein-Straße 6, Marburg 35043, Germany
| | - Martin C Göpfert
- Department of Cellular Neurobiology, Georg August University, Julia-Lermontowa-Weg 3, Göttingen 37077, Germany
| | - Fred Wolf
- Göttingen Campus Institute for Dynamics of Biological Networks (CIDBN), Georg August University, Heinrich-Düker-Weg 12, Göttingen 37073, Germany; Institute for Nonlinear Dynamics, Georg August University, Friedrich-Hund-Pl. 1, Göttingen 37077, Germany; Max Planck Institute for Dynamics and Self-Organization, Am Faßberg 17, Göttingen 37077, Germany; Max Planck Institute for Multidisciplinary Sciences, Hermann Rein Straße 3, Göttingen 37075, Germany.
| | - Jörg Großhans
- Department of Biology, Philipps University, Hans-Meerwein-Straße 6, Marburg 35043, Germany; Göttingen Campus Institute for Dynamics of Biological Networks (CIDBN), Georg August University, Heinrich-Düker-Weg 12, Göttingen 37073, Germany.
| | - Deqing Kong
- Department of Biology, Philipps University, Hans-Meerwein-Straße 6, Marburg 35043, Germany; Göttingen Campus Institute for Dynamics of Biological Networks (CIDBN), Georg August University, Heinrich-Düker-Weg 12, Göttingen 37073, Germany; Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Yushan Road 5, Qingdao 266003, China.
| |
Collapse
|
8
|
Wada H, Abe M, Wada N, Yoshimoto S, Fujii S, Moriyama M, Mori Y, Kido MA, Kiyoshima T. Piezo1 Is Related to the Enamel Matrix Formation in Mouse Tooth Germ Development. J Cell Physiol 2025; 240:e70036. [PMID: 40237554 DOI: 10.1002/jcp.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 03/23/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
Cellular responses to mechanical stimulation are involved in tissue development and the maintenance of biological functions. Teeth function as receptors for mastication and occlusal pressure. During tooth development, the tooth germ begins with an invagination of the epithelium, and its morphology matures through dynamic interactions between epithelial cells and mesenchymal cells, suggesting that mechanosensors may play an important role in this process. We analyzed the expression and function of Piezo1, a mechanically activated ion channel, during tooth development and clarified the involvement of Piezo1 in tooth morphogenesis. The expression of Piezo1 was observed in both the enamel organ and the surrounding mesenchymal cells at the early stage and in the ameloblasts and odontoblasts during enamel and dentin matrix formation. Yoda1, a Piezo1 activator, inhibited cell proliferation in mouse dental epithelial (mDE6) cells and E15 tooth germs, and suppressed cell migration in mDE6 cells. Meanwhile, GsMTx4, a Piezo1 inactivator, showed opposite results. Furthermore, in the organ culture of E15 tooth germs, the activation and inactivation of Piezo1 were found to affect the expression of ameloblast differentiation marker genes and control the arrangement of ameloblasts. Interestingly, the expression of E-cadherin was reduced in the cell membrane of ameloblasts at the cusp in the GsMTx4-treated tooth germs of organ culture, and enamel formation was significantly decreased. Yoda1-treated mDE6 cells showed upregulated E-cadherin expression, which was downregulated by calpain inhibitor. These findings suggest that Piezo1 may be involved in tooth morphogenesis during ameloblast development by playing an essential role in cell proliferation, migration, arrangement, differentiation, and mineralization.
Collapse
Affiliation(s)
- Hiroko Wada
- Section of Pathology, Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka, Japan
| | - Misaki Abe
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Naohisa Wada
- Department of General Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shohei Yoshimoto
- Section of Pathology, Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Shinsuke Fujii
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Masafumi Moriyama
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yoshihide Mori
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Mizuho A Kido
- Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Tamotsu Kiyoshima
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
9
|
Alibrandi S, Rinaldi C, Vinci SL, Conti A, Donato L, Scimone C, Sidoti A, D’Angelo R. Mechanotransduction in Development: A Focus on Angiogenesis. BIOLOGY 2025; 14:346. [PMID: 40282211 PMCID: PMC12024848 DOI: 10.3390/biology14040346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
Cells respond to external mechanical cues and transduce these forces into biological signals. This process is known as mechanotransduction and requires a group of proteins called mechanosensors. This peculiar class of receptors include extracellular matrix proteins, plasma membrane proteins, the cytoskeleton and the nuclear envelope. These cell components are responsive to a wide spectrum of physical cues including stiffness, tensile force, hydrostatic pressure and shear stress. Among mechanotransducers, the Transient Receptor Potential (TRP) and the PIEZO family members are mechanosensitive ion channels, coupling force transduction with intracellular cation transport. Their activity contributes to embryo development, tissue remodeling and repair, and cell homeostasis. In particular, vessel development is driven by hemodynamic cues such as flow direction and shear stress. Perturbed mechanotransduction is involved in several pathological vascular phenotypes including hereditary hemorrhagic telangiectasia. This review is conceived to summarize the most recent findings of mechanotransduction in development. We first collected main features of mechanosensitive proteins. However, we focused on the role of mechanical cues during development. Mechanosensitive ion channels and their function in vascular development are also discussed, with a focus on brain vessel morphogenesis.
Collapse
Affiliation(s)
- Simona Alibrandi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Istituto Euro-Mediterraneo di Scienza e Tecnologia (I.E.ME.S.T.), Street Michele Miraglia 20, 90139 Palermo, Italy
| | - Carmela Rinaldi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
| | - Sergio Lucio Vinci
- Neuroradiology Unit, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
| | - Alfredo Conti
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Street Altura 3, 40123 Bologna, Italy
- Department of Biomedical and NeuroMotor Sciences (DiBiNeM), Alma Mater Studiorum—University of Bologna, 40127 Bologna, Italy
| | - Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Istituto Euro-Mediterraneo di Scienza e Tecnologia (I.E.ME.S.T.), Street Michele Miraglia 20, 90139 Palermo, Italy
| | - Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Istituto Euro-Mediterraneo di Scienza e Tecnologia (I.E.ME.S.T.), Street Michele Miraglia 20, 90139 Palermo, Italy
| | - Rosalia D’Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
| |
Collapse
|
10
|
Beverley KM, Ahn SJ, Levitan I. Flow-sensitive ion channels in vascular endothelial cells: Mechanisms of activation and roles in mechanotransduction. Biophys J 2025:S0006-3495(25)00193-6. [PMID: 40156185 DOI: 10.1016/j.bpj.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/06/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
The purpose of this review is to evaluate the current knowledge about the mechanisms by which mechanosensitive ion channels are activated by fluid shear stress in endothelial cells. We focus on three classes of endothelial ion channels that are most well studied for their sensitivity to flow and roles in mechanotransduction: inwardly rectifying K+ channels, Piezo channels, and TRPV channels. We also discuss the mechanisms by which these channels initiate and contribute to mechanosensitive signaling pathways. Three types of mechanisms have been described for flow-induced activation of ion channels: 1) through interaction with apical membrane flow sensors, such as glycocalyx, which is likely to be deformed by flow, 2) directly by sensing membrane stretch that is induced by shear stress, or 3) via flow-sensitive channel-channel or lipid channel interactions. We also demonstrate the physiological role of these channels and how they are related to cardiovascular and neurological diseases. Further studies are needed to determine how these channels function cooperatively to mediate the endothelial response to flow.
Collapse
Affiliation(s)
- Katie M Beverley
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois.
| | - Sang Joon Ahn
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Irena Levitan
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
11
|
Zhang Y, Shan KZ, Liang P, Lowry AJ, Feng L, Yang H. PIEZO1 Drives Trophoblast Fusion and Placental Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645313. [PMID: 40196526 PMCID: PMC11974931 DOI: 10.1101/2025.03.25.645313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
PIEZO1, a mechanosensor1,2 in endothelial cells, plays a critical role in fetal vascular development during embryogenesis3,4. However, its expression and function in placental trophoblasts remain unexplored. Here, we demonstrate that PIEZO1 is expressed in placental villus trophoblasts, where it is essential for trophoblast fusion and placental development. Mice with trophoblast-specific PIEZO1 knockout exhibit embryonic lethality without obvious vascular defects. Instead, PIEZO1 deficiency disrupts the formation of the syncytiotrophoblast layer in the placenta. Mechanistically, PIEZO1-mediated calcium influx activates TMEM16F lipid scramblase, facilitating the externalization of phosphatidylserine, a key "fuse-me" signal for trophoblast fusion5,6. These findings reveal PIEZO1 as a crucial mechanosensor in trophoblasts and highlight its indispensable role in trophoblast fusion and placental development, expanding our understanding of PIEZO1's functions beyond endothelial cells during pregnancy.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biochemistry, Duke University School of Medicine, NC 27710, USA
| | - Ke Z. Shan
- Department of Biochemistry, Duke University School of Medicine, NC 27710, USA
| | - Pengfei Liang
- Department of Biochemistry, Duke University School of Medicine, NC 27710, USA
| | - Augustus J. Lowry
- Department of Biochemistry, Duke University School of Medicine, NC 27710, USA
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University School of Medicine, NC 27710, USA
| | - Huanghe Yang
- Department of Biochemistry, Duke University School of Medicine, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, NC 27710, USA
- Department of Neurobiology, Duke University School of Medicine, NC 27710, USA
| |
Collapse
|
12
|
Nakazawa N, Grenci G, Kameo Y, Takeda N, Sawada T, Kurisu J, Zhang Z, Toma K, Adachi T, Nonomura K, Kengaku M. PIEZO1-dependent mode switch of neuronal migration in heterogeneous microenvironments in the developing brain. Cell Rep 2025; 44:115405. [PMID: 40053456 DOI: 10.1016/j.celrep.2025.115405] [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: 05/11/2024] [Revised: 01/09/2025] [Accepted: 02/14/2025] [Indexed: 03/09/2025] Open
Abstract
The migration of newborn neurons is essential for brain morphogenesis and circuit formation, yet controversy exists regarding how neurons generate the driving force against strong mechanical stresses in crowded neural tissues. We found that cerebellar granule neurons employ a mechanosensing mechanism to switch the driving forces to maneuver in irregular brain tissue. In two-dimensional (2D) cultures, actomyosin is enriched in the leading process, exerting traction force on the cell soma. In tissue or 3D confinement, however, actomyosin concentrates at the posterior cell membrane, generating contractile forces that assist passage through narrow spaces, working alongside the traction force in the leading process. The 3D migration is initiated by the activation of a mechanosensitive channel, PIEZO1. PIEZO1-induced calcium influx in the soma triggers the PKC-ezrin cascade, which recruits actomyosin and transmits its contractile force to the posterior plasma membrane. Thus, migrating neurons adapt their motility modes in distinct extracellular environments in the developing brain.
Collapse
Affiliation(s)
- Naotaka Nakazawa
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Faculty of Science and Engineering, Kindai University, Osaka 577-8502, Japan.
| | - Gianluca Grenci
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; Biomedical Engineering Department, National University of Singapore, Singapore 117583, Singapore
| | - Yoshitaka Kameo
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan; Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8501, Japan; Graduate School of Engineering, Kyoto University, Kyoto 615-8530, Japan; College of Engineering, Shibaura Institute of Technology, Tokyo 135-8548, Japan
| | - Noriko Takeda
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Tsuyoshi Sawada
- Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8501, Japan; Graduate School of Engineering, Kyoto University, Kyoto 615-8530, Japan
| | - Junko Kurisu
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Zhejing Zhang
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Kenichi Toma
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Taiji Adachi
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan; Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8501, Japan; Graduate School of Engineering, Kyoto University, Kyoto 615-8530, Japan
| | - Keiko Nonomura
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan; Department of Life Science and Technology, Tokyo Institute of Technology, Kanagawa 226-8501, Japan; National Institute for Basic Biology, Aichi 444-8585, Japan
| | - Mineko Kengaku
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
13
|
Niu W, Liu X, Deng B, Hong T, Wang C, Yan Y, Liu J, Jiang Y, Li J. Piezo1 deletion mitigates diabetic cardiomyopathy by maintaining mitochondrial dynamics via ERK/Drp1 pathway. Cardiovasc Diabetol 2025; 24:127. [PMID: 40114130 PMCID: PMC11927149 DOI: 10.1186/s12933-025-02625-8] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE Increasing evidence highlights the critical role of Piezo1 in cardiovascular diseases, with its expression upregulated in diabetic heart. However, the involvement of Piezo1 in the pathogenesis of diabetic cardiomyopathy (DCM) remains unclear. This study aims to elucidate the regulatory role of Piezo1 in mitochondrial dynamics within the context of DCM and to investigate the underlying mechanisms. METHODS We constructed cardiac-specific knockout of Piezo1 (Piezo1∆Myh6) mice. Type 1 diabetes was induced using streptozotocin (STZ) injection while type 2 diabetes was established through a high-fat diet combined with STZ. Echocardiography assessed left ventricular function, histological evaluations used HE and Masson staining to examine cardiac pathology in Piezo1fl/fl controls, Piezo1∆Myh6 controls, Piezo1fl/fl diabetic and Piezo1∆Myh6 diabetic mice. Mitochondrial function including oxygen species level, mitochondrial morphology, and respiration rate were also assessed. RESULTS Our findings revealed that Piezo1 expression was upregulated in the myocardium of diabetic mice and in high-glucose-treated cells. Cardiac-specific knockout of Piezo1 improved cardiac dysfunction and ameliorated cardiac fibrosis in diabetic mice. Moreover, Piezo1 deficiency also attenuated mitochondrial impairment. Piezo1fl/fl diabetic mice exhibited increased calpain activity and excessive mitochondrial fission mediated by Drp1 and obvious reduced fusion; however, Piezo1 deficiency restored calpain levels and mitochondrial dysfunction. These observations were also corroborated in H9C2 cells and neonatal mouse cardiomyocytes. Cardiac-specific knockout of Piezo1 increased phosphorylation of Drp1 and ERK1/2 in vivo and in vitro. Piezo1 knockout or treatment with inhibitor improved mitochondrial function. CONCLUSIONS This study provides the first evidence that Piezo1 is elevated in DCM through the modulation of mitochondrial dynamics, which is reversed by Piezo1 deficiency. Thus, Piezo1 inhibition may provide a promising therapeutic strategy for the treatment of DCM.
Collapse
MESH Headings
- Animals
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/prevention & control
- Diabetic Cardiomyopathies/physiopathology
- Diabetic Cardiomyopathies/etiology
- Mitochondrial Dynamics
- Dynamins/metabolism
- Mice, Knockout
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/pathology
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/chemically induced
- Ion Channels/genetics
- Ion Channels/deficiency
- Ion Channels/metabolism
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Male
- Fibrosis
- Mice, Inbred C57BL
- Phosphorylation
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/enzymology
- Diabetes Mellitus, Type 1/complications
- Ventricular Function, Left
- Extracellular Signal-Regulated MAP Kinases/metabolism
- MAP Kinase Signaling System
- Rats
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/complications
- Mice
Collapse
Affiliation(s)
- Weipin Niu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Central Laboratory, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250014, China
| | - Xin Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Shandong Institute of Commerce and Technology, Jinan, 250103, China
| | - Bo Deng
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Tianying Hong
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Cuifen Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yameng Yan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jiali Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yuehua Jiang
- Central Laboratory, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250014, China.
| | - Jing Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
14
|
Moore Zajic EL, Zhao R, McKinney MC, Yi K, Wood C, Trainor PA. Cell extrusion drives neural crest cell delamination. Proc Natl Acad Sci U S A 2025; 122:e2416566122. [PMID: 40063802 PMCID: PMC11929498 DOI: 10.1073/pnas.2416566122] [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: 08/26/2024] [Accepted: 01/17/2025] [Indexed: 03/15/2025] Open
Abstract
Neural crest cells (NCC) comprise a heterogeneous population of cells with variable potency that contribute to nearly every tissue and organ throughout the body. Considered unique to vertebrates, NCC are transiently generated within the dorsolateral region of the neural plate or neural tube during neurulation. Their delamination and migration are crucial for embryo development as NCC differentiation is influenced by their final resting locations. Previous work in avian and aquatic species revealed that NCC delaminate via an epithelial-mesenchymal transition (EMT), which transforms these progenitor cells from static polarized epithelial cells into migratory mesenchymal cells with fluid front and back polarity. However, the cellular and molecular mechanisms facilitating NCC delamination in mammals are poorly understood. Through time-lapse imaging of NCC delamination in mouse embryos, we identified a subset of cells that exit the neuroepithelium as isolated round cells, which then halt for a short period prior to acquiring the mesenchymal migratory morphology classically associated with delaminating NCC. High-magnification imaging and protein localization analyses of the cytoskeleton, together with measurements of pressure and tension of delaminating NCC and neighboring neuroepithelial cells, revealed that round NCC are extruded from the neuroepithelium prior to completion of EMT. Furthermore, cranial NCC are extruded through activation of the mechanosensitive ion channel, PIEZO1. Our results support a model in which cell density, pressure, and tension in the neuroepithelium result in activation of the live cell extrusion pathway and delamination of a subpopulation of NCC in parallel with EMT, which has implications for cell delamination in development and disease.
Collapse
Affiliation(s)
| | - Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO64110
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS66160
| | | | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO64110
| | | | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, MO64110
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS66160
| |
Collapse
|
15
|
Li Z, Jiang Q, Wei J, Dang D, Meng Z, Wu H. Piezo1 promotes the progression of necrotizing enterocolitis by activating the Ca2(+)/CaMKII-dependent pathway. Commun Biol 2025; 8:417. [PMID: 40074811 PMCID: PMC11904196 DOI: 10.1038/s42003-025-07821-6] [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: 07/28/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating inflammatory bowel necrosis of preterm infants with limited therapeutic approaches. Mounting evidence supports the role of Piezo1, namely, a widely distributed mechanosensor in intestinal epithelial cells (IECs), in intestinal inflammation but its underlying mechanism in the development of NEC remains unexplored. In this study, we demonstrated that Piezo1 expression was higher in preterm infants with lower gestational age. C57BL/6J mice wherein Piezo1 was deleted in IECs (villin-specific Piezo1 knockout mice; Piezo1flox/floxVillinCre+) and Piezo1flox/flox littermates were subjected to induce NEC, and Piezo1 knockout regulated the intestinal barrier function, restricted cytokines secretion, and diminished the inflammatory response in NEC mouse models. Piezo1 elevated cytosolic Ca2+ levels and activated Ca2+/calmodulin-dependent protein kinase II (CaMKII) to promote the CaMKII/NF-κB interaction and NF-κB activation in vitro. Finally, the effects of a CaMKII inhibitor, KN93, were evaluated both in vitro and in vivo in NEC models, and the functions of Piezo1 in IECs were suppressed partially by KN93. In this study, we characterise the undefined role of Piezo1 in the development of NEC, which may partially be attributed to the differential role of calcium under pathophysiological conditions.
Collapse
Affiliation(s)
- Zhenyu Li
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
- The Child Health Clinical Research Center of Jilin Province, Changchun, Jilin, China
| | - Qinlei Jiang
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
- The Child Health Clinical Research Center of Jilin Province, Changchun, Jilin, China
| | - Jiaqi Wei
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
- The Child Health Clinical Research Center of Jilin Province, Changchun, Jilin, China
| | - Dan Dang
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
- The Child Health Clinical Research Center of Jilin Province, Changchun, Jilin, China
| | - Zhaoli Meng
- Laboratory of Tumor Immunology, The First Hospital of Jilin University, Changchun, China
| | - Hui Wu
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China.
- The Child Health Clinical Research Center of Jilin Province, Changchun, Jilin, China.
| |
Collapse
|
16
|
Yue L, Pang B, Li G, Zhang C, Li Q, Li K, Fang H, Zhu Z, Shao S, Dang E, Wang G, Qiao P. Endothelial Piezo1 Mediates Barrier Dysfunction and NLRP3 Inflammasomes Activation in Psoriasis. J Invest Dermatol 2025:S0022-202X(25)00303-3. [PMID: 40054572 DOI: 10.1016/j.jid.2025.01.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/02/2025] [Accepted: 01/20/2025] [Indexed: 03/28/2025]
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by abnormal dilation of microvessels in the dermal papillary layer. Multiple studies have demonstrated that vascular endothelial cells regulate vascular function rather than merely acting as passive barriers. Piezo1 serves as a critical mechanical switch, sensing mechanical changes induced by vasodilation. However, the precise pathological role of Piezo1 in psoriasis remains unclear. In this study, we observed significant upregulation of Piezo1 in endothelial cells from the psoriatic dermis. Piezo1 activation impaired endothelial barrier function and promoted the expression of inflammatory factors. These alterations activated the NLRP3 inflammasome, which secretes IL-1β and IL-18, contributing to the local immune dysregulation characteristic of psoriasis. In addition, Piezo1 promoted mitochondrial ROS production and the release of mitochondrial DNA into the cytoplasm, thereby activating the NLRP3 inflammasome in endothelial cells. Furthermore, in mice with imiquimod-induced psoriasis-like dermatitis, Piezo1 suppression significantly alleviated the psoriasis-like phenotype, microvascular dilation, inflammatory infiltration, and NLRP3 inflammasome activation. Our findings suggest that the tortuous and dilated microvasculature in psoriatic skin disrupts vascular barriers and promotes NLRP3 inflammasome activation through Piezo1, contributing to the onset and progression of psoriasis.
Collapse
Affiliation(s)
- Lixin Yue
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Bingyu Pang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guohao Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chen Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qingyang Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kang Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui Fang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhenlai Zhu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shuai Shao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Pei Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
17
|
Abello J, Yin Y, Zhao Y, Maurer J, Lee J, Bodell C, Richee J, Clevenger AJ, Burton Z, Goeckel ME, Lin M, Grainger S, Halabi CM, Raghavan SA, Sah R, Stratman AN. Endothelial cell Piezo1 promotes vascular smooth muscle cell differentiation on large arteries. Eur J Cell Biol 2025; 104:151473. [PMID: 39729736 DOI: 10.1016/j.ejcb.2024.151473] [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: 06/28/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024] Open
Abstract
Vascular stabilization is a mechanosensitive process, in part driven by blood flow. Here, we demonstrate the involvement of the mechanosensitive ion channel, Piezo1, in promoting arterial accumulation of vascular smooth muscle cells (vSMCs) during zebrafish development. Using a series of small molecule antagonists or agonists to temporally regulate Piezo1 activity, we identified a role for the Piezo1 channel in regulating klf2a, a blood flow responsive transcription factor, expression levels and altered targeting of vSMCs between arteries and veins. Increasing Piezo1 activity suppressed klf2a and increased vSMC association with the cardinal vein, while inhibition of Piezo1 activity increased klf2a levels and decreased vSMC association with arteries. We supported the small molecule findings with in vivo genetic suppression of piezo1 and 2 in zebrafish, resulting in loss of transgelin+ vSMCs on the dorsal aorta. Further, endothelial cell (EC)-specific Piezo1 knockout in mice was sufficient to decrease vSMC accumulation along the descending dorsal aorta during development, thus phenocopying our zebrafish data, and supporting functional conservation of Piezo1 in mammals. To determine the underlying mechanism, we used in vitro modeling assays to demonstrate that differential sensing of pulsatile versus laminar flow forces across endothelial cells changes the expression of mural cell differentiation genes. Together, our findings suggest a crucial role for EC Piezo1 in sensing force within large arteries to mediate mural cell differentiation and stabilization of the arterial vasculature.
Collapse
Affiliation(s)
- Javier Abello
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Ying Yin
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Yonghui Zhao
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Josh Maurer
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Jihui Lee
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Cherokee Bodell
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jahmiera Richee
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Abigail J Clevenger
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Zarek Burton
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Megan E Goeckel
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Michelle Lin
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephanie Grainger
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Carmen M Halabi
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Shreya A Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Rajan Sah
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA; Center for Cardiovascular Research, Washington University, St Louis, MO, USA
| | - Amber N Stratman
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
18
|
Peralta M, Dupas A, Larnicol A, Lefebvre O, Goswami R, Stemmelen T, Molitor A, Carapito R, Girardo S, Osmani N, Goetz JG. Endothelial calcium firing mediates the extravasation of metastatic tumor cells. iScience 2025; 28:111690. [PMID: 39898056 PMCID: PMC11787530 DOI: 10.1016/j.isci.2024.111690] [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: 03/13/2024] [Revised: 11/08/2024] [Accepted: 12/23/2024] [Indexed: 02/04/2025] Open
Abstract
Metastatic dissemination is driven by genetic, biochemical, and biophysical cues that favor the distant colonization of organs and the formation of life-threatening secondary tumors. We have previously demonstrated that endothelial cells (ECs) actively remodel during extravasation by enwrapping arrested tumor cells (TCs) and extruding them from the vascular lumen while maintaining perfusion. In this work, we dissect the cellular and molecular mechanisms driving endothelial remodeling. Using high-resolution intravital imaging in zebrafish embryos, we demonstrate that the actomyosin network of ECs controls tissue remodeling and subsequent TC extravasation. Furthermore, we uncovered that this cytoskeletal remodeling is driven by altered endothelial-calcium (Ca2+) signaling caused by arrested TCs. Accordingly, we demonstrated that the inhibition of voltage-dependent calcium L-type channels impairs extravasation. Lastly, we identified P2X4, TRP, and Piezo1 mechano-gated Ca2+ channels as key mediators of the process. These results further highlight the central role of endothelial remodeling during the extravasation of TCs and open avenues for successful therapeutic targeting.
Collapse
Affiliation(s)
- Marina Peralta
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Amandine Dupas
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Annabel Larnicol
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Olivier Lefebvre
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Ruchi Goswami
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Tristan Stemmelen
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d’ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091 Strasbourg, France
| | - Anne Molitor
- Laboratoire d’ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
| | - Raphael Carapito
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d’ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091 Strasbourg, France
| | - Salvatore Girardo
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Naël Osmani
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Jacky G. Goetz
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| |
Collapse
|
19
|
Lacroix JJ, Wijerathne TD. PIEZO channels as multimodal mechanotransducers. Biochem Soc Trans 2025; 53:BST20240419. [PMID: 39936392 PMCID: PMC12010695 DOI: 10.1042/bst20240419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/13/2025]
Abstract
All living beings experience a wide range of endogenous and exogenous mechanical forces. The ability to detect these forces and rapidly convert them into specific biological signals is essential to a wide range of physiological processes. In vertebrates, these fundamental tasks are predominantly achieved by two related mechanosensitive ion channels called PIEZO1 and PIEZO2. PIEZO channels are thought to sense mechanical forces through flexible transmembrane blade-like domains. Structural studies indeed show that these mechanosensory domains adopt a curved conformation in a resting membrane but become flattened in a membrane under tension, promoting an open state. Yet, recent studies suggest the intriguing possibility that distinct mechanical stimuli activate PIEZO channels through discrete molecular rearrangements of these domains. In addition, biological signals downstream of PIEZO channel activation vary as a function of the mechanical stimulus and of the cellular context. These unique features could explain how PIEZOs confer cells the ability to differentially interpret a complex landscape of mechanical cues.
Collapse
Affiliation(s)
- Jérôme J Lacroix
- Department of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, U.S.A
| | - Tharaka D Wijerathne
- Department of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, U.S.A
| |
Collapse
|
20
|
Jia BZ, Tang X, Rossmann MP, Zon LI, Engert F, Cohen AE. Swimming motions evoke Ca 2+ events in vascular endothelial cells of larval zebrafish via mechanical activation of Piezo1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636757. [PMID: 39975374 PMCID: PMC11839014 DOI: 10.1101/2025.02.05.636757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Calcium signaling in blood vessels regulates their growth1,2, immune response3, and vascular tone4. Vascular endothelial cells are known to be mechanosensitive5-7, and it has been assumed that this mechanosensation mediates calcium responses to pulsatile blood flow8-10. Here we show that in larval zebrafish, the dominant trigger for vascular endothelial Ca2+ events comes from body motion, not heartbeat-driven blood flow. Through a series of pharmacological and mechanical perturbations, we showed that body motion is necessary and sufficient to induce endothelial Ca2+ events, while neither neural activity nor blood circulation is either necessary or sufficient. Knockout and temporally restricted knockdown of piezo1 eliminated the motion-induced Ca2+ events. Our results demonstrate that swimming-induced tissue motion is an important driver of endothelial Ca2+ dynamics in larval zebrafish.
Collapse
Affiliation(s)
- Bill Z. Jia
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Xin Tang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Marlies P. Rossmann
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Leonard I. Zon
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Florian Engert
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Adam E. Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
21
|
Han N, Yu N, Yu L. The mRNA Stability of PIEZO1, Regulated by Methyltransferase-Like 3 via N 6-Methylation of Adenosine Modification in a YT521-B Homology Domain Family 2-Dependent Manner, Facilitates the Progression of Diabetic Retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:265-280. [PMID: 39476953 DOI: 10.1016/j.ajpath.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/29/2024] [Accepted: 10/10/2024] [Indexed: 11/09/2024]
Abstract
Diabetic retinopathy (DR) is the major ocular complication of diabetes caused by chronic hyperglycemia, which leads to incurable blindness. Currently, the effectiveness of therapeutic interventions is limited. This study aimed to investigate the function of piezo-type mechanosensitive ion channel component 1 (PIEZO1) and its potential regulatory mechanism in DR progression. PIEZO1 expression was up-regulated in the retinal tissues of streptozotocin-induced diabetic mice and high-glucose (HG)-triggered Müller cells. Functionally, the knockdown of PIEZO1 improved the abnormal retinal function of diabetic mice and impeded inflammatory cytokine secretion and gliosis of Müller cells under HG conditions. Mechanistic investigations using RNA immunoprecipitation-real-time quantitative PCR, methylation RNA immunoprecipitation-real-time quantitative PCR, and luciferase reporter assays demonstrated that PIEZO1 was a downstream target of methyltransferase-like 3 (METTL3). METTL3-mediated N6-methyladenosine (m6A) modification within the coding sequence of PIEZO1 mRNA significantly shortened its half-life. In HG-stimulated cells, there was a negative regulatory relationship between PIEZO1 and YTH (YT521-B homology) domain family 2 (YTHDF2), a recognized m6A reader. The loss of YTHDF2 resulted in an extended half-life of PIEZO1 in cells with overexpression of METTL3, indicating that the effect of METTL3 on the mRNA stability of PIEZO1 was dependent on YTHDF2. Taken together, this study demonstrated the protective role of the PIEZO1 silencing in DR development, and that the degradation of PIEZO1 mRNA is accelerated by METTL3/YTHDF2-mediated m6A modification.
Collapse
Affiliation(s)
- Ning Han
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Na Yu
- Department of Blood Transfusion, The Second Hospital of Jilin University, Changchun, China
| | - Li Yu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
22
|
He Z, Du Y, Peng C, Xu B, Tang J, Liu R, Yang K. Piezo1 Promotes Osteogenesis Through CaMKII Signalling in a Rat Maxillary Expansion Model. Orthod Craniofac Res 2025; 28:196-206. [PMID: 39350680 DOI: 10.1111/ocr.12862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 01/07/2025]
Abstract
OBJECTIVES Rapid maxillary expansion (RME) is a widely used technique to treat maxillary transverse deficiency. Piezo1 is a cation channel that is activated by mechanical force and regulates bone formation. This study aims to elucidate the role of Piezo1 in bone remodelling during the RME process. MATERIALS AND METHODS In this study, the periosteal-derived stem cells (PDSCs) were cultured and stretched by the Flexcell system. The effects of Piezo1 on osteogenesis were assessed via RNA sequencing, real-time quantitative PCR, and western blot analyses. Moreover, for in vivo analyses, the rat RME model was established. The function of Piezo1 in mid-palatal suture bone remodelling was evaluated using micro-CT, haematoxylin-eosin (HE) staining, and immunohistochemistry analyses. RESULTS It was revealed that under tension force, the osteogenic factors (Runt-related transcription factor 2, Osterix, and Alkaline Phosphatase) and Ca2+/calmodulin -dependent protein kinase (CaMKII) were significantly enhanced in PDSCs over time. Furthermore, these were also upregulated in the RME model with the expansion of the mid-palatal suture. However, Piezo1 inhibition by Grammostola spatulata mechanotoxin 4 downregulated the levels of these factors in the RME model. CONCLUSIONS This study indicated that Piezo1 is associated with the osteogenesis of PDSCs and bone remodelling in the RME process. CaMKII might also participate in this process.
Collapse
Affiliation(s)
- Zhengquan He
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
- Department of Orthodontics, Changsha Stomatological Hospital, Changsha, China
| | - Yugui Du
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Chuhan Peng
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Bowen Xu
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Jiayue Tang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Runxuan Liu
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Kai Yang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Xu H, Chen X, Luo S, Jiang J, Pan X, He Y, Deng B, Liu S, Wan R, Lin L, Tan Q, Chen X, Yao Y, He B, An Y, Li J. Cardiomyocyte-specific Piezo1 deficiency mitigates ischemia-reperfusion injury by preserving mitochondrial homeostasis. Redox Biol 2025; 79:103471. [PMID: 39740362 PMCID: PMC11750285 DOI: 10.1016/j.redox.2024.103471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 12/15/2024] [Indexed: 01/02/2025] Open
Abstract
Ca2+ overload and mitochondrial dysfunction play crucial roles in myocardial ischemia-reperfusion (I/R) injury. Piezo1, a mechanosensitive cation channel, is essential for intracellular Ca2+ homeostasis. The objective of this research was to explore the effects of Piezo1 on mitochondrial function during myocardial I/R injury. We showed that the expression of myocardial Piezo1 was elevated in the infracted area of I/R and cardiomyocyte-specific Piezo1 deficiency (Piezo1△Myh6) mice attenuated I/R by decreasing infarct size and cardiac dysfunction. Piezo1△Myh6 regulated mitochondrial fusion and fission to improve mitochondrial function and decrease inflammation and oxidative stress in vivo and in vitro. Mechanistically, myocardial Piezo1 knockout alleviated intracellular calcium overload to normalize calpain-associated mitochondrial homeostasis. Our findings indicated that Piezo1 depletion in cardiomyocytes partially restored mitochondrial homeostasis during cardiac ischemia/reperfusion (I/R) injury. This study suggests an innovative therapeutic strategy to alleviate cardiac I/R injury.
Collapse
Affiliation(s)
- Honglin Xu
- Innovation Research Center, Shandong University of Traditional Chinese Medicine, Jinan, 250307, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xin Chen
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Shangfei Luo
- Innovation Research Center, Shandong University of Traditional Chinese Medicine, Jinan, 250307, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jintao Jiang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xianmei Pan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yu He
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710049, China
| | - Bo Deng
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Silin Liu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Rentao Wan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Liwen Lin
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qiaorui Tan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiaoting Chen
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Youfen Yao
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Bin He
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yajuan An
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jing Li
- Innovation Research Center, Shandong University of Traditional Chinese Medicine, Jinan, 250307, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, LS2 9JT, UK.
| |
Collapse
|
24
|
Bertaccini GA, Casanellas I, Evans EL, Nourse JL, Dickinson GD, Liu G, Seal S, Ly AT, Holt JR, Wijerathne TD, Yan S, Hui EE, Lacroix JJ, Panicker MM, Upadhyayula S, Parker I, Pathak MM. Visualizing PIEZO1 Localization and Activity in hiPSC-Derived Single Cells and Organoids with HaloTag Technology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.22.573117. [PMID: 38187535 PMCID: PMC10769387 DOI: 10.1101/2023.12.22.573117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
PIEZO1 is critical to numerous physiological processes, transducing diverse mechanical stimuli into electrical and chemical signals. Recent studies underscore the importance of visualizing endogenous PIEZO1 activity and localization to understand its functional roles. To enable physiologically and clinically relevant studies on human PIEZO1, we genetically engineered human induced pluripotent stem cells (hiPSCs) to express a HaloTag fused to endogenous PIEZO1. Combined with advanced imaging, our chemogenetic platform allows precise visualization of PIEZO1 localization dynamics in various cell types. Furthermore, the PIEZO1-HaloTag hiPSC technology facilitates the non-invasive monitoring of channel activity across diverse cell types using Ca2+-sensitive HaloTag ligands, achieving temporal resolution approaching that of patch clamp electrophysiology. Finally, we used lightsheet imaging of hiPSC-derived neural organoids to achieve molecular scale imaging of PIEZO1 in three-dimensional tissue organoids. Our advances offer a novel platform for studying PIEZO1 mechanotransduction in human cells and tissues, with potential for elucidating disease mechanisms and targeted therapeutic development.
Collapse
Affiliation(s)
- Gabriella A Bertaccini
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Ignasi Casanellas
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Elizabeth L Evans
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Jamison L Nourse
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - George D Dickinson
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Gaoxiang Liu
- Advanced Bioimaging Center, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Sayan Seal
- Advanced Bioimaging Center, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Alan T Ly
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Jesse R Holt
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Tharaka D Wijerathne
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Shijun Yan
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - Elliot E Hui
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - Jerome J Lacroix
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Mitradas M Panicker
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Srigokul Upadhyayula
- Advanced Bioimaging Center, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Ian Parker
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Medha M Pathak
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| |
Collapse
|
25
|
Rashidi N, Harasymowicz NS, Savadipour A, Steward N, Tang R, Oswald S, Guilak F. PIEZO1-mediated mechanotransduction regulates collagen synthesis on nanostructured 2D and 3D models of fibrosis. Acta Biomater 2025; 193:242-254. [PMID: 39675497 DOI: 10.1016/j.actbio.2024.12.034] [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: 07/10/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
Progressive fibrosis can lead to tissue malfunction and organ failure due to the pathologic accumulation of a collagen-rich extracellular matrix. In vitro models provide useful tools for deconstructing the roles of specific biomechanical or biological mechanisms, such as substrate micro- and nanoscale architecture, in these processes for identifying potential therapeutic targets. Here, we investigated how the mechanosensitive ion channel PIEZO1 influences fibrotic gene and protein expression in adipose-derived stem cells (hASCs). Specifically, we examined the role of PIEZO1 and the mechanosensitive transcription factors YAP/TAZ in sensing aligned or non-aligned substrate architecture to regulate collagen formation. We utilized both 2D microphotopatterned substrates and 3D electrospun polycaprolactone (PCL) substrates to study the role of culture dimensionality. We found that PIEZO1 regulates collagen synthesis in hASCs in a manner that is sensitive to substrate architecture. Activation of PIEZO1 induced significant morphological changes in hASCs, particularly when cultured on aligned substrates, leading to a 30-40 % reduction in cell spreading area and increased cell elongation, in 3D-aligned cultures. Picrosirius Red staining and immunoblotting revealed that PIEZO1 activation reduced collagen accumulation in 3D culture. While YAP translocated to the cytoplasm following PIEZO1 activation, depleting YAP and TAZ did not change collagen expression significantly downstream of PIEZO1 activation, implying that YAP/TAZ translocation from the nucleus and decreased collagen synthesis may be independent consequences of PIEZO1 activation. Our studies demonstrate a role for PIEZO1 in cellular mechanosensing of substrate architecture and provide targetable pathways for treating fibrosis and for enhancing tissue-engineered and regenerative approaches for fibrous tissue repair. STATEMENT OF SIGNIFICANCE: This study examines how cells sense and respond to their physical environment via PIEZO1 mechanotransduction. We discovered that cells use PIEZO1 to detect the alignment of surrounding structures, influencing the production of collagen - a key component in fibrosis. Our study used both 2D and 3D models to mimic different tissue environments, providing new insights into how cellular responses change in more complex settings. Importantly, we found that activating PIEZO1 alters cell shape and collagen production, especially on aligned surfaces. Interestingly, while PIEZO1 activation caused YAP translocation to the cytoplasm, this translocation did not directly affect collagen production. This work advances our understanding of fibrosis development and identifies PIEZO1 as a potential target for new therapies.
Collapse
Affiliation(s)
- Neda Rashidi
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Mechanical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Natalia S Harasymowicz
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alireza Savadipour
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Mechanical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Nancy Steward
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ruhang Tang
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sara Oswald
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Mechanical Engineering, Washington University, St. Louis, MO 63130, USA; Cytex Therapeutics, Inc., Durham, NC 27704, USA.
| |
Collapse
|
26
|
Zhu Y, Chen J, Chen C, Tang R, Xu J, Shi S, Yu X. Deciphering mechanical cues in the microenvironment: from non-malignant settings to tumor progression. Biomark Res 2025; 13:11. [PMID: 39849659 PMCID: PMC11755887 DOI: 10.1186/s40364-025-00727-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/05/2025] [Indexed: 01/25/2025] Open
Abstract
The tumor microenvironment functions as a dynamic and intricate ecosystem, comprising a diverse array of cellular and non-cellular components that precisely orchestrate pivotal tumor behaviors, including invasion, metastasis, and drug resistance. While unraveling the intricate interplay between the tumor microenvironment and tumor behaviors represents a tremendous challenge, recent research illuminates a crucial biological phenomenon known as cellular mechanotransduction. Within the microenvironment, mechanical cues like tensile stress, shear stress, and stiffness play a pivotal role by activating mechanosensitive effectors such as PIEZO proteins, integrins, and Yes-associated protein. This activation initiates cascades of intrinsic signaling pathways, effectively linking the physical properties of tissues to their physiological and pathophysiological processes like morphogenesis, regeneration, and immunity. This mechanistic insight offers a novel perspective on how the mechanical cues within the tumor microenvironment impact tumor behaviors. While the intricacies of the mechanical tumor microenvironment are yet to be fully elucidated, it exhibits distinct physical attributes from non-malignant tissues, including elevated solid stresses, interstitial hypertension, augmented matrix stiffness, and enhanced viscoelasticity. These traits exert notable influences on tumor progression and treatment responses, enriching our comprehension of the multifaceted nature of the microenvironment. Through this innovative review, we aim to provide a new lens to decipher the mechanical attributes within the tumor microenvironment from non-malignant contexts, broadening our knowledge on how these factors promote or inhibit tumor behaviors, and thus offering valuable insights to identify potential targets for anti-tumor strategies.
Collapse
Affiliation(s)
- Yicheng Zhu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jiaoshun Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chen Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Rong Tang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
27
|
Gonzalez M, Clayton S, Wauson E, Christian D, Tran QK. Promotion of nitric oxide production: mechanisms, strategies, and possibilities. Front Physiol 2025; 16:1545044. [PMID: 39917079 PMCID: PMC11799299 DOI: 10.3389/fphys.2025.1545044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
The discovery of nitric oxide (NO) and the role of endothelial cells (ECs) in its production has revolutionized medicine. NO can be produced by isoforms of NO synthases (NOS), including the neuronal (nNOS), inducible (iNOS), and endothelial isoforms (eNOS), and via the non-classical nitrate-nitrite-NO pathway. In particular, endothelium-derived NO, produced by eNOS, is essential for cardiovascular health. Endothelium-derived NO activates soluble guanylate cyclase (sGC) in vascular smooth muscle cells (VSMCs), elevating cyclic GMP (cGMP), causing vasodilation. Over the past four decades, the importance of this pathway in cardiovascular health has fueled the search for strategies to enhance NO bioavailability and/or preserve the outcomes of NO's actions. Currently approved approaches operate in three directions: 1) providing exogenous NO, 2) promoting sGC activity, and 3) preventing degradation of cGMP by inhibiting phosphodiesterase 5 activity. Despite clear benefits, these approaches face challenges such as the development of nitrate tolerance and endothelial dysfunction. This highlights the need for sustainable options that promote endogenous NO production. This review will focus on strategies to promote endogenous NO production. A detailed review of the mechanisms regulating eNOS activity will be first provided, followed by a review of strategies to promote endogenous NO production based on the levels of available preclinical and clinical evidence, and perspectives on future possibilities.
Collapse
Affiliation(s)
| | | | | | | | - Quang-Kim Tran
- Department of Physiology and Pharmacology, Des Moines University Medicine and Health Sciences, West Des Moines, IA, United States
| |
Collapse
|
28
|
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
|
29
|
Wang F, Zou X, Zheng H, Kong T, Pei D. Human epicardial organoids from pluripotent stem cells resemble fetal stage with potential cardiomyocyte- transdifferentiation. Cell Biosci 2025; 15:4. [PMID: 39825425 PMCID: PMC11740338 DOI: 10.1186/s13578-024-01339-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/16/2024] [Indexed: 01/20/2025] Open
Abstract
Epicardium, the most outer mesothelium, exerts crucial functions in fetal heart development and adult heart regeneration. Here we use a three-step manipulation of WNT signalling entwined with BMP and RA signalling for generating a self-organized epicardial organoid that highly express with epicardium makers WT1 and TCF21 from human embryonic stem cells. After 8-days treatment of TGF-beta following by bFGF, cells enter into epithelium-mesenchymal transition and give rise to smooth muscle cells. Epicardium could also integrate and invade into mouse heart with SNAI1 expression, and give birth to numerous cardiomyocyte-like cells. Single-cell RNA seq unveils the heterogeneity and multipotency exhibited by epicardium-derived-cells and fetal-like epicardium. Meanwhile, extracellular matrix and growth factors secreted by epicardial organoid mimics the ecology of subepicardial space between the epicardium and cardiomyocytes. As such, this epicardial organoid offers a unique ground for investigating and exploring the potential of epicardium in heart development and regeneration.
Collapse
Affiliation(s)
- Fanwen Wang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Xinle Zou
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China.
| | - Huilin Zheng
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- College of Biological & Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Tianci Kong
- College of Biological & Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
| |
Collapse
|
30
|
Liu S, Wu J, Meng L, Liu Y, Yu J, Yue J, Hao D, Yu P, Wan Y, Li P, Jin P, Shi L. Piezo1-Induced Nasal Epithelial Barrier Dysfunction in Allergic Rhinitis. Inflammation 2025:10.1007/s10753-024-02234-9. [PMID: 39798033 DOI: 10.1007/s10753-024-02234-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 12/13/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
This study aimed to investigate the role of Piezo1 in nasal epithelial barrier dysfunction in allergic rhinitis (AR) using both in vitro and in vivo experimental methods. A total of 79 human nasal mucosal samples were collected, including 43 from AR patients and 36 from healthy controls. Additionally, 12 BALB/c mice were used for the in vivo experiments. Human nasal epithelial cells (HNEpCs) were employed for the in vitro studies. In the in vivo study, mice were sensitized with ovalbumin (OVA) to induce AR. In the in vitro experiments, Piezo1 expression in HNEpCs was silenced using shRNA, followed by stimulation with IL-13. The expression of Piezo1, ERK1/2, and tight junctions (TJs) components (including ZO-1, Occludin, and Claudin-1) was assessed using quantitative RT-PCR, immunofluorescence, and Western blotting. Statistical analyses included paired Student's t-test and one-way ANOVA. Piezo1 expression was significantly elevated in both AR patients and OVA-induced AR mice, while TJs components were significantly reduced (p < 0.05). Knockdown of Piezo1 in HNEpCs restored the levels of TJs and improved barrier integrity. A negative correlation between Piezo1 and ERK1/2 expression was observed. Piezo1 plays a crucial role in nasal epithelial barrier dysfunction in AR by modulating TJs and the ERK1/2 pathway. These findings suggest that Piezo1 may serve as a potential therapeutic target for AR.
Collapse
Affiliation(s)
- Shengyang Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Duanxing West Road, Jinan, 250000, Shandong, China
- Shandong Provincial Key Medical and Health Discipline of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, China
| | - Jianhua Wu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Duanxing West Road, Jinan, 250000, Shandong, China
- Department of Otolaryngology-Head and Neck Surgery, Binzhou People's Hospital, Binzhou, Shandong, China
| | - Linghui Meng
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Duanxing West Road, Jinan, 250000, Shandong, China
| | - Yuan Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Duanxing West Road, Jinan, 250000, Shandong, China
- Department of Otorhinolaryngology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, China
| | - Jinzhuang Yu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Duanxing West Road, Jinan, 250000, Shandong, China
- Shandong Provincial Key Medical and Health Discipline of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, China
| | - Jing Yue
- Shandong Provincial Key Medical and Health Discipline of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, China
- Department of Traditional Chinese Medicine, Shandong Second Provincial General Hospital, Jinan, Shandong, China
| | - Dingqian Hao
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Duanxing West Road, Jinan, 250000, Shandong, China
| | - Peng Yu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Duanxing West Road, Jinan, 250000, Shandong, China
- Shandong Provincial Key Medical and Health Discipline of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, China
| | - YuZhu Wan
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Duanxing West Road, Jinan, 250000, Shandong, China
- Shandong Provincial Key Medical and Health Discipline of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, China
| | - Ping Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Duanxing West Road, Jinan, 250000, Shandong, China
- Shandong Provincial Key Medical and Health Discipline of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, China
| | - Peng Jin
- Department of Otolaryngology, The Second Hospital of Shandong University, Beiyuan Street 247, Jinan City, Shandong Province, China.
| | - Li Shi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Duanxing West Road, Jinan, 250000, Shandong, China.
- Shandong Provincial Key Medical and Health Discipline of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, China.
- Department of Otolaryngology, The Second Hospital of Shandong University, Beiyuan Street 247, Jinan City, Shandong Province, China.
| |
Collapse
|
31
|
Shen YY, Jethe JV, Reid AP, Hehir J, Amaral MM, Ren C, Hao S, Zhou C, Fisher JAN. Label free, capillary-scale blood flow mapping in vivo reveals that low-intensity focused ultrasound evokes persistent dilation in cortical microvasculature. Commun Biol 2025; 8:12. [PMID: 39762513 PMCID: PMC11704147 DOI: 10.1038/s42003-024-07356-2] [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: 02/08/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Non-invasive, low intensity focused ultrasound is an emerging neuromodulation technique that offers the potential for precision, personalized therapy. An increasing body of research has identified mechanosensitive ion channels that can be modulated by FUS and support acute electrical activity in neurons. However, neuromodulatory effects that persist from hours to days have also been reported. The brain's ability to provide blood flow to electrically active regions involves a multitude of non-neuronal cell types and signaling pathways in the cerebral vasculature; an open question is whether persistent effects can be attributed, at least partly, to vascular mechanisms. Using an in vivo optical approach, we found that microvasculature, and not larger vessels, exhibit significant persistent dilation following sonication without the use of microbubbles. This finding reveals a heretofore unseen aspect of the effects of FUS in vivo and indicates that concurrent changes in neurovascular function may partially underly persistent neuromodulatory effects.
Collapse
Affiliation(s)
- YuBing Y Shen
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Jyoti V Jethe
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Ashlan P Reid
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Jacob Hehir
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Marcello Magri Amaral
- Department of Biomedical Engineering, Washington University in St Louis, St. Louis, MO, USA
- Biomedical Engineering, Universidade Brasil, San Paulo, SP, Brazil
| | - Chao Ren
- Imaging Science Ph.D. Program, Washington University in St Louis, St. Louis, MO, USA
| | - Senyue Hao
- Department of Electrical & Systems Engineering, Washington University in St Louis, St. Louis, MO, USA
| | - Chao Zhou
- Department of Biomedical Engineering, Washington University in St Louis, St. Louis, MO, USA
- Imaging Science Ph.D. Program, Washington University in St Louis, St. Louis, MO, USA
- Department of Electrical & Systems Engineering, Washington University in St Louis, St. Louis, MO, USA
| | | |
Collapse
|
32
|
Duan X, Liu R, Xi Y, Tian Z. The mechanisms of exercise improving cardiovascular function by stimulating Piezo1 and TRP ion channels: a systemic review. Mol Cell Biochem 2025; 480:119-137. [PMID: 38625513 DOI: 10.1007/s11010-024-05000-5] [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: 02/08/2024] [Accepted: 03/24/2024] [Indexed: 04/17/2024]
Abstract
Mechanosensitive ion channels are widely distributed in the heart, lung, bladder and other tissues, and plays an important role in exercise-induced cardiovascular function promotion. By reviewing the PubMed databases, the results were summarized using the terms "Exercise/Sport", "Piezo1", "Transient receptor potential (TRP)" and "Cardiovascular" as the keywords, 124-related papers screened were sorted and reviewed. The results showed that: (1) Piezo1 and TRP channels play an important role in regulating blood pressure and the development of cardiovascular diseases such as atherosclerosis, myocardial infarction, and cardiac fibrosis; (2) Exercise promotes cardiac health, inhibits the development of pathological heart to heart failure, regulating the changes in the characterization of Piezo1 and TRP channels; (3) Piezo1 activates downstream signaling pathways with very broad pathways, such as AKT/eNOS, NF-κB, p38MAPK and HIPPO-YAP signaling pathways. Piezo1 and Irisin regulate nuclear localization of YAP and are hypothesized to act synergistically to regulate tissue mechanical properties of the cardiovascular system and (4) The cardioprotective effects of exercise through the TRP family are mostly accomplished through Ca2+ and involve many signaling pathways. TRP channels exert their important cardioprotective effects by reducing the TRPC3-Nox2 complex and mediating Irisin-induced Ca2+ influx through TRPV4. It is proposed that exercise stimulates the mechanosensitive cation channel Piezo1 and TRP channels, which exerts cardioprotective effects. The activation of Piezo1 and TRP channels and their downstream targets to exert cardioprotective function by exercise may provide a theoretical basis for the prevention of cardiovascular diseases and the rehabilitation of clinical patients.
Collapse
Affiliation(s)
- Xinyan Duan
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Renhan Liu
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Yue Xi
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an, 710119, China.
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an, 710119, China
| |
Collapse
|
33
|
Garcia-Castorena JM, Riester R, Gamino-Ornelas M, Ada N, Guilak F, Danalache M. PIEZO1-mediated calcium influx transiently alters nuclear mechanical properties via actin remodeling in chondrocytes. Biochem Biophys Res Commun 2025; 742:151135. [PMID: 39667069 DOI: 10.1016/j.bbrc.2024.151135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 11/25/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024]
Abstract
Mechanosensation allows cells to generate intracellular signals in response to mechanical cues from their environment. Previous research has demonstrated that mechanical stress can alter the mechanical properties of the nucleus, affecting gene transcription, chromatin methylation, and nuclear mechanoprotection during mechanical loading. PIEZO1, a mechanically gated Ca2+ ion channel, has been shown to be important in sensing mechanical stress, however its signal transduction pathway is not thoroughly understood. In this study, we used primary porcine chondrocytes to determine whether PIEZO1 activation and subsequent Ca2+ influx altered nuclear mechanical properties, and whether these effects involved the actin cytoskeleton. We discovered that activating PIEZO1 with Yoda1, a specific small-molecule agonist, induces transient nuclear softening-a previously identified mechanoprotective response. This PIEZO1-mediated nuclear softening is abolished by inhibiting actin cytoskeleton remodeling with Latrunculin A or by removing extracellular Ca2+. Notably, PIEZO1-mediated nuclear softening did not lead to significant changes in gene expression or heterochromatin methylation. Our findings demonstrate that actin cytoskeleton remodeling following Ca2+ influx facilitates PIEZO1 signal transduction to the nucleus but does not induce lasting gene expression changes.
Collapse
Affiliation(s)
- Jaquelin M Garcia-Castorena
- Department of Orthopedic Surgery, Washington University, St. Louis, MO, 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA; Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO, 63110, USA
| | - Rosa Riester
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, Waldhörnlestraße 22, D-7207, Tübingen, Germany
| | - Miranda Gamino-Ornelas
- Department of Orthopedic Surgery, Washington University, St. Louis, MO, 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA; Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO, 63110, USA
| | - Nikitha Ada
- Department of Orthopedic Surgery, Washington University, St. Louis, MO, 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA; Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO, 63110, USA
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University, St. Louis, MO, 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA; Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO, 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO, 63110, USA.
| | - Marina Danalache
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, Waldhörnlestraße 22, D-7207, Tübingen, Germany
| |
Collapse
|
34
|
Kim OH, Tulip IJ, Kang H, Chang ES, Lee HJ. Compression force promotes glioblastoma progression through the Piezo1‑GDF15‑CTLA4 axis. Oncol Rep 2025; 53:2. [PMID: 39513613 PMCID: PMC11541303 DOI: 10.3892/or.2024.8835] [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: 05/27/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024] Open
Abstract
Glioma, a type of brain tumor, is influenced by mechanical forces in its microenvironment that affect cancer progression. However, our understanding of the contribution of compression and its associated mechanisms remains limited. The objective of the present study was to create an environment in which human brain glioma H4 cells experience pressure and thereby investigate the compressive mechanosensors and signaling pathways. Subsequent time‑lapse imaging and wound healing assays confirmed that 12 h of compression significantly increased cell migration, thereby linking compression with enhanced cell motility. Compression upregulated the expression of Piezo1, a mechanosensitive ion channel, and growth differentiation factor 15 (GDF15), a TGF‑β superfamily member. Knockdown experiments targeting PIEZO1 or GDF15 using small interfering RNA resulted in reduced cell motility, with Piezo1 regulating GDF15 expression. Compression also upregulated CTLA4, a critical immune checkpoint protein. The findings of the present study therefore suggest that compression enhances glioma progression by stimulating Piezo1, promoting GDF15 expression and increasing CTLA4 expression. Thus, these findings provide important insights into the influence of mechanical compression on glioma progression and highlight the involvement of the Piezo1‑GDF15 signaling pathway. Understanding tumor responses to mechanical forces in the brain microenvironment may guide the development of targeted therapeutic strategies to mitigate tumor progression and improve patient outcomes.
Collapse
Affiliation(s)
- Ok-Hyeon Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Israt Jahan Tulip
- Department of Global Innovative Drugs, Graduate School of Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Hana Kang
- Department of Global Innovative Drugs, Graduate School of Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Eun Seo Chang
- Department of Global Innovative Drugs, Graduate School of Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Hyun Jung Lee
- Department of Anatomy and Cell Biology, College of Medicine, Chung‑Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
35
|
Lim XR, Willemse L, Harraz OF. Amyloid beta Aβ 1-40 activates Piezo1 channels in brain capillary endothelial cells. Biophys J 2024:S0006-3495(24)04106-7. [PMID: 39722451 DOI: 10.1016/j.bpj.2024.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/12/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024] Open
Abstract
Amyloid beta (Aβ) peptide accumulation on blood vessels in the brain is a hallmark of neurodegeneration. While Aβ peptides constrict cerebral arteries and arterioles, their impact on capillaries is less understood. Aβ was recently shown to constrict brain capillaries through pericyte contraction, but whether-and if so how-Aβ affects endothelial cells (ECs) remains unknown. ECs represent the predominant vascular cell type in the cerebral circulation, and we recently showed that the mechanosensitive ion channel Piezo1 is functionally expressed in the plasma membrane of ECs. Since Aβ disrupts membrane structures, we hypothesized that Aβ1-40, the predominantly deposited isoform in the cerebral circulation, alters endothelial Piezo1 function. Using patch-clamp electrophysiology and freshly isolated capillary ECs, we assessed the impact of the Aβ1-40 peptide on single-channel Piezo1 activity. We show that Aβ1-40 increased Piezo1 open probability and channel open time. Aβ1-40 effects were absent when Piezo1 was genetically deleted or when a superoxide dismutase/catalase mimetic was used. Further, Aβ1-40 enhanced Piezo1 mechanosensitivity and lowered the pressure of half-maximal Piezo1 activation. Our data collectively suggest that Aβ1-40 facilitates higher Piezo1-mediated cation influx in brain ECs. These novel findings have the potential to unravel the possible involvement of Piezo1 modulation in the pathophysiology of neurodegenerative diseases characterized by Aβ accumulation.
Collapse
Affiliation(s)
- Xin Rui Lim
- Department of Pharmacology, Larner College of Medicine, Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont
| | - Luc Willemse
- Department of Pharmacology, Larner College of Medicine, Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont.
| |
Collapse
|
36
|
Yan S, Lu Y, An C, Hu W, Chen Y, Li Z, Wei W, Chen Z, Zeng X, Xu W, Lv Z, Pan F, Gao W, Wu Y. Biomechanical research using advanced micro-nano devices: In-Vitro cell Characterization focus. J Adv Res 2024:S2090-1232(24)00602-7. [PMID: 39701378 DOI: 10.1016/j.jare.2024.12.024] [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: 08/18/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Cells in the body reside in a dynamic microenvironment subjected to various physical stimuli, where mechanical stimulation plays a crucial role in regulating cellular physiological behaviors and functions. AIM OF REVIEW Investigating the mechanisms and interactions of mechanical transmission is essential for understanding the physiological and functional interplay between cells and physical stimuli. Therefore, establishing an in vitro biomechanical stimulation cell culture system holds significant importance for research related to cellular biomechanics. KEY SCIENTIFIC CONCEPTS OF REVIEW In this review, we primarily focused on various biomechanically relevant cell culture systems and highlighted the advancements and prospects in their preparation processes. Firstly, we discussed the types and characteristics of biomechanics present in the microenvironment within the human body. Subsequently, we introduced the research progress, working principles, preparation processes, potential advantages, applications, and challenges of various biomechanically relevant in vitro cell culture systems. Additionally, we summarized and categorized currently commercialized biomechanically relevant cell culture systems, offering a comprehensive reference for researchers in related fields.
Collapse
Affiliation(s)
- Shiqiang Yan
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China; Center of Cancer Immunology, Shenzhen Institute of Advanced Technology Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Yan Lu
- Department of Otolaryngology Head & Neck Surgery, The First Hospital, Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Changming An
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wanglai Hu
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Yaofeng Chen
- Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Ziwen Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Wenbo Wei
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
| | - Zongzheng Chen
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
| | - Xianhai Zeng
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China
| | - Wei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, Shandong, China
| | - Zhenghua Lv
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, Shandong, China.
| | - Fan Pan
- Center of Cancer Immunology, Shenzhen Institute of Advanced Technology Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China.
| | - Wei Gao
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China.
| | - Yongyan Wu
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China; Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen 518055, Guangdong, China.
| |
Collapse
|
37
|
Beech DJ, Fagnen C, Kalli AC. Biological sensing of fluid flow-lessons from PIEZO1. Biophys Rev 2024; 16:871-873. [PMID: 39830133 PMCID: PMC11735714 DOI: 10.1007/s12551-024-01246-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/01/2024] [Indexed: 01/22/2025] Open
Abstract
The flow sensing endothelial cell lining of blood and lymphatic vessels is essential in vertebrates. While the mechanisms are still mysterious in many regards, several critical components became apparent through molecular biology studies. In this article, we focus on PIEZO1, which forms unusual force-sensing ion channels capable of rapid transduction of force into biological effect. We describe current knowledge and emerging challenges. We suggest the idea of using computation to construct the flow sensing mechanism of endothelium to advance understanding, develop testable hypotheses and potentially design novel therapeutic strategies and synthetic flow sensing devices.
Collapse
Affiliation(s)
- David J. Beech
- School of Medicine, University of Leeds, Leeds, LS2 9JT UK
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Building, Clarendon Way, School of Medicine, University of Leeds, Leeds, LS2 9JT UK
| | | | | |
Collapse
|
38
|
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
|
39
|
Wang Y, Chatterjee E, Li G, Xu J, Xiao J. Force-sensing protein expression in response to cardiovascular mechanotransduction. EBioMedicine 2024; 110:105412. [PMID: 39481337 PMCID: PMC11554632 DOI: 10.1016/j.ebiom.2024.105412] [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: 07/12/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Force-sensing biophysical cues in microenvironment, including extracellular matrix performances, stretch-mediated mechanics, shear stress and flow-induced hemodynamics, have a significant influence in regulating vascular morphogenesis and cardiac remodeling by mechanotransduction. Once cells perceive these extracellular mechanical stimuli, Piezo activation promotes calcium influx by forming integrin-adhesion-coupling receptors. This induces robust contractility of cytoskeleton structures to further transmit biomechanical alternations into nuclei by regulating Hippo-Yes associated protein (YAP) signaling pathway between cytoplasmic and nuclear translocation. Although biomechanical stimuli are widely studied in cardiovascular diseases, the expression of force-sensing proteins in response to cardiovascular mechanotransduction has not been systematically concluded. Therefore, this review will summarize the force-sensing Piezo, cytoskeleton and YAP proteins to mediate extracellular mechanics, and also give the prominent emphasis on intrinsic connection of these mechanical proteins and cardiovascular mechanotransduction. Extensive insights into cardiovascular mechanics may provide some new strategies for cardiovascular clinical therapy.
Collapse
Affiliation(s)
- Yongtao Wang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China
| | - Emeli Chatterjee
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jiahong Xu
- Department of Cardiology, Shanghai Gongli Hospital, Shanghai 200135, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
40
|
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
|
41
|
Park H, Lee S, Furtado J, Robinson M, Schwartz M, Young L, Eichmann A. PIEZO1 overexpression in hereditary hemorrhagic telangiectasia arteriovenous malformations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625696. [PMID: 39651206 PMCID: PMC11623632 DOI: 10.1101/2024.11.27.625696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Background Hereditary hemorrhagic telangiectasia (HHT) is an inherited vascular disorder characterized by arteriovenous malformations (AVMs). Loss-of-function mutations in Activin receptor-like kinase 1 (ALK1) cause type 2 HHT and Alk1 knockout (KO) mice develop AVMs due to overactivation of VEGFR2/PI3K/AKT signaling pathways. However, the full spectrum of signaling alterations in Alk1 mutants remains unknown and means to combat AVM formation in patients are yet to be developed. Methods Single-cell RNA sequencing of endothelial-specific Alk1 KO mouse retinas and controls identified a cluster of endothelial cells (ECs) that was unique to Alk1 mutants and that overexpressed fluid shear stress (FSS) signaling signatures including upregulation of the mechanosensitive ion channel PIEZO1. PIEZO1 overexpression was confirmed in human HHT lesions, and genetic and pharmacological PIEZO1 inhibition was tested in Alk1 KO mice, as well as downstream PIEZO1 signaling. Results Pharmacological PIEZO1 inhibition, and genetic Piezo1 deletion in Alk1 -deficient mice effectively mitigated AVM formation. Furthermore, we identified that elevated VEGFR2/AKT, ERK5-p62-KLF4, hypoxia and proliferation signaling were significantly reduced in Alk1 - Piezo1 double ECKO mice. Conclusions PIEZO1 overexpression and signaling is integral to HHT2, and PIEZO1 blockade reduces AVM formation and alleviates cellular and molecular hallmarks of ALK1-deficient cells. This finding provides new insights into the mechanistic underpinnings of ALK1-related vascular diseases and identifies potential therapeutic targets to prevent AVMs.
Collapse
|
42
|
Chen H, Miao Y, Duan H, Yi S, Lin Z, Guo Y, Zou J, Niu L. The effect of combined ultrasound stimulation and gastrodin on seizures in mice. Front Neurosci 2024; 18:1499078. [PMID: 39649662 PMCID: PMC11621076 DOI: 10.3389/fnins.2024.1499078] [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: 09/20/2024] [Accepted: 11/12/2024] [Indexed: 12/11/2024] Open
Abstract
Both physiotherapy and medicine play essential roles in the treatment of epilepsy. The purpose of this research was to evaluate the efficacy of the combined therapy with focus ultrasound stimulation (FUS) and gastrodin (GTD) on seizures in a mouse model. Kainic acid-induced seizure mice were divided into five groups randomly: sham, FUS, saline + sham, GTD + sham and GTD + FUS. The results showed that combined therapy with ultrasound stimulation and gastrodin can significantly reduce the number and duration of seizures in GTD + FUS group. 9.4T magnetic resonance imaging and histologic staining results revealed the underlying mechanism of the combined therapy may be that ultrasound stimulation increases cell membrane permeability to increase GTD concentration in brain. In addition, we verified the safety of FUS combined with GTD therapy. This research provides a new strategy for neurological disorders combining treatment of physical neuromodulation and medicine.
Collapse
Affiliation(s)
- Houminji Chen
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuqing Miao
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Haowen Duan
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shasha Yi
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhengrong Lin
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yanwu Guo
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Junjie Zou
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lili Niu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
43
|
Zhang Z, Li X, Wang C, Zhang F, Liu J, Xu XZS. Shear stress sensing in C. elegans. Curr Biol 2024; 34:5382-5391.e3. [PMID: 39471806 PMCID: PMC11576262 DOI: 10.1016/j.cub.2024.09.075] [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: 04/24/2024] [Revised: 07/25/2024] [Accepted: 09/26/2024] [Indexed: 11/01/2024]
Abstract
Shear stress sensing represents a vital mode of mechanosensation.1 Previous efforts have mainly focused on characterizing how various cell types-for example, vascular endothelial cells-sense shear stress arising from fluid flow within the animal body.1,2 How animals sense shear stress derived from their external environment, however, is not well understood. Here, using C. elegans as a model, we show that external fluid flow triggers behavioral responses in C. elegans, facilitating their navigation of the environment during swimming. Such behavioral responses primarily result from shear stress generated by fluid flow. The sensory neurons AWC, ASH, and ASER are the major shear stress-sensitive neurons, among which AWC shows the most robust response to shear stress and is required for shear stress-induced behavior. Mechanistically, shear stress signals are transduced by G protein signaling in AWC, with cGMP as the second messenger, culminating in the opening of cGMP-sensitive cyclic nucleotide-gated (CNG) channels and neuronal excitation. These studies demonstrate that C. elegans senses and responds to shear stress and characterize the underlying neural and molecular mechanisms. Our work helps establish C. elegans as a genetic model for studying shear stress sensing.
Collapse
Affiliation(s)
- Zhiyong Zhang
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China; Life Sciences Institute, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xia Li
- Life Sciences Institute, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Can Wang
- Life Sciences Institute, Ann Arbor, MI, USA
| | - Fengfan Zhang
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China; Life Sciences Institute, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jianfeng Liu
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China.
| | - X Z Shawn Xu
- Life Sciences Institute, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
44
|
Schulz K, Hazelton-Cavill P, Alornyo KK, Edenhofer I, Lindenmeyer M, Lohr C, Huber TB, Denholm B, Koehler S. Piezo activity levels need to be tightly regulated to maintain normal morphology and function in pericardial nephrocytes. Sci Rep 2024; 14:28254. [PMID: 39548228 PMCID: PMC11568303 DOI: 10.1038/s41598-024-79352-9] [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: 08/29/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024] Open
Abstract
Due to their position on glomerular capillaries, podocytes are continuously counteracting biomechanical filtration forces. Most therapeutic interventions known to generally slow or prevent the progression of chronic kidney disease appear to lower these biomechanical forces on podocytes, highlighting the critical need to better understand podocyte mechano-signalling pathways. Here we investigated whether the mechanotransducer Piezo is involved in a mechanosensation pathway in Drosophila nephrocytes, the podocyte homologue in the fly. Loss of function analysis in Piezo depleted nephrocytes reveal a severe morphological and functional phenotype. Further, pharmacological activation of endogenous Piezo with Yoda1 causes a significant increase of intracellular Ca++ upon exposure to a mechanical stimulus in nephrocytes, as well as filtration disturbances. Elevated Piezo expression levels also result in a severe nephrocyte phenotype. Interestingly, expression of Piezo which lacks mechanosensitive channel activity, does not result in a severe nephrocyte phenotype, suggesting the observed changes in Piezo wildtype overexpressing cells are caused by the mechanosensitive channel activity. Moreover, blocking Piezo activity using the tarantula toxin GsMTx4 reverses the phenotypes observed in nephrocytes overexpressing Piezo. Taken together, here we provide evidence that Piezo activity levels need to be tightly regulated to maintain normal pericardial nephrocyte morphology and function.
Collapse
Affiliation(s)
- Kristina Schulz
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Campus Forschung II N25, Martinistraße 52, 20246, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paris Hazelton-Cavill
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Campus Forschung II N25, Martinistraße 52, 20246, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karl K Alornyo
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Campus Forschung II N25, Martinistraße 52, 20246, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ilka Edenhofer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Campus Forschung II N25, Martinistraße 52, 20246, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maja Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Campus Forschung II N25, Martinistraße 52, 20246, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Lohr
- Division of Neurophysiology, Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Campus Forschung II N25, Martinistraße 52, 20246, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Barry Denholm
- Biomedical Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Sybille Koehler
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Campus Forschung II N25, Martinistraße 52, 20246, Hamburg, Germany.
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Biomedical Sciences, University of Edinburgh, Edinburgh, Scotland, UK.
| |
Collapse
|
45
|
Zhang T, Bi C, Li Y, Zhao L, Cui Y, Ouyang K, Xiao B. Phosphorylation of Piezo1 at a single residue, serine-1612, regulates its mechanosensitivity and in vivo mechanotransduction function. Neuron 2024; 112:3618-3633.e6. [PMID: 39270653 DOI: 10.1016/j.neuron.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/15/2024]
Abstract
Piezo1 is a mechanically activated cation channel that converts mechanical force into diverse physiological processes. Owing to its large protein size of more than 2,500 amino acids and complex 38-transmembrane helix topology, how Piezo1 is post-translationally modified for regulating its in vivo mechanotransduction functions remains largely unexplored. Here, we show that PKA activation potentiates the mechanosensitivity and slows the inactivation kinetics of mouse Piezo1 and identify the major phosphorylation site, serine-1612 (S1612), that also responds to PKC activation and shear stress. Mutating S1612 abolishes PKA and PKC regulation of Piezo1 activities. Primary endothelial cells derived from the Piezo1-S1612A knockin mice lost PKA- and PKC-dependent phosphorylation and functional potentiation of Piezo1. The mutant mice show activity-dependent elevation of blood pressure and compromised exercise endurance, resembling endothelial-specific Piezo1 knockout mice. Taken together, we identify the major PKA and PKC phosphorylation site in Piezo1 and demonstrate its contribution to Piezo1-mediated physiological functions.
Collapse
Affiliation(s)
- Tingxin Zhang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Frontier Research Center of Biological, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Cheng Bi
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Frontier Research Center of Biological, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Yiran Li
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Frontier Research Center of Biological, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Lingyun Zhao
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yaxiong Cui
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Frontier Research Center of Biological, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, 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 of Biological, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
46
|
Lan Y, Lu J, Zhang S, Jie C, Chen C, Xiao C, Qin C, Cheng D. Piezo1-Mediated Mechanotransduction Contributes to Disturbed Flow-Induced Atherosclerotic Endothelial Inflammation. J Am Heart Assoc 2024; 13:e035558. [PMID: 39450718 PMCID: PMC11935715 DOI: 10.1161/jaha.123.035558] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/12/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Disturbed flow generates oscillatory shear stress (OSS), which in turn leads to endothelial inflammation and atherosclerosis. Piezo1, a biomechanical force sensor, plays a crucial role in the cardiovascular system. However, the specific role of Piezo1 in atherosclerosis remains to be fully elucidated. METHODS AND RESULTS We detected the expression of Piezo1 in atherosclerotic mice and endothelial cells from regions with disturbed blood flow. The pharmacological inhibitor Piezo1 inhibitor (GsMTx4) was used to evaluate the impact of Piezo1 on plaque progression and endothelial inflammation. We examined Piezo1's direct response to OSS in vitro and its effects on endothelial inflammation. Furthermore, mechanistic studies were conducted to explore the potential molecular cascade through which Piezo1 mediates endothelial inflammation in response to OSS. Our findings revealed the upregulation of Piezo1 in apoE-/- (apolipoprotein E) atherosclerotic mice, which is associated with disturbed flow. Treatment with GsMTx4 not only delayed plaque progression but also mitigated endothelial inflammation in both chronic and disturbed flow-induced atherosclerosis. Piezo1 was shown to facilitate calcium ions (Ca2+) influx in response to OSS, thereby activating endothelial inflammation. This inflammatory response was attenuated in the absence of Piezo1. Additionally, we identified that under OSS, Piezo1 activates the Ca2+/CaM/CaMKII (calmodulin/calmodulin-dependent protein kinases Ⅱ) pathways, which subsequently stimulate downstream kinases FAK (focal adhesion kinase) and Src. This leads to the activation of the OSS-sensitive YAP (yes-associated protein), ultimately triggering endothelial inflammation. CONCLUSIONS Our study highlights the key role of Piezo1 in atherosclerotic endothelial inflammation, proposing the Piezo1-Ca2+/CaM/CaMKII-FAK/Src-YAP axis as a previously unknown endothelial mechanotransduction pathway. Piezo1 is expected to become a potential therapeutic target for atherosclerosis and cardiovascular diseases.
Collapse
Affiliation(s)
- Yining Lan
- Department of NeurologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Jing Lu
- Department of NeurologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Shaohan Zhang
- The Second Affiliated Hospital of Qiqihar Medical CollegeQiqiharHeilongjiangChina
| | - Chunxiao Jie
- Department of NeurologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Chunyong Chen
- Department of NeurologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Chao Xiao
- Department of NeurologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Department of NeurologyLiuzhou People’s HospitalLiuzhouGuangxiChina
| | - Chao Qin
- Department of NeurologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Daobin Cheng
- Department of NeurologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| |
Collapse
|
47
|
Johnston J, Jeon H, Choi YY, Kim G, Shi T, Khong C, Chang HC, Myung NV, Wang Y. Stimulative piezoelectric nanofibrous scaffolds for enhanced small extracellular vesicle production in 3D cultures. Biomater Sci 2024; 12:5728-5741. [PMID: 39403853 PMCID: PMC11474809 DOI: 10.1039/d4bm00504j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
Small extracellular vesicles (sEVs) have great promise as effective carriers for drug delivery. However, the challenges associated with the efficient production of sEVs hinder their clinical applications. Herein, we report a stimulative 3D culture platform for enhanced sEV production. The proposed platform consists of a piezoelectric nanofibrous scaffold (PES) coupled with acoustic stimulation to enhance sEV production of cells in a 3D biomimetic microenvironment. Combining cell stimulation with a 3D culture platform in this stimulative PES enables a 15.7-fold increase in the production rate per cell with minimal deviations in particle size and protein composition compared with standard 2D cultures. We find that the enhanced sEV production is attributable to the activation and upregulation of crucial sEV production steps through the synergistic effect of stimulation and the 3D microenvironment. Moreover, changes in cell morphology lead to cytoskeleton redistribution through cell-matrix interactions in the 3D cultures. This in turn facilitates intracellular EV trafficking, which impacts the production rate. Overall, our work provides a promising 3D cell culture platform based on piezoelectric biomaterials for enhanced sEV production. This platform is expected to accelerate the potential use of sEVs for drug delivery and broad biomedical applications.
Collapse
Affiliation(s)
- James Johnston
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Hyunsu Jeon
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Yun Young Choi
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Gaeun Kim
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Tiger Shi
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Courtney Khong
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Nosang Vincent Myung
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Yichun Wang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
48
|
Fan Q, Hadla M, Peterson Z, Nelson G, Ye H, Wang X, Mardirossian JM, Harris PC, Alper SL, Prakash YS, Beyder A, Torres VE, Chebib FT. Activation of PIEZO1 Attenuates Kidney Cystogenesis In Vitro and Ex Vivo. KIDNEY360 2024; 5:1601-1612. [PMID: 39356563 DOI: 10.34067/kid.0000000598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Key Points
PIEZO1 activation reduces cystogenesis: Yoda1 activates PIEZO1, raising calcium and lowering cAMP, reducing cyst growth in autosomal dominant polycystic kidney disease models.Context-dependent role of PIEZO1: PIEZO1 knockout in mice with or without polycystic kidneys does not affect cyst formation, suggesting redundancy in mechanosensitive pathways.Therapeutic potential: Findings support PIEZO1 activation as a part of combination therapy to slow cyst growth in autosomal dominant polycystic kidney disease, needing more in vivo studies.
Background
The disruption of calcium signaling associated with polycystin deficiency is a key factor in abnormal epithelial growth in autosomal dominant polycystic kidney disease. Calcium homeostasis can be influenced by mechanotransduction. The mechanosensitive cation channel PIEZO1 has been implicated in sensing intrarenal pressure and regulating urinary osmoregulation, but its role in kidney cystogenesis is unclear.
Methods
We hypothesized that altered mechanotransduction contributes to cystogenesis in autosomal dominant polycystic kidney disease and that activation of mechanosensitive cation channels could be a therapeutic strategy.
Results
We demonstrate that Yoda1, a PIEZO1 activator, increases intracellular calcium and reduces forskolin-induced cAMP levels in mouse inner medullary collecting duct (mIMCD3) cells. Notably, knockout of polycystin-2 attenuated the efficacy of Yoda1 in reducing cAMP levels in mIMCD3 cells. Yoda1 also reduced forskolin-induced mIMCD3 cyst surface area in vitro and cystic index in mouse metanephros ex vivo in a dose-dependent manner. However, collecting duct–specific PIEZO1 knockout neither induced cystogenesis in wild-type mice nor altered cystogenesis in the Pkd1RC/RC mouse model.
Conclusions
These findings support the potential role of PIEZO1 agonists in mitigating cystogenesis by increasing intracellular calcium and reducing cAMP levels, but the unaltered in vivo cystic phenotype after PIEZO1 knockout in the collecting duct suggests possible redundancy in mechanotransductive pathways.
Collapse
Affiliation(s)
- Qingfeng Fan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Mohamad Hadla
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida
| | - Zack Peterson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Grace Nelson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Hong Ye
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Xiaofang Wang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | | | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
- Department of Biochemistry and Molecular biology, Mayo Clinic, Rochester, Minnesota
| | - Seth L Alper
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Arthur Beyder
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Fouad T Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
49
|
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
|
50
|
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
|