1
|
Kinsella JA, Debant M, Parsonage G, Morley LC, Bajarwan M, Revill C, Foster R, Beech DJ. Pharmacology of PIEZO1 channels. Br J Pharmacol 2024; 181:4714-4732. [PMID: 39402010 DOI: 10.1111/bph.17351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/15/2024] [Accepted: 08/25/2024] [Indexed: 11/05/2024] Open
Abstract
PIEZO1 is a eukaryotic membrane protein that assembles as trimers to form calcium-permeable, non-selective cation channels with exquisite capabilities for mechanical force sensing and transduction of force into effect in diverse cell types that include blood cells, endothelial cells, epithelial cells, fibroblasts and stem cells and diverse systems that include bone, lymphatics and muscle. The channel has wide-ranging roles and is considered as a target for novel therapeutics in ailments spanning cancers and cardiovascular, dental, gastrointestinal, hepatobiliary, infectious, musculoskeletal, nervous system, ocular, pregnancy, renal, respiratory and urological disorders. The identification of PIEZO1 modulators is in its infancy but useful experimental tools emerged for activating, and to a lesser extent inhibiting, the channels. Elementary structure-activity relationships are known for the Yoda series of small molecule agonists, which show the potential for diverse physicochemical and pharmacological properties. Intriguing effects of Yoda1 include the stimulated removal of excess cerebrospinal fluid. Despite PIEZO1's broad expression, opportunities are suggested for selective positive or negative modulation without intolerable adverse effects. Here we provide a focused, non-systematic, narrative review of progress with this pharmacology and discuss potential future directions for research in the area.
Collapse
Affiliation(s)
- Jacob A Kinsella
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
- School of Chemistry, University of Leeds, Leeds, UK
| | - Marjolaine Debant
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Gregory Parsonage
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Lara C Morley
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Muath Bajarwan
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | | | | | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
2
|
Huang Y, Mo H, Yang J, Gao L, Tao T, Shu Q, Guo W, Zhao Y, Lyu J, Wang Q, Guo J, Zhai H, Zhu L, Chen H, Xu G. Mechano-regulation of GLP-1 production by Piezo1 in intestinal L cells. eLife 2024; 13:RP97854. [PMID: 39509292 PMCID: PMC11542922 DOI: 10.7554/elife.97854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Abstract
Glucagon-like peptide 1 (GLP-1) is a gut-derived hormone secreted by intestinal L cells and vital for postprandial glycemic control. As open-type enteroendocrine cells, whether L cells can sense mechanical stimuli caused by chyme and thus regulate GLP-1 synthesis and secretion is unexplored. Molecular biology techniques revealed the expression of Piezo1 in intestinal L cells. Its level varied in different energy status and correlates with blood glucose and GLP-1 levels. Mice with L cell-specific loss of Piezo1 (Piezo1 IntL-CKO) exhibited impaired glucose tolerance, increased body weight, reduced GLP-1 production and decreased CaMKKβ/CaMKIV-mTORC1 signaling pathway under normal chow diet or high-fat diet. Activation of the intestinal Piezo1 by its agonist Yoda1 or intestinal bead implantation increased the synthesis and secretion of GLP-1, thus alleviated glucose intolerance in diet-induced-diabetic mice. Overexpression of Piezo1, Yoda1 treatment or stretching stimulated GLP-1 production and CaMKKβ/CaMKIV-mTORC1 signaling pathway, which could be abolished by knockdown or blockage of Piezo1 in primary cultured mouse L cells and STC-1 cells. These experimental results suggest a previously unknown regulatory mechanism for GLP-1 production in L cells, which could offer new insights into diabetes treatments.
Collapse
Affiliation(s)
- Yanling Huang
- Department of Physiology, School of Medicine, Jinan UniversityGuangzhouChina
| | - Haocong Mo
- Department of Physiology, School of Medicine, Jinan UniversityGuangzhouChina
| | - Jie Yang
- Department of Pathology, School of Basic Medicine, Guangzhou Medical UniversityGuangdongChina
| | - Luyang Gao
- Department of Physiology, School of Medicine, Jinan UniversityGuangzhouChina
| | - Tian Tao
- Department of Physiology, School of Medicine, Jinan UniversityGuangzhouChina
| | - Qing Shu
- Department of Physiology, School of Medicine, Jinan UniversityGuangzhouChina
| | - Wenying Guo
- Department of Physiology, School of Medicine, Jinan UniversityGuangzhouChina
| | - Yawen Zhao
- Department of Physiology, School of Medicine, Jinan UniversityGuangzhouChina
| | - Jingya Lyu
- Department of Physiology, School of Medicine, Jinan UniversityGuangzhouChina
| | - Qimeng Wang
- Biotherapy Center, Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhouChina
| | - Jinghui Guo
- School of Medicine, The Chinese University of Hong KongShenzhenChina
| | - Hening Zhai
- Endoscopy Center, The First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Linyan Zhu
- Department of Pharmacology, School of Medicine, Jinan UniversityGuangzhouChina
| | - Hui Chen
- Biotherapy Center, Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhouChina
| | - Geyang Xu
- Department of Physiology, School of Medicine, Jinan UniversityGuangzhouChina
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of EducationGuangzhouChina
| |
Collapse
|
3
|
Koster AK, Yarishkin O, Dubin AE, Kefauver JM, Pak RA, Cravatt BF, Patapoutian A. Chemical mapping of the surface interactome of PIEZO1 identifies CADM1 as a modulator of channel inactivation. Proc Natl Acad Sci U S A 2024; 121:e2415934121. [PMID: 39356664 PMCID: PMC11474052 DOI: 10.1073/pnas.2415934121] [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: 08/07/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
The propeller-shaped blades of the PIEZO1 and PIEZO2 ion channels partition into the plasma membrane and respond to indentation or stretching of the lipid bilayer, thus converting mechanical forces into signals that can be interpreted by cells, in the form of calcium flux and changes in membrane potential. While PIEZO channels participate in diverse physiological processes, from sensing the shear stress of blood flow in the vasculature to detecting touch through mechanoreceptors in the skin, the molecular details that enable these mechanosensors to tune their responses over a vast dynamic range of forces remain largely uncharacterized. To survey the molecular landscape surrounding PIEZO channels at the cell surface, we employed a mass spectrometry-based proteomic approach to capture and identify extracellularly exposed proteins in the vicinity of PIEZO1. This PIEZO1-proximal interactome was enriched in surface proteins localized to cell junctions and signaling hubs within the plasma membrane. Functional screening of these interaction candidates by calcium imaging and electrophysiology in an overexpression system identified the adhesion molecule CADM1/SynCAM that slows the inactivation kinetics of PIEZO1 with little effect on PIEZO2. Conversely, we found that CADM1 knockdown accelerates inactivation of endogenous PIEZO1 in Neuro-2a cells. Systematic deletion of CADM1 domains indicates that the transmembrane region is critical for the observed effects on PIEZO1, suggesting that modulation of inactivation is mediated by interactions in or near the lipid bilayer.
Collapse
Affiliation(s)
- Anna K. Koster
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
- Department of Chemistry, Scripps Research, La Jolla, CA92037
| | - Oleg Yarishkin
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | - Adrienne E. Dubin
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | - Jennifer M. Kefauver
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | - Ryan A. Pak
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | | | - Ardem Patapoutian
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| |
Collapse
|
4
|
Michelucci A, Catacuzzeno L. Piezo1, the new actor in cell volume regulation. Pflugers Arch 2024; 476:1023-1039. [PMID: 38581527 PMCID: PMC11166825 DOI: 10.1007/s00424-024-02951-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/29/2024] [Accepted: 03/20/2024] [Indexed: 04/08/2024]
Abstract
All animal cells control their volume through a complex set of mechanisms, both to counteract osmotic perturbations of the environment and to enable numerous vital biological processes, such as proliferation, apoptosis, and migration. The ability of cells to adjust their volume depends on the activity of ion channels and transporters which, by moving K+, Na+, and Cl- ions across the plasma membrane, generate the osmotic gradient that drives water in and out of the cell. In 2010, Patapoutian's group identified a small family of evolutionarily conserved, Ca2+-permeable mechanosensitive channels, Piezo1 and Piezo2, as essential components of the mechanically activated current that mediates mechanotransduction in vertebrates. Piezo1 is expressed in several tissues and its opening is promoted by a wide range of mechanical stimuli, including membrane stretch/deformation and osmotic stress. Piezo1-mediated Ca2+ influx is used by the cell to convert mechanical forces into cytosolic Ca2+ signals that control diverse cellular functions such as migration and cell death, both dependent on changes in cell volume and shape. The crucial role of Piezo1 in the regulation of cell volume was first demonstrated in erythrocytes, which need to reduce their volume to pass through narrow capillaries. In HEK293 cells, increased expression of Piezo1 was found to enhance the regulatory volume decrease (RVD), the process whereby the cell re-establishes its original volume after osmotic shock-induced swelling, and it does so through Ca2+-dependent modulation of the volume-regulated anion channels. More recently we reported that Piezo1 controls the RVD in glioblastoma cells via the modulation of Ca2+-activated K+ channels. To date, however, the mechanisms through which this mechanosensitive channel controls cell volume and maintains its homeostasis have been poorly investigated and are still far from being understood. The present review aims to provide a broad overview of the literature discussing the recent advances on this topic.
Collapse
Affiliation(s)
- A Michelucci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy.
| | - L Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy.
| |
Collapse
|
5
|
Kamat V, Sweet IR. Hypertonicity during a rapid rise in D-glucose mediates first-phase insulin secretion. Front Endocrinol (Lausanne) 2024; 15:1395028. [PMID: 38989001 PMCID: PMC11233695 DOI: 10.3389/fendo.2024.1395028] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Introduction Biphasic insulin secretion is an intrinsic characteristic of the pancreatic islet and has clinical relevance due to the loss of first-phase in patients with Type 2 diabetes. As it has long been shown that first-phase insulin secretion only occurs in response to rapid changes in glucose, we tested the hypothesis that islet response to an increase in glucose is a combination of metabolism plus an osmotic effect where hypertonicity is driving first-phase insulin secretion. Methods Experiments were performed using perifusion analysis of rat, mouse, and human islets. Insulin secretion rate (ISR) and other parameters associated with its regulation were measured in response to combinations of D-glucose and membrane-impermeable carbohydrates (L-glucose or mannitol) designed to dissect the effect of hypertonicity from that of glucose metabolism. Results Remarkably, the appearance of first-phase responses was wholly dependent on changes in tonicity: no first-phase in NAD(P)H, cytosolic calcium, cAMP secretion rate (cAMP SR), or ISR was observed when increased D-glucose concentration was counterbalanced by decreases in membrane-impermeable carbohydrates. When D-glucose was greater than 8 mM, rapid increases in L-glucose without any change in D-glucose resulted in first-phase responses in all measured parameters that were kinetically similar to D-glucose. First-phase ISR was completely abolished by H89 (a non-specific inhibitor of protein kinases) without affecting first-phase calcium response. Defining first-phase ISR as the difference between glucose-stimulated ISR with and without a change in hypertonicity, the peak of first-phase ISR occurred after second-phase ISR had reached steady state, consistent with the well-established glucose-dependency of mechanisms that potentiate glucose-stimulated ISR. Discussion The data collected in this study suggests a new model of glucose-stimulated biphasic ISR where first-phase ISR derives from (and after) a transitory amplification of second-phase ISR and driven by hypertonicity-induced rise in H89-inhibitable kinases likely driven by first-phase responses in cAMP, calcium, or a combination of both.
Collapse
Affiliation(s)
| | - Ian R. Sweet
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States
| |
Collapse
|
6
|
Guo W, Gao L, Mo H, Deng H, Zhao Y, Xu G. Mechano-sensor Piezo1 inhibits glucagon production in pancreatic α-cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167185. [PMID: 38653360 DOI: 10.1016/j.bbadis.2024.167185] [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: 11/27/2023] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
OBJECTIVE Glucagon is a critical hormone regulating glucose metabolism. It stimulates the liver to release glucose under low blood sugar conditions, thereby maintaining blood glucose stability. Excessive glucagon secretion and hyperglycemia is observed in individuals with diabetes. Precise modulation of glucagon is significant to maintain glucose homeostasis. Piezo1 is a mechanosensitive ion channel capable of converting extracellular mechanical forces into intracellular signals, thus regulating hormonal synthesis and secretion. This study aims to investigate the role of Piezo1 in regulating glucagon production in α cells. METHODS The effects of Piezo1 on glucagon production were examined in normal- or high-fat diet fed α cell-specific Piezo1 knockout mice (Gcg-Piezo1-/-), and the murine pancreatic α cell line αTC1-6. Expression of Proglucagon was investigated by real-time PCR and western blotting. Plasma glucagon and insulin were detected by enzyme immunoassay. RESULTS Under both normal- and high-fat diet conditions, Gcg-Piezo1-/- mice exhibited increased pancreatic α cell proportion, hyperglucagonemia, impaired glucose tolerance, and activated pancreatic mTORC1 signaling. Activation of Piezo1 by its agonist Yoda1 or overexpression of Piezo1 led to decreased glucagon synthesis and suppressed mTOR signaling pathway in αTC1-6 cells. Additionally, the levels of glucagon in the medium were also reduced. Conversely, knockdown of Piezo1 produced opposite effects. CONCLUSION Our study uncovers the regulatory role of the Piezo1 ion channel in α cells. Piezo1 influences glucagon production by affecting mTOR signaling pathway.
Collapse
Affiliation(s)
- Wenying Guo
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Luyang Gao
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Haocong Mo
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Handan Deng
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Yawen Zhao
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Geyang Xu
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, Guangdong, China.
| |
Collapse
|
7
|
Johansen CG, Holcomb K, Sela A, Morrall S, Park D, Farnsworth NL. Extracellular matrix stiffness mediates insulin secretion in pancreatic islets via mechanosensitive Piezo1 channel regulated Ca 2+ dynamics. Matrix Biol Plus 2024; 22:100148. [PMID: 38803329 PMCID: PMC11128509 DOI: 10.1016/j.mbplus.2024.100148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
The pancreatic islet is surrounded by ECM that provides both biochemical and mechanical cues to the islet β-cell to regulate cell survival and insulin secretion. Changes in ECM composition and mechanical properties drive β-cell dysfunction in many pancreatic diseases. While several studies have characterized changes in islet insulin secretion with changes in substrate stiffness, little is known about the mechanotransduction signaling driving altered islet function in response to mechanical cues. We hypothesized that increasing matrix stiffness will lead to insulin secretion dysfunction by opening the mechanosensitive ion channel Piezo1 and disrupting intracellular Ca2+ dynamics in mouse and human islets. To test our hypothesis, mouse and human cadaveric islets were encapsulated in a biomimetic reverse thermal gel (RTG) scaffold with tailorable stiffness that allows formation of islet focal adhesions with the scaffold and activation of Piezo1 in 3D. Our results indicate that increased scaffold stiffness causes insulin secretion dysfunction mediated by increases in Ca2+ influx and altered Ca2+ dynamics via opening of the mechanosensitive Piezo1 channel. Additionally, inhibition of Piezo1 rescued glucose-stimulated insulin secretion (GSIS) in islets in stiff scaffolds. Overall, our results emphasize the role mechanical properties of the islet microenvironment plays in regulating function. It also supports further investigation into the modulation of Piezo1 channel activity to restore islet function in diseases like type 2 diabetes (T2D) and pancreatic cancer where fibrosis of the peri-islet ECM leads to increased tissue stiffness and islet dysfunction.
Collapse
Affiliation(s)
- Chelsea G Johansen
- Department of Chemical & Biological Engineering, Colorado School of Mines, Golden, CO 80401, USA
| | - Keifer Holcomb
- Department of Chemical & Biological Engineering, Colorado School of Mines, Golden, CO 80401, USA
| | - Amit Sela
- Quantitative Biosciences & Engineering, Colorado School of Mines, Golden, CO 80401, USA
| | - Stephanie Morrall
- Quantitative Biosciences & Engineering, Colorado School of Mines, Golden, CO 80401, USA
| | - Daewon Park
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nikki L Farnsworth
- Department of Chemical & Biological Engineering, Colorado School of Mines, Golden, CO 80401, USA
- Quantitative Biosciences & Engineering, Colorado School of Mines, Golden, CO 80401, USA
| |
Collapse
|
8
|
Molano RD, Pileggi A, Tse HM, Stabler CL, Fraker CA. A static glucose-stimulated insulin secretion (sGSIS) assay that is significantly predictive of time to diabetes reversal in the human islet bioassay. BMJ Open Diabetes Res Care 2024; 12:e003897. [PMID: 38485229 PMCID: PMC10941118 DOI: 10.1136/bmjdrc-2023-003897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/08/2024] [Indexed: 03/17/2024] Open
Abstract
INTRODUCTION Static incubation (static glucose-stimulated insulin secretion, sGSIS) is a measure of islet secretory function. The Stimulation Index (SI; insulin produced in high glucose/insulin produced in low glucose) is currently used as a product release criterion of islet transplant potency. RESEARCH DESIGN AND METHODS Our hypothesis was that the Delta, insulin secreted in high glucose minus insulin secreted in low glucose, would be more predictive. To evaluate this hypothesis, sGSIS was performed on 32 consecutive human islet preparations, immobilizing the islets in a slurry of Sepharose beads to minimize mechanical perturbation. Simultaneous full-mass subrenal capsular transplants were performed in chemically induced diabetic immunodeficient mice. Logistic regression analysis was used to determine optimal cut-points for diabetes reversal time and the Fisher Exact Test was used to assess the ability of the Delta and the SI to accurately classify transplant outcomes. Receiver operating characteristic curve analysis was performed on cut-point grouped data, assessing the predictive power and optimal cut-point for each sGSIS potency metric. Finally, standard Kaplan-Meier-type survival analysis was conducted. RESULTS In the case of the sGSIS the Delta provided a superior islet potency metric relative to the SI.ConclusionsThe sGSIS Delta value is predicitive of time to diabetes reversal in the full mass human islet transplant bioassay.
Collapse
Affiliation(s)
- Ruth Damaris Molano
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Coral Gables, Florida, USA
| | - Antonello Pileggi
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Coral Gables, Florida, USA
| | - Hubert M Tse
- Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Cherie L Stabler
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
- J Crayton Pruitt Family Department of Biomedical Engineering, University of Florida Herbert Wertheim College of Engineering, Gainesville, Florida, USA
| | - Christopher A Fraker
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
9
|
Cao S, Buchholz KS, Tan P, Stowe JC, Wang A, Fowler A, Knaus KR, Khalilimeybodi A, Zambon AC, Omens JH, Saucerman JJ, McCulloch AD. Differential sensitivity to longitudinal and transverse stretch mediates transcriptional responses in mouse neonatal ventricular myocytes. Am J Physiol Heart Circ Physiol 2024; 326:H370-H384. [PMID: 38063811 PMCID: PMC11245882 DOI: 10.1152/ajpheart.00562.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024]
Abstract
To identify how cardiomyocyte mechanosensitive signaling pathways are regulated by anisotropic stretch, micropatterned mouse neonatal cardiomyocytes were stretched primarily longitudinally or transversely to the myofiber axis. Four hours of static, longitudinal stretch induced differential expression of 557 genes, compared with 30 induced by transverse stretch, measured using RNA-seq. A logic-based ordinary differential equation model of the cardiac myocyte mechanosignaling network, extended to include the transcriptional regulation and expression of 784 genes, correctly predicted measured expression changes due to anisotropic stretch with 69% accuracy. The model also predicted published transcriptional responses to mechanical load in vitro or in vivo with 63-91% accuracy. The observed differences between transverse and longitudinal stretch responses were not explained by differential activation of specific pathways but rather by an approximately twofold greater sensitivity to longitudinal stretch than transverse stretch. In vitro experiments confirmed model predictions that stretch-induced gene expression is more sensitive to angiotensin II and endothelin-1, via RhoA and MAP kinases, than to the three membrane ion channels upstream of calcium signaling in the network. Quantitative cardiomyocyte gene expression differs substantially with the axis of maximum principal stretch relative to the myofilament axis, but this difference is due primarily to differences in stretch sensitivity rather than to selective activation of mechanosignaling pathways.NEW & NOTEWORTHY Anisotropic stretch applied to micropatterned neonatal mouse ventricular myocytes induced markedly greater acute transcriptional responses when the major axis of stretch was parallel to the myofilament axis than when it was transverse. Analysis with a novel quantitative network model of mechanoregulated cardiomyocyte gene expression suggests that this difference is explained by higher cell sensitivity to longitudinal loading than transverse loading than by the activation of differential signaling pathways.
Collapse
Affiliation(s)
- Shulin Cao
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
| | - Kyle S Buchholz
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
| | - Philip Tan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States
| | - Jennifer C Stowe
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
| | - Ariel Wang
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
| | - Annabelle Fowler
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
| | - Katherine R Knaus
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
| | - Ali Khalilimeybodi
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, United States
| | - Alexander C Zambon
- Department of Biopharmaceutical Sciences, Keck Graduate Institute, Claremont, California, United States
| | - Jeffrey H Omens
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
- Department of Medicine, University of California San Diego, La Jolla, California, United States
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
- Department of Medicine, University of California San Diego, La Jolla, California, United States
| |
Collapse
|
10
|
Drobnik M, Smólski J, Grądalski Ł, Niemirka S, Młynarska E, Rysz J, Franczyk B. Mechanosensitive Cation Channel Piezo1 Is Involved in Renal Fibrosis Induction. Int J Mol Sci 2024; 25:1718. [PMID: 38338996 PMCID: PMC10855652 DOI: 10.3390/ijms25031718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Renal fibrosis, the result of different pathological processes, impairs kidney function and architecture, and usually leads to renal failure development. Piezo1 is a mechanosensitive cation channel highly expressed in kidneys. Activation of Piezo1 by mechanical stimuli increases cations influx into the cell with slight preference of calcium ions. Two different models of Piezo1 activation are considered: force through lipid and force through filament. Expression of Piezo1 on mRNA and protein levels was confirmed within the kidney. Their capacity is increased in the fibrotic kidney. The pharmacological tools for Piezo1 research comprise selective activators of the channels (Yoda1 and Jedi1/2) as well as non-selective inhibitors (spider peptide toxin) GsMTx4. Piezo1 is hypothesized to be the upstream element responsible for the activation of integrin. This pathway (calcium/calpain2/integrin beta1) is suggested to participate in profibrotic response induced by mechanical stimuli. Administration of the Piezo1 unspecific inhibitor or activators to unilateral ureter obstruction (UUO) mice or animals with folic acid-induced fibrosis modulates extracellular matrix deposition and influences kidney function. All in all, according to the recent data Piezo1 plays an important role in kidney fibrosis development. This channel has been selected as the target for pharmacotherapy of renal fibrosis.
Collapse
Affiliation(s)
- Marta Drobnik
- Department of Nephrocardiology, Medical University of Lodz, ul. Żeromskiego 113, 90-549 Lodz, Poland; (M.D.)
| | - Jakub Smólski
- Department of Nephrocardiology, Medical University of Lodz, ul. Żeromskiego 113, 90-549 Lodz, Poland; (M.D.)
| | - Łukasz Grądalski
- Department of Nephrocardiology, Medical University of Lodz, ul. Żeromskiego 113, 90-549 Lodz, Poland; (M.D.)
| | - Szymon Niemirka
- Department of Nephrocardiology, Medical University of Lodz, ul. Żeromskiego 113, 90-549 Lodz, Poland; (M.D.)
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Żeromskiego 113, 90-549 Lodz, Poland; (M.D.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Żeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Żeromskiego 113, 90-549 Lodz, Poland; (M.D.)
| |
Collapse
|
11
|
赵 川, 王 湘, 王 贵. [Hot Topics and Emerging Trends in Mechanobiology Research]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:1-5. [PMID: 38322522 PMCID: PMC10839494 DOI: 10.12182/20240160104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Indexed: 02/08/2024]
Abstract
Mechanobiology focuses on a series of important physiopathological processes, such as how cells perceive different mechanomechanical stimuli, the process of intracellular mechanotransduction, and how mechanical signals determine the behavior and fate of cells. From the initial stage of embryogenesis, to developmental biology and regenerative medicine, or even through the whole life process, mechanical signaling cascades and cellular mechanical responses in mechanobiology are of great significance in biomedical research. In recent years, research in the field of mechanobiology has undergone remarkable development. Several scientific consortia around the world have been analyzing mechanobiological processes from different perspectives, aiming to gain insights into the regulatory mechanisms by which mechanical factors affect cell fate determination. In this article, we summarized and reviewed the topics that have attracted more research interests in recent years in the field of mechanobiology, for example, arterial blood vessels, stem cell, and ion channel. We also discussed the potential trends that may emerge, such as nuclear deformation, fibrous extracellular matrix, tumor mechanobiology, cellular mechanotransduction, and piezo ion channels. In addition, we put forward new ideas concerning the limitations of mechanism research and the importance of big data analysis and mining in this field, thereby providing objective support and a systematic framework for grasping the hot research topics and exploring new research directions in the field of mechanobiology.
Collapse
Affiliation(s)
- 川榕 赵
- 重庆大学生物工程学院,生物流变科学与技术教育部重点实验室,血管植入物开发国家地方联合工程实验室 (重庆 400045)College of Bioengineering, Chongqing University, Key Laboratory of Biorheology Science and Technology (Chongqing University), Ministry of Education, and State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400045, China
- 金凤实验室 (重庆 401329)JinFeng Laboratory, Chongqing 401329, China
| | - 湘秀 王
- 重庆大学生物工程学院,生物流变科学与技术教育部重点实验室,血管植入物开发国家地方联合工程实验室 (重庆 400045)College of Bioengineering, Chongqing University, Key Laboratory of Biorheology Science and Technology (Chongqing University), Ministry of Education, and State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400045, China
- 金凤实验室 (重庆 401329)JinFeng Laboratory, Chongqing 401329, China
| | - 贵学 王
- 重庆大学生物工程学院,生物流变科学与技术教育部重点实验室,血管植入物开发国家地方联合工程实验室 (重庆 400045)College of Bioengineering, Chongqing University, Key Laboratory of Biorheology Science and Technology (Chongqing University), Ministry of Education, and State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400045, China
- 金凤实验室 (重庆 401329)JinFeng Laboratory, Chongqing 401329, China
| |
Collapse
|
12
|
Tranter JD, Kumar A, Nair VK, Sah R. Mechanosensing in Metabolism. Compr Physiol 2023; 14:5269-5290. [PMID: 38158369 DOI: 10.1002/cphy.c230005] [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] [Indexed: 01/03/2024]
Abstract
Electrical mechanosensing is a process mediated by specialized ion channels, gated directly or indirectly by mechanical forces, which allows cells to detect and subsequently respond to mechanical stimuli. The activation of mechanosensitive (MS) ion channels, intrinsically gated by mechanical forces, or mechanoresponsive (MR) ion channels, indirectly gated by mechanical forces, results in electrical signaling across lipid bilayers, such as the plasma membrane. While the functions of mechanically gated channels within a sensory context (e.g., proprioception and touch) are well described, there is emerging data demonstrating functions beyond touch and proprioception, including mechanoregulation of intracellular signaling and cellular/systemic metabolism. Both MR and MS ion channel signaling have been shown to contribute to the regulation of metabolic dysfunction, including obesity, insulin resistance, impaired insulin secretion, and inflammation. This review summarizes our current understanding of the contributions of several MS/MR ion channels in cell types implicated in metabolic dysfunction, namely, adipocytes, pancreatic β-cells, hepatocytes, and skeletal muscle cells, and discusses MS/MR ion channels as possible therapeutic targets. © 2024 American Physiological Society. Compr Physiol 14:5269-5290, 2024.
Collapse
Affiliation(s)
- John D Tranter
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ashutosh Kumar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Vinayak K Nair
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rajan Sah
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Cardiovascular Research, Washington University, St. Louis, Missouri, USA
- St. Louis VA Medical Center, St. Louis, Missouri, USA
| |
Collapse
|
13
|
Mitrokhin V, Bilichenko A, Kazanski V, Schobik R, Shileiko S, Revkova V, Kalsin V, Kamkina O, Kamkin A, Mladenov M. Transcriptomic profile of the mechanosensitive ion channelome in human cardiac fibroblasts. Exp Biol Med (Maywood) 2023; 248:2341-2350. [PMID: 38158807 PMCID: PMC10903254 DOI: 10.1177/15353702231218488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/27/2023] [Indexed: 01/03/2024] Open
Abstract
Human cardiac fibroblasts (HCFs) have mRNA transcripts that encode different mechanosensitive ion channels and channel regulatory proteins whose functions are not known yet. The primary goal of this work was to define the mechanosensitive ion channelome of HCFs. The most common type of cationic channel is the transient receptor potential (TRP) family, which is followed by the TWIK-related K+ channel (TREK), transmembrane protein 63 (TMEM63), and PIEZO channel (PIEZO) families. In the sodium-dependent NON-voltage-gated channel (SCNN) subfamily, only SCNN1D was shown to be highly expressed. Particular members of the acid-sensing ion channel (ASIC) (ASIC1 and ASIC3) subfamilies were also significantly expressed. The transcripts per kilobase million (TPMs) for Piezo 2 were almost 100 times less abundant than those for Piezo 1. The tandem of P domains in a weak inward rectifying K+ channel (TWIK)-2 channel, TWIK-related acid-sensitive K+ channel (TASK)-5, TASK-1, and the TWIK-related K1 (TREK-1) channel were the four most prevalent types in the K2P subfamily. The highest expression in the TRPP subfamily was found for PKD2 and PKD1, while in the TRPM subfamily, it was found for TRPM4, TRPM7, and TRPM3. TRPV2, TRPV4, TRPV3, and TRPV6 (all members of the TRPV subfamily) were also substantially expressed. A strong expression of the TRPC1, TRPC4, TRPC6, and TRPC2 channels and all members of the TRPML subfamily (MCOLN1, MCOLN2, and MCOLN3) was also shown. In terms of the transmembrane protein 16 (TMEM16) family, the HCFs demonstrated significant expression of the TMEM16H, TMEM16F, TMEM16J, TMEM16A, and TMEM16G channels. TMC3 is the most expressed channel in HCFs of all known members of the transmembrane channel-like protein (TMC) family. This analysis of the mechanosensitive ionic channel transcriptome in HCFs: (1) agrees with previously documented findings that all currently identified mechanosensitive channels play a significant and well recognized physiological function in elucidating the mechanosensitive characteristics of HCFs; (2) supports earlier preliminary reports that point to the most common expression of the TRP mechanosensitive family in HCFs; and (3) points to other new mechanosensitive channels (TRPC1, TRPC2, TWIK-2, TMEM16A, ASIC1, and ASIC3).
Collapse
Affiliation(s)
- Vadim Mitrokhin
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Andrei Bilichenko
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Viktor Kazanski
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Roman Schobik
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Stanislav Shileiko
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Veronika Revkova
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Vladimir Kalsin
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Olga Kamkina
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Andre Kamkin
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Mitko Mladenov
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia
| |
Collapse
|
14
|
Abstract
All cells in the body are exposed to physical force in the form of tension, compression, gravity, shear stress, or pressure. Cells convert these mechanical cues into intracellular biochemical signals; this process is an inherent property of all cells and is essential for numerous cellular functions. A cell's ability to respond to force largely depends on the array of mechanical ion channels expressed on the cell surface. Altered mechanosensing impairs conscious senses, such as touch and hearing, and unconscious senses, like blood pressure regulation and gastrointestinal (GI) activity. The GI tract's ability to sense pressure changes and mechanical force is essential for regulating motility, but it also underlies pain originating in the GI tract. Recent identification of the mechanically activated ion channels Piezo1 and Piezo2 in the gut and the effects of abnormal ion channel regulation on cellular function indicate that these channels may play a pathogenic role in disease. Here, we discuss our current understanding of mechanically activated Piezo channels in the pathogenesis of pancreatic and GI diseases, including pancreatitis, diabetes mellitus, irritable bowel syndrome, GI tumors, and inflammatory bowel disease. We also describe how Piezo channels could be important targets for treating GI diseases.
Collapse
|
15
|
He J, Xie X, Xiao Z, Qian W, Zhang L, Hou X. Piezo1 in Digestive System Function and Dysfunction. Int J Mol Sci 2023; 24:12953. [PMID: 37629134 PMCID: PMC10454946 DOI: 10.3390/ijms241612953] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Piezo1, a non-selective cation channel directly activated by mechanical forces, is widely expressed in the digestive system and participates in biological functions physiologically and pathologically. In this review, we summarized the latest insights on Piezo1's cellular effect across the entire digestive system, and discussed the role of Piezo1 in various aspects including ingestion and digestion, material metabolism, enteric nervous system, intestinal barrier, and inflammatory response within digestive system. The goal of this comprehensive review is to provide a solid foundation for future research about Piezo1 in digestive system physiologically and pathologically.
Collapse
Affiliation(s)
| | | | | | | | - Lei Zhang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.H.); (X.X.); (Z.X.); (W.Q.)
| | - Xiaohua Hou
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.H.); (X.X.); (Z.X.); (W.Q.)
| |
Collapse
|
16
|
Hatem A, Poussereau G, Gachenot M, Pérès L, Bouyer G, Egée S. Dual action of Dooku1 on PIEZO1 channel in human red blood cells. Front Physiol 2023; 14:1222983. [PMID: 37492641 PMCID: PMC10365639 DOI: 10.3389/fphys.2023.1222983] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/27/2023] [Indexed: 07/27/2023] Open
Abstract
PIEZO1 is a mechanosensitive non-selective cation channel, present in many cell types including Red Blood Cells (RBCs). Together with the Gárdos channel, PIEZO1 forms in RBCs a tandem that participates in the rapid adjustment of the cell volume. The pharmacology allowing functional studies of the roles of PIEZO1 has only recently been developed, with Yoda1 as a widely used PIEZO1 agonist. In 2018, Yoda1 analogues were developed, as a step towards an improved understanding of PIEZO1 roles and functions. Among these, Dooku1 was the most promising antagonist of Yoda1-induced effects, without having any ability to activate PIEZO1 channels. Since then, Dooku1 has been used in various cell types to antagonize Yoda1 effects. In the present study using RBCs, Dooku1 shows an apparent IC50 on Yoda1 effects of 90.7 µM, one order of magnitude above the previously reported data on other cell types. Unexpectedly, it was able, by itself, to produce entry of calcium sufficient to trigger Gárdos channel activation. Moreover, Dooku1 evoked a rise in intracellular sodium concentrations, suggesting that it targets a non-selective cation channel. Dooku1 effects were abolished upon using GsMTx4, a known mechanosensitive channel blocker, indicating that Dooku1 likely targets PIEZO1. Our observations lead to the conclusion that Dooku1 behaves as a PIEZO1 agonist in the RBC membrane, similarly to Yoda1 but with a lower potency. Taken together, these results show that the pharmacology of PIEZO1 in RBCs must be interpreted with care especially due to the unique characteristics of RBC membrane and associated cytoskeleton.
Collapse
Affiliation(s)
- Aline Hatem
- Sorbonne Université, CNRS, UMR8227 LBI2M, Station Biologique de Roscoff, Roscoff, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Gwendal Poussereau
- Sorbonne Université, CNRS, UMR8227 LBI2M, Station Biologique de Roscoff, Roscoff, France
| | - Martin Gachenot
- Sorbonne Université, CNRS, FR2424, Station Biologique de Roscoff, Roscoff, France
| | - Laurent Pérès
- Sorbonne Université, CNRS, UMR8227 LBI2M, Station Biologique de Roscoff, Roscoff, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Guillaume Bouyer
- Sorbonne Université, CNRS, UMR8227 LBI2M, Station Biologique de Roscoff, Roscoff, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Stéphane Egée
- Sorbonne Université, CNRS, UMR8227 LBI2M, Station Biologique de Roscoff, Roscoff, France
- Laboratory of Excellence GR-Ex, Paris, France
| |
Collapse
|
17
|
Yu X, Dai C, Zhao X, Huang Q, He X, Zhang R, Lin Z, Shen Y. Ruthenium red attenuates acute pancreatitis by inhibiting MCU and improving mitochondrial function. Biochem Biophys Res Commun 2022; 635:236-243. [DOI: 10.1016/j.bbrc.2022.10.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 09/28/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022]
|
18
|
Ponomareva S, Joisten H, François T, Naud C, Morel R, Hou Y, Myers T, Joumard I, Dieny B, Carriere M. Magnetic particles for triggering insulin release in INS-1E cells subjected to a rotating magnetic field. NANOSCALE 2022; 14:13274-13283. [PMID: 36056640 DOI: 10.1039/d2nr02009b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Diabetes is a major global health threat. Both academics and industry are striving to develop effective treatments for this disease. In this work, we present a new approach to induce insulin release from β-islet pancreatic cells (INS-1E) by mechanical stimulation. Two types of experiments were carried out. First, a local stimulation was performed by dispersing anisotropic magnetic particles within the cell medium, which settled down almost immediately on cell plasma membranes. Application of a low frequency magnetic field (up to 40 Hz) generated by a custom-made magnetic device resulted in oscillations of these particles, which then exerted a mechanical constraint on the cell plasma membranes. The second type of experiment consisted of a global stimulation, where cells were grown on magneto-elastic membranes composed of a biocompatible polymer with embedded magnetic particles. Upon application of a rotating magnetic field, magnetic particles within the membrane were attracted towards the field source, resulting in the membrane's vibrations being transmitted to the cells grown on it. In both experiments, the cell response to these mechanical stimulations caused by application of the variable magnetic field was quantified via the measurement of insulin release in the growth medium. We demonstrated that the mechanical action induced by the motion of magnetic particles or by membrane vibrations was an efficient stimulus for insulin granule secretion from β-cells. This opens a wide range of possible applications including the design of a system which triggers insulin secretion by β-islet pancreatic cells on demand.
Collapse
Affiliation(s)
- Svetlana Ponomareva
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SPINTEC, 38000 Grenoble, France.
| | - Helene Joisten
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SPINTEC, 38000 Grenoble, France.
- Univ. Grenoble Alpes, CEA, Leti, 38000 Grenoble, France
| | - Taina François
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SYMMES, 38000 Grenoble, France.
| | - Cecile Naud
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SPINTEC, 38000 Grenoble, France.
| | - Robert Morel
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SPINTEC, 38000 Grenoble, France.
| | - Yanxia Hou
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SYMMES, 38000 Grenoble, France.
| | - Thomas Myers
- Platform Kinetics, Pegholme, Wharfebank Mills, Otley, LS21 3JP, UK
| | - Isabelle Joumard
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SPINTEC, 38000 Grenoble, France.
| | - Bernard Dieny
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SPINTEC, 38000 Grenoble, France.
| | - Marie Carriere
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SYMMES, 38000 Grenoble, France.
| |
Collapse
|
19
|
Identifying interpretable gene-biomarker associations with functionally informed kernel-based tests in 190,000 exomes. Nat Commun 2022; 13:5332. [PMID: 36088354 PMCID: PMC9464252 DOI: 10.1038/s41467-022-32864-2] [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: 01/20/2022] [Accepted: 08/22/2022] [Indexed: 12/05/2022] Open
Abstract
Here we present an exome-wide rare genetic variant association study for 30 blood biomarkers in 191,971 individuals in the UK Biobank. We compare gene-based association tests for separate functional variant categories to increase interpretability and identify 193 significant gene-biomarker associations. Genes associated with biomarkers were ~ 4.5-fold enriched for conferring Mendelian disorders. In addition to performing weighted gene-based variant collapsing tests, we design and apply variant-category-specific kernel-based tests that integrate quantitative functional variant effect predictions for missense variants, splicing and the binding of RNA-binding proteins. For these tests, we present a computationally efficient combination of the likelihood-ratio and score tests that found 36% more associations than the score test alone while also controlling the type-1 error. Kernel-based tests identified 13% more associations than their gene-based collapsing counterparts and had advantages in the presence of gain of function missense variants. We introduce local collapsing by amino acid position for missense variants and use it to interpret associations and identify potential novel gain of function variants in PIEZO1. Our results show the benefits of investigating different functional mechanisms when performing rare-variant association tests, and demonstrate pervasive rare-variant contribution to biomarker variability.
Collapse
|
20
|
Tseng MC, Lim J, Chu YC, Chen CW, Feng CK, Wang JL. Dynamic Pressure Stimulation Upregulates Collagen II and Aggrecan in Nucleus Pulposus Cells Through Calcium Signaling. Spine (Phila Pa 1976) 2022; 47:1111-1119. [PMID: 34812197 DOI: 10.1097/brs.0000000000004286] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/25/2021] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN An in vitro study to investigate the effect of pressure stimulation on nucleus pulposus (NP) cells. OBJECTIVE The aim of this study was to investigate the question whether physical stimulation can be leveraged to enhance extracellular matrix (ECM) synthesis as a preventive measure for intervertebral disc (IVD) degeneration. SUMMARY OF BACKGROUND DATA ECM plays an important role in regulating hydration and pressure balance of the IVD. METHODS Cellular stimulation devices with different pressurizing protocols were used to create a pressurized environment to cells cultures. The setup was used to mimic the pressurized conditions within IVD to investigate the effect of pressure stimulation on NP cells. RESULTS Pressure stimulation at 300 kPa can enhance the synthesis of ECM proteins Collagen II and aggrecan in NP cells and the effect of dynamic pressure stimulation outperformed the static one. The difference between static and dynamic pressure stimulation was due primarily to calcium signaling activated by pressure fluctuation. The superior effect of dynamic pressure holds for a wide range of stimulation durations, relating to the range of spontaneous calcium oscillations in NP cells. CONCLUSION The results link mechanotransduction to the downstream ECM protein synthesis and suggest slow exercises that correspond with spontaneous calcium oscillations in NP cells can be effective to stimulate ECM synthesis in IVD.
Collapse
Affiliation(s)
- Mu-Cyun Tseng
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
21
|
A critical role of the mechanosensor PIEZO1 in glucose-induced insulin secretion in pancreatic β-cells. Nat Commun 2022; 13:4237. [PMID: 35869052 PMCID: PMC9307633 DOI: 10.1038/s41467-022-31103-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/06/2022] [Indexed: 11/08/2022] Open
Abstract
Glucose-induced insulin secretion depends on β-cell electrical activity. Inhibition of ATP-regulated potassium (KATP) channels is a key event in this process. However, KATP channel closure alone is not sufficient to induce β-cell electrical activity; activation of a depolarizing membrane current is also required. Here we examine the role of the mechanosensor ion channel PIEZO1 in this process. Yoda1, a specific PIEZO1 agonist, activates a small membrane current and thereby triggers β-cell electrical activity with resultant stimulation of Ca2+-influx and insulin secretion. Conversely, the PIEZO1 antagonist GsMTx4 reduces glucose-induced Ca2+-signaling, electrical activity and insulin secretion. Yet, PIEZO1 expression is elevated in islets from human donors with type-2 diabetes (T2D) and a rodent T2D model (db/db mouse), in which insulin secretion is reduced. This paradox is resolved by our finding that PIEZO1 translocates from the plasmalemma into the nucleus (where it cannot influence the membrane potential of the β-cell) under experimental conditions emulating T2D (high glucose culture). β-cell-specific Piezo1-knockout mice show impaired glucose tolerance in vivo and reduced glucose-induced insulin secretion, β-cell electrical activity and Ca2+ elevation in vitro. These results implicate mechanotransduction and activation of PIEZO1, via intracellular accumulation of glucose metabolites, as an important physiological regulator of insulin secretion.
Collapse
|
22
|
Wijerathne TD, Ozkan AD, Lacroix JJ. Yoda1's energetic footprint on Piezo1 channels and its modulation by voltage and temperature. Proc Natl Acad Sci U S A 2022; 119:e2202269119. [PMID: 35858335 PMCID: PMC9303978 DOI: 10.1073/pnas.2202269119] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/31/2022] [Indexed: 01/13/2023] Open
Abstract
Piezo1 channels are essential mechanically activated ion channels in vertebrates. Their selective activation by the synthetic chemical activator Yoda1 opened new avenues to probe their gating mechanisms and develop novel pharmaceuticals. Yet, the nature and extent of Piezo1 functions modulated by this small molecule remain unclear. Here we close this gap by conducting a comprehensive biophysical investigation of the effects of Yoda1 on mouse Piezo1 in mammalian cells. Using calcium imaging, we first show that cysteine bridges known to inhibit mechanically evoked Piezo1 currents also inhibit activation by Yoda1, suggesting Yoda1 acts by energetically modulating mechanosensory domains. The presence of Yoda1 alters single-channel dwell times and macroscopic kinetics consistent with a dual and reciprocal energetic modulation of open and shut states. Critically, we further discovered that the electrophysiological effects of Yoda1 depend on membrane potential and temperature, two other Piezo1 modulators. This work illuminates a complex interplay between physical and chemical modulators of Piezo1 channels.
Collapse
Affiliation(s)
- Tharaka D. Wijerathne
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766
| | - Alper D. Ozkan
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766
| | - Jérôme J. Lacroix
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766
| |
Collapse
|
23
|
Gao DD, Huang JH, Ding N, Deng WJ, Li PL, Mai YN, Wu JR, Hu M. Mechanosensitive Piezo1 channel in rat epididymal epithelial cells promotes transepithelial K+ secretion. Cell Calcium 2022; 104:102571. [DOI: 10.1016/j.ceca.2022.102571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/28/2022]
|
24
|
Li X, Hu J, Zhao X, Li J, Chen Y. Piezo channels in the urinary system. Exp Mol Med 2022; 54:697-710. [PMID: 35701561 PMCID: PMC9256749 DOI: 10.1038/s12276-022-00777-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/25/2022] [Accepted: 02/16/2022] [Indexed: 12/24/2022] Open
Abstract
The Piezo channel family, including Piezo1 and Piezo2, includes essential mechanosensitive transduction molecules in mammals. Functioning in the conversion of mechanical signals to biological signals to regulate a plethora of physiological processes, Piezo channels, which have a unique homotrimeric three-blade propeller-shaped structure, utilize a cap-motion and plug-and-latch mechanism to gate their ion-conducting pathways. Piezo channels have a wide range of biological roles in various human systems, both in vitro and in vivo. Currently, there is a lack of comprehensive understanding of their antagonists and agonists, and therefore further investigation is needed. Remarkably, increasingly compelling evidence demonstrates that Piezo channel function in the urinary system is important. This review article systematically summarizes the existing evidence of the importance of Piezo channels, including protein structure, mechanogating mechanisms, and pharmacological characteristics, with a particular focus on their physiological and pathophysiological roles in the urinary system. Collectively, this review aims to provide a direction for future clinical applications in urinary system diseases.
Collapse
Affiliation(s)
- Xu Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Junwei Hu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Xuedan Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Juanjuan Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yuelai Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
25
|
Kim HS, Suh JS, Jang YK, Ahn SH, Choi GH, Yang JY, Lim GH, Jung Y, Jiang J, Sun J, Suk M, Wang Y, Kim TJ. Förster Resonance Energy Transfer-Based Single-Cell Imaging Reveals Piezo1-Induced Ca 2+ Flux Mediates Membrane Ruffling and Cell Survival. Front Cell Dev Biol 2022; 10:865056. [PMID: 35646889 PMCID: PMC9136143 DOI: 10.3389/fcell.2022.865056] [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: 01/29/2022] [Accepted: 04/25/2022] [Indexed: 01/18/2023] Open
Abstract
A mechanosensitive ion channel, Piezo1 induces non-selective cation flux in response to various mechanical stresses. However, the biological interpretation and underlying mechanisms of cells resulting from Piezo1 activation remain elusive. This study elucidates Piezo1-mediated Ca2+ influx driven by channel activation and cellular behavior using novel Förster Resonance Energy Transfer (FRET)-based biosensors and single-cell imaging analysis. Results reveal that extracellular Ca2+ influx via Piezo1 requires intact caveolin, cholesterol, and cytoskeletal support. Increased cytoplasmic Ca2+ levels enhance PKA, ERK, Rac1, and ROCK activity, which have the potential to promote cancer cell survival and migration. Furthermore, we demonstrate that Piezo1-mediated Ca2+ influx upregulates membrane ruffling, a characteristic feature of cancer cell metastasis, using spatiotemporal image correlation spectroscopy. Thus, our findings provide new insights into the function of Piezo1, suggesting that Piezo1 plays a significant role in the behavior of cancer cells.
Collapse
Affiliation(s)
- Heon-Su Kim
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea,Institute of Systems Biology, Pusan National University, Pusan, South Korea
| | - Jung-Soo Suh
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Yoon-Kwan Jang
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Sang-Hyun Ahn
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Gyu-Ho Choi
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Jin-Young Yang
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Gah-Hyun Lim
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Youngmi Jung
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Jie Jiang
- Department of Cell Biology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jie Sun
- Department of Cell Biology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Myungeun Suk
- Department of Mechanical Engineering, Dong-Eui University, Pusan, South Korea
| | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Tae-Jin Kim
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea,Institute of Systems Biology, Pusan National University, Pusan, South Korea,Department of Biological Sciences, Pusan National University, Pusan, South Korea,*Correspondence: Tae-Jin Kim,
| |
Collapse
|
26
|
Zhang HB, Ding XB, Jin J, Guo WP, Yang QL, Chen PC, Yao H, Ruan L, Tao YT, Chen X. Predicted mouse interactome and network-based interpretation of differentially expressed genes. PLoS One 2022; 17:e0264174. [PMID: 35390003 PMCID: PMC8989236 DOI: 10.1371/journal.pone.0264174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 02/04/2022] [Indexed: 11/18/2022] Open
Abstract
The house mouse or Mus musculus has become a premier mammalian model for genetic research due to its genetic and physiological similarities to humans. It brought mechanistic insights into numerous human diseases and has been routinely used to assess drug efficiency and toxicity, as well as to predict patient responses. To facilitate molecular mechanism studies in mouse, we present the Mouse Interactome Database (MID, Version 1), which includes 155,887 putative functional associations between mouse protein-coding genes inferred from functional association evidence integrated from 9 public databases. These putative functional associations are expected to cover 19.32% of all mouse protein interactions, and 26.02% of these function associations may represent protein interactions. On top of MID, we developed a gene set linkage analysis (GSLA) web tool to annotate potential functional impacts from observed differentially expressed genes. Two case studies show that the MID/GSLA system provided precise and informative annotations that other widely used gene set annotation tools, such as PANTHER and DAVID, did not. Both MID and GSLA are accessible through the website http://mouse.biomedtzc.cn.
Collapse
Affiliation(s)
- Hai-Bo Zhang
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
| | - Xiao-Bao Ding
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
| | - Jie Jin
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
| | - Wen-Ping Guo
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
| | - Qiao-Lei Yang
- Institute of Pharmaceutical Biotechnology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Peng-Cheng Chen
- Institute of Pharmaceutical Biotechnology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Heng Yao
- Institute of Pharmaceutical Biotechnology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Li Ruan
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
| | - Yu-Tian Tao
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
- * E-mail: (YTT); (XC)
| | - Xin Chen
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
- Institute of Pharmaceutical Biotechnology, School of Medicine, Zhejiang University, Hangzhou, China
- Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University, Hangzhou, China
- * E-mail: (YTT); (XC)
| |
Collapse
|
27
|
Shinge SAU, Zhang D, Din AU, Yu F, Nie Y. Emerging Piezo1 signaling in inflammation and atherosclerosis; a potential therapeutic target. Int J Biol Sci 2022; 18:923-941. [PMID: 35173527 PMCID: PMC8771847 DOI: 10.7150/ijbs.63819] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/08/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose of Review: Atherosclerosis is the principal cause of cardiovascular diseases (CVDs) which are the major cause of death worldwide. Mechanical force plays an essential role in cardiovascular health and disease. To bring the awareness of mechanosensitive Piezo1 role in atherosclerosis and its therapeutic potentials we review recent literature to highlight its involvement in various mechanisms of the disease. Recent Findings: Recent studies reported Piezo1 channel as a sensor, and transducer of various mechanical forces into biochemical signals, which affect various cellular activities such as proliferation, migration, apoptosis and vascular remodeling including immune/inflammatory mechanisms fundamental phenomenon in atherogenesis. Summary: Numerous evidences suggest Piezo1 as a player in different mechanisms of cell biology, including immune/inflammatory and other cellular mechanisms correlated with atherosclerosis. This review discusses mechanistic insight about this matter and highlights the drugability and therapeutic potentials consistent with emerging functions Piezo1 in various mechanisms of atherosclerosis. Based on the recent works, we suggest Piezo1 as potential therapeutic target and a valid candidate for future research. Therefore, a deeper exploration of Piezo1 biology and translation towards the clinic will be a novel strategy for treating atherosclerosis and other CVDs.
Collapse
Affiliation(s)
- Shafiu A. Umar Shinge
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
| | - Daifang Zhang
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
- Clinical Research Center, Southwest Medical University, Luzhou, Sichuan PRC
| | - Ahmad Ud Din
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan PRC
| | - FengXu Yu
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, Sichuan PRC
| | - YongMei Nie
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, Sichuan PRC
| |
Collapse
|
28
|
Tentonin 3/TMEM150C regulates glucose-stimulated insulin secretion in pancreatic β-cells. Cell Rep 2021; 37:110067. [PMID: 34852221 DOI: 10.1016/j.celrep.2021.110067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/17/2021] [Accepted: 11/08/2021] [Indexed: 11/24/2022] Open
Abstract
Glucose homeostasis is initially regulated by the pancreatic hormone insulin. Glucose-stimulated insulin secretion in β-cells is composed of two cellular mechanisms: a high glucose concentration not only depolarizes the membrane potential of the β-cells by ATP-sensitive K+ channels but also induces cell inflation, which is sufficient to release insulin granules. However, the molecular identity of the stretch-activated cation channel responsible for the latter pathway remains unknown. Here, we demonstrate that Tentonin 3/TMEM150C (TTN3), a mechanosensitive channel, contributes to glucose-stimulated insulin secretion by mediating cation influx. TTN3 is expressed specifically in β-cells and mediates cation currents to glucose and hypotonic stimulations. The glucose-induced depolarization, firing activity, and Ca2+ influx of β-cells were significantly lower in Ttn3-/- mice. More importantly, Ttn3-/- mice show impaired glucose tolerance with decreased insulin secretion in vivo. We propose that TTN3, as a stretch-activated cation channel, contributes to glucose-stimulated insulin secretion.
Collapse
|
29
|
Bryniarska-Kubiak N, Kubiak A, Lekka M, Basta-Kaim A. The emerging role of mechanical and topographical factors in the development and treatment of nervous system disorders: dark and light sides of the force. Pharmacol Rep 2021; 73:1626-1641. [PMID: 34390472 PMCID: PMC8599311 DOI: 10.1007/s43440-021-00315-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/14/2022]
Abstract
Nervous system diseases are the subject of intensive research due to their association with high mortality rates and their potential to cause irreversible disability. Most studies focus on targeting the biological factors related to disease pathogenesis, e.g. use of recombinant activator of plasminogen in the treatment of stroke. Nevertheless, multiple diseases such as Parkinson’s disease and Alzheimer’s disease still lack successful treatment. Recently, evidence has indicated that physical factors such as the mechanical properties of cells and tissue and topography play a crucial role in homeostasis as well as disease progression. This review aims to depict these factors’ roles in the progression of nervous system diseases and consequently discusses the possibility of new therapeutic approaches. The literature is reviewed to provide a deeper understanding of the roles played by physical factors in nervous system disease development to aid in the design of promising new treatment approaches.
Collapse
Affiliation(s)
- Natalia Bryniarska-Kubiak
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| | - Andrzej Kubiak
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, 31342, Kraków, Poland
| | - Małgorzata Lekka
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, 31342, Kraków, Poland
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| |
Collapse
|
30
|
Jairaman A, Othy S, Dynes JL, Yeromin AV, Zavala A, Greenberg ML, Nourse JL, Holt JR, Cahalan SM, Marangoni F, Parker I, Pathak MM, Cahalan MD. Piezo1 channels restrain regulatory T cells but are dispensable for effector CD4 + T cell responses. SCIENCE ADVANCES 2021; 7:7/28/eabg5859. [PMID: 34233878 PMCID: PMC8262815 DOI: 10.1126/sciadv.abg5859] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/24/2021] [Indexed: 05/03/2023]
Abstract
T lymphocytes encounter complex mechanical cues during an immune response. The mechanosensitive ion channel, Piezo1, drives inflammatory responses to bacterial infections, wound healing, and cancer; however, its role in helper T cell function remains unclear. In an animal model for multiple sclerosis, experimental autoimmune encephalomyelitis (EAE), we found that mice with genetic deletion of Piezo1 in T cells showed diminished disease severity. Unexpectedly, Piezo1 was not essential for lymph node homing, interstitial motility, Ca2+ signaling, T cell proliferation, or differentiation into proinflammatory T helper 1 (TH1) and TH17 subsets. However, Piezo1 deletion in T cells resulted in enhanced transforming growth factor-β (TGFβ) signaling and an expanded pool of regulatory T (Treg) cells. Moreover, mice with deletion of Piezo1 specifically in Treg cells showed significant attenuation of EAE. Our results indicate that Piezo1 selectively restrains Treg cells, without influencing activation events or effector T cell functions.
Collapse
Affiliation(s)
- Amit Jairaman
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Joseph L Dynes
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Andriy V Yeromin
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Angel Zavala
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Milton L Greenberg
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Jamison L Nourse
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
| | - Jesse R Holt
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
| | - Stuart M Cahalan
- Howard Hughes Medical Institute, Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
- Vertex Pharmaceuticals, 3215 Merryfield Row, San Diego, CA 92121, USA
| | - Francesco Marangoni
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Ian Parker
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Medha M Pathak
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
- Center for Complex Systems Biology, University of California, Irvine, CA 92697, USA
| | - Michael D Cahalan
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA.
- Institute for Immunology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
31
|
Stewart L, Turner NA. Channelling the Force to Reprogram the Matrix: Mechanosensitive Ion Channels in Cardiac Fibroblasts. Cells 2021; 10:990. [PMID: 33922466 PMCID: PMC8145896 DOI: 10.3390/cells10050990] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/13/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac fibroblasts (CF) play a pivotal role in preserving myocardial function and integrity of the heart tissue after injury, but also contribute to future susceptibility to heart failure. CF sense changes to the cardiac environment through chemical and mechanical cues that trigger changes in cellular function. In recent years, mechanosensitive ion channels have been implicated as key modulators of a range of CF functions that are important to fibrotic cardiac remodelling, including cell proliferation, myofibroblast differentiation, extracellular matrix turnover and paracrine signalling. To date, seven mechanosensitive ion channels are known to be functional in CF: the cation non-selective channels TRPC6, TRPM7, TRPV1, TRPV4 and Piezo1, and the potassium-selective channels TREK-1 and KATP. This review will outline current knowledge of these mechanosensitive ion channels in CF, discuss evidence of the mechanosensitivity of each channel, and detail the role that each channel plays in cardiac remodelling. By better understanding the role of mechanosensitive ion channels in CF, it is hoped that therapies may be developed for reducing pathological cardiac remodelling.
Collapse
Affiliation(s)
| | - Neil A. Turner
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, UK;
| |
Collapse
|
32
|
Jiang L, Zhang Y, Lu D, Huang T, Yan K, Yang W, Gao J. Mechanosensitive Piezo1 channel activation promotes ventilator-induced lung injury via disruption of endothelial junctions in ARDS rats. Biochem Biophys Res Commun 2021; 556:79-86. [PMID: 33839418 DOI: 10.1016/j.bbrc.2021.03.163] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 03/29/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE This study aimed to investigate the role of endothelial Piezo1 in mediating ventilator-induced lung injury secondary to acute respiratory distress syndrome (ARDS). METHODS Rats and lung endothelial cells (ECs) were transfected with Piezo1 shRNA (shPiezo1) and Piezo1 siRNA, respectively, to knock down Piezo1. Intratracheal instillation or incubation with lipopolysaccharide (LPS) was used to establish an ARDS model, and high tidal volume (HVT) ventilation or 20% cyclic stretch (CS) was administered to simulate a two-hit injury. Lung injury, alterations in lung endothelial barrier, disruption of adherens junctions (AJs), and Ca2+ influx were assessed. RESULTS Lung vascular hyperpermeability was further increased in ARDS rats following HVT ventilation, which was abrogated in shPiezo1-treated rats. 20% CS led to severer rupture of AJs following LPS stimulation as indicated by immunofluorescence staining. The internalization and degradation of VE-cadherin were blocked by knockdown of Piezo1. Additionally, 20% CS induced Piezo1 activation, manifesting as elevated intracellular Ca2+ concentration in LPS-treated ECs, and subsequently increased calcium-dependent calpain activity. Pharmacological inhibition of calpain or Piezo1 knockdown prevented the loss of VE-cadherin, p120-catenin, and β-catenin in ARDS rats undergoing HVT ventilation and LPS-treated ECs exposed to 20% CS. CONCLUSION Excessive mechanical stretch during ARDS induces the activation of Piezo1 channel and its downstream target, calpain, via Ca2+ influx. This results in the disassembly of endothelial AJs and further facilitates lung endothelial barrier breakdown and vascular hyperpermeability.
Collapse
Affiliation(s)
- Lulu Jiang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yang Zhang
- Department of Anesthesiology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, 225001, China
| | - Dahao Lu
- Department of Anesthesiology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, 225001, China
| | - Tianfeng Huang
- Department of Anesthesiology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, 225001, China
| | - Keshi Yan
- Department of Anesthesiology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, 225001, China
| | - Wenjun Yang
- Department of Anesthesiology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, 225001, China
| | - Ju Gao
- Department of Anesthesiology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
33
|
Zhang M, Yan S, Xu X, Yu T, Guo Z, Ma M, Zhang Y, Gu Z, Feng Y, Du C, Wan M, Hu K, Han X, Gu N. Three-dimensional cell-culture platform based on hydrogel with tunable microenvironmental properties to improve insulin-secreting function of MIN6 cells. Biomaterials 2021; 270:120687. [PMID: 33540170 DOI: 10.1016/j.biomaterials.2021.120687] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/10/2020] [Accepted: 01/18/2021] [Indexed: 01/13/2023]
Abstract
Pancreatic β-cells have been reported to be mechanosensitive to cellular microenvironments, and subjecting the cells to more physiologically relevant microenvironments can produce better results than when subjecting them to the conventional two-dimensional (2D) cell-culture conditions. In this work, we propose a novel three-dimensional (3D) strategy for inducing multicellular spheroid formation based on hydrogels with tunable mechanical and interfacial properties. The results indicate that MIN6 cells can sense the substrates and form tightly clustered monolayers or multicellular spheroids on hydrogels with tunable physical properties. Compared to the conventional 2D cell-culture system, the glucose sensitivities of the MIN6 cells cultured in the 3D culture model is enhanced greatly and their insulin content (relative to the amount of protein) is increased 7.3-7.9 folds. Moreover, the relative gene and protein expression levels of some key factors such as Pdx1, NeuroD1, Piezo1, and Rac1 in the MIN6 cells are significantly higher on the 3D platform, compared to the 2D control group. We believe that this 3D cell-culture system developed for the generation of multicellular spheroids will be a promising platform for diabetes treatment in clinical islet transplantation.
Collapse
Affiliation(s)
- Miao Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Sen Yan
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xueqin Xu
- Key Laboratory of Clinical and Medical Engineering, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Tingting Yu
- Department of Medical Genetics, School of Basic Medical Science & Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Ming Ma
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yi Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Zhuxiao Gu
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yiwei Feng
- Key Laboratory of Clinical and Medical Engineering, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Chunyue Du
- Key Laboratory of Clinical and Medical Engineering, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Mengqi Wan
- Key Laboratory of Clinical and Medical Engineering, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ke Hu
- Key Laboratory of Clinical and Medical Engineering, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
34
|
Zhang Q, Dou H, Rorsman P. 'Resistance is futile?' - paradoxical inhibitory effects of K ATP channel closure in glucagon-secreting α-cells. J Physiol 2020; 598:4765-4780. [PMID: 32716554 PMCID: PMC7689873 DOI: 10.1113/jp279775] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
By secreting insulin and glucagon, the β- and α-cells of the pancreatic islets play a central role in the regulation of systemic metabolism. Both cells are equipped with ATP-regulated potassium (KATP ) channels that are regulated by the intracellular ATP/ADP ratio. In β-cells, KATP channels are active at low (non-insulin-releasing) glucose concentrations. An increase in glucose leads to KATP channel closure, membrane depolarization and electrical activity that culminates in elevation of [Ca2+ ]i and initiation of exocytosis of the insulin-containing secretory granules. The α-cells are also equipped with KATP channels but they are under strong tonic inhibition at low glucose, explaining why α-cells are electrically active under hypoglycaemic conditions and generate large Na+ - and Ca2+ -dependent action potentials. Closure of residual KATP channel activity leads to membrane depolarization and an increase in action potential firing but this stimulation of electrical activity is associated with inhibition rather than acceleration of glucagon secretion. This paradox arises because membrane depolarization reduces the amplitude of the action potentials by voltage-dependent inactivation of the Na+ channels involved in action potential generation. Exocytosis in α-cells is tightly linked to the opening of voltage-gated P/Q-type Ca2+ channels, the activation of which is steeply voltage-dependent. Accordingly, the inhibitory effect of the reduced action potential amplitude exceeds the stimulatory effect resulting from the increased action potential frequency. These observations highlight a previously unrecognised role of the action potential amplitude as a key regulator of pancreatic islet hormone secretion.
Collapse
Affiliation(s)
- Quan Zhang
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Haiqiang Dou
- Metabolic Physiology Unit, Institute of Neuroscience and Physiology, University of Göteborg, PO Box 430, Göteborg, SE-405 30, Sweden
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK.,Metabolic Physiology Unit, Institute of Neuroscience and Physiology, University of Göteborg, PO Box 430, Göteborg, SE-405 30, Sweden
| |
Collapse
|