1
|
Prudinnik DS, Kussanova A, Vorobjev IA, Tikhonov A, Ataullakhanov FI, Barteneva NS. Deformability of Heterogeneous Red Blood Cells in Aging and Related Pathologies. Aging Dis 2025:AD.2024.0526. [PMID: 39012672 DOI: 10.14336/ad.2024.0526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 06/19/2024] [Indexed: 07/17/2024] Open
Abstract
Aging is interrelated with changes in red blood cell parameters and functionality. In this article, we focus on red blood cells (RBCs) and provide a review of the known changes associated with the characterization of RBC deformability in aging and related pathologies. The biophysical parameters complement the commonly used biochemical parameters and may contribute to a better understanding of the aging process. The power of the deformability measurement approach is well established in clinical settings. Measuring RBCs' deformability has the advantage of relative simplicity, and it reflects the complex effects developing in erythrocytes during aging. However, aging and related pathological conditions also promote heterogeneity of RBC features and have a certain impact on the variance in erythrocyte cell properties. The possible applications of deformability as an early biophysical biomarker of pathological states are discussed, and modulating PIEZO1 as a therapeutic target is suggested. The changes in RBCs' shape can serve as a proxy for deformability evaluation, leveraging single-cell analysis with imaging flow cytometry and artificial intelligence algorithms. The characterization of biophysical parameters of RBCs is in progress in humans and will provide a better understanding of the complex dynamics of aging.
Collapse
Affiliation(s)
- Dmitry S Prudinnik
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
| | - Aigul Kussanova
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
| | - Ivan A Vorobjev
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
| | - Alexander Tikhonov
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
| | - Fazly I Ataullakhanov
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Natasha S Barteneva
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
| |
Collapse
|
2
|
Yang X, Wang X, Yang Z, Lu H. Iron-Mediated Regulation in Adipose Tissue: A Comprehensive Review of Metabolism and Physiological Effects. Curr Obes Rep 2025; 14:4. [PMID: 39753935 DOI: 10.1007/s13679-024-00600-0] [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] [Accepted: 12/11/2024] [Indexed: 01/14/2025]
Abstract
PURPOSE OF REVIEW Review the latest data regarding the intersection of adipose tissue (AT) and iron to meet the needs of AT metabolism and the progression of related diseases. RECENT FINDINGS Iron is involved in fundamental biological metabolic processes and is precisely fine-tuned within the body to maintain cellular, tissue and even systemic iron homeostasis. AT not only serves as an energy storage depot but also represents the largest endocrine organ in the human body, maintaining systemic metabolic homeostasis. It is involved in physiological processes such as energy storage, insulin sensitivity regulation and lipid metabolism. As a unique iron-sensing tissue, AT expresses related regulatory factors, including the classic hepcidin, ferroportin (FPN), iron regulatory protein/iron responsive element (IRP/IRE) and ferritin. Consequently, the interaction between AT and iron is intricately intertwined. Imbalance of iron homeostasis produces the potential risks of steatosis, impaired glucose tolerance and insulin resistance, leading to AT dysfunction diseases, including obesity, type 2 diabetes and metabolic dysfunction-associated steatotic liver disease (MASLD). Despite the role of AT iron has garnered increasing attention in recent years, a comprehensive review that systematically organizes the connection between iron and AT remains lacking. Given the necessity of iron homeostasis, emphasizing its potential impact on AT function and metabolism regulation provides valuable insights into physiological effects such as adipocyte differentiation and thermogenesis. Futhermore, regulators including adipokines, mitochondria and macrophages have been mentioned, along with analyzing the novel perspective of iron as a key mediator influencing the fat-gut crosstalk.
Collapse
Affiliation(s)
- Xinyu Yang
- Department of Endocrinology and Metabolism, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Xianghong Wang
- Department of Endocrinology and Metabolism, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Zhe Yang
- Department of Endocrinology and Metabolism, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Hongyun Lu
- Department of Endocrinology and Metabolism, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China.
| |
Collapse
|
3
|
Ren Y, Huang P, Huang X, Zhang L, Liu L, Xiang W, Liu L, He X. Alterations of DNA methylation profile in peripheral blood of children with simple obesity. Health Inf Sci Syst 2024; 12:26. [PMID: 38505098 PMCID: PMC10948706 DOI: 10.1007/s13755-024-00275-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/12/2024] [Indexed: 03/21/2024] Open
Abstract
Purpose To investigate the association between DNA methylation and childhood simple obesity. Methods Genome-wide analysis of DNA methylation was conducted on peripheral blood samples from 41 children with simple obesity and 31 normal controls to identify differentially methylated sites (DMS). Subsequently, gene functional analysis of differentially methylated genes (DMGs) was carried out. After screening the characteristic DMGs based on specific conditions, the methylated levels of these DMS were evaluated and verified by pyrosequencing. Receiver operating characteristic (ROC) curve analysis assessed the predictive efficacy of corresponding DMGs. Finally, Pearson correlation analysis revealed the correlation between specific DMS and clinical data. Results The overall DNA methylation level in the obesity group was significantly lower than in normal. A total of 241 DMS were identified. Functional pathway analysis revealed that DMGs were primarily involved in lipid metabolism, carbohydrate metabolism, amino acid metabolism, human diseases, among other pathways. The characteristic DMS within the genes Transcription factor A mitochondrial (TFAM) and Piezo type mechanosensitive ion channel component 1(PIEZO1) were recognized as CpG-cg05831083 and CpG-cg14926485, respectively. Furthermore, the methylation level of CpG-cg05831083 significantly correlated with body mass index (BMI) and vitamin D. Conclusions Abnormal DNA methylation is closely related to childhood simple obesity. The altered methylation of CpG-cg05831083 and CpG-cg14926485 could potentially serve as biomarkers for childhood simple obesity. Supplementary Information The online version contains supplementary material available at 10.1007/s13755-024-00275-w.
Collapse
Affiliation(s)
- Yi Ren
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011 China
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011 China
- Department of Pediatrics, Haikou Maternal and Child Health Hospital, Haikou, 570100 China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011 China
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011 China
| | - Xiaoyan Huang
- Department of Genetics, Metabolism, and Endocrinology, Hainan Women and Children’s Medical Center, Haikou, 570100 China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011 China
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011 China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011 China
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011 China
| | - Wei Xiang
- Hainan Women and Children’s Medical Center, Haikou, 570100 China
- Children’s Hospital of Fudan University at Hainan, Haikou, 570100 China
- Children’s Hospital of Hainan Medical University, Haikou, 570100 China
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011 China
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011 China
| | - Xiaojie He
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011 China
- Laboratory of Pediatric Nephrology, Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011 China
| |
Collapse
|
4
|
Zhu M, Fang Y, Sun Y, Li S, Yu J, Xiong B, Wen C, Zhou B, Huang B, Yin H, Xu H. Sonogenetics in the Treatment of Chronic Diseases: A New Method for Cell Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407373. [PMID: 39488795 DOI: 10.1002/advs.202407373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/21/2024] [Indexed: 11/04/2024]
Abstract
Sonogenetics is an innovative technology that integrates ultrasound with genetic editing to precisely modulate cellular activities in a non-invasive manner. This method entails introducing and activating mechanosensitive channels on the cell membrane of specific cells using gene delivery vectors. When exposed to ultrasound, these channels can be manipulated to open or close, thereby impacting cellular functions. Sonogenetics is currently being used extensively in the treatment of various chronic diseases, including Parkinson's disease, vision restoration, and cancer therapy. This paper provides a comprehensive review of key components of sonogenetics and focuses on evaluating its prospects and potential challenges in the treatment of chronic disease.
Collapse
Affiliation(s)
- Mingrui Zhu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Yan Fang
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yikang Sun
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Shaoyue Li
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Jifeng Yu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Bing Xiong
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Congjian Wen
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Boyang Zhou
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Bin Huang
- Zhejiang Hospital, Hangzhou, 310013, P. R. China
| | - Haohao Yin
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Huixiong Xu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| |
Collapse
|
5
|
Atcha H, Kulkarni D, Meli VS, Veerasubramanian PK, Wang Y, Cahalan MD, Pathak MM, Liu WF. Piezo1-mediated mechanotransduction enhances macrophage oxidized low-density lipoprotein uptake and atherogenesis. PNAS NEXUS 2024; 3:pgae436. [PMID: 39544498 PMCID: PMC11563038 DOI: 10.1093/pnasnexus/pgae436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/21/2024] [Indexed: 11/17/2024]
Abstract
Macrophages in the vascular wall ingest and clear lipids, but abundant lipid accumulation leads to foam cell formation and atherosclerosis, a pathological condition often characterized by tissue stiffening. While the role of biochemical stimuli in the modulation of macrophage function is well studied, the role of biophysical cues and the molecules involved in mechanosensation are less well understood. Here, we use genetic and pharmacological tools to show extracellular oxidized low-density lipoproteins (oxLDLs) stimulate Ca2+ signaling through activation of the mechanically gated ion channel Piezo1. Moreover, macrophage Piezo1 expression is critical in the transduction of environmental stiffness and channel deletion suppresses, whereas a gain-of-function mutation exacerbates oxLDL uptake. Additionally, we find that depletion of myeloid Piezo1 protects from atherosclerotic plaque formation in vivo. Together, our study highlights an important role for Piezo1 and its respective mutations in macrophage mechanosensing, lipid uptake, and cardiovascular disease.
Collapse
Affiliation(s)
- Hamza Atcha
- Department of Bioengineering, University of California, San Diego, La Jolla 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla 92037, USA
| | - Daanish Kulkarni
- Department of Biomedical Engineering, University of California, Irvine, Irvine 92697, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine 92697, USA
| | - Vijaykumar S Meli
- Department of Biomedical Engineering, University of California, Irvine, Irvine 92697, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine 92697, USA
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine 92697, USA
| | - Praveen Krishna Veerasubramanian
- Department of Biomedical Engineering, University of California, Irvine, Irvine 92697, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine 92697, USA
| | - Yuchun Wang
- Department of Biomedical Engineering, University of California, Irvine, Irvine 92697, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine 92697, USA
| | - Michael D Cahalan
- Department of Physiology and Biophysics, University of California Irvine, Irvine 92697, USA
| | - Medha M Pathak
- Department of Biomedical Engineering, University of California, Irvine, Irvine 92697, USA
- Department of Physiology and Biophysics, University of California Irvine, Irvine 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine 92697, USA
| | - Wendy F Liu
- Department of Biomedical Engineering, University of California, Irvine, Irvine 92697, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine 92697, USA
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine 92697, USA
| |
Collapse
|
6
|
Xiao B. Mechanisms of mechanotransduction and physiological roles of PIEZO channels. Nat Rev Mol Cell Biol 2024; 25:886-903. [PMID: 39251883 DOI: 10.1038/s41580-024-00773-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/11/2024]
Abstract
Mechanical force is an essential physical element that contributes to the formation and function of life. The discovery of the evolutionarily conserved PIEZO family, including PIEZO1 and PIEZO2 in mammals, as bona fide mechanically activated cation channels has transformed our understanding of how mechanical forces are sensed and transduced into biological activities. In this Review, I discuss recent structure-function studies that have illustrated how PIEZO1 and PIEZO2 adopt their unique structural design and curvature-based gating dynamics, enabling their function as dedicated mechanotransduction channels with high mechanosensitivity and selective cation conductivity. I also discuss our current understanding of the physiological and pathophysiological roles mediated by PIEZO channels, including PIEZO1-dependent regulation of development and functional homeostasis and PIEZO2-dominated mechanosensation of touch, tactile pain, proprioception and interoception of mechanical states of internal organs. Despite the remarkable progress in PIEZO research, this Review also highlights outstanding questions in the field.
Collapse
Affiliation(s)
- Bailong Xiao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
- Beijing Frontier Research Center of Biological Structure, Tsinghua University, Beijing, China.
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
| |
Collapse
|
7
|
Nagase T, Nagase M. Piezo ion channels: long-sought-after mechanosensors mediating hypertension and hypertensive nephropathy. Hypertens Res 2024; 47:2786-2799. [PMID: 39103520 DOI: 10.1038/s41440-024-01820-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/03/2024] [Accepted: 07/07/2024] [Indexed: 08/07/2024]
Abstract
Recent advances in mechanobiology and the discovery of mechanosensitive ion channels have opened a new era of research on hypertension and related diseases. Piezo1 and Piezo2, first reported in 2010, are regarded as bona fide mechanochannels that mediate various biological and pathophysiological phenomena in multiple tissues and organs. For example, Piezo channels have pivotal roles in blood pressure control, triggering shear stress-induced nitric oxide synthesis and vasodilation, regulating baroreflex in the carotid sinus and aorta, and releasing renin from renal juxtaglomerular cells. Herein, we provide an overview of recent literature on the roles of Piezo channels in the pathogenesis of hypertension and related kidney damage, including our experimental data on the involvement of Piezo1 in podocyte injury and that of Piezo2 in renin expression and renal fibrosis in animal models of hypertensive nephropathy. The mechanosensitive ion channels Piezo1 and Piezo2 play various roles in the pathogenesis of systemic hypertension by acting on vascular endothelial cells, baroreceptors in the carotid artery and aorta, and the juxtaglomerular apparatus. Piezo channels also contribute to hypertensive nephropathy by acting on mesangial cells, podocytes, and perivascular mesenchymal cells.
Collapse
Affiliation(s)
- Takashi Nagase
- Kunitachi Aoyagien Tachikawa Geriatric Health Services Facility, Tokyo, Japan
| | - Miki Nagase
- Department of Anatomy, Kyorin University School of Medicine, Tokyo, Japan.
| |
Collapse
|
8
|
Lichtenstein L, Cheng CW, Bajarwan M, Evans EL, Gaunt HJ, Bartoli F, Chuntharpursat-Bon E, Patel S, Konstantinou C, Futers TS, Reay M, Parsonage G, Moore JB, Bertrand-Michel J, Sukumar P, Roberts LD, Beech DJ. Endothelial force sensing signals to parenchymal cells to regulate bile and plasma lipids. SCIENCE ADVANCES 2024; 10:eadq3075. [PMID: 39331703 PMCID: PMC11430402 DOI: 10.1126/sciadv.adq3075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/21/2024] [Indexed: 09/29/2024]
Abstract
How cardiovascular activity interacts with lipid homeostasis is incompletely understood. We postulated a role for blood flow acting at endothelium in lipid regulatory organs. Transcriptome analysis was performed on livers from mice engineered for deletion of the flow-sensing PIEZO1 channel in endothelium. This revealed unique up-regulation of Cyp7a1, which encodes the rate-limiting enzyme for bile synthesis from cholesterol in hepatocytes. Consistent with this effect were increased gallbladder and plasma bile acids and lowered hepatic and plasma cholesterol. Elevated portal fluid flow acting via endothelial PIEZO1 and genetically enhanced PIEZO1 conversely suppressed Cyp7a1. Activation of hepatic endothelial PIEZO1 channels promoted phosphorylation of nitric oxide synthase 3, and portal flow-mediated suppression of Cyp7a1 depended on nitric oxide synthesis, suggesting endothelium-to-hepatocyte coupling via nitric oxide. PIEZO1 variants in people were associated with hepatobiliary disease and dyslipidemia. The data suggest an endothelial force sensing mechanism that controls lipid regulation in parenchymal cells to modulate whole-body lipid homeostasis.
Collapse
Affiliation(s)
- Laeticia Lichtenstein
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK
| | - Chew W. Cheng
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Muath Bajarwan
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | | | | | - Fiona Bartoli
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | | | - Shaili Patel
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
- Department of Hepatobiliary and Transplant Surgery, St James's University Hospital, Leeds LS9 7TF, UK
| | - Charalampos Konstantinou
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
- Department of Hepatobiliary and Transplant Surgery, St James's University Hospital, Leeds LS9 7TF, UK
| | | | - Melanie Reay
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | | | - J. Bernadette Moore
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK
| | - Justine Bertrand-Michel
- MetaToul-Lipidomics Facility, INSERM UMR1048, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, INSERM, Toulouse, France
| | | | - Lee D. Roberts
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - David J. Beech
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
9
|
Bertaccini GA, Casanellas I, Evans EL, Nourse JL, Dickinson GD, Liu G, Seal S, Ly AT, Holt JR, Wijerathne TD, Yan S, Hui EE, Lacroix JJ, Panicker MM, Upadhyayula S, Parker I, Pathak MM. Visualizing PIEZO1 Localization and Activity in hiPSC-Derived Single Cells and Organoids with HaloTag Technology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.573117. [PMID: 38187535 PMCID: PMC10769387 DOI: 10.1101/2023.12.22.573117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
PIEZO1 is critical to numerous physiological processes, transducing diverse mechanical stimuli into electrical and chemical signals. Recent studies underscore the importance of visualizing endogenous PIEZO1 activity and localization to understand its functional roles. To enable physiologically and clinically relevant studies on human PIEZO1, we genetically engineered human induced pluripotent stem cells (hiPSCs) to express a HaloTag fused to endogenous PIEZO1. Combined with advanced imaging, our chemogenetic platform allows precise visualization of PIEZO1 localization dynamics in various cell types. Furthermore, the PIEZO1-HaloTag hiPSC technology facilitates the non-invasive monitoring of channel activity across diverse cell types using Ca2+-sensitive HaloTag ligands, achieving temporal resolution approaching that of patch clamp electrophysiology. Finally, we used lightsheet imaging of hiPSC-derived neural organoids to achieve molecular scale imaging of PIEZO1 in three-dimensional tissue organoids. Our advances offer a novel platform for studying PIEZO1 mechanotransduction in human cells and tissues, with potential for elucidating disease mechanisms and targeted therapeutic development.
Collapse
Affiliation(s)
- Gabriella A Bertaccini
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Ignasi Casanellas
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Elizabeth L Evans
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Jamison L Nourse
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - George D Dickinson
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Gaoxiang Liu
- Advanced Bioimaging Center, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Sayan Seal
- Advanced Bioimaging Center, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Alan T Ly
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Jesse R Holt
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Tharaka D Wijerathne
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Shijun Yan
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - Elliot E Hui
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - Jerome J Lacroix
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Mitradas M Panicker
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Srigokul Upadhyayula
- Advanced Bioimaging Center, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Ian Parker
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Medha M Pathak
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| |
Collapse
|
10
|
Niu H, Maruoka M, Noguchi Y, Kosako H, Suzuki J. Phospholipid scrambling induced by an ion channel/metabolite transporter complex. Nat Commun 2024; 15:7566. [PMID: 39217145 PMCID: PMC11366033 DOI: 10.1038/s41467-024-51939-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Cells establish the asymmetrical distribution of phospholipids and alter their distribution by phospholipid scrambling (PLS) to adapt to environmental changes. Here, we demonstrate that a protein complex, consisting of the ion channel Tmem63b and the thiamine transporter Slc19a2, induces PLS upon calcium (Ca2+) stimulation. Through revival screening using a CRISPR sgRNA library on high PLS cells, we identify Tmem63b as a PLS-inducing factor. Ca2+ stimulation-mediated PLS is suppressed by deletion of Tmem63b, while human disease-related Tmem63b mutants induce constitutive PLS. To search for a molecular link between Ca2+ stimulation and PLS, we perform revival screening on Tmem63b-overexpressing cells, and identify Slc19a2 and the Ca2+-activated K+ channel Kcnn4 as PLS-regulating factors. Deletion of either of these genes decreases PLS activity. Biochemical screening indicates that Tmem63b and Slc19a2 form a heterodimer. These results demonstrate that a Tmem63b/Slc19a2 heterodimer induces PLS upon Ca2+ stimulation, along with Kcnn4 activation.
Collapse
Affiliation(s)
- Han Niu
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, Japan
- Graduate School of Biostudies, Kyoto University, Konoe-cho, Yoshida, Sakyoku, Kyoto, Japan
| | - Masahiro Maruoka
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, Japan
- Center for Integrated Biosystems, Institute for Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuki Noguchi
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, Japan
| | - Hidetaka Kosako
- Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Jun Suzuki
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, Japan.
- Graduate School of Biostudies, Kyoto University, Konoe-cho, Yoshida, Sakyoku, Kyoto, Japan.
- Center for Integrated Biosystems, Institute for Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan.
| |
Collapse
|
11
|
Nakamichi R, Asahara H. The role of mechanotransduction in tendon. J Bone Miner Res 2024; 39:814-820. [PMID: 38795012 PMCID: PMC11301520 DOI: 10.1093/jbmr/zjae074] [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: 01/16/2024] [Revised: 04/04/2024] [Accepted: 05/24/2024] [Indexed: 05/27/2024]
Abstract
Tendons play an important role in the maintenance of motor function by connecting muscles and bones and transmitting forces. Particularly, the role of mechanical stress has primarily focused on the key mechanism of tendon homeostasis, with much research on this topic. With the recent development of molecular biological techniques, the mechanisms of mechanical stress sensing and signal transduction have been gradually elucidated with the identification of mechanosensor in tendon cells and the master regulator in tendon development. This review provides a comprehensive overview of the structure and function of tendon tissue, including the role for physical performance and the detailed mechanism of mechanotransduction in its regulation. An important lesson is that the role of mechanotransduction in tendon tissue is only partially clarified, indicating the complexity of the mechanisms of motor function and fueling increasing interest in uncovering these mechanisms.
Collapse
Affiliation(s)
- Ryo Nakamichi
- Department of Molecular and Cellular Biology, Scripps Research, 10550 North Torrey Pines Road, MBB-102, La Jolla, CA 92037, United States
- Department of Systems Biomedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
- Department of Orthopaedic Surgery, Okayama University hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama 700-8558, Japan
| | - Hiroshi Asahara
- Department of Molecular and Cellular Biology, Scripps Research, 10550 North Torrey Pines Road, MBB-102, La Jolla, CA 92037, United States
- Department of Systems Biomedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| |
Collapse
|
12
|
Zhang Q, Pan RL, Wang H, Wang JJ, Lu SH, Zhang M. Nanoporous Titanium Implant Surface Accelerates Osteogenesis via the Piezo1/Acetyl-CoA/β-Catenin Pathway. NANO LETTERS 2024; 24:8257-8267. [PMID: 38920296 PMCID: PMC11247543 DOI: 10.1021/acs.nanolett.4c01101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 06/27/2024]
Abstract
Osseointegration is the most important factor determining implant success. The surface modification of TiO2 nanotubes prepared by anodic oxidation has remarkable advantages in promoting bone formation. However, the mechanism behind this phenomenon is still unintelligible. Here we show that the nanomorphology exhibited open and clean nanotube structure and strong hydrophilicity, and the nanomorphology significantly facilitated the adhesion, proliferation, and osteogenesis differentiation of stem cells. Exploring the mechanism, we found that the nanomorphology can enhance mitochondrial oxidative phosphorylation (OxPhos) by activating Piezo1 and increasing intracellular Ca2+. The increase in OxPhos can significantly uplift the level of acetyl-CoA in the cytoplasm but not significantly raise the level of acetyl-CoA in the nucleus, which was beneficial for the acetylation and stability of β-catenin and ultimately promoted osteogenesis. This study provides a new interpretation for the regulatory mechanism of stem cell osteogenesis by nanomorphology.
Collapse
Affiliation(s)
- Qian Zhang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration
& National Clinical Research Center for Oral Diseases & Shaanxi
International Joint Research Center for Oral Diseases, Department
of General Dentistry and Emergency, School of Stomatology, Air Force Medical University, Xi’an 710032, China
| | - Run-Long Pan
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration
& National Clinical Research Center for Oral Diseases & Shaanxi
International Joint Research Center for Oral Diseases, Department
of General Dentistry and Emergency, School of Stomatology, Air Force Medical University, Xi’an 710032, China
| | - Hui Wang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration
& National Clinical Research Center for Oral Diseases & Shaanxi
International Joint Research Center for Oral Diseases, Department
of General Dentistry and Emergency, School of Stomatology, Air Force Medical University, Xi’an 710032, China
| | - Jun-Jun Wang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration
& National Clinical Research Center for Oral Diseases & Shaanxi
International Joint Research Center for Oral Diseases, Department
of General Dentistry and Emergency, School of Stomatology, Air Force Medical University, Xi’an 710032, China
| | - Song-He Lu
- Scientific
Research Department, Air Force Medical University, Xi’an 710032, China
| | - Min Zhang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration
& National Clinical Research Center for Oral Diseases & Shaanxi
International Joint Research Center for Oral Diseases, Department
of General Dentistry and Emergency, School of Stomatology, Air Force Medical University, Xi’an 710032, China
| |
Collapse
|
13
|
Isik E, Aydinok Y, Albayrak C, Durmus B, Karakas Z, Orhan MF, Sarper N, Aydın S, Unal S, Oymak Y, Karadas N, Turedi A, Albayrak D, Tayfun F, Tugcu D, Karaman S, Tobu M, Unal E, Ozcan A, Unal S, Aksu T, Unuvar A, Bilici M, Azik F, Ay Y, Gelen SA, Zengin E, Albudak E, Eker I, Karakaya T, Cogulu O, Ozkinay F, Atik T. Identification of the molecular etiology in rare congenital hemolytic anemias using next-generation sequencing with exome-based copy number variant analysis. Eur J Haematol 2024; 113:82-89. [PMID: 38556258 DOI: 10.1111/ejh.14194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 02/29/2024] [Accepted: 03/09/2024] [Indexed: 04/02/2024]
Abstract
OBJECTIVES In congenital hemolytic anemias (CHA), it is not always possible to determine the specific diagnosis by evaluating clinical findings and conventional laboratory tests. The aim of this study is to evaluate the utility of next-generation sequencing (NGS) and clinical-exome-based copy number variant (CNV) analysis in patients with CHA. METHODS One hundred and forty-three CHA cases from 115 unrelated families referred for molecular analysis were enrolled in the study. Molecular analysis was performed using two different clinical exome panels in 130 patients, and whole-exome sequencing in nine patients. Exome-based CNV calling was incorporated into the traditional single-nucleotide variant and small insertion/deletion analysis pipeline for NGS data in 92 cases. In four patients from the same family, the PK Gypsy variant was investigated using long-range polymerase chain reaction. RESULTS Molecular diagnosis was established in 86% of the study group. The most frequently mutated genes were SPTB (31.7%) and PKLR (28.5%). CNV analysis of 92 cases revealed that three patients had different sizes of large deletions in the SPTB and six patients had a deletion in the PKLR. CONCLUSIONS In this study, NGS provided a high molecular diagnostic rate in cases with rare CHA. Analysis of the CNVs contributed to the diagnostic success.
Collapse
Affiliation(s)
- Esra Isik
- Division of Pediatric Genetics, Department of Pediatrics, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Yesim Aydinok
- Division of Pediatric Hematology, Department of Pediatrics, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Canan Albayrak
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Basak Durmus
- Division of Pediatric Genetics, Department of Pediatrics, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Zeynep Karakas
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mehmet Fatih Orhan
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Sakarya University, Sakarya, Turkey
| | - Nazan Sarper
- Division of Pediatric Hematology, Department of Pediatrics, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Sultan Aydın
- Division of Pediatric Hematology and Oncology, Antalya Training and Research Hospital, Antalya, Turkey
| | - Selma Unal
- Division of Pediatric Hematology, Department of Pediatrics, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Yesim Oymak
- Division of Pediatric Hematology, Dr. Behcet Uz Children's Hospital, Izmir, Turkey
| | - Nihal Karadas
- Division of Pediatric Genetics, Department of Pediatrics, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Aysen Turedi
- Division of Pediatric Hematology, Department of Pediatrics, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Davut Albayrak
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Medical Park Samsun Hospital, Samsun, Turkey
| | - Funda Tayfun
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Deniz Tugcu
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Serap Karaman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mahmut Tobu
- Department of Hematology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Ekrem Unal
- Division of Pediatric Hematology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Alper Ozcan
- Division of Pediatric Hematology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Sule Unal
- Division of Pediatric Hematology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Tekin Aksu
- Division of Pediatric Hematology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Aysegul Unuvar
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mustafa Bilici
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Fatih Azik
- Department of Pediatrics, Division of Pediatric Hematology, Faculty of Medicine, Muğla Sıtkı Koçman University, Mugla, Turkey
| | - Yilmaz Ay
- Division of Pediatric Hematology and Oncology, Kartal Dr Lütfi Kırdar Training and Research Hospital, Istanbul, Turkey
| | - Sema Aylan Gelen
- Division of Pediatric Hematology, Department of Pediatrics, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Emine Zengin
- Division of Pediatric Hematology, Department of Pediatrics, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Esin Albudak
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Ibrahim Eker
- Department of Pediatric Hematology and Oncology and Pediatric Hematopoietic Stem Cell Transplantation Unit, Afyonkarahisar Health Science University Faculty of Medicine, Afyon, Turkey
| | - Taner Karakaya
- Department of Medical Genetics, Samsun Education and Research Hospital, Samsun, Turkey
| | - Ozgur Cogulu
- Division of Pediatric Genetics, Department of Pediatrics, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Ferda Ozkinay
- Division of Pediatric Genetics, Department of Pediatrics, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Tahir Atik
- Division of Pediatric Genetics, Department of Pediatrics, Faculty of Medicine, Ege University, Izmir, Turkey
| |
Collapse
|
14
|
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
|
15
|
Yuan X, Zhao X, Wang W, Li C. Mechanosensing by Piezo1 and its implications in the kidney. Acta Physiol (Oxf) 2024; 240:e14152. [PMID: 38682304 DOI: 10.1111/apha.14152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/27/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
Piezo1 is an essential mechanosensitive transduction ion channel in mammals. Its unique structure makes it capable of converting mechanical cues into electrical and biological signals, modulating biological and (patho)physiological processes in a wide variety of cells. There is increasing evidence demonstrating that the piezo1 channel plays a vital role in renal physiology and disease conditions. This review summarizes the current evidence on the structure and properties of Piezo1, gating modulation, and pharmacological characteristics, with special focus on the distribution and (patho)physiological significance of Piezo1 in the kidney, which may provide insights into potential treatment targets for renal diseases involving this ion channel.
Collapse
Affiliation(s)
- Xi Yuan
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoduo Zhao
- Department of Pathology, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Weidong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chunling Li
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
16
|
Zhang Z, Liu X, Hu B, Chen K, Yu Y, Sun C, Zhu D, Bai H, Palli SR, Tan A. The mechanoreceptor Piezo is required for spermatogenesis in Bombyx mori. BMC Biol 2024; 22:118. [PMID: 38769528 PMCID: PMC11106986 DOI: 10.1186/s12915-024-01916-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: 12/15/2023] [Accepted: 05/10/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND The animal sperm shows high diversity in morphology, components, and motility. In the lepidopteran model insect, the silkworm Bombyx mori, two types of sperm, including nucleate fertile eupyrene sperm and anucleate unfertile apyrene sperm, are generated. Apyrene sperm assists fertilization by facilitating the migration of eupyrene spermatozoa from the bursa copulatrix to the spermatheca. During spermatogenesis, eupyrene sperm bundles extrude the cytoplasm by peristaltic squeezing, while the nuclei of the apyrene sperm bundles are discarded with the same process, forming matured sperm. RESULTS In this study, we describe that a mechanoreceptor BmPiezo, the sole Piezo ortholog in B. mori, plays key roles in larval feeding behavior and, more importantly, is essential for eupyrene spermatogenesis and male fertility. CRISPR/Cas9-mediated loss of BmPiezo function decreases larval appetite and subsequent body size and weight. Immunofluorescence analyses reveal that BmPiezo is intensely localized in the inflatable point of eupyrene sperm bundle induced by peristaltic squeezing. BmPiezo is also enriched in the middle region of apyrene sperm bundle before peristaltic squeezing. Cytological analyses of dimorphic sperm reveal developmental arrest of eupyrene sperm bundles in BmPiezo mutants, while the apyrene spermatogenesis is not affected. RNA-seq analysis and q-RT-PCR analyses demonstrate that eupyrene spermatogenic arrest is associated with the dysregulation of the actin cytoskeleton. Moreover, we show that the deformed eupyrene sperm bundles fail to migrate from the testes, resulting in male infertility due to the absence of eupyrene sperm in the bursa copulatrix and spermatheca. CONCLUSIONS In conclusion, our studies thus uncover a new role for Piezo in regulating spermatogenesis and male fertility in insects.
Collapse
Affiliation(s)
- Zhongjie Zhang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China.
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China.
| | - Xiaojing Liu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Bo Hu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Kai Chen
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Ye Yu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Chenxin Sun
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Dalin Zhu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Hua Bai
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Subba Reddy Palli
- Department of Entomology, University of Kentucky, Lexington, KY, 40546-0091, USA
| | - Anjiang Tan
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China.
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China.
| |
Collapse
|
17
|
Karkempetzaki AI, Ravid K. Piezo1 and Its Function in Different Blood Cell Lineages. Cells 2024; 13:482. [PMID: 38534326 PMCID: PMC10969519 DOI: 10.3390/cells13060482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Mechanosensation is a fundamental function through which cells sense mechanical stimuli by initiating intracellular ion currents. Ion channels play a pivotal role in this process by orchestrating a cascade of events leading to the activation of downstream signaling pathways in response to particular stimuli. Piezo1 is a cation channel that reacts with Ca2+ influx in response to pressure sensation evoked by tension on the cell lipid membrane, originating from cell-cell, cell-matrix, or hydrostatic pressure forces, such as laminar flow and shear stress. The application of such forces takes place in normal physiological processes of the cell, but also in the context of different diseases, where microenvironment stiffness or excessive/irregular hydrostatic pressure dysregulates the normal expression and/or activation of Piezo1. Since Piezo1 is expressed in several blood cell lineages and mutations of the channel have been associated with blood cell disorders, studies have focused on its role in the development and function of blood cells. Here, we review the function of Piezo1 in different blood cell lineages and related diseases, with a focus on megakaryocytes and platelets.
Collapse
Affiliation(s)
- Anastasia Iris Karkempetzaki
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA;
- Whitaker Cardiovascular Institute, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
- School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Katya Ravid
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA;
- Whitaker Cardiovascular Institute, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| |
Collapse
|
18
|
Carrisoza-Gaytan R, Mutchler SM, Carattino F, Soong J, Dalghi MG, Wu P, Wang W, Apodaca G, Satlin LM, Kleyman TR. PIEZO1 is a distal nephron mechanosensor and is required for flow-induced K+ secretion. J Clin Invest 2024; 134:e174806. [PMID: 38426496 PMCID: PMC10904061 DOI: 10.1172/jci174806] [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: 08/14/2023] [Accepted: 01/02/2024] [Indexed: 03/02/2024] Open
Abstract
Ca2+-activated BK channels in renal intercalated cells (ICs) mediate luminal flow-induced K+ secretion (FIKS), but how ICs sense increased flow remains uncertain. We examined whether PIEZO1, a mechanosensitive Ca2+-permeable channel expressed in the basolateral membranes of ICs, is required for FIKS. In isolated cortical collecting ducts (CCDs), the mechanosensitive cation-selective channel inhibitor GsMTx4 dampened flow-induced increases in intracellular Ca2+ concentration ([Ca2+]i), whereas the PIEZO1 activator Yoda1 increased [Ca2+]i and BK channel activity. CCDs from mice fed a high-K+ (HK) diet exhibited a greater Yoda1-dependent increase in [Ca2+]i than CCDs from mice fed a control K+ diet. ICs in CCDs isolated from mice with a targeted gene deletion of Piezo1 in ICs (IC-Piezo1-KO) exhibited a blunted [Ca2+]i response to Yoda1 or increased flow, with an associated loss of FIKS in CCDs. Male IC-Piezo1-KO mice selectively exhibited an increased blood [K+] in response to an oral K+ bolus and blunted urinary K+ excretion following a volume challenge. Whole-cell expression of BKα subunit was reduced in ICs of IC-Piezo1-KO mice fed an HK diet. We conclude that PIEZO1 mediates flow-induced basolateral Ca2+ entry into ICs, is upregulated in the CCD in response to an HK diet, and is necessary for FIKS.
Collapse
Affiliation(s)
| | | | - Francisco Carattino
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Joanne Soong
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marianela G. Dalghi
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Peng Wu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - WenHui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Gerard Apodaca
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Cell Biology and
| | - Lisa M. Satlin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Thomas R. Kleyman
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Cell Biology and
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
19
|
Cilek N, Ugurel E, Goksel E, Yalcin O. Signaling mechanisms in red blood cells: A view through the protein phosphorylation and deformability. J Cell Physiol 2024; 239:e30958. [PMID: 36748950 DOI: 10.1002/jcp.30958] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 01/07/2023] [Accepted: 01/19/2023] [Indexed: 02/08/2023]
Abstract
Intracellular signaling mechanisms in red blood cells (RBCs) involve various protein kinases and phosphatases and enable rapid adaptive responses to hypoxia, metabolic requirements, oxidative stress, or shear stress by regulating the physiological properties of the cell. Protein phosphorylation is a ubiquitous mechanism for intracellular signal transduction, volume regulation, and cytoskeletal organization in RBCs. Spectrin-based cytoskeleton connects integral membrane proteins, band 3 and glycophorin C to junctional proteins, ankyrin and Protein 4.1. Phosphorylation leads to a conformational change in the protein structure, weakening the interactions between proteins in the cytoskeletal network that confers a more flexible nature for the RBC membrane. The structural organization of the membrane and the cytoskeleton determines RBC deformability that allows cells to change their ability to deform under shear stress to pass through narrow capillaries. The shear stress sensing mechanisms and oxygenation-deoxygenation transitions regulate cell volume and mechanical properties of the membrane through the activation of ion transporters and specific phosphorylation events mediated by signal transduction. In this review, we summarize the roles of Protein kinase C, cAMP-Protein kinase A, cGMP-nitric oxide, RhoGTPase, and MAP/ERK pathways in the modulation of RBC deformability in both healthy and disease states. We emphasize that targeting signaling elements may be a therapeutic strategy for the treatment of hemoglobinopathies or channelopathies. We expect the present review will provide additional insights into RBC responses to shear stress and hypoxia via signaling mechanisms and shed light on the current and novel treatment options for pathophysiological conditions.
Collapse
Affiliation(s)
- Neslihan Cilek
- Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul, Turkey
- School of Medicine, Koc University, Istanbul, Turkey
- Graduate School of Health Sciences, Koc University, Istanbul, Turkey
| | - Elif Ugurel
- Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul, Turkey
- School of Medicine, Koc University, Istanbul, Turkey
| | - Evrim Goksel
- Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul, Turkey
- School of Medicine, Koc University, Istanbul, Turkey
- Graduate School of Health Sciences, Koc University, Istanbul, Turkey
| | - Ozlem Yalcin
- Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul, Turkey
- School of Medicine, Koc University, Istanbul, Turkey
| |
Collapse
|
20
|
Coste B, Delmas P. PIEZO Ion Channels in Cardiovascular Functions and Diseases. Circ Res 2024; 134:572-591. [PMID: 38422173 DOI: 10.1161/circresaha.123.322798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The cardiovascular system provides blood supply throughout the body and as such is perpetually applying mechanical forces to cells and tissues. Thus, this system is primed with mechanosensory structures that respond and adapt to changes in mechanical stimuli. Since their discovery in 2010, PIEZO ion channels have dominated the field of mechanobiology. These have been proposed as the long-sought-after mechanosensitive excitatory channels involved in touch and proprioception in mammals. However, more and more pieces of evidence point to the importance of PIEZO channels in cardiovascular activities and disease development. PIEZO channel-related cardiac functions include transducing hemodynamic forces in endothelial and vascular cells, red blood cell homeostasis, platelet aggregation, and arterial blood pressure regulation, among others. PIEZO channels contribute to pathological conditions including cardiac hypertrophy and pulmonary hypertension and congenital syndromes such as generalized lymphatic dysplasia and xerocytosis. In this review, we highlight recent advances in understanding the role of PIEZO channels in cardiovascular functions and diseases. Achievements in this quickly expanding field should open a new road for efficient control of PIEZO-related diseases in cardiovascular functions.
Collapse
Affiliation(s)
- Bertrand Coste
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| | - Patrick Delmas
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| |
Collapse
|
21
|
Bai X, Smith HE, Romero LO, Bell B, Vásquez V, Golden A. A mutation in F-actin polymerization factor suppresses the distal arthrogryposis type 5 PIEZO2 pathogenic variant in Caenorhabditis elegans. Development 2024; 151:dev202214. [PMID: 38349741 PMCID: PMC10911111 DOI: 10.1242/dev.202214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 01/15/2024] [Indexed: 02/15/2024]
Abstract
The mechanosensitive PIEZO channel family has been linked to over 26 disorders and diseases. Although progress has been made in understanding these channels at the structural and functional levels, the underlying mechanisms of PIEZO-associated diseases remain elusive. In this study, we engineered four PIEZO-based disease models using CRISPR/Cas9 gene editing. We performed an unbiased chemical mutagen-based genetic suppressor screen to identify putative suppressors of a conserved gain-of-function variant pezo-1[R2405P] that in human PIEZO2 causes distal arthrogryposis type 5 (DA5; p. R2718P). Electrophysiological analyses indicate that pezo-1(R2405P) is a gain-of-function allele. Using genomic mapping and whole-genome sequencing approaches, we identified a candidate suppressor allele in the C. elegans gene gex-3. This gene is an ortholog of human NCKAP1 (NCK-associated protein 1), a subunit of the Wiskott-Aldrich syndrome protein (WASP)-verprolin homologous protein (WAVE/SCAR) complex, which regulates F-actin polymerization. Depletion of gex-3 by RNAi, or with the suppressor allele gex-3(av259[L353F]), significantly increased brood size and ovulation rate, as well as alleviating the crushed oocyte phenotype of the pezo-1(R2405P) mutant. Expression of GEX-3 in the soma is required to rescue the brood size defects in pezo-1(R2405P) animals. Actin organization and orientation were disrupted and distorted in the pezo-1 mutants. Mutation of gex-3(L353F) partially alleviated these defects. The identification of gex-3 as a suppressor of the pathogenic variant pezo-1(R2405P) suggests that the PIEZO coordinates with the cytoskeleton regulator to maintain the F-actin network and provides insight into the molecular mechanisms of DA5 and other PIEZO-associated diseases.
Collapse
Affiliation(s)
- Xiaofei Bai
- Department of Biology, University of Florida, Gainesville, FL 32610, USA
- Genetics Institute, University of Florida, Gainesville, FL 32610, USA
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Harold E. Smith
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Luis O. Romero
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, Memphis, TN 38163, USA
| | - Briar Bell
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, Memphis, TN 38163, USA
| | - Valeria Vásquez
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Andy Golden
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Claude-Taupin A, Dupont N. To squeeze or not: Regulation of cell size by mechanical forces in development and human diseases. Biol Cell 2024; 116:e2200101. [PMID: 38059665 DOI: 10.1111/boc.202200101] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/08/2023]
Abstract
Physical constraints, such as compression, shear stress, stretching and tension play major roles during development and tissue homeostasis. Mechanics directly impact physiology, and their alteration is also recognized as having an active role in driving human diseases. Recently, growing evidence has accumulated on how mechanical forces are translated into a wide panel of biological responses, including metabolism and changes in cell morphology. The aim of this review is to summarize and discuss our knowledge on the impact of mechanical forces on cell size regulation. Other biological consequences of mechanical forces will not be covered by this review. Moreover, wherever possible, we also discuss mechanosensors and molecular and cellular signaling pathways upstream of cell size regulation. We finally highlight the relevance of mechanical forces acting on cell size in physiology and human diseases.
Collapse
Affiliation(s)
- Aurore Claude-Taupin
- Institut Necker Enfants Malades (INEM), INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Paris, France
| | - Nicolas Dupont
- Institut Necker Enfants Malades (INEM), INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Paris, France
| |
Collapse
|
23
|
Liang P, Zhang Y, Wan YCS, Ma S, Dong P, Lowry AJ, Francis SJ, Khandelwal S, Delahunty M, Telen MJ, Strouse JJ, Arepally GM, Yang H. Deciphering and disrupting PIEZO1-TMEM16F interplay in hereditary xerocytosis. Blood 2024; 143:357-369. [PMID: 38033286 PMCID: PMC10862370 DOI: 10.1182/blood.2023021465] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/07/2023] [Accepted: 11/19/2023] [Indexed: 12/02/2023] Open
Abstract
ABSTRACT Cell-surface exposure of phosphatidylserine (PS) is essential for phagocytic clearance and blood clotting. Although a calcium-activated phospholipid scramblase (CaPLSase) has long been proposed to mediate PS exposure in red blood cells (RBCs), its identity, activation mechanism, and role in RBC biology and disease remain elusive. Here, we demonstrate that TMEM16F, the long-sought-after RBC CaPLSase, is activated by calcium influx through the mechanosensitive channel PIEZO1 in RBCs. PIEZO1-TMEM16F functional coupling is enhanced in RBCs from individuals with hereditary xerocytosis (HX), an RBC disorder caused by PIEZO1 gain-of-function channelopathy. Enhanced PIEZO1-TMEM16F coupling leads to an increased propensity to expose PS, which may serve as a key risk factor for HX clinical manifestations including anemia, splenomegaly, and postsplenectomy thrombosis. Spider toxin GsMTx-4 and antigout medication benzbromarone inhibit PIEZO1, preventing force-induced echinocytosis, hemolysis, and PS exposure in HX RBCs. Our study thus reveals an activation mechanism of TMEM16F CaPLSase and its pathophysiological function in HX, providing insights into potential treatment.
Collapse
Affiliation(s)
- Pengfei Liang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
| | - Yang Zhang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
| | - Yui Chun S. Wan
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
| | - Shang Ma
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX
| | - Ping Dong
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
| | - Augustus J. Lowry
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
| | - Samuel J. Francis
- Department of Surgery, Duke University School of Medicine, Durham, NC
| | - Sanjay Khandelwal
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Martha Delahunty
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Marilyn J. Telen
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | - John J. Strouse
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | | | - Huanghe Yang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
- Department of Neurobiology, Duke University School of Medicine, Durham, NC
| |
Collapse
|
24
|
Guizouarn H. Mechanosensation and lipid scrambling news. Blood 2024; 143:300-301. [PMID: 38270947 DOI: 10.1182/blood.2023023367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024] Open
|
25
|
Petkova-Kirova P, Murciano N, Iacono G, Jansen J, Simionato G, Qiao M, Van der Zwaan C, Rotordam MG, John T, Hertz L, Hoogendijk AJ, Becker N, Wagner C, Von Lindern M, Egee S, Van den Akker E, Kaestner L. The Gárdos Channel and Piezo1 Revisited: Comparison between Reticulocytes and Mature Red Blood Cells. Int J Mol Sci 2024; 25:1416. [PMID: 38338693 PMCID: PMC10855361 DOI: 10.3390/ijms25031416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 02/12/2024] Open
Abstract
The Gárdos channel (KCNN4) and Piezo1 are the best-known ion channels in the red blood cell (RBC) membrane. Nevertheless, the quantitative electrophysiological behavior of RBCs and its heterogeneity are still not completely understood. Here, we use state-of-the-art biochemical methods to probe for the abundance of the channels in RBCs. Furthermore, we utilize automated patch clamp, based on planar chips, to compare the activity of the two channels in reticulocytes and mature RBCs. In addition to this characterization, we performed membrane potential measurements to demonstrate the effect of channel activity and interplay on the RBC properties. Both the Gárdos channel and Piezo1, albeit their average copy number of activatable channels per cell is in the single-digit range, can be detected through transcriptome analysis of reticulocytes. Proteomics analysis of reticulocytes and mature RBCs could only detect Piezo1 but not the Gárdos channel. Furthermore, they can be reliably measured in the whole-cell configuration of the patch clamp method. While for the Gárdos channel, the activity in terms of ion currents is higher in reticulocytes compared to mature RBCs, for Piezo1, the tendency is the opposite. While the interplay between Piezo1 and Gárdos channel cannot be followed using the patch clamp measurements, it could be proved based on membrane potential measurements in populations of intact RBCs. We discuss the Gárdos channel and Piezo1 abundance, interdependencies and interactions in the context of their proposed physiological and pathophysiological functions, which are the passing of small constrictions, e.g., in the spleen, and their active participation in blood clot formation and thrombosis.
Collapse
Affiliation(s)
- Polina Petkova-Kirova
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
- Department of Biochemistry, Saarland University, 66123 Saarbrücken, Germany
| | - Nicoletta Murciano
- Nanion Technologies, 80339 Munich, Germany; (N.M.); (M.G.R.); (N.B.)
- Theoretical Medicine and Biosciences, Campus University Hospital, Saarland University, 66421 Homburg, Germany; (J.J.); (M.Q.); (L.H.)
| | - Giulia Iacono
- Department of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; (G.I.); (C.V.d.Z.); (A.J.H.); (M.V.L.); (E.V.d.A.)
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Julia Jansen
- Theoretical Medicine and Biosciences, Campus University Hospital, Saarland University, 66421 Homburg, Germany; (J.J.); (M.Q.); (L.H.)
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany (T.J.); (C.W.)
| | - Greta Simionato
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany (T.J.); (C.W.)
- Department of Experimental Surgery, Campus University Hospital, Saarland University, 66421 Homburg, Germany
| | - Min Qiao
- Theoretical Medicine and Biosciences, Campus University Hospital, Saarland University, 66421 Homburg, Germany; (J.J.); (M.Q.); (L.H.)
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany (T.J.); (C.W.)
| | - Carmen Van der Zwaan
- Department of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; (G.I.); (C.V.d.Z.); (A.J.H.); (M.V.L.); (E.V.d.A.)
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands
| | | | - Thomas John
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany (T.J.); (C.W.)
| | - Laura Hertz
- Theoretical Medicine and Biosciences, Campus University Hospital, Saarland University, 66421 Homburg, Germany; (J.J.); (M.Q.); (L.H.)
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany (T.J.); (C.W.)
| | - Arjan J. Hoogendijk
- Department of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; (G.I.); (C.V.d.Z.); (A.J.H.); (M.V.L.); (E.V.d.A.)
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Nadine Becker
- Nanion Technologies, 80339 Munich, Germany; (N.M.); (M.G.R.); (N.B.)
| | - Christian Wagner
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany (T.J.); (C.W.)
- Physics and Materials Science Research Unit, University of Luxembourg, L-1511 Luxembourg, Luxembourg
| | - Marieke Von Lindern
- Department of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; (G.I.); (C.V.d.Z.); (A.J.H.); (M.V.L.); (E.V.d.A.)
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Stephane Egee
- Biological Station Roscoff, Sorbonne University, CNRS, UMR8227 LBI2M, F-29680 Roscoff, France;
- Laboratory of Excellence GR-Ex, F-75015 Paris, France
| | - Emile Van den Akker
- Department of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; (G.I.); (C.V.d.Z.); (A.J.H.); (M.V.L.); (E.V.d.A.)
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Lars Kaestner
- Theoretical Medicine and Biosciences, Campus University Hospital, Saarland University, 66421 Homburg, Germany; (J.J.); (M.Q.); (L.H.)
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany (T.J.); (C.W.)
| |
Collapse
|
26
|
Stommen A, Ghodsi M, Cloos AS, Conrard L, Dumitru AC, Henriet P, Pierreux CE, Alsteens D, Tyteca D. Piezo1 Regulation Involves Lipid Domains and the Cytoskeleton and Is Favored by the Stomatocyte-Discocyte-Echinocyte Transformation. Biomolecules 2023; 14:51. [PMID: 38254651 PMCID: PMC10813235 DOI: 10.3390/biom14010051] [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: 11/15/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024] Open
Abstract
Piezo1 is a mechanosensitive ion channel required for various biological processes, but its regulation remains poorly understood. Here, we used erythrocytes to address this question since they display Piezo1 clusters, a strong and dynamic cytoskeleton and three types of submicrometric lipid domains, respectively enriched in cholesterol, GM1 ganglioside/cholesterol and sphingomyelin/cholesterol. We revealed that Piezo1 clusters were present in both the rim and the dimple erythrocyte regions. Upon Piezo1 chemical activation by Yoda1, the Piezo1 cluster proportion mainly increased in the dimple area. This increase was accompanied by Ca2+ influx and a rise in echinocytes, in GM1/cholesterol-enriched domains in the dimple and in cholesterol-enriched domains in the rim. Conversely, the effects of Piezo1 activation were abrogated upon membrane cholesterol depletion. Furthermore, upon Piezo1-independent Ca2+ influx, the above changes were not observed. In healthy donors with a high echinocyte proportion, Ca2+ influx, lipid domains and Piezo1 fluorescence were high even at resting state, whereas the cytoskeleton membrane occupancy was lower. Accordingly, upon decreases in cytoskeleton membrane occupancy and stiffness in erythrocytes from patients with hereditary spherocytosis, Piezo1 fluorescence was increased. Altogether, we showed that Piezo1 was differentially controlled by lipid domains and the cytoskeleton and was favored by the stomatocyte-discocyte-echinocyte transformation.
Collapse
Affiliation(s)
- Amaury Stommen
- CELL Unit and PICT Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (A.S.); (M.G.); (A.-S.C.); (P.H.); (C.E.P.)
| | - Marine Ghodsi
- CELL Unit and PICT Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (A.S.); (M.G.); (A.-S.C.); (P.H.); (C.E.P.)
| | - Anne-Sophie Cloos
- CELL Unit and PICT Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (A.S.); (M.G.); (A.-S.C.); (P.H.); (C.E.P.)
| | - Louise Conrard
- Center for Microscopy and Molecular Imaging (CMMI), Biopark Charleroi, Université Libre de Bruxelles, 6041 Gosselies, Belgium;
| | - Andra C. Dumitru
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, 1348 Louvain-la-Neuve, Belgium (D.A.)
| | - Patrick Henriet
- CELL Unit and PICT Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (A.S.); (M.G.); (A.-S.C.); (P.H.); (C.E.P.)
| | - Christophe E. Pierreux
- CELL Unit and PICT Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (A.S.); (M.G.); (A.-S.C.); (P.H.); (C.E.P.)
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, 1348 Louvain-la-Neuve, Belgium (D.A.)
| | - Donatienne Tyteca
- CELL Unit and PICT Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (A.S.); (M.G.); (A.-S.C.); (P.H.); (C.E.P.)
| |
Collapse
|
27
|
Murciano N, Rotordam MG, Becker N, Ludlow MJ, Parsonage G, Darras A, Kaestner L, Beech DJ, George M, Fertig N, Rapedius M, Brüggemann A. A high-throughput electrophysiology assay to study the response of PIEZO1 to mechanical stimulation. J Gen Physiol 2023; 155:e202213132. [PMID: 37801066 PMCID: PMC10558326 DOI: 10.1085/jgp.202213132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 07/17/2023] [Accepted: 09/17/2023] [Indexed: 10/07/2023] Open
Abstract
PIEZO1 channels are mechanically activated cation channels that play a pivotal role in sensing mechanical forces in various cell types. Their dysfunction has been associated with numerous pathophysiological states, including generalized lymphatic dysplasia, varicose vein disease, and hereditary xerocytosis. Given their physiological relevance, investigating PIEZO1 is crucial for the pharmaceutical industry, which requires scalable techniques to allow for drug discovery. In this regard, several studies have used high-throughput automated patch clamp (APC) combined with Yoda1, a specific gating modifier of PIEZO1 channels, to explore the function and properties of PIEZO1 in heterologous expression systems, as well as in primary cells. However, a combination of solely mechanical stimulation (M-Stim) and high-throughput APC has not yet been available for the study of PIEZO1 channels. Here, we show that optimization of pipetting parameters of the SyncroPatch 384 coupled with multihole NPC-384 chips enables M-Stim of PIEZO1 channels in high-throughput electrophysiology. We used this approach to explore differences between the response of mouse and human PIEZO1 channels to mechanical and/or chemical stimuli. Our results suggest that applying solutions on top of the cells at elevated pipetting flows is crucial for activating PIEZO1 channels by M-Stim on the SyncroPatch 384. The possibility of comparing and combining mechanical and chemical stimulation in a high-throughput patch clamp assay facilitates investigations on PIEZO1 channels and thereby provides an important experimental tool for drug development.
Collapse
Affiliation(s)
- Nicoletta Murciano
- Nanion Technologies GmbH, München, Germany
- Theoretical Medicine and Biosciences, Saarland University, Homburg, Germany
| | | | | | - Melanie J. Ludlow
- 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
| | - Alexis Darras
- Experimental Physics, Saarland University, Saarbrücken, Germany
| | - Lars Kaestner
- Theoretical Medicine and Biosciences, Saarland University, Homburg, Germany
- Experimental Physics, Saarland University, Saarbrücken, Germany
| | - David J. Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | | | | | | | | |
Collapse
|
28
|
Allegrini B, NGuyen LD, Mignotet M, Etchebest C, Fenneteau O, Platon J, Lambilliotte A, Guizouarn H, Da Costa L. Next generation sequencing (NGS) interest in deciphering erythrocyte molecular defects' association in red cell disorders: Clinical and erythrocyte phenotypes of patients with mutations inheritance in PIEZO1, Spectrin ß1, RhAG and SLC4A1. Blood Cells Mol Dis 2023; 103:102780. [PMID: 37516005 DOI: 10.1016/j.bcmd.2023.102780] [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: 04/24/2023] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 07/31/2023]
Abstract
We report here an instructive case referred at 16 months-old for exploration of hemolysis without anemia (compensated anemia with reticulocytosis). The biology tests confirmed the hemolysis with increased total and indirect bilirubin. The usual hemolysis diagnosis tests were normal (DAT, G6PD, PK, Hb electrophoresis) except cytology and ektacytometry suggesting an association of multiple red blood cell (RBC) membrane disorders. This led us to propose a molecular screening analysis using targeted-Next Generation Sequencing (t-NGS) with a capture technique on 93 genes involved in RBC and erythropoiesis defects. We identified 4 missense heterozygous allelic variations, all of them were described without any significance (VUS) in the SLC4A1, RhAG, PIEZO1 and SPTB genes. The study of the familial cosegregation and research functional tests allowed to decipher the role of at least two by two genes in the phenotype and the hemolytic disease of this young patient. Specialized t-NGS panel (or virtual exome/genome sequencing) in a disease-referent laboratory and the motivated collaboration of clinicians, biologists and scientists should be the gold standard for improving the diagnosis of the patients affected with RBC diseases or rare inherited anemias.
Collapse
Affiliation(s)
- Benoit Allegrini
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France
| | | | - Morgane Mignotet
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France
| | - Catherine Etchebest
- Inserm U1134, France; Laboratory of Excellence for RBCs, LABEX GR-Ex, 75015 Paris, France
| | - Odile Fenneteau
- AP-HP, Service Hématologie Biologique, Hôpital R. Debré, Paris, France
| | - Jessica Platon
- HEMATIM EA4666, Université Picardie Jules Vernes, Amiens, France
| | | | - Hélène Guizouarn
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France; Laboratory of Excellence for RBCs, LABEX GR-Ex, 75015 Paris, France.
| | - Lydie Da Costa
- AP-HP, Service Hématologie Biologique, Hôpital R. Debré, Paris, France; HEMATIM EA4666, Université Picardie Jules Vernes, Amiens, France; Université Paris, Paris, France; Inserm U1134, France
| |
Collapse
|
29
|
Stewart GW, Gibson JS, Rees DC. The cation-leaky hereditary stomatocytosis syndromes: A tale of six proteins. Br J Haematol 2023; 203:509-522. [PMID: 37679660 DOI: 10.1111/bjh.19093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/13/2023] [Accepted: 08/22/2023] [Indexed: 09/09/2023]
Abstract
This review concerns a series of dominantly inherited haemolytic anaemias in which the membrane of the erythrocyte 'leaks' the univalent cations, compromising the osmotic stability of the cell. The majority of the conditions are explained by mutations in one of six genes, coding for multispanning membrane proteins of different structure and function. These are: RhAG, coding for an ammonium carrier; SLC4A1, coding for the band 3 anion exchanger; PIEZO1, coding for a mechanosensitive cation channel; GLUT1, coding for a glucose transporter; KCNN4, coding for an internal-calcium-activated potassium channel; and ABCB6, coding for a porphyrin transporter. This review describes the five clinical syndromes associated with genetic defects in these genes and their variable genotype/phenotype relationships.
Collapse
Affiliation(s)
- Gordon W Stewart
- Division of Medicine, Faculty of Medical Sciences, University College London, London, UK
| | - John S Gibson
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - David C Rees
- Haematological Medicine, Kings College London, London, UK
| |
Collapse
|
30
|
Jamwal M, Singh N, Sharma P, Duseja A, Lad DP, Malhotra P, Das R. Dehydrated hereditary stomatocytosis masquerading as primary haemochromatosis: a diagnostic challenge. Pathology 2023; 55:876-879. [PMID: 37393146 DOI: 10.1016/j.pathol.2023.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/15/2023] [Accepted: 03/28/2023] [Indexed: 07/03/2023]
Affiliation(s)
- Manu Jamwal
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Namrata Singh
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Prashant Sharma
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ajay Duseja
- Department of Hepatology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepesh P Lad
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Malhotra
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Reena Das
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
31
|
Zhou Z, Martinac B. Mechanisms of PIEZO Channel Inactivation. Int J Mol Sci 2023; 24:14113. [PMID: 37762415 PMCID: PMC10531961 DOI: 10.3390/ijms241814113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
PIEZO channels PIEZO1 and PIEZO2 are the newly identified mechanosensitive, non-selective cation channels permeable to Ca2+. In higher vertebrates, PIEZO1 is expressed ubiquitously in most tissues and cells while PIEZO2 is expressed more specifically in the peripheral sensory neurons. PIEZO channels contribute to a wide range of biological behaviors and developmental processes, therefore driving significant attention in the effort to understand their molecular properties. One prominent property of PIEZO channels is their rapid inactivation, which manifests itself as a decrease in channel open probability in the presence of a sustained mechanical stimulus. The lack of the PIEZO channel inactivation is linked to various mechanopathologies emphasizing the significance of studying this PIEZO channel property and the factors affecting it. In the present review, we discuss the mechanisms underlying the PIEZO channel inactivation, its modulation by the interaction of the channels with lipids and/or proteins, and how the changes in PIEZO inactivation by the channel mutations can cause a variety of diseases in animals and humans.
Collapse
Affiliation(s)
- Zijing Zhou
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Darlinghurst, NSW 2010, Australia;
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Darlinghurst, NSW 2010, Australia;
- St Vincent’s Clinical School, University of New South Wales, Darlinghurst, NSW 2010, Australia
| |
Collapse
|
32
|
Evtugina NG, Peshkova AD, Khabirova AI, Andrianova IA, Abdullayeva S, Ayombil F, Shepeliuk T, Grishchuk EL, Ataullakhanov FI, Litvinov RI, Weisel JW. Activation of Piezo1 channels in compressed red blood cells augments platelet-driven contraction of blood clots. J Thromb Haemost 2023; 21:2418-2429. [PMID: 37268065 PMCID: PMC10949619 DOI: 10.1016/j.jtha.2023.05.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND Piezo1 is a mechanosensitive cationic channel that boosts intracellular [Ca2+]i. Compression of red blood cells (RBCs) during platelet-driven contraction of blood clots may cause the activation of Piezo1. OBJECTIVES To establish relationships between Piezo1 activity and blood clot contraction. METHODS Effects of a Piezo1 agonist, Yoda1, and antagonist, GsMTx-4, on clot contraction in vitro were studied in human blood containing physiological [Ca2+]. Clot contraction was induced by exogenous thrombin. Activation of Piezo1 was assessed by Ca2+ influx in RBCs and with other functional and morphologic features. RESULTS Piezo1 channels in compressed RBCs are activated naturally during blood clot contraction and induce an upsurge in the intracellular [Ca2+]i, followed by phosphatidylserine exposure. Adding the Piezo1 agonist Yoda1 to whole blood increased the extent of clot contraction due to Ca2+-dependent volumetric shrinkage of RBCs and increased platelet contractility due to their hyperactivation by the enhanced generation of endogenous thrombin on activated RBCs. Addition of rivaroxaban, the inhibitor of thrombin formation, or elimination of Ca2+ from the extracellular space abrogated the stimulating effect of Yoda1 on clot contraction. The Piezo1 antagonist, GsMTx-4, caused a decrease in the extent of clot contraction relative to the control both in whole blood and in platelet-rich plasma. Activated Piezo1 in compressed and deformed RBCs amplified the platelet contractility as a positive feedback mechanism during clot contraction. CONCLUSION The results obtained demonstrate that the Piezo1 channel expressed on RBCs comprises a mechanochemical modulator of blood clotting that may be considered a potential therapeutic target to correct hemostatic disorders.
Collapse
Affiliation(s)
- Natalia G Evtugina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | - Alina D Peshkova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation; Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Alina I Khabirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | - Izabella A Andrianova
- Department of Internal Medicine, Division of Hematology and Program in Molecular Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Shahnoza Abdullayeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | - Francis Ayombil
- Division of Hematology and the Raymond G. Perelman Center for Cellular and Molecular Therapeutics, the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Taisia Shepeliuk
- Department of Physiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ekaterina L Grishchuk
- Department of Physiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Fazoil I Ataullakhanov
- Department of Physiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
33
|
Parsonage G, Cuthbertson K, Endesh N, Murciano N, Hyman AJ, Revill CH, Povstyan OV, Chuntharpursat‐Bon E, Debant M, Ludlow MJ, Futers TS, Lichtenstein L, Kinsella JA, Bartoli F, Rotordam MG, Becker N, Brüggemann A, Foster R, Beech DJ. Improved PIEZO1 agonism through 4-benzoic acid modification of Yoda1. Br J Pharmacol 2023; 180:2039-2063. [PMID: 36457143 PMCID: PMC10952572 DOI: 10.1111/bph.15996] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/03/2022] [Accepted: 11/06/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND AND PURPOSE The protein PIEZO1 forms mechanically activated, calcium-permeable, non-selective cation channels in numerous cell types from several species. Options for pharmacological modulation are limited and so we modified a small-molecule agonist at PIEZO1 channels (Yoda1) to increase the ability to modulate these channels. EXPERIMENTAL APPROACH Medicinal chemistry generated Yoda1 analogues that were tested in intracellular calcium and patch-clamp assays on cultured cells exogenously expressing human or mouse PIEZO1 or mouse PIEZO2. Physicochemical assays and wire myography assays on veins from mice with genetic disruption of PIEZO1. KEY RESULTS A Yoda1 analogue (KC159) containing 4-benzoic acid instead of the pyrazine of Yoda1 and its potassium salt (KC289) have equivalent or improved reliability, efficacy and potency, compared with Yoda1 in functional assays. Tested against overexpressed mouse PIEZO1 in calcium assays, the order of potency (as EC50 values, nM) was KC289, 150 > KC159 280 > Yoda1, 600). These compounds were selective for PIEZO1 over other membrane proteins, and the physicochemical properties were more suited to physiological conditions than those of Yoda1. The vasorelaxant effects were consistent with PIEZO1 agonism. In contrast, substitution with 2-benzoic acid failed to generate a modulator. CONCLUSION AND IMPLICATIONS 4-Benzoic acid modification of Yoda1 improves PIEZO1 agonist activity at PIEZO1 channels. We suggest naming this new modulator Yoda2. It should be a useful tool compound in physiological assays and facilitate efforts to identify a binding site. Such compounds may have therapeutic potential, for example, in diseases linked genetically to PIEZO1 such as lymphatic dysplasia.
Collapse
Affiliation(s)
- Gregory Parsonage
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of MedicineUniversity of LeedsLeedsUK
| | | | - Naima Endesh
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of MedicineUniversity of LeedsLeedsUK
| | - Nicoletta Murciano
- Nanion Technologies GmbHMunichGermany
- Theoretical Medicine and BiosciencesSaarland UniversityHomburgGermany
| | - Adam J. Hyman
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of MedicineUniversity of LeedsLeedsUK
| | | | - Oleksandr V. Povstyan
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of MedicineUniversity of LeedsLeedsUK
| | | | - Marjolaine Debant
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of MedicineUniversity of LeedsLeedsUK
| | - Melanie J. Ludlow
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of MedicineUniversity of LeedsLeedsUK
| | - Timothy Simon Futers
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of MedicineUniversity of LeedsLeedsUK
| | - Laeticia Lichtenstein
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of MedicineUniversity of LeedsLeedsUK
| | - Jacob A. Kinsella
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of MedicineUniversity of LeedsLeedsUK
| | - Fiona Bartoli
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of MedicineUniversity of LeedsLeedsUK
| | - Maria Giustina Rotordam
- Nanion Technologies GmbHMunichGermany
- Theoretical Medicine and BiosciencesSaarland UniversityHomburgGermany
| | | | | | | | - David J. Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of MedicineUniversity of LeedsLeedsUK
| |
Collapse
|
34
|
Bai X, Smith HE, Romero LO, Bell B, Vásquez V, Golden A. Mutation in F-actin Polymerization Factor Suppresses Distal Arthrogryposis Type 5 (DA5) PIEZO2 Pathogenic Variant in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550416. [PMID: 37546771 PMCID: PMC10402071 DOI: 10.1101/2023.07.24.550416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The mechanosensitive PIEZO channel family has been linked to over 26 disorders and diseases. Although progress has been made in understanding these channels at the structural and functional levels, the underlying mechanisms of PIEZO-associated diseases remain elusive. In this study, we engineered four PIEZO-based disease models using CRISPR/Cas9 gene editing. We performed an unbiased chemical mutagen-based genetic suppressor screen to identify putative suppressors of a conserved gain-of-function variant pezo-1[R2405P] that in human PIEZO2 causes distal arthrogryposis type 5 (DA5; p. R2718P). Electrophysiological analyses indicate that pezo-1(R2405P) is a gain-of-function allele. Using genomic mapping and whole genome sequencing approaches, we identified a candidate suppressor allele in the C. elegans gene gex-3. This gene is an ortholog of human NCKAP1 (NCK-associated protein 1), a subunit of the Wiskott-Aldrich syndrome protein (WASP)-verprolin homologous protein (WAVE/SCAR) complex, which regulates F-actin polymerization. Depletion of gex-3 by RNAi, or with the suppressor allele gex-3(av259[L353F]) , significantly restored the small brood size and low ovulation rate, as well as alleviated the crushed oocyte phenotype of the pezo-1(R2405P) mutant. Auxin-inducible degradation of GEX-3 revealed that only somatic-specific degradation of GEX-3 restored the reduced brood size in the pezo-1(R2405P) mutants. Additionally, actin organization and orientation were disrupted and distorted in the pezo-1 mutants. Mutation of gex-3(L353F) partially alleviated these defects. The identification of gex-3 as a suppressor of the pathogenic variant pezo-1(R2405P) suggests that the cytoskeleton plays an important role in regulating PIEZO channel activity and provides insight into the molecular mechanisms of DA5 and other PIEZO-associated diseases.
Collapse
|
35
|
Yaganoglu S, Kalyviotis K, Vagena-Pantoula C, Jülich D, Gaub BM, Welling M, Lopes T, Lachowski D, Tang SS, Del Rio Hernandez A, Salem V, Müller DJ, Holley SA, Vermot J, Shi J, Helassa N, Török K, Pantazis P. Highly specific and non-invasive imaging of Piezo1-dependent activity across scales using GenEPi. Nat Commun 2023; 14:4352. [PMID: 37468521 PMCID: PMC10356793 DOI: 10.1038/s41467-023-40134-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 07/11/2023] [Indexed: 07/21/2023] Open
Abstract
Mechanosensing is a ubiquitous process to translate external mechanical stimuli into biological responses. Piezo1 ion channels are directly gated by mechanical forces and play an essential role in cellular mechanotransduction. However, readouts of Piezo1 activity are mainly examined by invasive or indirect techniques, such as electrophysiological analyses and cytosolic calcium imaging. Here, we introduce GenEPi, a genetically-encoded fluorescent reporter for non-invasive optical monitoring of Piezo1-dependent activity. We demonstrate that GenEPi has high spatiotemporal resolution for Piezo1-dependent stimuli from the single-cell level to that of the entire organism. GenEPi reveals transient, local mechanical stimuli in the plasma membrane of single cells, resolves repetitive contraction-triggered stimulation of beating cardiomyocytes within microtissues, and allows for robust and reliable monitoring of Piezo1-dependent activity in vivo. GenEPi will enable non-invasive optical monitoring of Piezo1 activity in mechanochemical feedback loops during development, homeostatic regulation, and disease.
Collapse
Affiliation(s)
- Sine Yaganoglu
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | | | | | - Dörthe Jülich
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Benjamin M Gaub
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | - Maaike Welling
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
- Department of Bioengineering, Imperial College London, London, UK
| | - Tatiana Lopes
- Section of Investigative Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
| | | | - See Swee Tang
- Department of Bioengineering, Imperial College London, London, UK
| | | | - Victoria Salem
- Department of Bioengineering, Imperial College London, London, UK
| | - Daniel J Müller
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | - Scott A Holley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Julien Vermot
- Department of Bioengineering, Imperial College London, London, UK
| | - Jian Shi
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, University of Leeds, Leeds, UK
| | - Nordine Helassa
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London, UK
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Katalin Török
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London, UK
| | - Periklis Pantazis
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland.
- Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
36
|
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: 2.5] [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
|
37
|
Andolfo I, Monaco V, Cozzolino F, Rosato BE, Marra R, Cerbone V, Pinto VM, Forni GL, Unal S, Iolascon A, Monti M, Russo R. Proteome alterations in erythrocytes with PIEZO1 gain-of-function mutations. Blood Adv 2023; 7:2681-2693. [PMID: 36595486 PMCID: PMC10333744 DOI: 10.1182/bloodadvances.2022008673] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023] Open
Abstract
Gain-of-function mutations in PIEZO1 cause dehydrated hereditary stomatocytosis (DHS) or hereditary xerocytosis, an autosomal dominant hemolytic anemia characterized by high reticulocyte count, a tendency to macrocytosis, and mild jaundice, as well as by other variably penetrant clinical features, such as perinatal edema, severe thromboembolic complications after splenectomy, and hepatic iron overload. PIEZO1 mutations in DHS lead to slowed inactivation kinetics of the ion channel and/or facilitation of channel opening in response to physiological stimuli. To characterize the alterations of red blood cell proteome in patients with mutated PIEZO1, we used a differential approach to compare the proteome of patients with DHS (16 patients from 13 unrelated ancestries) vs healthy individuals. We identified new components in the regulation of the complex landscape of erythrocytes ion and volume balance mediated by PIEZO1. Specifically, the main impaired processes in patients with DHS were ion homeostasis, transmembrane transport, regulation of vesicle-mediated transport, and the proteasomal catabolic process. Functional assays demonstrated coexpression of PIEZO1 and band 3 when PIEZO1 was activated. Moreover, the alteration of the vesicle-mediated transport was functionally demonstrated by an increased vesiculation rate in patients with DHS compared with healthy controls. This finding also provides an explanation of the pathogenetic mechanism underlying the increased thrombotic rate observed in these patients. Finally, the newly identified proteins, involved in the intracellular signaling pathways altered by PIEZO1 mutations, could be used in the future as potential druggable targets in DHS.
Collapse
Affiliation(s)
- Immacolata Andolfo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate, Franco Salvatore, Napoli, Italy
| | - Vittoria Monaco
- CEINGE Biotecnologie Avanzate, Franco Salvatore, Napoli, Italy
- Dipartimento di Scienze Chimiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Flora Cozzolino
- CEINGE Biotecnologie Avanzate, Franco Salvatore, Napoli, Italy
- Dipartimento di Scienze Chimiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Barbara Eleni Rosato
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate, Franco Salvatore, Napoli, Italy
| | - Roberta Marra
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate, Franco Salvatore, Napoli, Italy
| | | | - Valeria Maria Pinto
- Centro della Microcitemia, delle Anemie Congenite e dei Disordini del Metabolismo del Ferro, EO Ospedali Galliera, Genoa, Italy
| | - Gian Luca Forni
- Centro della Microcitemia, delle Anemie Congenite e dei Disordini del Metabolismo del Ferro, EO Ospedali Galliera, Genoa, Italy
| | - Sule Unal
- Research Center for Fanconi Anemia and Other Inherited Bone Marrow Failure Syndromes, Department of Pediatric Hematology, Hacettepe University, Ankara, Turkey
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate, Franco Salvatore, Napoli, Italy
| | - Maria Monti
- CEINGE Biotecnologie Avanzate, Franco Salvatore, Napoli, Italy
- Dipartimento di Scienze Chimiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Roberta Russo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate, Franco Salvatore, Napoli, Italy
| |
Collapse
|
38
|
Yu D, Ahmed A, Jayasi J, Womac A, Sally O, Bae C. Inflammation condition sensitizes Piezo1 mechanosensitive channel in mouse cerebellum astrocyte. Front Cell Neurosci 2023; 17:1200946. [PMID: 37305437 PMCID: PMC10248153 DOI: 10.3389/fncel.2023.1200946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
Piezo1 mechanosensitive ion channel (MSC) plays a significant role in human physiology. Despite several research on the function and expression of Piezo1 in the nervous system, its electrophysiological properties in neuroinflammatory astrocytes remain unknown. We tested whether astrocytic neuroinflammatory state regulates Piezo1 using electrical recordings, calcium imaging, and wound healing assays on cultured astrocytes. In this study, we determined whether neuroinflammatory condition regulates astrocytic Piezo1 currents in astrocytes. First, we performed electrophysiological recordings on the mouse cerebellum astrocytes (C8-S) under lipopolysaccharide (LPS)-induced neuroinflammatory condition. We found that LPS treatment significantly increased MSC currents in C8-S. The half-maximal pressure of LPS treated MSC currents was left-shifted but the slope sensitivity was not altered by LPS treatment. LPS-induced increase of MSC currents were further augmented by Piezo1 agonist, Yoda1 but were normalized by Piezo1 inhibitor, GsMTx4. Furthermore, silencing Piezo1 in LPS treated C8-S normalized not only MSC currents but also calcium influx and cell migration velocity. Together, our results show that LPS sensitized Piezo1 channel in C8-S astrocytes. These findings will suggest that astrocytic Piezo1 is a determinant of neuroinflammation pathogenesis and may in turn become the foundation of further research into curing several neuronal illnesses and injury related inflammation of neuronal cells.
Collapse
|
39
|
Lötsch J, Mayer B, Kringel D. Machine learning analysis predicts a person's sex based on mechanical but not thermal pain thresholds. Sci Rep 2023; 13:7332. [PMID: 37147321 PMCID: PMC10163041 DOI: 10.1038/s41598-023-33337-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/11/2023] [Indexed: 05/07/2023] Open
Abstract
Sex differences in pain perception have been extensively studied, but precision medicine applications such as sex-specific pain pharmacology have barely progressed beyond proof-of-concept. A data set of pain thresholds to mechanical (blunt and punctate pressure) and thermal (heat and cold) stimuli applied to non-sensitized and sensitized (capsaicin, menthol) forearm skin of 69 male and 56 female healthy volunteers was analyzed for data structures contingent with the prior sex structure using unsupervised and supervised approaches. A working hypothesis that the relevance of sex differences could be approached via reversibility of the association, i.e., sex should be identifiable from pain thresholds, was verified with trained machine learning algorithms that could infer a person's sex in a 20% validation sample not seen to the algorithms during training, with balanced accuracy of up to 79%. This was only possible with thresholds for mechanical stimuli, but not for thermal stimuli or sensitization responses, which were not sufficient to train an algorithm that could assign sex better than by guessing or when trained with nonsense (permuted) information. This enabled the translation to the molecular level of nociceptive targets that convert mechanical but not thermal information into signals interpreted as pain, which could eventually be used for pharmacological precision medicine approaches to pain. By exploiting a key feature of machine learning, which allows for the recognition of data structures and the reduction of information to the minimum relevant, experimental human pain data could be characterized in a way that incorporates "non" logic that could be translated directly to the molecular pharmacological level, pointing toward sex-specific precision medicine for pain.
Collapse
Affiliation(s)
- Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596, Frankfurt, Germany.
| | - Benjamin Mayer
- Institute of Clinical Pharmacology, Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Dario Kringel
- Institute of Clinical Pharmacology, Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| |
Collapse
|
40
|
Zhou T, Chen Y, Liao Z, Zhang L, Su D, Li Z, Yang X, Ke X, Liu H, Chen Y, Weng R, Shen H, Xu C, Wan Y, Xu R, Su P. Spatiotemporal Characterization of Human Early Intervertebral Disc Formation at Single-Cell Resolution. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206296. [PMID: 36965031 DOI: 10.1002/advs.202206296] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/28/2023] [Indexed: 05/18/2023]
Abstract
The intervertebral disc (IVD) acts as a fibrocartilaginous joint to anchor adjacent vertebrae. Although several studies have demonstrated the cellular heterogeneity of adult mature IVDs, a single-cell transcriptomic atlas mapping early IVD formation is still lacking. Here, the authors generate a spatiotemporal and single cell-based transcriptomic atlas of human IVD formation at the embryonic stage and a comparative mouse transcript landscape. They identify two novel human notochord (NC)/nucleus pulposus (NP) clusters, SRY-box transcription factor 10 (SOX10)+ and cathepsin K (CTSK)+ , that are distributed in the early and late stages of IVD formation and they are validated by lineage tracing experiments in mice. Matrisome NC/NP clusters, T-box transcription factor T (TBXT)+ and CTSK+ , are responsible for the extracellular matrix homeostasis. The IVD atlas suggests that a subcluster of the vertebral chondrocyte subcluster might give rise to an inner annulus fibrosus of chondrogenic origin, while the fibroblastic outer annulus fibrosus preferentially expresseds transgelin and fibromodulin . Through analyzing intercellular crosstalk, the authors further find that notochordal secreted phosphoprotein 1 (SPP1) is a novel cue in the IVD microenvironment, and it is associated with IVD development and degeneration. In conclusion, the single-cell transcriptomic atlas will be leveraged to develop preventative and regenerative strategies for IVD degeneration.
Collapse
Affiliation(s)
- Taifeng Zhou
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yu Chen
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Zhiheng Liao
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Long Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Deying Su
- Guangdong Provincial Key Laboratory of Proteomics and State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhuling Li
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaoming Yang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaona Ke
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Hengyu Liu
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuyu Chen
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ricong Weng
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Huimin Shen
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Caixia Xu
- Research Center for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yong Wan
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ren Xu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Peiqiang Su
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
41
|
Alfhili MA, Alsughayyir J. Metabolic exhaustion and casein kinase 1α drive deguelin-induced premature red blood cell death. Xenobiotica 2023; 53:445-453. [PMID: 37590011 DOI: 10.1080/00498254.2023.2248492] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 08/18/2023]
Abstract
1. Deguelin (DGN), a retinoid isolated from many plants, exhibits a potent anticancer activity against a wide spectrum of tumour cells. There is a dearth of evidence, however, regarding the toxicity of DGN to red blood cells (RBCs). This is relevant given the prevalent chemotherapy-associated anaemia observed in cancer patients.2. RBCs were exposed to 1-100 μM of DGN for 24 h at 37 °C. Haemolysis and related markers were photometrically measured while flow cytometry was employed to detect phosphatidylserine exposure through Annexin-V-FITC binding and light scatter properties. Additionally, cytosolic Ca2+ and reactive oxygen species were quantified using Fluo4/AM and H2DCFDA, respectively. DGN was also tested against specific signalling inhibitors in addition to vitamin C and ATP.3. DGN caused a significant increase in Annexin-V-positive cells which was accompanied by cell shrinkage without Ca2+ elevation or oxidative stress. DGN also elicited dose-responsive haemolysis which was ameliorated by preventing KCl efflux and in the presence of sucrose, D4476, and ATP. In whole blood, DGN significantly reduced the reticulocyte count and increased platelet distribution width and large cell count.4. DGN triggers premature RBC eryptosis and haemolysis through casein kinase 1α and ATP depletion, and exhibits a specific toxicity towards reticulocytes and platelets.
Collapse
Affiliation(s)
- Mohammad A Alfhili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Jawaher Alsughayyir
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
42
|
Lohia R, Allegrini B, Berry L, Guizouarn H, Cerdan R, Abkarian M, Douguet D, Honoré E, Wengelnik K. Pharmacological activation of PIEZO1 in human red blood cells prevents Plasmodium falciparum invasion. Cell Mol Life Sci 2023; 80:124. [PMID: 37071200 PMCID: PMC10113305 DOI: 10.1007/s00018-023-04773-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023]
Abstract
An inherited gain-of-function variant (E756del) in the mechanosensitive cationic channel PIEZO1 was shown to confer a significant protection against severe malaria. Here, we demonstrate in vitro that human red blood cell (RBC) infection by Plasmodium falciparum is prevented by the pharmacological activation of PIEZO1. Yoda1 causes an increase in intracellular calcium associated with rapid echinocytosis that inhibits RBC invasion, without affecting parasite intraerythrocytic growth, division or egress. Notably, Yoda1 treatment significantly decreases merozoite attachment and subsequent RBC deformation. Intracellular Na+/K+ imbalance is unrelated to the mechanism of protection, although delayed RBC dehydration observed in the standard parasite culture medium RPMI/albumax further enhances the resistance to malaria conferred by Yoda1. The chemically unrelated Jedi2 PIEZO1 activator similarly causes echinocytosis and RBC dehydration associated with resistance to malaria invasion. Spiky outward membrane projections are anticipated to reduce the effective surface area required for both merozoite attachment and internalization upon pharmacological activation of PIEZO1. Globally, our findings indicate that the loss of the typical biconcave discoid shape of RBCs, together with an altered optimal surface to volume ratio, induced by PIEZO1 pharmacological activation prevent efficient P. falciparum invasion.
Collapse
Affiliation(s)
- Rakhee Lohia
- LPHI, University of Montpellier, CNRS UMR5294, Montpellier, France
| | | | - Laurence Berry
- LPHI, University of Montpellier, CNRS UMR5294, Montpellier, France
| | | | - Rachel Cerdan
- LPHI, University of Montpellier, CNRS UMR5294, Montpellier, France
| | - Manouk Abkarian
- Centre de Biologie Structurale, CNRS UMR5048, INSERM U1054, University of Montpellier, Montpellier, France
| | - Dominique Douguet
- IPMC, University Côte d'Azur, CNRS, INSERM, UMR7275, Labex ICST, Valbonne, France
| | - Eric Honoré
- IPMC, University Côte d'Azur, CNRS, INSERM, UMR7275, Labex ICST, Valbonne, France.
| | - Kai Wengelnik
- LPHI, University of Montpellier, CNRS UMR5294, INSERM, Montpellier, France.
| |
Collapse
|
43
|
Zhou J, Lan Q, Li W, Ji LN, Wang K, Xia XH. Single Molecule Protein Segments Sequencing by a Plasmonic Nanopore. NANO LETTERS 2023; 23:2800-2807. [PMID: 36927001 DOI: 10.1021/acs.nanolett.3c00086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Obtaining sequential and conformational information on proteins is vital to understand their functions. Although the nanopore-based electrical detection can sense single molecule (SM) protein and distinguish among different amino acids, this approach still faces difficulties in slowing down protein translocation and improving ionic current signal-to-noise ratio. Here, we observe the unfolding and multistep sequential translocation of SM cytochrome c (cyt c) through a surface enhanced Raman scattering (SERS) active conical gold nanopore. High bias voltage unfolds SM protein causing more exposure of amino acid residues to the nanopore, which slows down the protein translocation. Specific SERS traces of different SM cyt c segments are then recorded sequentially when they pass through the hotspot inside the gold nanopore. This study shows that the combination of SM SERS with a nanopore can provide a direct insight into protein segments and expedite the development of nanopore toward SM protein sequencing.
Collapse
Affiliation(s)
- Juan Zhou
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Qing Lan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Wang Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Li-Na Ji
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Kang Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xing-Hua Xia
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
44
|
Rolland L, Torrente AG, Bourinet E, Maskini D, Drouard A, Chevalier P, Jopling C, Faucherre A. Prolonged Piezo1 Activation Induces Cardiac Arrhythmia. Int J Mol Sci 2023; 24:ijms24076720. [PMID: 37047693 PMCID: PMC10094979 DOI: 10.3390/ijms24076720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/29/2023] [Accepted: 04/01/2023] [Indexed: 04/14/2023] Open
Abstract
The rhythmical nature of the cardiovascular system constantly generates dynamic mechanical forces. At the centre of this system is the heart, which must detect these changes and adjust its performance accordingly. Mechanoelectric feedback provides a rapid mechanism for detecting even subtle changes in the mechanical environment and transducing these signals into electrical responses, which can adjust a variety of cardiac parameters such as heart rate and contractility. However, pathological conditions can disrupt this intricate mechanosensory system and manifest as potentially life-threatening cardiac arrhythmias. Mechanosensitive ion channels are thought to be the main proponents of mechanoelectric feedback as they provide a rapid response to mechanical stimulation and can directly affect cardiac electrical activity. Here, we demonstrate that the mechanosensitive ion channel PIEZO1 is expressed in zebrafish cardiomyocytes. Furthermore, chemically prolonging PIEZO1 activation in zebrafish results in cardiac arrhythmias. indicating that this ion channel plays an important role in mechanoelectric feedback. This also raises the possibility that PIEZO1 gain of function mutations could be linked to heritable cardiac arrhythmias in humans.
Collapse
Affiliation(s)
- Laura Rolland
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, LabEx ICST, 34094 Montpellier, France
| | - Angelo Giovanni Torrente
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, LabEx ICST, 34094 Montpellier, France
| | - Emmanuel Bourinet
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, LabEx ICST, 34094 Montpellier, France
| | - Dounia Maskini
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, LabEx ICST, 34094 Montpellier, France
| | - Aurélien Drouard
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, LabEx ICST, 34094 Montpellier, France
| | - Philippe Chevalier
- Neuromyogene Institut, Claude Bernard University, Lyon 1, 69008 Villeurbanne, France
- Service de Rythmologie, Hospices Civils de Lyon, 69500 Lyon, France
| | - Chris Jopling
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, LabEx ICST, 34094 Montpellier, France
| | - Adèle Faucherre
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, LabEx ICST, 34094 Montpellier, France
| |
Collapse
|
45
|
Nader E, Conran N, Leonardo FC, Hatem A, Boisson C, Carin R, Renoux C, Costa FF, Joly P, Brito PL, Esperti S, Bernard J, Gauthier A, Poutrel S, Bertrand Y, Garcia C, Saad STO, Egée S, Connes P. Piezo1 activation augments sickling propensity and the adhesive properties of sickle red blood cells in a calcium-dependent manner. Br J Haematol 2023. [PMID: 37011913 DOI: 10.1111/bjh.18799] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/07/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023]
Abstract
Haemoglobin S polymerization in the red blood cells (RBCs) of individuals with sickle cell anaemia (SCA) can cause RBC sickling and cellular alterations. Piezo1 is a mechanosensitive protein that modulates intracellular calcium (Ca2+ ) influx, and its activation has been associated with increased RBC surface membrane phosphatidylserine (PS) exposure. Hypothesizing that Piezo1 activation, and ensuing Gárdos channel activity, alter sickle RBC properties, RBCs from patients with SCA were incubated with the Piezo1 agonist, Yoda1 (0.1-10 μM). Oxygen-gradient ektacytometry and membrane potential measurement showed that Piezo1 activation significantly decreased sickle RBC deformability, augmented sickling propensity, and triggered pronounced membrane hyperpolarization, in association with Gárdos channel activation and Ca2+ influx. Yoda1 induced Ca2+ -dependent adhesion of sickle RBCs to laminin, in microfluidic assays, mediated by increased BCAM binding affinity. Furthermore, RBCs from SCA patients that were homo-/heterozygous for the rs59446030 gain-of-function Piezo1 variant demonstrated enhanced sickling under deoxygenation and increased PS exposure. Thus, Piezo1 stimulation decreases sickle RBC deformability, and increases the propensities of these cells to sickle upon deoxygenation and adhere to laminin. Results support a role of Piezo1 in some of the RBC properties that contribute to SCA vaso-occlusion, indicating that Piezo1 may represent a potential therapeutic target molecule for this disease.
Collapse
Affiliation(s)
- Elie Nader
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
| | - Nicola Conran
- Hematology and Transfusion Center, University of Campinas, Campinas, Brazil
| | - Flavia C Leonardo
- Hematology and Transfusion Center, University of Campinas, Campinas, Brazil
| | - Aline Hatem
- Sorbonne Université, CNRS, UMR 8227 LBI2M, Station Biologique de Roscoff SBR, Roscoff, France
| | - Camille Boisson
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi-site, Hospices Civils de Lyon, Lyon, France
| | - Romain Carin
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
| | - Céline Renoux
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi-site, Hospices Civils de Lyon, Lyon, France
| | - Fernando F Costa
- Hematology and Transfusion Center, University of Campinas, Campinas, Brazil
| | - Philippe Joly
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi-site, Hospices Civils de Lyon, Lyon, France
| | - Pamela L Brito
- Hematology and Transfusion Center, University of Campinas, Campinas, Brazil
| | - Sofia Esperti
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
- Erytech Pharma, Lyon, France
| | - Joelle Bernard
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi-site, Hospices Civils de Lyon, Lyon, France
| | - Alexandra Gauthier
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
- Institut d'Hématologique et d'Oncologique Pédiatrique, Hospices Civils de Lyon, Lyon, France
| | - Solene Poutrel
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
- Service de Médecine Interne, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Yves Bertrand
- Institut d'Hématologique et d'Oncologique Pédiatrique, Hospices Civils de Lyon, Lyon, France
| | - Caroline Garcia
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi-site, Hospices Civils de Lyon, Lyon, France
| | - Sara T O Saad
- Hematology and Transfusion Center, University of Campinas, Campinas, Brazil
| | - Stéphane Egée
- Sorbonne Université, CNRS, UMR 8227 LBI2M, Station Biologique de Roscoff SBR, Roscoff, France
| | - Philippe Connes
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
| |
Collapse
|
46
|
Chuntharpursat-Bon E, Povstyan OV, Ludlow MJ, Carrier DJ, Debant M, Shi J, Gaunt HJ, Bauer CC, Curd A, Simon Futers T, Baxter PD, Peckham M, Muench SP, Adamson A, Humphreys N, Tumova S, Bon RS, Cubbon R, Lichtenstein L, Beech DJ. PIEZO1 and PECAM1 interact at cell-cell junctions and partner in endothelial force sensing. Commun Biol 2023; 6:358. [PMID: 37005489 PMCID: PMC10067937 DOI: 10.1038/s42003-023-04706-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 03/14/2023] [Indexed: 04/04/2023] Open
Abstract
Two prominent concepts for the sensing of shear stress by endothelium are the PIEZO1 channel as a mediator of mechanically activated calcium ion entry and the PECAM1 cell adhesion molecule as the apex of a triad with CDH5 and VGFR2. Here, we investigated if there is a relationship. By inserting a non-disruptive tag in native PIEZO1 of mice, we reveal in situ overlap of PIEZO1 with PECAM1. Through reconstitution and high resolution microscopy studies we show that PECAM1 interacts with PIEZO1 and directs it to cell-cell junctions. PECAM1 extracellular N-terminus is critical in this, but a C-terminal intracellular domain linked to shear stress also contributes. CDH5 similarly drives PIEZO1 to junctions but unlike PECAM1 its interaction with PIEZO1 is dynamic, increasing with shear stress. PIEZO1 does not interact with VGFR2. PIEZO1 is required in Ca2+-dependent formation of adherens junctions and associated cytoskeleton, consistent with it conferring force-dependent Ca2+ entry for junctional remodelling. The data suggest a pool of PIEZO1 at cell junctions, the coming together of PIEZO1 and PECAM1 mechanisms and intimate cooperation of PIEZO1 and adhesion molecules in tailoring junctional structure to mechanical requirement.
Collapse
Affiliation(s)
| | | | | | - David J Carrier
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | | | - Jian Shi
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Hannah J Gaunt
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | | | - Alistair Curd
- School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - T Simon Futers
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Paul D Baxter
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Michelle Peckham
- School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Stephen P Muench
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Antony Adamson
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Manchester, M13 9PT, UK
| | - Neil Humphreys
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Manchester, M13 9PT, UK
| | - Sarka Tumova
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Robin S Bon
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Richard Cubbon
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | | | - David J Beech
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
47
|
Hertz L, Flormann D, Birnbaumer L, Wagner C, Laschke MW, Kaestner L. Evidence of in vivo exogen protein uptake by red blood cells: a putative therapeutic concept. Blood Adv 2023; 7:1033-1039. [PMID: 36490356 PMCID: PMC10036505 DOI: 10.1182/bloodadvances.2022008404] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/31/2022] [Accepted: 11/20/2022] [Indexed: 12/14/2022] Open
Abstract
For some molecular players in red blood cells (RBCs), the functional indications and molecular evidence are discrepant. One such protein is transient receptor potential channel of canonical subfamily, member 6 (TRPC6). Transcriptome analysis of reticulocytes revealed the presence of TRPC6 in mouse RBCs and its absence in human RBCs. We transfused TRPC6 knockout RBCs into wild-type mice and performed functional tests. We observed the "rescue" of TRPC6 within 10 days; however, the "rescue" was slower in splenectomized mice. The latter finding led us to mimic the mechanical challenge with the cantilever of an atomic force microscope and simultaneously carry out imaging by confocal (3D) microscopy. We observed the strong interaction of RBCs with the opposed surface at around 200 pN and the formation of tethers. The results of both the transfusion experiments and the atomic force spectroscopy suggest mechanically stimulated protein transfer to RBCs as a protein source in the absence of the translational machinery. This protein transfer mechanism has the potential to be utilized in therapeutic contexts, especially for hereditary diseases involving RBCs, such as hereditary xerocytosis or Gárdos channelopathy.
Collapse
Affiliation(s)
- Laura Hertz
- Theoretical Medicine and Biosciences, Medical Faculty, Saarland University, Homburg, Germany
| | - Daniel Flormann
- Dynamics of Fluids, Experimental Physics, Saarland University, Saarbruecken, Germany
| | - Lutz Birnbaumer
- Institute of Biomedical Research (BIOMED), Catholic University of Argentina, Buenos Aires, Argentina
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, NC
| | - Christian Wagner
- Dynamics of Fluids, Experimental Physics, Saarland University, Saarbruecken, Germany
- Physics and Materials Science Research Unit, University of Luxembourg, Luxembourg City, Luxembourg
| | - Matthias W. Laschke
- Medical Faculty, Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Lars Kaestner
- Theoretical Medicine and Biosciences, Medical Faculty, Saarland University, Homburg, Germany
- Dynamics of Fluids, Experimental Physics, Saarland University, Saarbruecken, Germany
| |
Collapse
|
48
|
Goodman MB, Haswell ES, Vásquez V. Mechanosensitive membrane proteins: Usual and unusual suspects in mediating mechanotransduction. J Gen Physiol 2023; 155:e202213248. [PMID: 36696153 PMCID: PMC9930137 DOI: 10.1085/jgp.202213248] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
This Viewpoint, which accompanies a Special Issue focusing on membrane mechanosensors, discusses unifying and unique features of both established and emerging mechanosensitive (MS) membrane proteins, their distribution across protein families and phyla, and current and future challenges in the study of these important proteins and their partners. MS membrane proteins are essential for tissue development, cellular motion, osmotic homeostasis, and sensing external and self-generated mechanical cues like those responsible for touch and proprioception. Though researchers' attention and this Viewpoint focus on a few famous ion channels that are considered the usual suspects as MS mechanosensors, we also discuss some of the more unusual suspects, such as G-protein coupled receptors. As the field continues to grow, so too will the list of proteins suspected to function as mechanosensors and the diversity of known MS membrane proteins.
Collapse
Affiliation(s)
- Miriam B. Goodman
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Elizabeth S. Haswell
- Department of Biology, Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Valeria Vásquez
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
49
|
Nakahara E, Yamamoto KS, Ogura H, Aoki T, Utsugisawa T, Azuma K, Akagawa H, Watanabe K, Muraoka M, Nakamura F, Kamei M, Tatebayashi K, Shinozuka J, Yamane T, Hibino M, Katsura Y, Nakano-Akamatsu S, Kadowaki N, Maru Y, Ito E, Ohga S, Yagasaki H, Morioka I, Yamamoto T, Kanno H. Variant spectrum of PIEZO1 and KCNN4 in Japanese patients with dehydrated hereditary stomatocytosis. Hum Genome Var 2023; 10:8. [PMID: 36864026 PMCID: PMC9981561 DOI: 10.1038/s41439-023-00235-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/26/2023] [Accepted: 02/01/2023] [Indexed: 03/04/2023] Open
Abstract
Hereditary stomatocytosis (HSt) is a type of congenital hemolytic anemia caused by abnormally increased cation permeability of erythrocyte membranes. Dehydrated HSt (DHSt) is the most common subtype of HSt and is diagnosed based on clinical and laboratory findings related to erythrocytes. PIEZO1 and KCNN4 have been recognized as causative genes, and many related variants have been reported. We analyzed the genomic background of 23 patients from 20 Japanese families suspected of having DHSt using a target capture sequence and identified pathogenic/likely pathogenic variants of PIEZO1 or KCNN4 in 12 families.
Collapse
Affiliation(s)
- Erina Nakahara
- Department of Transfusion Medicine and Cell Processing, Tokyo Women's Medical University, Tokyo, Japan
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Keiko Shimojima Yamamoto
- Department of Transfusion Medicine and Cell Processing, Tokyo Women's Medical University, Tokyo, Japan.
- Institute for Comprehensive Medical Sciences, Tokyo Women's Medical University, Tokyo, Japan.
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan.
| | - Hiromi Ogura
- Department of Transfusion Medicine and Cell Processing, Tokyo Women's Medical University, Tokyo, Japan
| | - Takako Aoki
- Department of Transfusion Medicine and Cell Processing, Tokyo Women's Medical University, Tokyo, Japan
| | - Taiju Utsugisawa
- Department of Transfusion Medicine and Cell Processing, Tokyo Women's Medical University, Tokyo, Japan
| | - Kenko Azuma
- Institute for Comprehensive Medical Sciences, Tokyo Women's Medical University, Tokyo, Japan
| | - Hiroyuki Akagawa
- Institute for Comprehensive Medical Sciences, Tokyo Women's Medical University, Tokyo, Japan
| | - Kenichiro Watanabe
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Michiko Muraoka
- Department of Pediatrics, Fukuyama Medical Center, Okayama, Japan
| | - Fumihiko Nakamura
- Department of Laboratory Medicine, Nara Prefecture General Medical Center, Nara, Japan
| | - Michi Kamei
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Koji Tatebayashi
- Department of Neonatology, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Jun Shinozuka
- Department of Pediatrics, Uji-Tokushukai Medical Center, Kyoto, Japan
| | - Takahisa Yamane
- Department of Hematology, Osaka City General Hospital, Osaka, Japan
| | - Makoto Hibino
- Department of Respiratory Medicine, Shonan Fujisawa Tokushukai Hospital, Fujisawa, Kanagawa, Japan
| | - Yoshiya Katsura
- Department of Metabolism and Endocrinology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | | | - Norimitsu Kadowaki
- Department of Internal Medicine, Division of Hematology, Rheumatology and Respiratory Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yoshiro Maru
- Department of Pharmacology, Tokyo Women's Medical University, Tokyo, Japan
| | - Etsuro Ito
- Department of Pediatrics, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Hiroshi Yagasaki
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Ichiro Morioka
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Toshiyuki Yamamoto
- Institute for Comprehensive Medical Sciences, Tokyo Women's Medical University, Tokyo, Japan
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan
| | - Hitoshi Kanno
- Department of Transfusion Medicine and Cell Processing, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
50
|
Qiu X, Deng Z, Wang M, Feng Y, Bi L, Li L. Piezo protein determines stem cell fate by transmitting mechanical signals. Hum Cell 2023; 36:540-553. [PMID: 36580272 DOI: 10.1007/s13577-022-00853-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/21/2022] [Indexed: 12/30/2022]
Abstract
Piezo ion channel is a mechanosensitive protein on the cell membrane, which contains Piezo1 and Piezo2. Piezo channels are activated by mechanical forces, including stretch, matrix stiffness, static pressure, and shear stress. Piezo channels transmit mechanical signals that cause different downstream responses in the differentiation process, including integrin signaling pathway, ERK1/2 MAPK signaling pathway, Notch signaling, and WNT signaling pathway. In the fate of stem cell differentiation, scientists found differences in Piezo channel expression and found that Piezo channel expression is related to developmental diseases. Here, we briefly review the structure and function of Piezo channels and the relationship between Piezo and mechanical signals, discussing the current understanding of the role of Piezo channels in stem cell fate and associated molecules and developmental diseases. Ultimately, we believe this review will help identify the association between Piezo channels and stem cell fate.
Collapse
Affiliation(s)
- Xiaolei Qiu
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Zhuoyue Deng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Meijing Wang
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Yuqi Feng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Lintao Bi
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| |
Collapse
|