1
|
Kamkin AG, Kamkina OV, Kazansky VE, Mitrokhin VM, Bilichenko A, Nasedkina EA, Shileiko SA, Rodina AS, Zolotareva AD, Zolotarev VI, Sutyagin PV, Mladenov MI. Identification of RNA reads encoding different channels in isolated rat ventricular myocytes and the effect of cell stretching on L-type Ca 2+current. Biol Direct 2023; 18:70. [PMID: 37899484 PMCID: PMC10614344 DOI: 10.1186/s13062-023-00427-0] [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: 06/19/2023] [Accepted: 10/13/2023] [Indexed: 10/31/2023] Open
Abstract
BACKGROUND The study aimed to identify transcripts of specific ion channels in rat ventricular cardiomyocytes and determine their potential role in the regulation of ionic currents in response to mechanical stimulation. The gene expression levels of various ion channels in freshly isolated rat ventricular cardiomyocytes were investigated using the RNA-seq technique. We also measured changes in current through CaV1.2 channels under cell stretching using the whole-cell patch-clamp method. RESULTS Among channels that showed mechanosensitivity, significant amounts of TRPM7, TRPC1, and TRPM4 transcripts were found. We suppose that the recorded L-type Ca2+ current is probably expressed through CaV1.2. Furthermore, stretching cells by 6, 8, and 10 μm, which increases ISAC through the TRPM7, TRPC1, and TRPM4 channels, also decreased ICa,L through the CaV1.2 channels in K+ in/K+ out, Cs+ in/K+ out, K+ in/Cs+ out, and Cs+ in/Cs+ out solutions. The application of a nonspecific ISAC blocker, Gd3+, during cell stretching eliminated ISAC through nonselective cation channels and ICa,L through CaV1.2 channels. Since the response to Gd3+ was maintained in Cs+ in/Cs+ out solutions, we suggest that voltage-gated CaV1.2 channels in the ventricular myocytes of adult rats also exhibit mechanosensitive properties. CONCLUSIONS Our findings suggest that TRPM7, TRPC1, and TRPM4 channels represent stretch-activated nonselective cation channels in rat ventricular myocytes. Probably the CaV1.2 channels in these cells exhibit mechanosensitive properties. Our results provide insight into the molecular mechanisms underlying stretch-induced responses in rat ventricular myocytes, which may have implications for understanding cardiac physiology and pathophysiology.
Collapse
Affiliation(s)
- Andre G Kamkin
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Olga V Kamkina
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Viktor E Kazansky
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Vadim M Mitrokhin
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Andrey Bilichenko
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Elizaveta A Nasedkina
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Stanislav A Shileiko
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Anastasia S Rodina
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Alexandra D Zolotareva
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Valentin I Zolotarev
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Pavel V Sutyagin
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Mitko I Mladenov
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russian Federation.
- Faculty of Natural Sciences and Mathematics, Institute of Biology, "Ss. Cyril and Methodius" University, Skopje, North, Macedonia.
| |
Collapse
|
2
|
Li H, Korcari A, Ciufo D, Mendias CL, Rodeo SA, Buckley MR, Loiselle AE, Pitt GS, Cao C. Increased Ca 2+ signaling through Ca V 1.2 induces tendon hypertrophy with increased collagen fibrillogenesis and biomechanical properties. FASEB J 2023; 37:e23007. [PMID: 37261735 PMCID: PMC10254118 DOI: 10.1096/fj.202300607r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/03/2023] [Accepted: 05/17/2023] [Indexed: 06/02/2023]
Abstract
Tendons are tension-bearing tissues transmitting force from muscle to bone for body movement. This mechanical loading is essential for tendon development, homeostasis, and healing after injury. While Ca2+ signaling has been studied extensively for its roles in mechanotransduction, regulating muscle, bone, and cartilage development and homeostasis, knowledge about Ca2+ signaling and the source of Ca2+ signals in tendon fibroblast biology are largely unknown. Here, we investigated the function of Ca2+ signaling through CaV 1.2 voltage-gated Ca2+ channel in tendon formation. Using a reporter mouse, we found that CaV 1.2 is highly expressed in tendon during development and downregulated in adult homeostasis. To assess its function, we generated ScxCre;CaV 1.2TS mice that express a gain-of-function mutant CaV 1.2 in tendon. We found that mutant tendons were hypertrophic, with more tendon fibroblasts but decreased cell density. TEM analyses demonstrated increased collagen fibrillogenesis in the hypertrophic tendons. Biomechanical testing revealed that the hypertrophic tendons display higher peak load and stiffness, with no changes in peak stress and elastic modulus. Proteomic analysis showed no significant difference in the abundance of type I and III collagens, but mutant tendons had about two-fold increase in other ECM proteins such as tenascin C, tenomodulin, periostin, type XIV and type VIII collagens, around 11-fold increase in the growth factor myostatin, and significant elevation of matrix remodeling proteins including Mmp14, Mmp2, and cathepsin K. Taken together, these data highlight roles for increased Ca2+ signaling through CaV 1.2 on regulating expression of myostatin growth factor and ECM proteins for tendon collagen fibrillogenesis during tendon formation.
Collapse
Affiliation(s)
- Haiyin Li
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopeadics, University of Rochester Medical Center, Rochester, NY, USA
| | - Antonion Korcari
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - David Ciufo
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopeadics, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Scott A. Rodeo
- Sports Medicine and Shoulder Service, Hospital for Special Surgery, New York, NY, USA
| | - Mark R. Buckley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - Alayna E. Loiselle
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopeadics, University of Rochester Medical Center, Rochester, NY, USA
| | - Geoffrey S. Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Chike Cao
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopeadics, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
3
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
4
|
Cavin JB, Wongkrasant P, Glover JB, Balemba OB, MacNaughton WK, Sharkey KA. Intestinal distension orchestrates neuronal activity in the enteric nervous system of adult mice. J Physiol 2023; 601:1183-1206. [PMID: 36752210 PMCID: PMC10319177 DOI: 10.1113/jp284171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/17/2023] [Indexed: 02/09/2023] Open
Abstract
The enteric nervous system (ENS) regulates the motor, secretory and defensive functions of the gastrointestinal tract. Enteric neurons integrate mechanical and chemical inputs from the gut lumen to generate complex motor outputs. How intact enteric neural circuits respond to changes in the gut lumen is not well understood. We recorded intracellular calcium in live-cell confocal recordings in neurons from intact segments of mouse intestine in order to investigate neuronal response to luminal mechanical and chemical stimuli. Wnt1-, ChAT- and Calb1-GCaMP6 mice were used to record neurons from the jejunum and colon. We measured neuronal calcium response to KCl (75 mM), veratridine (10 μM), 1,1-dimethyl-4-phenylpiperazinium (DMPP; 100 μM) or luminal nutrients (Ensure®), in the presence or absence of intraluminal distension. In the jejunum and colon, distension generated by the presence of luminal content (chyme and faecal pellets, respectively) renders the underlying enteric circuit unresponsive to depolarizing stimuli. In the distal colon, high levels of distension inhibit neuronal response to KCl, while intermediate levels of distension reorganize Ca2+ response in circumferentially propagating slow waves. Mechanosensitive channel inhibition suppresses distension-induced Ca2+ elevations, and calcium-activated potassium channel inhibition restores neuronal response to KCl, but not DMPP in the distended colon. In the jejunum, distension prevents a previously unknown tetrodotoxin-resistant neuronal response to luminal nutrient stimulation. Our results demonstrate that intestinal distension regulates the excitability of ENS circuits via mechanosensitive channels. Physiological levels of distension locally silence or synchronize neurons, dynamically regulating the excitability of enteric neural circuits based on the content of the intestinal lumen. KEY POINTS: How the enteric nervous system of the gastrointestinal tract responds to luminal distension remains to be fully elucidated. Here it is shown that intestinal distension modifies intracellular calcium levels in the underlying enteric neuronal network, locally and reversibly silencing neurons in the distended regions. In the distal colon, luminal distension is integrated by specific mechanosensitive channels and coordinates the dynamics of neuronal activation within the enteric network. In the jejunum, distension suppresses the neuronal calcium responses induced by luminal nutrients. Physiological levels of distension dynamically regulate the excitability of enteric neuronal circuits.
Collapse
Affiliation(s)
- Jean-Baptiste Cavin
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Preedajit Wongkrasant
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Joel B Glover
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Live Cell Imaging Laboratory, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Onesmo B Balemba
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - Wallace K MacNaughton
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Keith A Sharkey
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
5
|
Manoj P, Kim JA, Kim S, Li T, Sewani M, Chelu MG, Li N. Sinus node dysfunction: current understanding and future directions. Am J Physiol Heart Circ Physiol 2023; 324:H259-H278. [PMID: 36563014 PMCID: PMC9886352 DOI: 10.1152/ajpheart.00618.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
The sinoatrial node (SAN) is the primary pacemaker of the heart. Normal SAN function is crucial in maintaining proper cardiac rhythm and contraction. Sinus node dysfunction (SND) is due to abnormalities within the SAN, which can affect the heartbeat frequency, regularity, and the propagation of electrical pulses through the cardiac conduction system. As a result, SND often increases the risk of cardiac arrhythmias. SND is most commonly seen as a disease of the elderly given the role of degenerative fibrosis as well as other age-dependent changes in its pathogenesis. Despite the prevalence of SND, current treatment is limited to pacemaker implantation, which is associated with substantial medical costs and complications. Emerging evidence has identified various genetic abnormalities that can cause SND, shedding light on the molecular underpinnings of SND. Identification of these molecular mechanisms and pathways implicated in the pathogenesis of SND is hoped to identify novel therapeutic targets for the development of more effective therapies for this disease. In this review article, we examine the anatomy of the SAN and the pathophysiology and epidemiology of SND. We then discuss in detail the most common genetic mutations correlated with SND and provide our perspectives on future research and therapeutic opportunities in this field.
Collapse
Affiliation(s)
- Pavan Manoj
- School of Public Health, Texas A&M University, College Station, Texas
| | - Jitae A Kim
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Stephanie Kim
- Department of BioSciences, Rice University, Houston, Texas
| | - Tingting Li
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Maham Sewani
- Department of BioSciences, Rice University, Houston, Texas
| | - Mihail G Chelu
- Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Na Li
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
6
|
Li H, Korcari A, Ciufo D, Mendias CL, Rodeo SA, Buckley MR, Loiselle AE, Pitt GS, Cao C. Increased Ca 2+ signaling through Ca V 1.2 induces tendon hypertrophy with increased collagen fibrillogenesis and biomechanical properties. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525119. [PMID: 36747837 PMCID: PMC9900778 DOI: 10.1101/2023.01.24.525119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Tendons are tension-bearing tissues transmitting force from muscle to bone for body movement. This mechanical loading is essential for tendon development, homeostasis, and healing after injury. While Ca 2+ signaling has been studied extensively for its roles in mechanotransduction, regulating muscle, bone and cartilage development and homeostasis, knowledge about Ca 2+ signaling and the source of Ca 2+ signals in tendon fibroblast biology are largely unknown. Here, we investigated the function of Ca 2+ signaling through Ca V 1.2 voltage-gated Ca 2+ channel in tendon formation. Using a reporter mouse, we found that Ca V 1.2 is highly expressed in tendon during development and downregulated in adult homeostasis. To assess its function, we generated ScxCre;Ca V 1.2 TS mice that express a gain-of-function mutant Ca V 1.2 channel (Ca V 1.2 TS ) in tendon. We found that tendons in the mutant mice were approximately 2/3 larger and had more tendon fibroblasts, but the cell density of the mutant mice decreased by around 22%. TEM analyses demonstrated increased collagen fibrillogenesis in the hypertrophic tendon. Biomechanical testing revealed that the hypertrophic Achilles tendons display higher peak load and stiffness, with no changes in peak stress and elastic modulus. Proteomics analysis reveals no significant difference in the abundance of major extracellular matrix (ECM) type I and III collagens, but mutant mice had about 2-fold increase in other ECM proteins such as tenascin C, tenomodulin, periostin, type XIV and type VIII collagens, around 11-fold increase in the growth factor of TGF-β family myostatin, and significant elevation of matrix remodeling proteins including Mmp14, Mmp2 and cathepsin K. Taken together, these data highlight roles for increased Ca 2+ signaling through Ca V 1.2 on regulating expression of myostatin growth factor and ECM proteins for tendon collagen fibrillogenesis during tendon formation.
Collapse
|
7
|
Kola JB, Docsa T, Uray K. Mechanosensing in the Physiology and Pathology of the Gastrointestinal Tract. Int J Mol Sci 2022; 24:ijms24010177. [PMID: 36613619 PMCID: PMC9820522 DOI: 10.3390/ijms24010177] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/10/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Normal gastrointestinal function relies on sensing and transducing mechanical signals into changes in intracellular signaling pathways. Both specialized mechanosensing cells, such as certain enterochromaffin cells and enteric neurons, and non-specialized cells, such as smooth muscle cells, interstitial cells of Cajal, and resident macrophages, participate in physiological and pathological responses to mechanical signals in the gastrointestinal tract. We review the role of mechanosensors in the different cell types of the gastrointestinal tract. Then, we provide several examples of the role of mechanotransduction in normal physiology. These examples highlight the fact that, although these responses to mechanical signals have been known for decades, the mechanosensors involved in these responses to mechanical signals are largely unknown. Finally, we discuss several diseases involving the overstimulation or dysregulation of mechanotransductive pathways. Understanding these pathways and identifying the mechanosensors involved in these diseases may facilitate the identification of new drug targets to effectively treat these diseases.
Collapse
Affiliation(s)
- Job Baffin Kola
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Center of Excellence, The Hungarian Academy of Sciences, 4032 Debrecen, Hungary
| | - Tibor Docsa
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Center of Excellence, The Hungarian Academy of Sciences, 4032 Debrecen, Hungary
| | - Karen Uray
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Center of Excellence, The Hungarian Academy of Sciences, 4032 Debrecen, Hungary
- Correspondence:
| |
Collapse
|
8
|
Mercado-Perez A, Beyder A. Gut feelings: mechanosensing in the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2022; 19:283-296. [PMID: 35022607 PMCID: PMC9059832 DOI: 10.1038/s41575-021-00561-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 12/11/2022]
Abstract
The primary function of the gut is to procure nutrients. Synchronized mechanical activities underlie nearly all its endeavours. Coordination of mechanical activities depends on sensing of the mechanical forces, in a process called mechanosensation. The gut has a range of mechanosensory cells. They function either as specialized mechanoreceptors, which convert mechanical stimuli into coordinated physiological responses at the organ level, or as non-specialized mechanosensory cells that adjust their function based on the mechanical state of their environment. All major cell types in the gastrointestinal tract contain subpopulations that act as specialized mechanoreceptors: epithelia, smooth muscle, neurons, immune cells, and others. These cells are tuned to the physical properties of the surrounding tissue, so they can discriminate mechanical stimuli from the baseline mechanical state. The importance of gastrointestinal mechanosensation has long been recognized, but the latest discoveries of molecular identities of mechanosensors and technical advances that resolve the relevant circuitry have poised the field to make important intellectual leaps. This Review describes the mechanical factors relevant for normal function, as well as the molecules, cells and circuits involved in gastrointestinal mechanosensing. It concludes by outlining important unanswered questions in gastrointestinal mechanosensing.
Collapse
Affiliation(s)
- Arnaldo Mercado-Perez
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA
- Medical Scientist Training Program (MSTP), Mayo Clinic, Rochester, MN, USA
| | - Arthur Beyder
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA.
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
9
|
Ahmed ASI, Sheng MHC, Lau KHW, Wilson SM, Wongworawat MD, Tang X, Ghahramanpouri M, Nehme A, Xu Y, Abdipour A, Zhang XB, Wasnik S, Baylink DJ. Calcium released by osteoclastic resorption stimulates autocrine/paracrine activities in local osteogenic cells to promote coupled bone formation. Am J Physiol Cell Physiol 2022; 322:C977-C990. [PMID: 35385325 PMCID: PMC9109806 DOI: 10.1152/ajpcell.00413.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A major cause of osteoporosis is impaired coupled bone formation. Mechanistically, both osteoclast-derived and bone-derived growth factors have been previously implicated. We hypothesize that the release of bone calcium during osteoclastic bone resorption is essential for coupled bone formation. Osteoclastic resorption increases interstitial fluid calcium locally from the normal 1.8 mM up to 5 mM. MC3T3-E1 osteoprogenitors, cultured in a 3.6 mM calcium medium, demonstrated that calcium signaling stimulated osteogenic cell proliferation, differentiation, and migration. Calcium channel knockdown studies implicated calcium channels, Cav1.2, store-operated calcium entry (SOCE), and calcium-sensing receptor (CaSR) in regulating bone cell anabolic activities. MC3T3-E1 cultured in a 3.6 mM calcium medium expressed increased gene expression of Wnt signaling and growth factors platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), and bone morphogenic protein-2 (BMP 2). Our coupling model of bone formation, the Receptor activator of nuclear factor-kappa-Β ligand (RANKL) treated mouse calvaria, confirmed the role of calcium signaling in coupled bone formation by exhibiting increased gene expression for osterix and osteocalcin. Critically, dual immunocytochemistry showed that RANKL treatment increased osterix positive cells and increased fluorescence intensity of Cav1.2 and CaSR protein expression per osterix positive cell. The data established that calcium released by osteoclasts contributed to the regulation of coupled bone formation. CRISPR/Cas-9 knockout of Cav1.2 in osteoprogenitors cultured in basal calcium medium caused a >80% decrease in the expression of downstream osteogenic genes, emphasizing the large magnitude of the effect of calcium signaling. Thus, calcium signaling is a major regulator of coupled bone formation.
Collapse
Affiliation(s)
- Abu Shufian Ishtiaq Ahmed
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, California, United States.,The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Matilda H C Sheng
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, California, United States.,Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, California, United States
| | - Kin-Hing William Lau
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, California, United States
| | - Sean M Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - M Daniel Wongworawat
- Department of Orthopaedic Surgery, Loma Linda University, Loma Linda, California, United States
| | - Xiaolei Tang
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY, United States
| | - Mahdis Ghahramanpouri
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, California, United States
| | - Antoine Nehme
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, California, United States
| | - Yi Xu
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, California, United States.,Division of Hematology and Oncology, Department of Medicine, Loma Linda University and Loma Linda University Cancer Center, Loma Linda, CA, United States
| | - Amir Abdipour
- Division of Nephrology, Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Xiao-Bing Zhang
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, United States
| | - Samiksha Wasnik
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, California, United States
| | - David J Baylink
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, California, United States
| |
Collapse
|
10
|
Application of piconewton forces to individual filopodia reveals mechanosensory role of L-type Ca 2+ channels. Biomaterials 2022; 284:121477. [PMID: 35395455 DOI: 10.1016/j.biomaterials.2022.121477] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/18/2022] [Indexed: 11/02/2022]
Abstract
Filopodia are ubiquitous membrane projections that play crucial role in guiding cell migration on rigid substrates and through extracellular matrix by utilizing yet unknown mechanosensing molecular pathways. As recent studies show that Ca2+ channels localized to filopodia play an important role in regulation of their formation and since some Ca2+ channels are known to be mechanosensitive, force-dependent activity of filopodial Ca2+ channels might be linked to filopodia's mechanosensing function. We tested this hypothesis by monitoring changes in the intra-filopodial Ca2+ level in response to application of stretching force to individual filopodia of several cell types using optical tweezers. Results show that stretching forces of tens of pN strongly promote Ca2+ influx into filopodia, causing persistent Ca2+ oscillations that last for minutes even after the force is released. Several known mechanosensitive Ca2+ channels, such as Piezo 1, Piezo 2 and TRPV4, were found to be dispensable for the observed force-dependent Ca2+ influx, while L-type Ca2+ channels appear to be a key player in the discovered phenomenon. As previous studies have shown that intra-filopodial transient Ca2+ signals play an important role in guidance of cell migration, our results suggest that the force-dependent activation of L-type Ca2+ channels may contribute to this process. Overall, our study reveals an intricate interplay between mechanical forces and Ca2+ signaling in filopodia, providing novel mechanistic insights for the force-dependent filopodia functions in guidance of cell migration.
Collapse
|
11
|
What keeps us ticking? Sinoatrial node mechano-sensitivity: the grandfather clock of cardiac rhythm. Biophys Rev 2021; 13:707-716. [PMID: 34777615 DOI: 10.1007/s12551-021-00831-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 08/17/2021] [Indexed: 01/01/2023] Open
Abstract
The rhythmic and spontaneously generated electrical excitation that triggers the heartbeat originates in the sinoatrial node (SAN). SAN automaticity has been thoroughly investigated, which has uncovered fundamental mechanisms involved in cardiac pacemaking that are generally categorised into two interacting and overlapping systems: the 'membrane' and 'Ca2+ clock'. The principal focus of research has been on these two systems of oscillators, which have been studied primarily in single cells and isolated tissue, experimental preparations that do not consider mechanical factors present in the whole heart. SAN mechano-sensitivity has long been known to be a contributor to SAN pacemaking-both as a driver and regulator of automaticity-but its essential nature has been underappreciated. In this review, following a description of the traditional 'clocks' of SAN automaticity, we describe mechanisms of SAN mechano-sensitivity and its vital role for SAN function, making the argument that the 'mechanics oscillator' is, in fact, the 'grandfather clock' of cardiac rhythm.
Collapse
|
12
|
Li Q, Duan Z, Sun C, Zheng J, Xu G, Yang N. Genetic variations for the eggshell crystal structure revealed by genome-wide association study in chickens. BMC Genomics 2021; 22:786. [PMID: 34727889 PMCID: PMC8565016 DOI: 10.1186/s12864-021-08103-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 10/18/2021] [Indexed: 11/10/2022] Open
Abstract
Background Eggshell is a bio-ceramic material comprising columnar calcite (CaCO3) crystals and organic proteinaceous matrix. The size, shape and orientation of the CaCO3 crystals influence the microstructural properties of chicken eggshells. However, the genetic architecture underlying eggshell crystal polymorphism remains to be elucidated. Results The integral intensity of the nine major diffraction peaks, total integral intensity and degree of orientation of the crystals were measured followed by a genome-wide association study in 839 F2 hens. The results showed that the total integral intensity was positively correlated with the eggshell strength, eggshell thickness, eggshell weight, mammillary layer thickness and effective layer thickness. The SNP-based heritabilities of total integral intensity and degree of orientation were 0.23 and 0.06, respectively. The 621 SNPs located in the range from 55.6 to 69.1 Mb in GGA1 were significantly associated with TA. PLCZ1, ABCC9, ITPR2, KCNJ8, CACNA1C and IAPP, which are involved in the biological process of regulating cytosolic calcium ion concentration, can be suggested as key genes regulating the total integral intensity. Conclusions The findings greatly advance the understanding of the genetic basis underlying the crystal ultrastructure of eggshell quality and thus will have practical significance in breeding programs for improving eggshell quality. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08103-1.
Collapse
Affiliation(s)
- Quanlin Li
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, 100193, Beijing, China
| | - Zhongyi Duan
- National Animal Husbandry Service, 100125, Beijing, China
| | - Congjiao Sun
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, 100193, Beijing, China
| | - Jiangxia Zheng
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, 100193, Beijing, China
| | - Guiyun Xu
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, 100193, Beijing, China
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, 100193, Beijing, China.
| |
Collapse
|
13
|
Prospero AG, Pinto LA, Matos RVR, Soares GA, Oliveira RB, Mascarenhas S, Miranda JRDA. New device for active gastric mechanical stimulation. Neurogastroenterol Motil 2021; 33:e14169. [PMID: 33969918 DOI: 10.1111/nmo.14169] [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] [Revised: 04/16/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Gastroparesis is a chronic stomach disorder and effective treatment is the aim of different strategies. Alternative therapies consist of an electrical stimulation of the stomach to evoke a response in the gastric activity. We present the development and in vivo application of an electromagnet system to induce a mechanical stimulus in the stomach aiming for gastric contractile responses. METHODS The electromagnet system consisted of an implantable magnet and an external drive coil. We implanted the magnet at the greater curvature of the gastric body in rats. We applied an alternating current to the drive coils, inducing mechanical stimulation of the gastric wall. We measured the gastric contraction activity and gastric electrical activity in response to the stimulus using AC biosusceptometry and electrogastrography. Moreover, we used the phenol red to evaluate the stimulus effects on gastrointestinal transit. KEY RESULTS The stimulus increased the spectral intensity and signal-to-noise ratio significantly of gastric contraction activity and gastric electrical activity. Furthermore, we found a lower phenol red retention in the stomach in rats without stimulus. No significant differences were found in frequency and root mean square amplitude. CONCLUSIONS & INFERENCES We developed a new simple electromagnet system that evoked a contraction and gastric electrical response using a mechanical stimulus and decreased gastric emptying time. The system is an accessible tool and may contribute to gastroparesis studies in animals.
Collapse
Affiliation(s)
- Andre Gonçalves Prospero
- Departamento de Biofísica e Farmacologia, Laboratório de Biomagnetismo, UNESP/São Paulo State University, Botucatu, Brazil
| | - Leonardo Antonio Pinto
- Departamento de Biofísica e Farmacologia, Laboratório de Biomagnetismo, UNESP/São Paulo State University, Botucatu, Brazil
| | - Ronaldo Vitor Reis Matos
- Departamento de Biofísica e Farmacologia, Laboratório de Biomagnetismo, UNESP/São Paulo State University, Botucatu, Brazil
| | - Guilherme Augusto Soares
- Departamento de Biofísica e Farmacologia, Laboratório de Biomagnetismo, UNESP/São Paulo State University, Botucatu, Brazil
| | - Ricardo Brandt Oliveira
- Faculdade de Medicina de Ribeirão Preto, USP/University of São Paulo, Ribeirão Preto, Brazil
| | | | - José Ricardo de Arruda Miranda
- Departamento de Biofísica e Farmacologia, Laboratório de Biomagnetismo, UNESP/São Paulo State University, Botucatu, Brazil
| |
Collapse
|
14
|
Caprini D, Schwartz S, Lanza E, Milanetti E, Lucente V, Ferrarese G, Chiodo L, Nicoletti M, Folli V. A Shearless Microfluidic Device Detects a Role in Mechanosensitivity for AWC ON Neuron in Caenorhabditis elegans. Adv Biol (Weinh) 2021; 5:e2100927. [PMID: 34423577 DOI: 10.1002/adbi.202100927] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/03/2021] [Indexed: 11/08/2022]
Abstract
AWC olfactory neurons are fundamental for chemotaxis toward volatile attractants in Caenorhabditis elegans. Here, it is shown that AWCON responds not only to chemicals but also to mechanical stimuli caused by fluid flow changes in a microfluidic device. The dynamics of calcium events are correlated with the stimulus amplitude. It is further shown that the mechanosensitivity of AWCON neurons has an intrinsic nature rather than a synaptic origin, and the calcium transient response is mediated by TAX-4 cGMP-gated cation channel, suggesting the involvement of one or more "odorant" receptors in AWCON mechano-transduction. In many cases, the responses show plateau properties resembling bistable calcium dynamics where neurons can switch from one stable state to the other. To investigate the unprecedentedly observed mechanosensitivity of AWCON neurons, a novel microfluidic device is designed to minimize the fluid shear flow in the arena hosting the nematodes. Animals in this device show reduced neuronal activation of AWCON neurons. The results observed indicate that the tangential component of the mechanical stress is the main contributor to the mechanosensitivity of AWCON . Furthermore, the microfluidic platform, integrating shearless perfusion and calcium imaging, provides a novel and more controlled solution for in vivo analysis both in micro-organisms and cultured cells.
Collapse
Affiliation(s)
- Davide Caprini
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy
| | - Silvia Schwartz
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy
| | - Enrico Lanza
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy
| | - Edoardo Milanetti
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy.,Department of Physics, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy
| | - Valeria Lucente
- CREST OPTICS S.p.A., Via di Torre Rossa 66, Rome, 00165, Italy
| | - Giuseppe Ferrarese
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy.,Department of Engineering, Campus Bio-Medico University, Via Álvaro del Portillo 21, Rome, 00128, Italy
| | - Letizia Chiodo
- Department of Engineering, Campus Bio-Medico University, Via Álvaro del Portillo 21, Rome, 00128, Italy
| | - Martina Nicoletti
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy.,Department of Engineering, Campus Bio-Medico University, Via Álvaro del Portillo 21, Rome, 00128, Italy
| | - Viola Folli
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy
| |
Collapse
|
15
|
Hegyi B, Shimkunas R, Jian Z, Izu LT, Bers DM, Chen-Izu Y. Mechanoelectric coupling and arrhythmogenesis in cardiomyocytes contracting under mechanical afterload in a 3D viscoelastic hydrogel. Proc Natl Acad Sci U S A 2021; 118:e2108484118. [PMID: 34326268 PMCID: PMC8346795 DOI: 10.1073/pnas.2108484118] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The heart pumps blood against the mechanical afterload from arterial resistance, and increased afterload may alter cardiac electrophysiology and contribute to life-threatening arrhythmias. However, the cellular and molecular mechanisms underlying mechanoelectric coupling in cardiomyocytes remain unclear. We developed an innovative patch-clamp-in-gel technology to embed cardiomyocytes in a three-dimensional (3D) viscoelastic hydrogel that imposes an afterload during regular myocyte contraction. Here, we investigated how afterload affects action potentials, ionic currents, intracellular Ca2+ transients, and cell contraction of adult rabbit ventricular cardiomyocytes. We found that afterload prolonged action potential duration (APD), increased transient outward K+ current, decreased inward rectifier K+ current, and increased L-type Ca2+ current. Increased Ca2+ entry caused enhanced Ca2+ transients and contractility. Moreover, elevated afterload led to discordant alternans in APD and Ca2+ transient. Ca2+ alternans persisted under action potential clamp, indicating that the alternans was Ca2+ dependent. Furthermore, all these afterload effects were significantly attenuated by inhibiting nitric oxide synthase 1 (NOS1). Taken together, our data reveal a mechano-chemo-electrotransduction (MCET) mechanism that acutely transduces afterload through NOS1-nitric oxide signaling to modulate the action potential, Ca2+ transient, and contractility. The MCET pathway provides a feedback loop in excitation-Ca2+ signaling-contraction coupling, enabling autoregulation of contractility in cardiomyocytes in response to afterload. This MCET mechanism is integral to the individual cardiomyocyte (and thus the heart) to intrinsically enhance its contractility in response to the load against which it has to do work. While this MCET is largely compensatory for physiological load changes, it may also increase susceptibility to arrhythmias under excessive pathological loading.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Rafael Shimkunas
- Department of Pharmacology, University of California, Davis, CA 95616
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Zhong Jian
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Leighton T Izu
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Ye Chen-Izu
- Department of Pharmacology, University of California, Davis, CA 95616;
- Department of Biomedical Engineering, University of California, Davis, CA 95616
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| |
Collapse
|
16
|
Lu Y, Chen Y, Lin Y, Chen S, Chen Y. Mechanoelectrical feedback in pulmonary vein arrhythmogenesis: Clinical challenges and therapeutic opportunities. J Arrhythm 2020; 36:608-614. [PMID: 32782628 PMCID: PMC7411213 DOI: 10.1002/joa3.12391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/28/2020] [Accepted: 06/04/2020] [Indexed: 12/24/2022] Open
Abstract
Mechanoelectrical feedback is an important factor in the pathophysiology of atrial fibrillation (AF). Ectopic electrical activity originating from pulmonary vein (PV) myocardial sleeves has been found to trigger and maintain paroxysmal AF. Dilated PVs by high stretching force may activate mechanoelectrical feedback, which induces calcium overload and produces afterdepolarization. These results, in turn, increase PV arrhythmogenesis and contribute to initiation of AF. Paracrine factors, effectors of the renin-angiotensin system, membranous channels, or cytoskeleton of PV myocytes may modulate PV arrhythmogenesis directly through mechanoelectrical feedback or indirectly through endocardial/myocardial cross-talk. The purpose of this review is to present laboratory and translational relevance of mechanoelectrical feedback in PV arrhythmogenesis. Targeting mechanoelectrical feedback in PV arrhythmogenesis may shed light on potential opportunities and clinical concerns of AF treatment.
Collapse
Affiliation(s)
- Yen‐Yu Lu
- Division of CardiologyDepartment of Internal MedicineSijhih Cathay General HospitalNew Taipei CityTaiwan
- School of MedicineFu‐Jen Catholic UniversityNew Taipei CityTaiwan
| | - Yao‐Chang Chen
- Department of Biomedical Engineering and Institute of PhysiologyNational Defense Medical CenterTaipeiTaiwan
| | - Yung‐Kuo Lin
- Division of Cardiovascular MedicineDepartment of Internal MedicineWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
- Cardiovacular Research CenterWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Shih‐Ann Chen
- Heart Rhythm Center and Division of CardiologyDepartment of MedicineTaipei Veterans General HospitalTaipeiTaiwan
| | - Yi‐Jen Chen
- Division of Cardiovascular MedicineDepartment of Internal MedicineWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
- Cardiovacular Research CenterWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
- Graduate Institute of Clinical MedicineCollege of MedicineTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
17
|
Quinn TA, Kohl P. Cardiac Mechano-Electric Coupling: Acute Effects of Mechanical Stimulation on Heart Rate and Rhythm. Physiol Rev 2020; 101:37-92. [PMID: 32380895 DOI: 10.1152/physrev.00036.2019] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The heart is vital for biological function in almost all chordates, including humans. It beats continually throughout our life, supplying the body with oxygen and nutrients while removing waste products. If it stops, so does life. The heartbeat involves precise coordination of the activity of billions of individual cells, as well as their swift and well-coordinated adaption to changes in physiological demand. Much of the vital control of cardiac function occurs at the level of individual cardiac muscle cells, including acute beat-by-beat feedback from the local mechanical environment to electrical activity (as opposed to longer term changes in gene expression and functional or structural remodeling). This process is known as mechano-electric coupling (MEC). In the current review, we present evidence for, and implications of, MEC in health and disease in human; summarize our understanding of MEC effects gained from whole animal, organ, tissue, and cell studies; identify potential molecular mediators of MEC responses; and demonstrate the power of computational modeling in developing a more comprehensive understanding of ‟what makes the heart tick.ˮ.
Collapse
Affiliation(s)
- T Alexander Quinn
- Department of Physiology and Biophysics and School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Medical Faculty of the University of Freiburg, Freiburg, Germany; and CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Peter Kohl
- Department of Physiology and Biophysics and School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Medical Faculty of the University of Freiburg, Freiburg, Germany; and CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
18
|
Saucerman JJ, Tan PM, Buchholz KS, McCulloch AD, Omens JH. Mechanical regulation of gene expression in cardiac myocytes and fibroblasts. Nat Rev Cardiol 2019; 16:361-378. [PMID: 30683889 PMCID: PMC6525041 DOI: 10.1038/s41569-019-0155-8] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The intact heart undergoes complex and multiscale remodelling processes in response to altered mechanical cues. Remodelling of the myocardium is regulated by a combination of myocyte and non-myocyte responses to mechanosensitive pathways, which can alter gene expression and therefore function in these cells. Cellular mechanotransduction and its downstream effects on gene expression are initially compensatory mechanisms during adaptations to the altered mechanical environment, but under prolonged and abnormal loading conditions, they can become maladaptive, leading to impaired function and cardiac pathologies. In this Review, we summarize mechanoregulated pathways in cardiac myocytes and fibroblasts that lead to altered gene expression and cell remodelling under physiological and pathophysiological conditions. Developments in systems modelling of the networks that regulate gene expression in response to mechanical stimuli should improve integrative understanding of their roles in vivo and help to discover new combinations of drugs and device therapies targeting mechanosignalling in heart disease.
Collapse
Affiliation(s)
- Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Philip M Tan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kyle S Buchholz
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| | - Andrew D McCulloch
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Jeffrey H Omens
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
19
|
Pourfarhangi KE, Bergman A, Gligorijevic B. ECM Cross-Linking Regulates Invadopodia Dynamics. Biophys J 2019; 114:1455-1466. [PMID: 29590602 DOI: 10.1016/j.bpj.2018.01.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/22/2017] [Accepted: 01/22/2018] [Indexed: 12/26/2022] Open
Abstract
Invadopodia are membrane protrusions dynamically assembled by invasive cancer cells in contact with the extracellular matrix (ECM). Invadopodia are enriched by the structural proteins actin and cortactin as well as metalloproteases such as MT1-MMP, whose function is to degrade the surrounding ECM. During metastasis, invadopodia are necessary for cancer cell intravasation and extravasation. Although signaling pathways involved in the assembly and function of invadopodia are well studied, few studies address invadopodia dynamics and how the cell-ECM interactions contribute to cell invasion. Using iterative analysis based on time-lapse microscopy and mathematical modeling of invasive cancer cells, we found that cells oscillate between invadopodia presence and cell stasis-termed the "invadopodia state"-and invadopodia absence during cell translocation-termed the "migration state." Our data suggest that β1-integrin-ECM binding and ECM cross-linking control the duration of each of the two states. By changing the concentration of cross-linkers in two-dimensional and three-dimensional cultures, we generate an ECM in which 0-0.92 of total lysine residues are cross-linked. Using an ECM with a range of cross-linking degrees, we demonstrate that the dynamics of invadopodia-related functions have a biphasic relationship to ECM cross-linking. At intermediate levels of ECM cross-linking (0.39), cells exhibit rapid invadopodia protrusion-retraction cycles and rapid calcium spikes, which lead to more frequent MT1-MMP delivery, causing maximal invadopodia-mediated ECM degradation. In contrast, both extremely high or low levels of cross-linking lead to slower invadopodia-related dynamics and lower ECM degradation. Additionally, β1-integrin inhibition modifies the dynamics of invadopodia-related functions as well as the length of time cells spend in either of the states. Collectively, these data suggest that β1-integrin-ECM binding nonlinearly translates small physical differences in the extracellular environment to differences in the dynamics of cancer cell behaviors. Understanding the conditions under which invadopodia can be reduced by subtle environment-targeting treatments may lead to combination therapies for preventing metastatic spread.
Collapse
Affiliation(s)
| | - Aviv Bergman
- Systems & Computational Biology Department, Albert Einstein College of Medicine, New York, New York; Santa Fe Institute, Santa Fe, New Mexico
| | - Bojana Gligorijevic
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, Pennsylvania; Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| |
Collapse
|
20
|
Takahashi K, Hayashi S, Miyajima M, Omori M, Wang J, Kaihara K, Morimatsu M, Wang C, Chen J, Iribe G, Naruse K, Sokabe M. L-type calcium channel modulates mechanosensitivity of the cardiomyocyte cell line H9c2. Cell Calcium 2019; 79:68-74. [PMID: 30836292 DOI: 10.1016/j.ceca.2019.02.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 02/01/2019] [Accepted: 02/18/2019] [Indexed: 01/08/2023]
Abstract
The application of mechanical stimuli to cells often induce increases in intracellular calcium, affecting the regulation of a variety of cell functions. Although the mechanism of mechanotransduction-induced calcium increases has not been fully resolved, the involvement of mechanosensitive ion channels in the plasma membrane and the endoplasmic reticulum has been reported. Here, we demonstrate that voltage-gated L-type calcium channels play a critical role in the mechanosensitive calcium response in H9c2 rat cardiomyocytes. The intracellular calcium level in H9c2 cells increased in a reproducible dose-dependent manner in response to uniaxial stretching. The stretch-activated calcium response (SICR) completely disappeared in calcium-free medium, whereas thapsigargin and cyclopiazonic acid, inhibitors of sarcoendoplasmic reticulum calcium ATPase, partially reduced the SICR. These findings suggest that both calcium influx across the cell membrane and calcium release from the sarcoendoplasmic reticulum are involved in the SICR. Nifedipine, diltiazem, and verapamil, inhibitors of L-type calcium channels, reduced the SICR in a dose-dependent manner. Furthermore, small interfering RNA against the L-type calcium channel α1c subunit diminished the SICR dramatically. Nifedipine also diminished the mechanosensitivity of Langendorff-perfused rat heart. These results suggest that the SICR in H9c2 cardiomyocytes involves the activation of L-type calcium channels and subsequent calcium release from the sarcoendoplasmic reticulum.
Collapse
Affiliation(s)
- Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan; Department of Physiology, Nagoya University School of Medicine, Nagoya, 466-8550, Japan.
| | - Shogo Hayashi
- Department of Physiology, Nagoya University School of Medicine, Nagoya, 466-8550, Japan
| | - Mari Miyajima
- Department of Physiology, Nagoya University School of Medicine, Nagoya, 466-8550, Japan
| | - Marei Omori
- Department of Physiology, Nagoya University School of Medicine, Nagoya, 466-8550, Japan
| | - Jing Wang
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan; Department of Cardiology, Qingdao Municipal Hospital, Qingdao, 266001, China
| | - Keiko Kaihara
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masatoshi Morimatsu
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Chen Wang
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Jian Chen
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan; Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Gentaro Iribe
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Keiji Naruse
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masahiro Sokabe
- Department of Physiology, Nagoya University School of Medicine, Nagoya, 466-8550, Japan; Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| |
Collapse
|
21
|
Tan PM, Buchholz KS, Omens JH, McCulloch AD, Saucerman JJ. Predictive model identifies key network regulators of cardiomyocyte mechano-signaling. PLoS Comput Biol 2017; 13:e1005854. [PMID: 29131824 PMCID: PMC5703578 DOI: 10.1371/journal.pcbi.1005854] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/27/2017] [Accepted: 10/26/2017] [Indexed: 12/11/2022] Open
Abstract
Mechanical strain is a potent stimulus for growth and remodeling in cells. Although many pathways have been implicated in stretch-induced remodeling, the control structures by which signals from distinct mechano-sensors are integrated to modulate hypertrophy and gene expression in cardiomyocytes remain unclear. Here, we constructed and validated a predictive computational model of the cardiac mechano-signaling network in order to elucidate the mechanisms underlying signal integration. The model identifies calcium, actin, Ras, Raf1, PI3K, and JAK as key regulators of cardiac mechano-signaling and characterizes crosstalk logic imparting differential control of transcription by AT1R, integrins, and calcium channels. We find that while these regulators maintain mostly independent control over distinct groups of transcription factors, synergy between multiple pathways is necessary to activate all the transcription factors necessary for gene transcription and hypertrophy. We also identify a PKG-dependent mechanism by which valsartan/sacubitril, a combination drug recently approved for treating heart failure, inhibits stretch-induced hypertrophy, and predict further efficacious pairs of drug targets in the network through a network-wide combinatorial search. Common stresses such as high blood pressure or heart attack can lead to heart failure, which afflicts over 25 million people worldwide. These stresses cause cardiomyocytes to grow and remodel, which may initially be beneficial but ultimately worsen heart function. Current heart failure drugs such as beta-blockers counteract biochemical cues prompting cardiomyocyte growth, yet mechanical cues to cardiomyocytes such as stretch are just as important in driving cardiac dysfunction. However, no pharmacological treatments have yet been approved that specifically target mechano-signaling, in part because it is not clear how cardiomyocytes integrate signals from multiple mechano-responsive sensors and pathways into their decision to grow. To address this challenge, we built a systems-level computational model that represents 125 interactions between 94 stretch-responsive signaling molecules. The model correctly predicts 134 of 172 previous independent experimental observations, and identifies the key regulators of stretch-induced cardiomyocyte remodeling. Although cardiomyocytes have many mechano-signaling pathways that function largely independently, we find that cooperation between them is necessary to cause growth and remodeling. We identify mechanisms by which a recently approved heart failure drug pair affects mechano-signaling, and we further predict additional pairs of drug targets that could be used to help reverse heart failure.
Collapse
Affiliation(s)
- Philip M. Tan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Kyle S. Buchholz
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Jeffrey H. Omens
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Andrew D. McCulloch
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
22
|
MacDonald EA, Stoyek MR, Rose RA, Quinn TA. Intrinsic regulation of sinoatrial node function and the zebrafish as a model of stretch effects on pacemaking. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 130:198-211. [PMID: 28743586 DOI: 10.1016/j.pbiomolbio.2017.07.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/17/2017] [Accepted: 07/21/2017] [Indexed: 12/18/2022]
Abstract
Excitation of the heart occurs in a specialised region known as the sinoatrial node (SAN). Tight regulation of SAN function is essential for the maintenance of normal heart rhythm and the response to (patho-)physiological changes. The SAN is regulated by extrinsic (central nervous system) and intrinsic (neurons, peptides, mechanics) factors. The positive chronotropic response to stretch in particular is essential for beat-by-beat adaptation to changes in hemodynamic load. Yet, the mechanism of this stretch response is unknown, due in part to the lack of an appropriate experimental model for targeted investigations. We have been investigating the zebrafish as a model for the study of intrinsic regulation of SAN function. In this paper, we first briefly review current knowledge of the principal components of extrinsic and intrinsic SAN regulation, derived primarily from experiments in mammals, followed by a description of the zebrafish as a novel experimental model for studies of intrinsic SAN regulation. This mini-review is followed by an original investigation of the response of the zebrafish isolated SAN to controlled stretch. Stretch causes an immediate and continuous increase in beating rate in the zebrafish isolated SAN. This increase reaches a maximum part way through a period of sustained stretch, with the total change dependent on the magnitude and direction of stretch. This is comparable to what occurs in isolated SAN from most mammals (including human), suggesting that the zebrafish is a novel experimental model for the study of mechanisms involved in the intrinsic regulation of SAN function by mechanical effects.
Collapse
Affiliation(s)
- Eilidh A MacDonald
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Matthew R Stoyek
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Robert A Rose
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada; School of Biomedical Engineering, Dalhousie University, Halifax, Canada.
| |
Collapse
|
23
|
Park SW, Shin KC, Park HJ, Yoou SK, Park JY, Kang YS, Sung DJ, Kim JG, Park SH, Kim B, Cho H, Bae YM. Caveolar remodeling is a critical mechanotransduction mechanism of the stretch-induced L-type Ca 2+ channel activation in vascular myocytes. Pflugers Arch 2017; 469:829-842. [PMID: 28303387 DOI: 10.1007/s00424-017-1957-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 01/26/2017] [Accepted: 02/13/2017] [Indexed: 12/11/2022]
Abstract
Activation of L-type voltage-dependent Ca2+ channels (VDCCL) by membrane stretch contributes to many biological responses such as myogenic contraction of arteries. However, mechanism for the stretch-induced VDCCL activation is unclear. In this study, we examined the hypothesis that caveolar remodeling and its related signaling cascade contribute to the stretch-induced activation of VDCCL in rat mesenteric arterial smooth muscle cells. The VDCCL currents were recorded with nystatin-perforated or with conventional whole-cell patch-clamp technique. Hypotonic (~230 mOsm) swelling-induced membrane stretch reversibly increased the VDCCL currents. Electron microscope and confocal imaging analysis revealed that both hypotonic swelling and cholesterol depletion by methyl-β-cychlodextrin (MβCD) similarly disrupted the caveolae structure and translocated caveolin-1 (Cav-1) from membrane to cytosolic space. Accordingly, MβCD also increased VDCCL currents. Moreover, subsequent hypotonic swelling after MβCD treatment failed to increase the VDCCL currents further. Western blotting experiments revealed that hypotonic swelling phosphorylated Cav-1 and JNK. Inhibitors of tyrosine kinases (genistein) and JNK (SP00125) prevented the swelling-induced facilitation of VDCCL currents. Knockdown of Cav-1 by small interfering RNA blocked both the VDCCL current facilitation by stretch and the related phosphorylation of JNK. Taken together, the results suggest that membrane stretch is transduced to the facilitation of VDCCL currents via caveolar structure-dependent tyrosine phosphorylation of Cav-1 and subsequent activation of JNK in rat mesenteric arterial myocytes.
Collapse
Affiliation(s)
- Sang Woong Park
- Department of Emergency Medical Services, Eulji University, Seongnam, Gyeonggi-do, 461-713, South Korea
| | - Kyung Chul Shin
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, Chungbuk, 380-701, South Korea
| | - Hyun Ji Park
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, Chungbuk, 380-701, South Korea
| | - Soon-Kyu Yoou
- Department of Emergency Medical Services, Eulji University, Seongnam, Gyeonggi-do, 461-713, South Korea
| | - Jin-Yeon Park
- Colleage of Veterinary Medicine, Department of Biomedical Science & Technology (DBST), Konkuk University, Seoul, 143-701, South Korea
| | - Young-Sun Kang
- Colleage of Veterinary Medicine, Department of Biomedical Science & Technology (DBST), Konkuk University, Seoul, 143-701, South Korea
| | - Dong Jun Sung
- Division of Sport Science, College of Science and Technology, Konkuk University, Chungju, Chungbuk, 380-701, South Korea
| | - Jae Gon Kim
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, Chungbuk, 380-701, South Korea
| | - Seung Hwa Park
- Department of Anatomy, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, Chungbuk, 380-701, South Korea
| | - BoKyung Kim
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, Chungbuk, 380-701, South Korea
| | - Hana Cho
- Department of Physiology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Young Min Bae
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, Chungbuk, 380-701, South Korea.
| |
Collapse
|
24
|
Alcaino C, Farrugia G, Beyder A. Mechanosensitive Piezo Channels in the Gastrointestinal Tract. CURRENT TOPICS IN MEMBRANES 2017; 79:219-244. [PMID: 28728818 PMCID: PMC5606247 DOI: 10.1016/bs.ctm.2016.11.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Sensation of mechanical forces is critical for normal function of the gastrointestinal (GI) tract and abnormalities in mechanosensation are linked to GI pathologies. In the GI tract there are several mechanosensitive cell types-epithelial enterochromaffin cells, intrinsic and extrinsic enteric neurons, smooth muscle cells and interstitial cells of Cajal. These cells use mechanosensitive ion channels that respond to mechanical forces by altering transmembrane ionic currents in a process called mechanoelectrical coupling. Several mechanosensitive ionic conductances have been identified in the mechanosensory GI cells, ranging from mechanosensitive voltage-gated sodium and calcium channels to the mechanogated ion channels, such as the two-pore domain potassium channels K2P (TREK-1) and nonselective cation channels from the transient receptor potential family. The recently discovered Piezo channels are increasingly recognized as significant contributors to cellular mechanosensitivity. Piezo1 and Piezo2 are nonselective cationic ion channels that are directly activated by mechanical forces and have well-defined biophysical and pharmacologic properties. The role of Piezo channels in the GI epithelium is currently under investigation and their role in the smooth muscle syncytium and enteric neurons is still not known. In this review, we outline the current state of knowledge on mechanosensitive ion channels in the GI tract, with a focus on the known and potential functions of the Piezo channels.
Collapse
Affiliation(s)
- C Alcaino
- Mayo Clinic College of Medicine, Rochester, MN, United States
| | - G Farrugia
- Mayo Clinic College of Medicine, Rochester, MN, United States
| | - A Beyder
- Mayo Clinic College of Medicine, Rochester, MN, United States
| |
Collapse
|
25
|
Jensen LJ, Nielsen MS, Salomonsson M, Sørensen CM. T-type Ca 2+ channels and autoregulation of local blood flow. Channels (Austin) 2017; 11:183-195. [PMID: 28055302 DOI: 10.1080/19336950.2016.1273997] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
L-type voltage gated Ca2+ channels are considered to be the primary source of calcium influx during the myogenic response. However, many vascular beds also express T-type voltage gated Ca2+ channels. Recent studies suggest that these channels may also play a role in autoregulation. At low pressures (40-80 mmHg) T-type channels affect myogenic responses in cerebral and mesenteric vascular beds. T-type channels also seem to be involved in skeletal muscle autoregulation. This review discusses the expression and role of T-type voltage gated Ca2+ channels in the autoregulation of several different vascular beds. Lack of specific pharmacological inhibitors has been a huge challenge in the field. Now the research has been strengthened by genetically modified models such as mice lacking expression of T-type voltage gated Ca2+ channels (CaV3.1 and CaV3.2). Hopefully, these new tools will help further elucidate the role of voltage gated T-type Ca2+ channels in autoregulation and vascular function.
Collapse
Affiliation(s)
- Lars Jørn Jensen
- a Departments of Veterinary Clinical and Animal Sciences , University of Copenhagen , Copenhagen , Denmark
| | - Morten Schak Nielsen
- b Department of Biomedical Sciences, Faculty of Health and Medical Sciences , University of Copenhagen , Copenhagen , Denmark
| | - Max Salomonsson
- b Department of Biomedical Sciences, Faculty of Health and Medical Sciences , University of Copenhagen , Copenhagen , Denmark
| | - Charlotte Mehlin Sørensen
- b Department of Biomedical Sciences, Faculty of Health and Medical Sciences , University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
26
|
Arterial Myogenic Activation through Smooth Muscle Filamin A. Cell Rep 2016; 14:2050-2058. [PMID: 26923587 DOI: 10.1016/j.celrep.2016.02.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 01/11/2016] [Accepted: 01/28/2016] [Indexed: 11/24/2022] Open
Abstract
Mutations in the filamin A (FlnA) gene are frequently associated with severe arterial abnormalities, although the physiological role for this cytoskeletal element remains poorly understood in vascular cells. We used a conditional mouse model to selectively delete FlnA in smooth muscle (sm) cells at the adult stage, thus avoiding the developmental effects of the knockout. Basal blood pressure was significantly reduced in conscious smFlnA knockout mice. Remarkably, pressure-dependent tone of the resistance caudal artery was lost, whereas reactivity to vasoconstrictors was preserved. Impairment of the myogenic behavior was correlated with a lack of calcium influx in arterial myocytes upon an increase in intraluminal pressure. Notably, the stretch activation of CaV1.2 was blunted in the absence of smFlnA. In conclusion, FlnA is a critical upstream element of the signaling cascade underlying the myogenic tone. These findings allow a better understanding of the molecular basis of arterial autoregulation and associated disease states.
Collapse
|
27
|
Latrunculin B modulates electrophysiological characteristics and arrhythmogenesis in pulmonary vein cardiomyocytes. Clin Sci (Lond) 2016; 130:721-32. [PMID: 26839418 DOI: 10.1042/cs20150593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 02/02/2016] [Indexed: 01/28/2023]
Abstract
AF (atrial fibrillation) is the most common sustained arrhythmia, and the PVs (pulmonary veins) play a critical role in triggering AF. Stretch causes structural remodelling, including cytoskeleton rearrangement, which may play a role in the genesis of AF. Lat-B (latrunculin B), an inhibitor of actin polymerization, is involved in Ca(2+) regulation. However, it is unclear whether Lat-B directly modulates the electrophysiological characteristics and Ca(2+) homoeostasis of the PVs. Conventional microelectrodes, whole-cell patch-clamp, and the fluo-3 fluorimetric ratio technique were used to record ionic currents and intracellular Ca(2+) within isolated rabbit PV preparations, or within isolated single PV cardiomyocytes, before and after administration of Lat-B (100 nM). Langendorff-perfused rabbit hearts were exposed to acute and continuous atrial stretch, and we studied PV electrical activity. Lat-B (100 nM) decreased the spontaneous electrical activity by 16±4% in PV preparations. Lat-B (100 nM) decreased the late Na(+) current, L-type Ca(2+) current, Na(+)/Ca(2+) exchanger current, and stretch-activated BKCa current, but did not affect the Na(+) current in PV cardiomyocytes. Lat-B reduced the transient outward K(+) current and ultra-rapid delayed rectifier K(+) current, but increased the delayed rectifier K(+) current in isolated PV cardiomyocytes. In addition, Lat-B (100 nM) decreased intracellular Ca(2+) transient and sarcoplasmic reticulum Ca(2+) content in PV cardiomyocytes. Moreover, Lat-B attenuated stretch-induced increased spontaneous electrical activity and trigger activity. The effects of Lat-B on the PV spontaneous electrical activity were attenuated in the presence of Y-27632 [10 μM, a ROCK (Rho-associated kinase) inhibitor] and cytochalasin D (10 μM, an actin polymerization inhibitor). In conclusion, Lat-B regulates PV electrophysiological characteristics and attenuates stretch-induced arrhythmogenesis.
Collapse
|
28
|
Vincent PF, Bouleau Y, Petit C, Dulon D. A synaptic F-actin network controls otoferlin-dependent exocytosis in auditory inner hair cells. eLife 2015; 4. [PMID: 26568308 PMCID: PMC4714970 DOI: 10.7554/elife.10988] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/12/2015] [Indexed: 12/04/2022] Open
Abstract
We show that a cage-shaped F-actin network is essential for maintaining a tight spatial organization of Cav1.3 Ca2+ channels at the synaptic ribbons of auditory inner hair cells. This F-actin network is also found to provide mechanosensitivity to the Cav1.3 channels when varying intracellular hydrostatic pressure. Furthermore, this F-actin mesh network attached to the synaptic ribbons directly influences the efficiency of otoferlin-dependent exocytosis and its sensitivity to intracellular hydrostatic pressure, independently of its action on the Cav1.3 channels. We propose a new mechanistic model for vesicle exocytosis in auditory hair cells where the rate of vesicle recruitment to the ribbons is directly controlled by a synaptic F-actin network and changes in intracellular hydrostatic pressure. DOI:http://dx.doi.org/10.7554/eLife.10988.001 To hear a sound, the pressure produced by sound waves must be converted into an electrical nerve signal. The cells inside the ear that perform this transformation are called hair cells, which are so named because they have hundreds of hair-like structures on their upper surface. Pressure from sound waves causes movements in the inner ear that bend these ‘hairs’. This causes the hair cells to release chemical signals to neighboring nerve cell terminals that ultimately transmit information about the sound to the brain. The chemical signals are stored inside the hair cells in bubble-like compartments called vesicles. To release the chemicals from the cell, the vesicles merge with the membrane that surrounds the hair cell. Most cells that communicate in this way are limited in how long they can transmit such messages. However, hair cells can continuously fuse vesicles to the membrane even when a sound lasts for a long time. This suggests that the hair cells have a different way of producing vesicles and getting them to the membrane than other cell types. Inside the hair cells, vesicles are stored in regions called active zones. Each active zone contains a “ribbon” (attached to which are hundreds of vesicles) and also ion channels that allow calcium ions to flow into the cell. (An increase in calcium ion concentration inside the cell is necessary for the vesicle to fuse with the cell membrane and so release its chemical content). Now, Vincent et al. show that in hair cells, a cage-like network made from a protein called actin surrounds each active zone. This network helps to position the calcium ion channels. Treating the hair cells with a compound that disorganized the actin networks speed up the process of vesicle movement, which suggests that the actin network also controls the rate at which vesicles reach the membrane. Next, it will be important to identify how the actin network interacts with other molecules that help vesicles to release their contents; in particular a protein called otoferlin, which is thought to act as a calcium ion sensor. DOI:http://dx.doi.org/10.7554/eLife.10988.002
Collapse
Affiliation(s)
- Philippe Fy Vincent
- Bordeaux Neurocampus, Equipe Neurophysiologie de la Synapse Auditive, Université de Bordeaux, Bordeaux, France
| | - Yohan Bouleau
- Bordeaux Neurocampus, Equipe Neurophysiologie de la Synapse Auditive, Université de Bordeaux, Bordeaux, France
| | - Christine Petit
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Sorbonne Universités, UPMC Université Paris, Paris, France.,Syndrome de Usher et Autres Atteintes Rétino-Cochléaires, Institut de la Vision, Paris, France.,Collège de France, Paris, France
| | - Didier Dulon
- Bordeaux Neurocampus, Equipe Neurophysiologie de la Synapse Auditive, Université de Bordeaux, Bordeaux, France.,UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| |
Collapse
|
29
|
Neshatian L, Strege PR, Rhee PL, Kraichely RE, Mazzone A, Bernard CE, Cima RR, Larson DW, Dozois EJ, Kline CF, Mohler PJ, Beyder A, Farrugia G. Ranolazine inhibits voltage-gated mechanosensitive sodium channels in human colon circular smooth muscle cells. Am J Physiol Gastrointest Liver Physiol 2015; 309:G506-12. [PMID: 26185330 PMCID: PMC4572410 DOI: 10.1152/ajpgi.00051.2015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 07/09/2015] [Indexed: 01/31/2023]
Abstract
Human jejunum smooth muscle cells (SMCs) and interstitial cells of Cajal (ICCs) express the SCN5A-encoded voltage-gated, mechanosensitive sodium channel NaV1.5. NaV1.5 contributes to small bowel excitability, and NaV1.5 inhibitor ranolazine produces constipation by an unknown mechanism. We aimed to determine the presence and molecular identity of Na(+) current in the human colon smooth muscle and to examine the effects of ranolazine on Na(+) current, mechanosensitivity, and smooth muscle contractility. Inward currents were recorded by whole cell voltage clamp from freshly dissociated human colon SMCs at rest and with shear stress. SCN5A mRNA and NaV1.5 protein were examined by RT-PCR and Western blots, respectively. Ascending human colon strip contractility was examined in a muscle bath preparation. SCN5A mRNA and NaV1.5 protein were identified in human colon circular muscle. Freshly dissociated human colon SMCs had Na(+) currents (-1.36 ± 0.36 pA/pF), shear stress increased Na(+) peaks by 17.8 ± 1.8% and accelerated the time to peak activation by 0.7 ± 0.3 ms. Ranolazine (50 μM) blocked peak Na(+) current by 43.2 ± 9.3% and inhibited shear sensitivity by 25.2 ± 3.2%. In human ascending colon strips, ranolazine decreased resting tension (31%), reduced the frequency of spontaneous events (68%), and decreased the response to smooth muscle electrical field stimulation (61%). In conclusion, SCN5A-encoded NaV1.5 is found in human colonic circular smooth muscle. Ranolazine blocks both peak amplitude and mechanosensitivity of Na(+) current in human colon SMCs and decreases contractility of human colon muscle strips. Our data provide a likely mechanistic explanation for constipation induced by ranolazine.
Collapse
Affiliation(s)
- Leila Neshatian
- 1Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; ,2Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Peter R. Strege
- 1Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; ,2Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Poong-Lyul Rhee
- 4Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; and
| | - Robert E. Kraichely
- 1Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; ,2Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Amelia Mazzone
- 1Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; ,2Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Cheryl E. Bernard
- 1Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; ,2Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Robert R. Cima
- 3Department of Colon and Rectal Surgery, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - David W. Larson
- 3Department of Colon and Rectal Surgery, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Eric J. Dozois
- 3Department of Colon and Rectal Surgery, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Crystal F. Kline
- 5The Dorothy M. Davis Heart and Lung Research Institute and Departments of Internal Medicine and Physiology & Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Peter J. Mohler
- 5The Dorothy M. Davis Heart and Lung Research Institute and Departments of Internal Medicine and Physiology & Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Arthur Beyder
- 1Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; ,2Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Gianrico Farrugia
- Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| |
Collapse
|
30
|
Lee A, Wang S, Williams B, Hagen J, Scheetz TE, Haeseleer F. Characterization of Cav1.4 complexes (α11.4, β2, and α2δ4) in HEK293T cells and in the retina. J Biol Chem 2014; 290:1505-21. [PMID: 25468907 DOI: 10.1074/jbc.m114.607465] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In photoreceptor synaptic terminals, voltage-gated Cav1.4 channels mediate Ca(2+) signals required for transmission of visual stimuli. Like other high voltage-activated Cav channels, Cav1.4 channels are composed of a main pore-forming Cav1.4 α1 subunit and auxiliary β and α2δ subunits. Of the four distinct classes of β and α2δ, β2 and α2δ4 are thought to co-assemble with Cav1.4 α1 subunits in photoreceptors. However, an understanding of the functional properties of this combination of Cav subunits is lacking. Here, we provide evidence that Cav1.4 α1, β2, and α2δ4 contribute to Cav1.4 channel complexes in the retina and describe their properties in electrophysiological recordings. In addition, we identified a variant of β2, named here β2X13, which, along with β2a, is present in photoreceptor terminals. Cav1.4 α1, β2, and α2δ4 were coimmunoprecipitated from lysates of transfected HEK293 cells and mouse retina and were found to interact in the outer plexiform layer of the retina containing the photoreceptor synaptic terminals, by proximity ligation assays. In whole-cell patch clamp recordings of transfected HEK293T cells, channels (Cav1.4 α1 + β2X13) containing α2δ4 exhibited weaker voltage-dependent activation than those with α2δ1. Moreover, compared with channels (Cav1.4 α1 + α2δ4) with β2a, β2X13-containing channels exhibited greater voltage-dependent inactivation. The latter effect was specific to Cav1.4 because it was not seen for Cav1.2 channels. Our results provide the first detailed functional analysis of the Cav1.4 subunits that form native photoreceptor Cav1.4 channels and indicate potential heterogeneity in these channels conferred by β2a and β2X13 variants.
Collapse
Affiliation(s)
- Amy Lee
- From the Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, and Neurology, University of Iowa, Iowa City, Iowa 52242
| | - Shiyi Wang
- From the Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, and Neurology, University of Iowa, Iowa City, Iowa 52242
| | - Brittany Williams
- From the Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, and Neurology, University of Iowa, Iowa City, Iowa 52242
| | - Jussara Hagen
- From the Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, and Neurology, University of Iowa, Iowa City, Iowa 52242
| | - Todd E Scheetz
- the Departments of Ophthalmology and Visual Sciences and Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242, and
| | - Françoise Haeseleer
- the Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195
| |
Collapse
|
31
|
Mazet B. Gastrointestinal motility and its enteric actors in mechanosensitivity: past and present. Pflugers Arch 2014; 467:191-200. [PMID: 25366494 DOI: 10.1007/s00424-014-1635-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 10/14/2014] [Accepted: 10/19/2014] [Indexed: 12/14/2022]
Abstract
Coordinated contractions of the smooth muscle layers of the gastrointestinal (GI) tract are required to produce motor patterns that ensure normal GI motility. The crucial role of the enteric nervous system (ENS), the intrinsic ganglionated network located within the GI wall, has long been recognized in the generation of the main motor patterns. However, devising an appropriate motility requires the integration of informations emanating from the lumen of the GI tract. As already found more than half a century ago, the ability of the GI tract to respond to mechanical forces such as stretch is not restricted to neuronal mechanisms. Instead, mechanosensitivity is now recognized as a property of several non-neuronal cell types, the excitability of which is probably involved in shaping the motor patterns. This brief review gives an overview on how mechanosensitivity of different cell types in the GI tract has been established and, whenever available, on what ionic conductances are involved in mechanotransduction and their potential impact on normal GI motility.
Collapse
Affiliation(s)
- Bruno Mazet
- Aix Marseille Université, CNRS, CRN2M UMR 7286, CS80011 Bd Pierre Dramard, 13344, Marseille Cedex 15, France,
| |
Collapse
|
32
|
Aviner B, Gradwohl G, Mor Aviner M, Levy S, Grossman Y. Selective modulation of cellular voltage-dependent calcium channels by hyperbaric pressure-a suggested HPNS partial mechanism. Front Cell Neurosci 2014; 8:136. [PMID: 24904281 PMCID: PMC4034351 DOI: 10.3389/fncel.2014.00136] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 04/30/2014] [Indexed: 11/30/2022] Open
Abstract
Professional deep sea divers experience motor and cognitive impairment, known as High Pressure Neurological Syndrome (HPNS), when exposed to pressures of 100 msw (1.1 MPa) and above, considered to be the result of synaptic transmission alteration. Previous studies have indicated modulation of presynaptic Ca2+ currents at high pressure. We directly measured for the first time pressure effects on the currents of voltage dependent Ca2+ channels (VDCCs) expressed in Xenopus oocytes. Pressure selectivity augmented the current in CaV1.2 and depressed it in CaV3.2 channels. Pressure application also affected the channels' kinetics, such as ƮRise, ƮDecay. Pressure modulation of VDCCs seems to play an important role in generation of HPNS signs and symptoms.
Collapse
Affiliation(s)
- Ben Aviner
- Department of Physiology and Neurobiology, Ben Gurion University of the Negev Beer Sheva, Israel
| | - Gideon Gradwohl
- Department of Physics, Jerusalem College of Technology Jerusalem, Israel
| | - Merav Mor Aviner
- Department of Physiology and Neurobiology, Ben Gurion University of the Negev Beer Sheva, Israel
| | - Shiri Levy
- Department of Physiology and Neurobiology, Ben Gurion University of the Negev Beer Sheva, Israel
| | - Yoram Grossman
- Department of Physiology and Neurobiology, Ben Gurion University of the Negev Beer Sheva, Israel
| |
Collapse
|
33
|
Takahashi K, Naruse K. Stretch-activated BK channel and heart function. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2013; 110:239-44. [PMID: 23281538 DOI: 10.1016/j.pbiomolbio.2012.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The heart is an organ that is exposed to extreme dynamic mechanical stimuli. From birth till death, the heart indefinitely repeats periodic contraction and dilation, i.e., shortening and elongation of cardiomyocytes. Mechanical stretch elicits a change in heart rate and may cause arrhythmia if it is excessive. Thus, mechanosensitivity is crucial to heart function. The molecule that is substantially involved in mechanosensitivity is a stretch-activated ion channel. Among several ion channels believed to be activated by stretch in the heart, the stretch-activated KCa (SAKCA) channel, a member of the group of large conductance (Big Potassium, BK) channels, shows a mechanosensitive (MS) response to membrane stretch. As BK channels respond to voltage and intracellular calcium concentration with large conductance, they are considered to be involved in repolarization after depolarization. Some BK channels are known to be activated by stretch and are expressed in a number of cells, including human osteoblasts and guinea pig intestinal neurons. The SAKCA channel was found to be sensitive to stretch in the chick heart. Given that the cardiomyocyte is unremittingly exposed to contraction and dilation and that it generates action potential and its contractility is modulated by intracellular calcium concentration, the SAKCA channel, which is dependent voltage and calcium, may be involved in action potential generation. It was recently reported that a BK channel is involved in the modulation of heart rate in the mouse. Further studies regarding the role of MS BK channels, including SAKCA, in the modulation of heart rate and contractility are expected.
Collapse
Affiliation(s)
- Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
| | | |
Collapse
|
34
|
Distribution of voltage-dependent and intracellular Ca2+ channels in submucosal neurons from rat distal colon. Cell Tissue Res 2013; 353:355-66. [DOI: 10.1007/s00441-013-1643-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 04/18/2013] [Indexed: 10/26/2022]
|
35
|
Won KJ, Sanders KM, Ward SM. Stretch-dependent sensitization of post-junctional neural effectors in colonic muscles. Neurogastroenterol Motil 2013; 25:e101-13. [PMID: 23279087 PMCID: PMC3552106 DOI: 10.1111/nmo.12059] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND The colon undergoes distension-induced changes in motor activity as luminal contents or feces increase wall pressure. Input from enteric motor neurons regulates this motility. Here we examined stretch-dependent responses in circular muscle strips of murine colon. METHODS Length ramps (6-31μm s(-1) ) were applied in the axis of the circular muscle layer in a controlled manner until 5 mN isometric force was reached. KEY RESULTS Length ramps produced transient membrane potential hyperpolarizations and attenuation of action potential (AP) complexes. Responses were reproducible when ramps were applied every 30 s. Stretch-dependent hyperpolarization was blocked by TTX, suggesting AP-dependent release of inhibitory neurotransmitter(s). Atropine did not potentiate stretch-induced hyperpolarizations, but increased compliance of the circular layer. N(ω)-nitro-L-arginine (L-NNA) inhibited stretch-dependent hyperpolarization and decreased muscle compliance, suggesting release of NO mediates stretch-dependent inhibition. Control membrane potential was restored by the NO donor sodium nitorprusside. Stretch-dependent hyperpolarizations were blocked by L-methionine, an inhibitor of stretch-dependent K(+) (SDK) channels in colonic muscles. Loss of interstitial cells of Cajal, elicited by Kit neutralizing antibody, also inhibited responses to stretch. In presence of L-NNA and apamin, stretch responses became excitatory and were characterized by membrane depolarization and increased AP firing. A neurokinin-1 receptor antagonist inhibited this stretch-dependent increase in excitability. CONCLUSIONS AND INFERENCES Our data show that stretch-dependent responses in colonic muscles require tonic firing of enteric inhibitory neurons, but reflex activation of neurons does not appear to be necessary. NO causes activation of SDK channels, and stretch of muscles further activates these channels, explaining the inhibitory response to stretch in colonic muscle strips.
Collapse
Affiliation(s)
- Kyung-Jong Won
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA, (775) 784-6061 or FAX (775) 784-6903,Department of Physiology, College of Medicine, Konkuk University, 322 Danwol-dong, Chungju 380-701, South Korea
| | - Kenton M. Sanders
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA, (775) 784-6061 or FAX (775) 784-6903
| | - Sean M. Ward
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA, (775) 784-6061 or FAX (775) 784-6903
| |
Collapse
|
36
|
Takahashi K, Kakimoto Y, Toda K, Naruse K. Mechanobiology in cardiac physiology and diseases. J Cell Mol Med 2013; 17:225-32. [PMID: 23441631 PMCID: PMC3822585 DOI: 10.1111/jcmm.12027] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 01/11/2013] [Indexed: 11/28/2022] Open
Abstract
Mechanosensitivity is essential for heart function just as for all other cells and organs in the body, and it is involved in both normal physiology and diseases processes of the cardiovascular system. In this review, we have outlined the relationship between mechanosensitivity and heart physiology, including the Frank-Starling law of the heart and mechanoelectric feedback. We then focused on molecules involved in mechanotransduction, particularly mechanosensitive ion channels. We have also discussed the involvement of mechanosensitivity in heart diseases, such as arrhythmias, hypertrophy and ischaemic heart disease. Finally, mechanobiology in cardiogenesis is described with regard to regenerative medicine.
Collapse
Affiliation(s)
- Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
| | | | | | | |
Collapse
|
37
|
Rosa AO, Yamaguchi N, Morad M. Mechanical regulation of native and the recombinant calcium channel. Cell Calcium 2013; 53:264-74. [PMID: 23357406 DOI: 10.1016/j.ceca.2012.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 12/25/2012] [Indexed: 11/30/2022]
Abstract
L-type calcium channels are modulated by a host of mechanisms that include voltage, calcium ions (Ca(2+) dependent inactivation and facilitation), cytosolic proteins (CAM, CAMKII, PKA, PKC, etc.), and oxygen radicals. Here we describe yet another Ca(2+) channel regulatory mechanism that is induced by pressure-flow (PF) forces of ∼25dyn/cm(2) producing 35-60% inhibition of channel current. Only brief periods (300ms) of such PF pulses were required to suppress reversibly the current. Recombinant Ca(2+) channels (α1c77/β2a/α2δ and α1c77/β1/α2δ), expressed in HEK293 cells, were similarly suppressed by PF pulses. To examine whether Ca(2+) released by PF pulses triggered from different sub-cellular compartments (SR, ER, mitochondria) underlies the inhibitory effect of PF on the channel current, pharmacological agents and ionic substitutions were employed to probe this possibility. No significant difference in effectiveness of PF pulses to suppress ICa or IBa (used to inhibit CICR) was found between control cells and those exposed to U73122 and 2-APB (PLC and IP3R pathway modulators), thapsigargin and BAPTA (SERCA2a modulator), dinitrophenol, FCCP and Ru360 (mitochondrial inhibitors), l-NAME (NOS inhibitor signaling), cAMP and Pertussis toxin (Gi protein modulator). We concluded that the rapid and reversible modulation of the Ca(2+) channel by PF pulses is independent of intracellular release of Ca(2+) and Ca(2+) dependent inactivation of the channel and may represent direct mechanical regulatory effect on the channel protein in addition to previously reported Ca(2+)-release or entry dependent mechanism.
Collapse
Affiliation(s)
- Angelo O Rosa
- Cardiac Signaling Center, University of South Carolina, Charleston, SC 29425, USA
| | | | | |
Collapse
|
38
|
Strege P, Beyder A, Bernard C, Crespo-Diaz R, Behfar A, Terzic A, Ackerman M, Farrugia G. Ranolazine inhibits shear sensitivity of endogenous Na+ current and spontaneous action potentials in HL-1 cells. Channels (Austin) 2012; 6:457-62. [PMID: 23018927 PMCID: PMC3536731 DOI: 10.4161/chan.22017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Na(V)1.5 is a mechanosensitive voltage-gated Na(+) channel encoded by the gene SCN5A, expressed in cardiac myocytes and required for phase 0 of the cardiac action potential (AP). In the cardiomyocyte, ranolazine inhibits depolarizing Na(+) current and delayed rectifier (I(Kr)) currents. Recently, ranolazine was also shown to be an inhibitor of Na(V)1.5 mechanosensitivity. Stretch also accelerates the firing frequency of the SA node, and fluid shear stress increases the beating rate of cultured cardiomyocytes in vitro. However, no cultured cell platform exists currently for examination of spontaneous electrical activity in response to mechanical stimulation. In the present study, flow of solution over atrial myocyte-derived HL-1 cultured cells was used to study shear stress mechanosensitivity of Na(+) current and spontaneous, endogenous rhythmic action potentials. In voltage-clamped HL-1 cells, bath flow increased peak Na(+) current by 14 ± 5%. In current-clamped cells, bath flow increased the frequency and decay rate of AP by 27 ± 12% and 18 ± 4%, respectively. Ranolazine blocked both responses to shear stress. This study suggests that cultured HL-1 cells are a viable in vitro model for detailed study of the effects of mechanical stimulation on spontaneous cardiac action potentials. Inhibition of the frequency and decay rate of action potentials in HL-1 cells are potential mechanisms behind the antiarrhythmic effect of ranolazine.
Collapse
Affiliation(s)
- Peter Strege
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Quinn TA, Kohl P. Mechano-sensitivity of cardiac pacemaker function: pathophysiological relevance, experimental implications, and conceptual integration with other mechanisms of rhythmicity. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 110:257-68. [PMID: 23046620 PMCID: PMC3526794 DOI: 10.1016/j.pbiomolbio.2012.08.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 08/09/2012] [Indexed: 12/11/2022]
Abstract
Cardiac pacemaker cells exhibit spontaneous, rhythmic electrical excitation, termed automaticity. This automatic initiation of action potentials requires spontaneous diastolic depolarisation, whose rate determines normal rhythm generation in the heart. Pacemaker mechanisms have been split recently into: (i) cyclic changes in trans-sarcolemmal ion flows (termed the ‘membrane-clock’), and (ii) rhythmic intracellular calcium cycling (the ‘calcium-clock’). These two ‘clocks’ undoubtedly interact, as trans-sarcolemmal currents involved in pacemaking include calcium-carrying mechanisms, while intracellular calcium cycling requires trans-sarcolemmal ion flux as the mechanism by which it affects membrane potential. The split into separate ‘clocks’ is, therefore, somewhat arbitrary. Nonetheless, the ‘clock’ metaphor has been conceptually stimulating, in particular since there is evidence to support the view that either ‘clock’ could be sufficient in principle to set the rate of pacemaker activation. Of course, the same has also been shown for sub-sets of ‘membrane-clock’ ion currents, illustrating the redundancy of mechanisms involved in maintaining such basic functionality as the heartbeat, a theme that is common for vital physiological systems. Following the conceptual path of identifying individual groups of sub-mechanisms, it is important to remember that the heart is able to adapt pacemaker rate to changes in haemodynamic load, even after isolation or transplantation, and on a beat-by-beat basis. Neither the ‘membrane-’ nor the ‘calcium-clock’ do, as such, inherently account for this rapid adaptation to circulatory demand (cellular Ca2+ balance changes over multiple beats, while variation of sarcolemmal ion channel presence takes even longer). This suggests that a third set of mechanisms must be involved in setting the pace. These mechanisms are characterised by their sensitivity to the cyclically changing mechanical environment, and – in analogy to the above terminology – this might be considered a ‘mechanics-clock’. In this review, we discuss possible roles of mechano-sensitive mechanisms for the entrainment of membrane current dynamics and calcium-handling. This can occur directly via stretch-activation of mechano-sensitive ion channels in the sarcolemma and/or in intracellular membrane compartments, as well as by modulation of ‘standard’ components of the ‘membrane-’ or ‘calcium-clock’. Together, these mechanisms allow rapid adaptation to changes in haemodynamic load, on a beat-by-beat basis. Additional relevance arises from the fact that mechano-sensitivity of pacemaking may help to explain pacemaker dysfunction in mechanically over- or under-loaded tissue. As the combined contributions of the various underlying oscillatory mechanisms are integrated at the pacemaker cell level into a single output – a train of pacemaker action potentials – we will not adhere to a metaphor that implies separate time-keeping units (‘clocks’), and rather focus on cardiac pacemaking as the result of interactions of a set of coupled oscillators, whose individual contributions vary depending on the pathophysiological context. We conclude by considering the utility and limitations of viewing the pacemaker as a coupled system of voltage-, calcium-, and mechanics-modulated oscillators that, by integrating a multitude of inputs, offers the high level of functional redundancy that is vitally important for cardiac automaticity.
Collapse
Affiliation(s)
- T Alexander Quinn
- National Heart and Lung Institute, Imperial College London, London, UK.
| | | |
Collapse
|
40
|
Poh YC, Corrias A, Cheng N, Buist ML. A quantitative model of human jejunal smooth muscle cell electrophysiology. PLoS One 2012; 7:e42385. [PMID: 22912702 PMCID: PMC3422293 DOI: 10.1371/journal.pone.0042385] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 07/04/2012] [Indexed: 11/19/2022] Open
Abstract
Recently, a number of ion channel mutations have been identified in the smooth muscle cells of the human jejunum. Although these are potentially significant in understanding diseases that are currently of unknown etiology, no suitable computational cell model exists to evaluate the effects of such mutations. Here, therefore, a biophysically based single cell model of human jejunal smooth muscle electrophysiology is presented. The resulting cellular description is able to reproduce experimentally recorded slow wave activity and produces realistic responses to a number of perturbations, providing a solid platform on which the causes of intestinal myopathies can be investigated.
Collapse
Affiliation(s)
- Yong Cheng Poh
- Department of Bioengineering, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Centre for Life Sciences (CeLS), Singapore, Singapore
| | - Alberto Corrias
- Department of Bioengineering, National University of Singapore, Singapore, Singapore
| | - Nicholas Cheng
- Department of Bioengineering, National University of Singapore, Singapore, Singapore
| | - Martin Lindsay Buist
- Department of Bioengineering, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Centre for Life Sciences (CeLS), Singapore, Singapore
| |
Collapse
|
41
|
Beyder A, Farrugia G. Targeting ion channels for the treatment of gastrointestinal motility disorders. Therap Adv Gastroenterol 2012; 5:5-21. [PMID: 22282704 PMCID: PMC3263980 DOI: 10.1177/1756283x11415892] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gastrointestinal (GI) functional and motility disorders are highly prevalent and responsible for long-term morbidity and sometimes mortality in the affected patients. It is estimated that one in three persons has a GI functional or motility disorder. However, diagnosis and treatment of these widespread conditions remains challenging. This partly stems from the multisystem pathophysiology, including processing abnormalities in the central and peripheral (enteric) nervous systems and motor dysfunction in the GI wall. Interstitial cells of Cajal (ICCs) are central to the generation and propagation of the cyclical electrical activity and smooth muscle cells (SMCs) are responsible for electromechanical coupling. In these and other excitable cells voltage-sensitive ion channels (VSICs) are the main molecular units that generate and regulate electrical activity. Thus, VSICs are potential targets for intervention in GI motility disorders. Research in this area has flourished with advances in the experimental methods in molecular and structural biology and electrophysiology. However, our understanding of the molecular mechanisms responsible for the complex and variable electrical behavior of ICCs and SMCs remains incomplete. In this review, we focus on the slow waves and action potentials in ICCs and SMCs. We describe the constituent VSICs, which include voltage-gated sodium (Na(V)), calcium (Ca(V)), potassium (K(V), K(Ca)), chloride (Cl(-)) and nonselective ion channels (transient receptor potentials [TRPs]). VSICs have significant structural homology and common functional mechanisms. We outline the approaches and limitations and provide examples of targeting VSICs at the pores, voltage sensors and alternatively spliced sites. Rational drug design can come from an integrated view of the structure and mechanisms of gating and activation by voltage or mechanical stress.
Collapse
Affiliation(s)
- Arthur Beyder
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
42
|
Lees-Green R, Du P, O'Grady G, Beyder A, Farrugia G, Pullan AJ. Biophysically based modeling of the interstitial cells of cajal: current status and future perspectives. Front Physiol 2011; 2:29. [PMID: 21772822 PMCID: PMC3131535 DOI: 10.3389/fphys.2011.00029] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 06/13/2011] [Indexed: 12/29/2022] Open
Abstract
Gastrointestinal motility research is progressing rapidly, leading to significant advances in the last 15 years in understanding the cellular mechanisms underlying motility, following the discovery of the central role played by the interstitial cells of Cajal (ICC). As experimental knowledge of ICC physiology has expanded, biophysically based modeling has become a valuable tool for integrating experimental data, for testing hypotheses on ICC pacemaker mechanisms, and for applications in in silico studies including in multiscale models. This review is focused on the cellular electrophysiology of ICC. Recent evidence from both experimental and modeling domains have called aspects of the existing pacemaker theories into question. Therefore, current experimental knowledge of ICC pacemaker mechanisms is examined in depth, and current theories of ICC pacemaking are evaluated and further developed. Existing biophysically based ICC models and their physiological foundations are then critiqued in light of the recent advances in experimental knowledge, and opportunities to improve these models are identified. The review concludes by examining several potential clinical applications of biophysically based ICC modeling from the subcellular through to the organ level, including ion channelopathies and ICC network degradation.
Collapse
Affiliation(s)
- Rachel Lees-Green
- Auckland Bioengineering Institute, The University of Auckland Auckland, New Zealand
| | | | | | | | | | | |
Collapse
|
43
|
Strege PR, Bernard CE, Kraichely RE, Mazzone A, Sha L, Beyder A, Gibbons SJ, Linden DR, Kendrick ML, Sarr MG, Szurszewski JH, Farrugia G. Hydrogen sulfide is a partially redox-independent activator of the human jejunum Na+ channel, Nav1.5. Am J Physiol Gastrointest Liver Physiol 2011; 300:G1105-14. [PMID: 21393430 PMCID: PMC3119119 DOI: 10.1152/ajpgi.00556.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hydrogen sulfide (H(2)S) is produced endogenously by L-cysteine metabolism. H(2)S modulates several ion channels with an unclear mechanism of action. A possible mechanism is through reduction-oxidation reactions attributable to the redox potential of the sulfur moiety. The aims of this study were to determine the effects of the H(2)S donor NaHS on Na(V)1.5, a voltage-dependent sodium channel expressed in the gastrointestinal tract in human jejunum smooth muscle cells and interstitial cells of Cajal, and to elucidate whether H(2)S acts on Na(V)1.5 by redox reactions. Whole cell Na(+) currents were recorded in freshly dissociated human jejunum circular myocytes and Na(V)1.5-transfected human embryonic kidney-293 cells. RT-PCR amplified mRNA for H(2)S enzymes cystathionine β-synthase and cystathionine γ-lyase from the human jejunum. NaHS increased native Na(+) peak currents and shifted the half-point (V(1/2)) of steady-state activation and inactivation by +21 ± 2 mV and +15 ± 3 mV, respectively. Similar effects were seen on the heterologously expressed Na(V)1.5 α subunit with EC(50)s in the 10(-4) to 10(-3) M range. The reducing agent dithiothreitol (DTT) mimicked in part the effects of NaHS by increasing peak current and positively shifting steady-state activation. DTT together with NaHS had an additive effect on steady-state activation but not on peak current, suggesting that the latter may be altered via reduction. Pretreatment with the Hg(2+)-conjugated oxidizer thimerosal or the alkylating agent N-ethylmaleimide inhibited or decreased NaHS induction of Na(V)1.5 peak current. These studies show that H(2)S activates the gastrointestinal Na(+) channel, and the mechanism of action of H(2)S is partially redox independent.
Collapse
Affiliation(s)
- Peter R. Strege
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Cheryl E. Bernard
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Robert E. Kraichely
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Amelia Mazzone
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Lei Sha
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Arthur Beyder
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Simon J. Gibbons
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - David R. Linden
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Michael L. Kendrick
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Michael G. Sarr
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Joseph H. Szurszewski
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
44
|
|
45
|
Abstract
BACKGROUND Gastrointestinal inflammation significantly affects the electrical excitability of smooth muscle cells. Considerable progress over the last few years have been made to establish the mechanisms by which ion channel function is altered in the setting of gastrointestinal inflammation. Details have begun to emerge on the molecular basis by which ion channel function may be regulated in smooth muscle following inflammation. These include changes in protein and gene expression of the smooth muscle isoform of L-type Ca(2+) channels and ATP-sensitive K(+) channels. Recent attention has also focused on post-translational modifications as a primary means of altering ion channel function in the absence of changes in protein/gene expression. Protein phosphorylation of serine/theronine or tyrosine residues, cysteine thiol modifications, and tyrosine nitration are potential mechanisms affected by oxidative/nitrosative stress that alter the gating kinetics of ion channels. Collectively, these findings suggest that inflammation results in electrical remodeling of smooth muscle cells in addition to structural remodeling. PURPOSE The purpose of this review is to synthesize our current understanding regarding molecular mechanisms that result in altered ion channel function during gastrointestinal inflammation and to address potential areas that can lead to targeted new therapies.
Collapse
Affiliation(s)
- H I Akbarali
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, VCU Program in Enteric Neuromuscular Sciences (VPENS), Virginia Commonwealth University, Richmond, VA 23298, USA.
| | | | | | | |
Collapse
|
46
|
Gil V, Gallego D, Grasa L, Martín MT, Jiménez M. Purinergic and nitrergic neuromuscular transmission mediates spontaneous neuronal activity in the rat colon. Am J Physiol Gastrointest Liver Physiol 2010; 299:G158-69. [PMID: 20395536 DOI: 10.1152/ajpgi.00448.2009] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nitric oxide (NO) and ATP mediate smooth muscle relaxation in the gastrointestinal tract. However, the involvement of these neurotransmitters in spontaneous neuronal activity is unknown. The aim of the present work was to study spontaneous neuromuscular transmission in the rat midcolon. Microelectrode experiments were performed under constant stretch both in circular and longitudinal directions. Spontaneous inhibitory junction potentials (sIJP) were recorded. Tetrodotoxin (1 microM) and apamin (1 microM) depolarized smooth muscle cells and inhibited sIJP. N(omega)-nitro-l-arginine (l-NNA, 1 mM) depolarized smooth muscle cells but did not modify sIJP. In contrast, the P2Y(1) antagonist MRS-2500 (1 microM) did not modify the resting membrane potential (RMP) but reduced sIJP (IC(50) = 3.1 nM). Hexamethonium (200 microM), NF-023 (10 microM), and ondansetron (1 microM) did not modify RMP and sIJP. These results correlate with in vitro (muscle bath) and in vivo (strain gauges) data where l-NNA but not MRS-2500 induced a sustained increase of spontaneous motility. We concluded that, in the rat colon, inhibitory neurons regulate smooth muscle RMP and cause sIJP. In vitro, the release of inhibitory neurotransmitters is independent of nicotinic, P2X, and 5-hydroxytryptamine type 3 receptors. Neuronal NO causes a sustained smooth muscle hyperpolarization that is responsible for a constant inhibition of spontaneous motility. In contrast, ATP acting on P2Y(1) receptors is responsible for sIJP but does not mediate inhibitory neural tone. ATP and NO have complementary physiological functions in the regulation of gastrointestinal motility.
Collapse
Affiliation(s)
- Víctor Gil
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | | | | | | | | |
Collapse
|
47
|
Gallego D, Aulí M, Aleu J, Martínez E, Rofes L, Martí-Ragué J, Jiménez M, Clavé P. Effect of otilonium bromide on contractile patterns in the human sigmoid colon. Neurogastroenterol Motil 2010; 22:e180-91. [PMID: 20367799 DOI: 10.1111/j.1365-2982.2010.01495.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The mechanism of action of the spasmolytic compound otilonium bromide (OB) on human colonic motility is not understood. The aim of our study was to characterize the pharmacological effects of OB on contractile patterns in the human sigmoid colon. METHODS Circular sigmoid strips were studied in organ baths. Isolated smooth muscle cells from human sigmoid colon were examined using the calcium imaging technique. KEY RESULTS Otilonium bromide inhibited by 85% spontaneous non-neural rhythmic phasic contractions (RPCs), (IC(50) = 49.9 nmol L(-1)) and stretch-induced tone (IC(50) = 10.7 nmol L(-1)) with maximum effects at micromolar range. OB also inhibited by 50% both on- (IC(50) = 38.0 nmol L(-1)) and off-contractions induced by electrical stimulation of excitatory motor neurons. In contrast, the inhibitory latency period prior to off-contractions was unaffected by OB. OB inhibited acetylcholine-, substance P-, and neurokinin A-induced contractions. The L-type Ca(2+) channel agonist BayK8644 reversed the effects of OB on RPCs, on- and off-contractions. Hexamethonium, atropine, the NK(2) antagonist, or depletion of intracellular Ca(2+) stores by thapsigargin did not prevent the inhibitory effect of OB on RPCs and electrical contractions. KCl-induced calcium transients in isolated smooth muscle cells were also inhibited by OB (IC(50) = 0.2 micromol L(-1)). CONCLUSIONS & INFERENCES Otilonium bromide strongly inhibited the main patterns of human sigmoid motility in vitro by blocking calcium influx through L-type calcium channels on smooth muscle cells. This pharmacological profile may mediate the clinically observed effects of the drug in patients with irritable bowel syndrome.
Collapse
Affiliation(s)
- D Gallego
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Instituto de Salud Carlos III, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Ben-Zeev G, Telias M, Nussinovitch I. Lysophospholipids modulate voltage-gated calcium channel currents in pituitary cells; effects of lipid stress. Cell Calcium 2010; 47:514-24. [PMID: 20510448 DOI: 10.1016/j.ceca.2010.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Revised: 04/28/2010] [Accepted: 04/28/2010] [Indexed: 01/26/2023]
Abstract
Voltage-gated calcium channels (VGCCs) are osmosensitive. The hypothesis that this property of VGCCs stems from their susceptibility to alterations in the mechanical properties of the bilayer was tested on VGCCs in pituitary cells using cone-shaped lysophospholipids (LPLs) to perturb bilayer lipid stress. LPLs of different head group size and charge were used: lysophosphatidylcholine (LPC), lysophosphatidylinositol (LPI), lysophosphatidylserine (LPS) and lysophosphatidylethanolamine (LPE). Phosphatidylcholine (PC) and LPC (C6:0) were used as controls. We show that partition of both LPC and LPI into the membrane of pituitary cells suppressed L-type calcium channel currents (I(L)). This suppression of I(L) was slow in onset, reversible upon washout with BSA and associated with a depolarizing shift in activation ( approximately 8mV). In contrast to these effects of LPC and LPI on I(L), LPS, LPE, PC and LPC (C6:0) exerted minimal or insignificant effects. This difference may be attributed to the prominent conical shape of LPC and LPI compared to the shapes of LPS and LPE (which have smaller headgroups), and to PC (which is cylindrical). The similar effects of LPC and LPI on I(L), despite differences in the structure and charge of their headgroups suggest a common lipid stress dependent mechanism in their action on VGCCs.
Collapse
Affiliation(s)
- Galia Ben-Zeev
- Department of Medical Neurobiology, Institute for Medical Research-Israel-Canada, Jerusalem, Israel
| | | | | |
Collapse
|
49
|
Iribe G, Jin H, Kaihara K, Naruse K. Effects of axial stretch on sarcolemmal BKCachannels in post-hatch chick ventricular myocytes. Exp Physiol 2010; 95:699-711. [DOI: 10.1113/expphysiol.2009.051896] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
50
|
Simon A, Shenton F, Hunter I, Banks RW, Bewick GS. Amiloride-sensitive channels are a major contributor to mechanotransduction in mammalian muscle spindles. J Physiol 2009; 588:171-85. [PMID: 19917568 DOI: 10.1113/jphysiol.2009.182683] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We investigated whether channels of the epithelial sodium/amiloride-sensitive degenerin (ENaC/DEG) family are a major contributor to mechanosensory transduction in primary mechanosensory afferents, using adult rat muscle spindles as a model system. Stretch-evoked afferent discharge was reduced in a dose-dependent manner by amiloride and three analogues - benzamil, 5-(N-ethyl-N-isopropyl) amiloride (EIPA) and hexamethyleneamiloride (HMA), reaching > or = 85% inhibition at 1 mm. Moreover, firing was slightly but significantly increased by ENaC delta subunit agonists (icilin and capsazepine). HMA's profile of effects was distinct from that of the other drugs. Amiloride, benzamil and EIPA significantly decreased firing (P < 0.01 each) at 1 microm, while 10 microm HMA was required for highly significant inhibition (P < 0.0001). Conversely, amiloride, benzamil and EIPA rarely blocked firing entirely at 1 mm, whereas 1 mm HMA blocked 12 of 16 preparations. This pharmacology suggests low-affinity ENaCs are the important spindle mechanotransducer. In agreement with this, immunoreactivity to ENaC alpha, beta and gamma subunits was detected both by Western blot and immunocytochemistry. Immunofluorescence intensity ratios for ENaC alpha, beta or gamma relative to the vesicle marker synaptophysin in the same spindle all significantly exceeded controls (P < 0.001). Ratios for the related brain sodium channel ASIC2 (BNaC1alpha) were also highly significantly greater (P < 0.005). Analysis of confocal images showed strong colocalisation within the terminal of ENaC/ASIC2 subunits and synaptophysin. This study implicates ENaC and ASIC2 in mammalian mechanotransduction. Moreover, within the terminals they colocalise with synaptophysin, a marker for the synaptic-like vesicles which regulate afferent excitability in these mechanosensitive endings.
Collapse
Affiliation(s)
- Anna Simon
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| | | | | | | | | |
Collapse
|