1
|
Vasilev F, Ezhova Y, Chun JT. Signaling Enzymes and Ion Channels Being Modulated by the Actin Cytoskeleton at the Plasma Membrane. Int J Mol Sci 2021; 22:ijms221910366. [PMID: 34638705 PMCID: PMC8508623 DOI: 10.3390/ijms221910366] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
A cell should deal with the changing external environment or the neighboring cells. Inevitably, the cell surface receives and transduces a number of signals to produce apt responses. Typically, cell surface receptors are activated, and during this process, the subplasmalemmal actin cytoskeleton is often rearranged. An intriguing point is that some signaling enzymes and ion channels are physically associated with the actin cytoskeleton, raising the possibility that the subtle changes of the local actin cytoskeleton can, in turn, modulate the activities of these proteins. In this study, we reviewed the early and new experimental evidence supporting the notion of actin-regulated enzyme and ion channel activities in various cell types including the cells of immune response, neurons, oocytes, hepatocytes, and epithelial cells, with a special emphasis on the Ca2+ signaling pathway that depends on the synthesis of inositol 1,4,5-trisphosphate. Some of the features that are commonly found in diverse cells from a wide spectrum of the animal species suggest that fine-tuning of the activities of the enzymes and ion channels by the actin cytoskeleton may be an important strategy to inhibit or enhance the function of these signaling proteins.
Collapse
Affiliation(s)
- Filip Vasilev
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Rue St Denis, Montreal, QC H2X 0A9, Canada
- Correspondence: (F.V.); (J.T.C.); Tel.: +1-514-249-5862 (F.V.); +39-081-583-3407 (J.T.C.)
| | - Yulia Ezhova
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada;
| | - Jong Tai Chun
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
- Correspondence: (F.V.); (J.T.C.); Tel.: +1-514-249-5862 (F.V.); +39-081-583-3407 (J.T.C.)
| |
Collapse
|
2
|
Sundararaj S, Ravindran A, Casarotto MG. AHNAK: The quiet giant in calcium homeostasis. Cell Calcium 2021; 96:102403. [PMID: 33813182 DOI: 10.1016/j.ceca.2021.102403] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 10/21/2022]
Abstract
The phosphoprotein AHNAK is a large, ubiquitously expressed scaffolding protein involved in mediating a host of protein-protein interactions. This enables AHNAK to participate in various multi-protein complexes thereby orchestrating a range of diverse biological processes, including tumour suppression, immune regulation and cell architecture maintenance. A less studied but nonetheless equally important function occurs in calcium homeostasis. It does so by largely interacting with the L-type voltage-gated calcium channel (LVGCC) present in the plasma membrane of excitable cells such as muscles and neurons. Several studies have characterized the underlying basis of AHNAK's functional role in calcium channel modulation, which has led to a greater understanding of this cellular process and its associated pathologies. In this article we review and examine recent advances relating to the physiological aspects of AHNAK in calcium regulation. Specifically, we will provide a broad overview of AHNAK including its structural makeup and its interaction with several isoforms of LVGCC, and how these molecular interactions regulate calcium modulation across various tissues and their implication in muscle and neuronal function.
Collapse
Affiliation(s)
- Srinivasan Sundararaj
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra, Australia.
| | - Agin Ravindran
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Marco G Casarotto
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra, Australia.
| |
Collapse
|
3
|
Lai WF, Wong WT. Roles of the actin cytoskeleton in aging and age-associated diseases. Ageing Res Rev 2020; 58:101021. [PMID: 31968269 DOI: 10.1016/j.arr.2020.101021] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/06/2020] [Accepted: 01/17/2020] [Indexed: 12/12/2022]
Abstract
The integrity of the cytoskeleton is essential to diverse cellular processes such as phagocytosis and intracellular trafficking. Disruption of the organization and dynamics of the actin cytoskeleton leads to age-associated symptoms and diseases, ranging from cancer to neurodegeneration. In addition, changes in the integrity of the actin cytoskeleton disrupt the functioning of not only somatic and stem cells but also gametes, resulting in aberrant embryonic development. Strategies to preserve the integrity and dynamics of the cytoskeleton are, therefore, potentially therapeutic to age-related disorders. The objective of this article is to revisit the current understanding of the roles played by the actin cytoskeleton in aging, and to review the opportunities and challenges for the transition of basic research into intervention development. It is hoped that, with the snapshot of evidence regarding changes in actin dynamics with advanced age, insights into future research directions can be attained.
Collapse
Affiliation(s)
- Wing-Fu Lai
- School of Pharmaceutical Sciences, Shenzhen University, PR China; School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, PR China; Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, PR China.
| | - Wing-Tak Wong
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, PR China
| |
Collapse
|
4
|
Wenceslau CF, McCarthy CG, Szasz T, Calmasini FB, Mamenko M, Webb RC. Formyl peptide receptor-1 activation exerts a critical role for the dynamic plasticity of arteries via actin polymerization. Pharmacol Res 2019; 141:276-290. [PMID: 30639374 DOI: 10.1016/j.phrs.2019.01.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 12/20/2022]
Abstract
Several human diseases, include cancer and stroke are characterized by changes in immune system activation and vascular contractility. However, the mechanistic foundation of a vascular immuno-physiology network is still largely unknown. Formyl peptide receptor-1 (FPR-1), which plays a vital role in the function of the innate immune system, is widely expressed in arteries, but its role in vascular plasticity is unclear. We questioned why a receptor that is crucial for immune defense, and cell motility in leukocytes, would be expressed in vascular smooth muscle cells (VSMCs). We hypothesized that activation of FPR-1 in arteries is important for the temporal reorganization of actin filaments, and consequently, changes in vascular function, similar to what is observed in neutrophils. To address our hypothesis, we used FPR-1 knockout and VSMCs lacking FPR-1. We observed that FPR-1 activation induces actin polymerization in wild type VSMCs. Absence of FPR-1 in the vasculature significantly decreased vascular contraction and induced loss of myogenic tone to elevated intraluminal pressures via disruption of actin polymerization. Actin polymerization activator ameliorated these responses. In conclusion, we have established a novel role for FPR-1 in VSMC contractility and motility, similar to the one observed in sentinel cells of the innate immune system. This discovery is fundamental for vascular immuno-pathophysiology, given that FPR-1 in VSMCs not only functions as an immune system receptor, but it also has an important role for the dynamic plasticity of arteries.
Collapse
Affiliation(s)
- Camilla F Wenceslau
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA; Department of Physiology, Augusta University, Augusta, GA, USA.
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA; Department of Physiology, Augusta University, Augusta, GA, USA
| | - Theodora Szasz
- Department of Physiology, Augusta University, Augusta, GA, USA
| | | | - Mykola Mamenko
- Department of Physiology, Augusta University, Augusta, GA, USA
| | - R Clinton Webb
- Department of Physiology, Augusta University, Augusta, GA, USA
| |
Collapse
|
5
|
Soto C, Bergado G, Blanco R, Griñán T, Rodríguez H, Ros U, Pazos F, Lanio ME, Hernández AM, Álvarez C. Sticholysin II-mediated cytotoxicity involves the activation of regulated intracellular responses that anticipates cell death. Biochimie 2018; 148:18-35. [DOI: 10.1016/j.biochi.2018.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 02/07/2018] [Indexed: 12/12/2022]
|
6
|
Hori M, Yazama F, Matsuura Y, Yoshimoto R, Kaneda T, Yasumoto T, Ozaki H, Karaki H. Inhibition of actin polymerization by marine toxin pectenotoxin-2. J Vet Med Sci 2017; 80:225-234. [PMID: 29279465 PMCID: PMC5836757 DOI: 10.1292/jvms.17-0654] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pectenotoxin-2 (PCTX-2) is one of the polyether macrolide toxins isolated from scallops
involved in diarrheic shellfish poisoning via actin depolymerization. In the present
study, we examined the bioactive mechanism of PCTX-2 in smooth muscle cells and clarify
mode of action of the PCTX-2-induced actin depolymerization using purified skeletal actin.
PCTX-2 (300 nM-3 µM) non-selectively inhibited vascular smooth muscle
contractions elicited by high K+ or phenylephrine in a dose-dependent manner.
However, elevated cytosolic Ca2+ and myosin light chain phosphorylation
stimulated by high K+ were only slightly inhibited by PCTX-2. By monitoring the
fluorescent intensity of pyrenyl-actin, PCTX-2 was found to inhibit both the velocity and
degree of actin polymerization. The critical concentration of G-actin was linearly
increased in accordance with the concentration of PCTX-2, indicating sequestration of
G-actin with 1 to 1 ratio. The kinetics of F-actin depolymerization by dilution assay
indicated that PCTX-2 does not sever F-actin. Transmission electron microscopic and
confocal microscopic observations demonstrated that PCTX-2 selectively depolymerized
filamentous actin without affecting tublin. In conclusion, PCTX-2 is a potent natural
actin depolymerizer which sequesters G-actin without severing F-actin.
Collapse
Affiliation(s)
- Masatoshi Hori
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Futoshi Yazama
- Laboratory of Cell Biology and Morphology, School of Bioresources Hiroshima Prefectural University, Shoubara-shi, Hiroshima 727-0023, Japan
| | - Yasuhiro Matsuura
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Ryo Yoshimoto
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Takeharu Kaneda
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino, Tokyo 180-8602, Japan
| | - Takeshi Yasumoto
- Japan Food Research Laboratories, 6-11-10 Nagayama, Tama, Tokyo 206-0025, Japan
| | - Hiroshi Ozaki
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Hideaki Karaki
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
7
|
Xu Q, Huff LP, Fujii M, Griendling KK. Redox regulation of the actin cytoskeleton and its role in the vascular system. Free Radic Biol Med 2017; 109:84-107. [PMID: 28285002 PMCID: PMC5497502 DOI: 10.1016/j.freeradbiomed.2017.03.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/17/2017] [Accepted: 03/06/2017] [Indexed: 12/17/2022]
Abstract
The actin cytoskeleton is critical for form and function of vascular cells, serving mechanical, organizational and signaling roles. Because many cytoskeletal proteins are sensitive to reactive oxygen species, redox regulation has emerged as a pivotal modulator of the actin cytoskeleton and its associated proteins. Here, we summarize work implicating oxidants in altering actin cytoskeletal proteins and focus on how these alterations affect cell migration, proliferation and contraction of vascular cells. Finally, we discuss the role of oxidative modification of the actin cytoskeleton in vivo and highlight its importance for vascular diseases.
Collapse
Affiliation(s)
- Qian Xu
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States; Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Lauren P Huff
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States
| | - Masakazu Fujii
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States.
| |
Collapse
|
8
|
Tseng PY, Henderson PB, Hergarden AC, Patriarchi T, Coleman AM, Lillya MW, Montagut-Bordas C, Lee B, Hell JW, Horne MC. α-Actinin Promotes Surface Localization and Current Density of the Ca 2+ Channel Ca V1.2 by Binding to the IQ Region of the α1 Subunit. Biochemistry 2017; 56:3669-3681. [PMID: 28613835 DOI: 10.1021/acs.biochem.7b00359] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The voltage-gated L-type Ca2+ channel CaV1.2 is crucial for initiating heartbeat and control of a number of neuronal functions such as neuronal excitability and long-term potentiation. Mutations of CaV1.2 subunits result in serious health problems, including arrhythmia, autism spectrum disorders, immunodeficiency, and hypoglycemia. Thus, precise control of CaV1.2 surface expression and localization is essential. We previously reported that α-actinin associates and colocalizes with neuronal CaV1.2 channels and that shRNA-mediated depletion of α-actinin significantly reduces localization of endogenous CaV1.2 in dendritic spines in hippocampal neurons. Here we investigated the hypothesis that direct binding of α-actinin to CaV1.2 supports its surface expression. Using two-hybrid screens and pull-down assays, we identified three point mutations (K1647A, Y1649A, and I1654A) in the central, pore-forming α11.2 subunit of CaV1.2 that individually impaired α-actinin binding. Surface biotinylation and flow cytometry assays revealed that CaV1.2 channels composed of the corresponding α-actinin-binding-deficient mutants result in a 35-40% reduction in surface expression compared to that of wild-type channels. Moreover, the mutant CaV1.2 channels expressed in HEK293 cells exhibit a 60-75% decrease in current density. The larger decrease in current density as compared to surface expression imparted by these α11.2 subunit mutations hints at the possibility that α-actinin not only stabilizes surface localization of CaV1.2 but also augments its ion conducting activity.
Collapse
Affiliation(s)
- Pang-Yen Tseng
- Department of Pharmacology, School of Medicine, University of California , Davis, California 95615-8636, United States
| | - Peter B Henderson
- Department of Pharmacology, School of Medicine, University of California , Davis, California 95615-8636, United States
| | - Anne C Hergarden
- Department of Pharmacology, School of Medicine, University of California , Davis, California 95615-8636, United States
| | - Tommaso Patriarchi
- Department of Pharmacology, School of Medicine, University of California , Davis, California 95615-8636, United States
| | - Andrea M Coleman
- Department of Pharmacology, School of Medicine, University of California , Davis, California 95615-8636, United States
| | - Mark W Lillya
- Department of Pharmacology, School of Medicine, University of California , Davis, California 95615-8636, United States
| | - Carlota Montagut-Bordas
- Department of Pharmacology, School of Medicine, University of California , Davis, California 95615-8636, United States
| | - Boram Lee
- Department of Pharmacology, School of Medicine, University of California , Davis, California 95615-8636, United States
| | - Johannes W Hell
- Department of Pharmacology, School of Medicine, University of California , Davis, California 95615-8636, United States
| | - Mary C Horne
- Department of Pharmacology, School of Medicine, University of California , Davis, California 95615-8636, United States
| |
Collapse
|
9
|
Gonnissen D, Qu Y, Langer K, Öztürk C, Zhao Y, Chen C, Seebohm G, Düfer M, Fuchs H, Galla HJ, Riehemann K. Comparison of cellular effects of starch-coated SPIONs and poly(lactic-co-glycolic acid) matrix nanoparticles on human monocytes. Int J Nanomedicine 2016; 11:5221-5236. [PMID: 27789942 PMCID: PMC5072557 DOI: 10.2147/ijn.s106540] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Within the last years, progress has been made in the knowledge of the properties of medically used nanoparticles and their toxic effects, but still, little is known about their influence on cellular processes of immune cells. The aim of our comparative study was to present the influence of two different nanoparticle types on subcellular processes of primary monocytes and the leukemic monocyte cell line MM6. We used core-shell starch-coated superparamagnetic iron oxide nanoparticles (SPIONs) and matrix poly(lactic-co-glycolic acid) (PLGA) nanoparticles for our experiments. In addition to typical biocompatibility testing like the detection of necrosis or secretion of interleukins (ILs), we investigated the impact of these nanoparticles on the actin cytoskeleton and the two voltage-gated potassium channels Kv1.3 and Kv7.1. Induction of necrosis was not seen for PLGA nanoparticles and SPIONs in primary monocytes and MM6 cells. Likewise, no alteration in secretion of IL-1β and IL-10 was detected under the same experimental conditions. In contrast, IL-6 secretion was exclusively downregulated in primary monocytes after contact with both nanoparticles. Two-electrode voltage clamp experiments revealed that both nanoparticles reduce currents of the aforementioned potassium channels. The two nanoparticles differed significantly in their impact on the actin cytoskeleton, demonstrated via atomic force microscopy elasticity measurement and phalloidin staining. While SPIONs led to the disruption of the respective cytoskeleton, PLGA did not show any influence in both experimental setups. The difference in the effects on ion channels and the actin cytoskeleton suggests that nanoparticles affect these subcellular components via different pathways. Our data indicate that the alteration of the cytoskeleton and the effect on ion channels are new parameters that describe the influence of nanoparticles on cells. The results are highly relevant for medical application and further evaluation of nanomaterial biosafety.
Collapse
Affiliation(s)
- Dominik Gonnissen
- Center for Nanotechnology, Institute of Physics, University of Münster, Münster, Germany
| | - Ying Qu
- Center for Nanotechnology, Institute of Physics, University of Münster, Münster, Germany; National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Klaus Langer
- Institute of Pharmaceutical Technology and Biopharmacy, University of Münster, Münster
| | | | - Yuliang Zhao
- National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Chunying Chen
- National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Guiscard Seebohm
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases, University Hospital Münster
| | - Martina Düfer
- Department of Pharmacology, Institute of Pharmaceutical and Medicinal Chemistry
| | - Harald Fuchs
- Center for Nanotechnology, Institute of Physics, University of Münster, Münster, Germany
| | - Hans-Joachim Galla
- Department of Cell Biology/Biophysics, Institute of Biochemistry, University of Münster, Münster, Germany
| | - Kristina Riehemann
- Center for Nanotechnology, Institute of Physics, University of Münster, Münster, Germany
| |
Collapse
|
10
|
Viola H, Johnstone V, Cserne Szappanos H, Richman T, Tsoutsman T, Filipovska A, Semsarian C, Hool L. The L-type Ca(2+) channel facilitates abnormal metabolic activity in the cTnI-G203S mouse model of hypertrophic cardiomyopathy. J Physiol 2016; 594:4051-70. [PMID: 27062056 DOI: 10.1113/jp271681] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/01/2016] [Indexed: 12/31/2022] Open
Abstract
KEY POINTS Genetic mutations in cardiac troponin I (cTnI) are associated with development of hypertrophic cardiomyopathy characterized by myocyte remodelling, disorganization of cytoskeletal proteins and altered energy metabolism. The L-type Ca(2+) channel is the main route for calcium influx and is crucial to cardiac excitation and contraction. The channel also regulates mitochondrial function in the heart by a functional communication between the channel and mitochondria via the cytoskeletal network. We find that L-type Ca(2+) channel kinetics are altered in cTnI-G203S cardiac myocytes and that activation of the channel causes a significantly greater increase in mitochondrial membrane potential and metabolic activity in cTnI-G203S cardiac myocytes. These responses occur as a result of impaired communication between the L-type Ca(2+) channel and cytoskeletal protein F-actin, involving decreased movement of actin-myosin and block of the mitochondrial voltage-dependent anion channel, resulting in a 'hypermetabolic' mitochondrial state. We propose that L-type Ca(2+) channel antagonists, such as diltiazem, might be effective in reducing the cardiomyopathy by normalizing mitochondrial metabolic activity. ABSTRACT Genetic mutations in cardiac troponin I (cTnI) account for 5% of families with hypertrophic cardiomyopathy. Hypertrophic cardiomyopathy is associated with disorganization of cytoskeletal proteins and altered energy metabolism. The L-type Ca(2+) channel (ICa-L ) plays an important role in regulating mitochondrial function. This involves a functional communication between the channel and mitochondria via the cytoskeletal network. We investigate the role of ICa-L in regulating mitochondrial function in 25- to 30-week-old cardiomyopathic mice expressing the human disease-causing mutation Gly203Ser in cTnI (cTnI-G203S). The inactivation rate of ICa-L is significantly faster in cTnI-G203S myocytes [cTnI-G203S: τ1 = 40.68 ± 3.22, n = 10 vs. wild-type (wt): τ1 = 59.05 ± 6.40, n = 6, P < 0.05]. Activation of ICa-L caused a greater increase in mitochondrial membrane potential (Ψm , 29.19 ± 1.85%, n = 15 vs. wt: 18.84 ± 2.01%, n = 10, P < 0.05) and metabolic activity (24.40 ± 6.46%, n = 8 vs. wt: 9.98 ± 1.57%, n = 9, P < 0.05). The responses occurred because of impaired communication between ICa-L and F-actin, involving lack of dynamic movement of actin-myosin and block of the mitochondrial voltage-dependent anion channel. Similar responses were observed in precardiomyopathic mice. ICa-L antagonists nisoldipine and diltiazem decreased Ψm to basal levels. We conclude that the Gly203Ser mutation leads to impaired functional communication between ICa-L and mitochondria, resulting in a 'hypermetabolic' state. This might contribute to development of cTnI-G203S cardiomyopathy because the response is present in young precardiomyopathic mice. ICa-L antagonists might be effective in reducing the cardiomyopathy by altering mitochondrial function.
Collapse
Affiliation(s)
- Helena Viola
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, WA, Australia
| | - Victoria Johnstone
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, WA, Australia
| | - Henrietta Cserne Szappanos
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, WA, Australia
| | - Tara Richman
- The Harry Perkins Institute for Medical Research, The University of Western Australia, Crawley, WA, Australia
| | - Tatiana Tsoutsman
- Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, Sydney, NSW, Australia.,Sydney Medical School, University of Sydney, NSW, Australia
| | - Aleksandra Filipovska
- The Harry Perkins Institute for Medical Research, The University of Western Australia, Crawley, WA, Australia
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, Sydney, NSW, Australia.,Sydney Medical School, University of Sydney, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Livia Hool
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, WA, Australia.,Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| |
Collapse
|
11
|
Viola HM, Johnstone VPA, Cserne Szappanos H, Richman TR, Tsoutsman T, Filipovska A, Semsarian C, Seidman JG, Seidman CE, Hool LC. The Role of the L-Type Ca 2+ Channel in Altered Metabolic Activity in a Murine Model of Hypertrophic Cardiomyopathy. JACC Basic Transl Sci 2016; 1:61-72. [PMID: 30167506 PMCID: PMC6113168 DOI: 10.1016/j.jacbts.2015.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 12/31/2015] [Indexed: 01/08/2023]
Abstract
Heterozygous mice (αMHC403/+) expressing the human disease-causing mutation Arg403Gln exhibit cardinal features of hypertrophic cardiomyopathy (HCM) including hypertrophy, myocyte disarray, and increased myocardial fibrosis. Treatment of αMHC403/+mice with the L-type calcium channel (ICa-L) antagonist diltiazem has been shown to decrease left ventricular anterior wall thickness, cardiac myocyte hypertrophy, disarray, and fibrosis. However, the role of the ICa-L in the development of HCM is not known. In addition to maintaining cardiac excitation and contraction in myocytes, the ICa-L also regulates mitochondrial function through transmission of movement of ICa-L via cytoskeletal proteins to mitochondrial voltage-dependent anion channel. Here, the authors investigated the role of ICa-L in regulating mitochondrial function in αMHC403/+mice. Whole-cell patch clamp studies showed that ICa-L current inactivation kinetics were significantly increased in αMHC403/+cardiac myocytes, but that current density and channel expression were similar to wild-type cardiac myocytes. Activation of ICa-L caused a significantly greater increase in mitochondrial membrane potential and metabolic activity in αMHC403/+. These increases were attenuated with ICa-L antagonists and following F-actin or β-tubulin depolymerization. The authors observed increased levels of fibroblast growth factor-21 in αMHC403/+mice, and altered mitochondrial DNA copy number consistent with altered mitochondrial activity and the development of cardiomyopathy. These studies suggest that the Arg403Gln mutation leads to altered functional communication between ICa-L and mitochondria that is associated with increased metabolic activity, which may contribute to the development of cardiomyopathy. ICa-L antagonists may be effective in reducing the cardiomyopathy in HCM by altering metabolic activity. Heterozygous mice (αMHC403/+) expressing the human hypertrophic cardiomyopathy (HCM) disease causing mutation Arg403Gln exhibit cardinal features of HCM. This study investigated the role of L-type Ca2+ channel (ICa-L) in regulating mitochondrial function in Arg403Gln (αMHC403/+) mice. Activation of ICa-L in αMHC403/+mice caused a significantly greater increase in mitochondrial membrane potential and metabolic activity when compared to wild-type mice. Increases in mitochondrial membrane potential and metabolic activity were attenuated with ICa-L antagonists and when F-actin or β-tubulin were depolymerized. ICa-L antagonists may be effective in reducing the cardiomyopathy in HCM by altering metabolic activity.
Collapse
Affiliation(s)
- Helena M Viola
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Australia
| | - Victoria P A Johnstone
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Australia
| | - Henrietta Cserne Szappanos
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Australia
| | - Tara R Richman
- The Harry Perkins Institute for Medical Research, The University of Western Australia, Crawley, Australia
| | - Tatiana Tsoutsman
- Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, Sydney, Australia.,Sydney Medical School, University of Sydney, Australia
| | - Aleksandra Filipovska
- The Harry Perkins Institute for Medical Research, The University of Western Australia, Crawley, Australia
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, Sydney, Australia.,Sydney Medical School, University of Sydney, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | | | | | - Livia C Hool
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Australia.,Victor Chang Cardiac Research Institute, Sydney, Australia
| |
Collapse
|
12
|
Martinsen A, Dessy C, Morel N. Regulation of calcium channels in smooth muscle: new insights into the role of myosin light chain kinase. Channels (Austin) 2015; 8:402-13. [PMID: 25483583 DOI: 10.4161/19336950.2014.950537] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Smooth muscle myosin light chain kinase (MLCK) plays a crucial role in artery contraction, which regulates blood pressure and blood flow distribution. In addition to this role, MLCK contributes to Ca(2+) flux regulation in vascular smooth muscle (VSM) and in non-muscle cells, where cytoskeleton has been suggested to help Ca(2+) channels trafficking. This conclusion is based on the use of pharmacological inhibitors of MLCK and molecular and cellular techniques developed to down-regulate the enzyme. Dissimilarities have been observed between cells and whole tissues, as well as between large conductance and small resistance arteries. A differential expression in MLCK and ion channels (either voltage-dependent Ca(2+) channels or non-selective cationic channels) could account for these observations, and is in line with the functional properties of the arteries. A potential involvement of MLCK in the pathways modulating Ca(2+) entry in VSM is described in the present review.
Collapse
Key Words
- CaM, calmodulin
- ER, endoplasmic reticulum
- MLCK, myosin light chain kinase
- Myosin light chain kinase
- ROC, receptor-operated Ca2+ (channel)
- SMC, smooth muscle cell
- SOC, store-operated Ca2+ (channel)
- SR, sarcoplasmic reticulum
- TRP
- TRP, transient receptor potential (channel)
- VOC, voltage-operated Ca2+ (channel)
- VSM, vascular smooth muscle
- VSMC, vascular smooth muscle cell
- [Ca2+]cyt, cytosolic Ca2+ concentration
- siRNA, small interfering RNA
- vascular smooth muscle
- voltage-dependent calcium channels
Collapse
Affiliation(s)
- A Martinsen
- a Cell physiology; IoNS; UCLouvain ; Brussels , Belgium
| | | | | |
Collapse
|
13
|
Viola HM, Hool LC. Role of the cytoskeleton in communication between L-type Ca(2+) channels and mitochondria. Clin Exp Pharmacol Physiol 2015; 40:295-304. [PMID: 23551128 DOI: 10.1111/1440-1681.12072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 02/25/2013] [Accepted: 02/26/2013] [Indexed: 12/15/2022]
Abstract
The L-type Ca(2+) channel is the main route for Ca(2+) entry into cardiac myocytes, which is essential for the maintenance of cardiac excitation and contraction. Alterations in L-type Ca(2+) channel activity and Ca(2+) homeostasis have been implicated in the development of cardiomyopathies. Cardiac excitation and contraction is fuelled by ATP, synthesized predominantly by the mitochondria via the Ca(2+)-dependent process oxidative phosphorylation. Mitochondrial reactive oxygen species (ROS) are by-products of oxidative phosphorylation and are associated with the development of cardiac pathology. The cytoskeleton plays a role in the communication of signals from the plasma membrane to intracellular organelles. There is good evidence that both L-type Ca(2+) channel activity and mitochondrial function can be modulated by changes in the cytoskeletal network. Activation of the L-type Ca(2+) channel can regulate mitochondrial function through cytoskeletal proteins as a result of transmission of movement from the β(2)-subunit of the channel that occurs during activation and inactivation of the channel. An association between cytoskeletal proteins and the mitochondrial voltage-dependent anion channel (VDAC) may play a role in this response. The L-type Ca(2+) channel is the initiator of contraction in cardiac muscle and the VDAC is responsible for regulating mitochondrial ATP/ADP trafficking. This article presents evidence that a functional coupling between L-type Ca(2+) channels and mitochondria may assist in meeting myocardial energy demand on a beat-to-beat basis.
Collapse
Affiliation(s)
- Helena M Viola
- Cardiovascular Electrophysiology Laboratory, School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, WA, Australia
| | | |
Collapse
|
14
|
Davis TA, Loos B, Engelbrecht AM. AHNAK: the giant jack of all trades. Cell Signal 2014; 26:2683-93. [PMID: 25172424 DOI: 10.1016/j.cellsig.2014.08.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/08/2014] [Accepted: 08/18/2014] [Indexed: 12/19/2022]
Abstract
The nucleoprotein AHNAK is an unusual and somewhat mysterious scaffolding protein characterised by its large size of approximately 700 kDa. Several aspects of this protein remain uncertain, including its exact molecular function and regulation on both the gene and protein levels. Various studies have attempted to annotate AHNAK and, notably, protein interaction and expression analyses have contributed greatly to our current understanding of the protein. The implicated biological processes are, however, very diverse, ranging from a role in the formation of the blood-brain barrier, cell architecture and migration, to the regulation of cardiac calcium channels and muscle membrane repair. In addition, recent evidence suggests that AHNAK might be yet another accomplice in the development of tumour metastasis. This review will discuss the different functional roles of AHNAK, highlighting recent advancements that have added foundation to the proposed roles while identifying ties between them. Implications for related fields of research are noted and suggestions for future research that will assist in unravelling the function of AHNAK are offered.
Collapse
Affiliation(s)
- T A Davis
- Department of Physiological Sciences, University of Stellenbosch, Mike de Vries Building, c/o Merriman Avenue and Bosman Street, Stellenbosch 7600, South Africa.
| | - B Loos
- Department of Physiological Sciences, University of Stellenbosch, Mike de Vries Building, c/o Merriman Avenue and Bosman Street, Stellenbosch 7600, South Africa
| | - A-M Engelbrecht
- Department of Physiological Sciences, University of Stellenbosch, Mike de Vries Building, c/o Merriman Avenue and Bosman Street, Stellenbosch 7600, South Africa
| |
Collapse
|
15
|
Impaired functional communication between the L-type calcium channel and mitochondria contributes to metabolic inhibition in the mdx heart. Proc Natl Acad Sci U S A 2014; 111:E2905-14. [PMID: 24969422 DOI: 10.1073/pnas.1402544111] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Duchenne muscular dystrophy is a fatal X-linked disease characterized by the absence of dystrophin. Approximately 20% of boys will die of dilated cardiomyopathy that is associated with cytoskeletal protein disarray, contractile dysfunction, and reduced energy production. However, the mechanisms for altered energy metabolism are not yet fully clarified. Calcium influx through the L-type Ca(2+) channel is critical for maintaining cardiac excitation and contraction. The L-type Ca(2+) channel also regulates mitochondrial function and metabolic activity via transmission of movement of the auxiliary beta subunit through intermediate filament proteins. Here, we find that activation of the L-type Ca(2+) channel is unable to induce increases in mitochondrial membrane potential and metabolic activity in intact cardiac myocytes from the murine model of Duchenne muscular dystrophy (mdx) despite robust increases recorded in wt myocytes. Treatment of mdx mice with morpholino oligomers to induce exon skipping of dystrophin exon 23 (that results in functional dystrophin accumulation) or application of a peptide that resulted in block of voltage-dependent anion channel (VDAC) "rescued" mitochondrial membrane potential and metabolic activity in mdx myocytes. The mitochondrial VDAC coimmunoprecipitated with the L-type Ca(2+) channel. We conclude that the absence of dystrophin in the mdx ventricular myocyte leads to impaired functional communication between the L-type Ca(2+) channel and mitochondrial VDAC. This appears to contribute to metabolic inhibition. These findings provide new mechanistic and functional insight into cardiomyopathy associated with Duchenne muscular dystrophy.
Collapse
|
16
|
How does calcium regulate mitochondrial energetics in the heart? - new insights. Heart Lung Circ 2014; 23:602-9. [PMID: 24657282 DOI: 10.1016/j.hlc.2014.02.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 02/17/2014] [Indexed: 02/07/2023]
Abstract
Maintenance of cellular calcium homeostasis is critical to regulating mitochondrial ATP production and cardiac contraction. The ion channel known as the L-type calcium channel is the main route for calcium entry into cardiac myocytes. The channel associates with cytoskeletal proteins that assist with the communication of signals from the plasma membrane to intracellular organelles, including mitochondria. This article explores the roles of calcium and the cytoskeleton in regulation of mitochondrial function in response to alterations in L-type calcium channel activity. Direct activation of the L-type calcium channel results in an increase in intracellular calcium and increased mitochondrial calcium uptake. As a result, mitochondrial NADH production, oxygen consumption and reactive oxygen species production increase. In addition the L-type calcium channel is able to regulate mitochondrial membrane potential via cytoskeletal proteins when conformational changes in the channel occur during activation and inactivation. Since the L-type calcium channel is the initiator of contraction, a functional coupling between the channel and mitochondria via the cytoskeleton may represent a synchronised process by which mitochondrial function is regulated in addition to calcium influx to meet myocardial energy demand on a beat to beat basis.
Collapse
|
17
|
Hall DD, Dai S, Tseng PY, Malik Z, Nguyen M, Matt L, Schnizler K, Shephard A, Mohapatra DP, Tsuruta F, Dolmetsch RE, Christel CJ, Lee A, Burette A, Weinberg RJ, Hell JW. Competition between α-actinin and Ca²⁺-calmodulin controls surface retention of the L-type Ca²⁺ channel Ca(V)1.2. Neuron 2013; 78:483-97. [PMID: 23664615 DOI: 10.1016/j.neuron.2013.02.032] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2013] [Indexed: 12/30/2022]
Abstract
Regulation of neuronal excitability and cardiac excitation-contraction coupling requires the proper localization of L-type Ca²⁺ channels. We show that the actin-binding protein α-actinin binds to the C-terminal surface targeting motif of α11.2, the central pore-forming Ca(V)1.2 subunit, in order to foster its surface expression. Disruption of α-actinin function by dominant-negative or small hairpin RNA constructs reduces Ca(V)1.2 surface localization in human embryonic kidney 293 and neuronal cultures and dendritic spine localization in neurons. We demonstrate that calmodulin displaces α-actinin from their shared binding site on α11.2 upon Ca²⁺ influx through L-type channels, but not through NMDAR, thereby triggering loss of Ca(V)1.2 from spines. Coexpression of a Ca²⁺-binding-deficient calmodulin mutant does not affect basal Ca(V)1.2 surface expression but inhibits its internalization upon Ca²⁺ influx. We conclude that α-actinin stabilizes Ca(V)1.2 at the plasma membrane and that its displacement by Ca²⁺-calmodulin triggers Ca²⁺-induced endocytosis of Ca(V)1.2, thus providing an important negative feedback mechanism for Ca²⁺ influx.
Collapse
Affiliation(s)
- Duane D Hall
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Takeshita D, Tanaka M, Mitsuyama S, Yoshikawa Y, Zhang GX, Obata K, Ito H, Taniguchi S, Takaki M. A new calpain inhibitor protects left ventricular dysfunction induced by mild ischemia-reperfusion in in situ rat hearts. J Physiol Sci 2013; 63:113-23. [PMID: 23242912 PMCID: PMC10717469 DOI: 10.1007/s12576-012-0243-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 11/25/2012] [Indexed: 10/27/2022]
Abstract
We have previously indicated that a new soluble calpain inhibitor, SNJ-1945 (SNJ), attenuates cardiac dysfunction after cardioplegia arrest-reperfusion by inhibiting the proteolysis of α-fodrin in in vitro study. Nevertheless, the in vivo study design is indispensable to explore realistic therapeutic approaches for clinical use. The aim of the present in situ study was to investigate whether SNJ attenuated left ventricular (LV) dysfunction (stunning) after mild ischemic-reperfusion (mI-R) in rat hearts. SNJ (60 μmol/l, 5 ml i.p.) was injected 30 min before gradual and partial coronary occlusion at proximal left anterior descending artery. To investigate LV function, we obtained curvilinear end-systolic pressure-volume relationship by increasing afterload 60 min after reperfusion. In the mI-R group, specific LV functional indices at midrange LV volume (mLVV), end-systolic pressure (ESP(mLVV)), and pressure-volume area (PVA(mLVV): a total mechanical energy per beat, linearly related to oxygen consumption) significantly decreased, but SNJ reversed these decreases to time control level. Furthermore, SNJ prevented the α-fodrin degradation and attenuated degradation of Ca(2+) handling proteins after mI-R. Our results indicate that improvements in LV function following mI-R injury are associated with inhibition of the proteolysis of α-fodrin in in situ rat hearts. In conclusion, SNJ should be a promising tool to protect the heart from the stunning.
Collapse
Affiliation(s)
- D. Takeshita
- Department of Physiology II, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8521 Japan
| | - M. Tanaka
- Department of Physiology II, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8521 Japan
- Faculty of Health Care Science, Himeji Dokkyo University, Himeji, Japan
| | - S. Mitsuyama
- Department of Physiology II, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8521 Japan
| | - Y. Yoshikawa
- Department of Thoracic and Cardiovascular Surgery, Nara Medical University, Nara, Japan
| | - G. -X. Zhang
- Department of Physiology II, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8521 Japan
- Department of Physiology, Medical College of Soochow University, Dushu Lake Campus, Suzhou Industrial Park, Suzhou, 215123 People’s Republic of China
| | - K. Obata
- Department of Physiology II, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8521 Japan
| | - H. Ito
- Department of Physiology II, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8521 Japan
| | - S. Taniguchi
- Department of Thoracic and Cardiovascular Surgery, Nara Medical University, Nara, Japan
| | - Miyako Takaki
- Department of Physiology II, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8521 Japan
| |
Collapse
|
19
|
Viola HM, Davies SMK, Filipovska A, Hool LC. L-type Ca(2+) channel contributes to alterations in mitochondrial calcium handling in the mdx ventricular myocyte. Am J Physiol Heart Circ Physiol 2013; 304:H767-75. [PMID: 23335798 DOI: 10.1152/ajpheart.00700.2012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The L-type Ca(2+) channel is the main route for calcium entry into cardiac myocytes, and it is essential for contraction. Alterations in whole cell L-type Ca(2+) channel current and Ca(2+) homeostasis have been implicated in the development of cardiomyopathies. Cytoskeletal proteins can influence whole cell L-type Ca(2+) current and mitochondrial function. Duchenne muscular dystrophy is a fatal X-linked disease that leads to progressive muscle weakness due to the absence of cytoskeletal protein dystrophin. This includes dilated cardiomyopathy, but the mechanisms are not well understood. We sought to identify the effect of alterations in whole cell L-type Ca(2+) channel current on mitochondrial function in the murine model of Duchenne muscular dystrophy (mdx). Activation of the L-type Ca(2+) channel with the dihydropyridine agonist BayK(-) caused a significantly larger increase in cytosolic Ca(2+) in mdx vs. wild-type (wt) ventricular myocytes. Consistent with elevated cytosolic Ca(2+), resting mitochondrial Ca(2+), NADH, and mitochondrial superoxide were significantly greater in mdx vs. wt myocytes. Activation of the channel with BayK(-) caused a further increase in mitochondrial Ca(2+), NADH, and superoxide in mdx myocytes. The ratios of the increases were similar to the ratios recorded in wt myocytes. In mitochondria isolated from 8-wk-old mdx hearts, respiration and mitochondrial electron transport chain complex activity were similar to mitochondria isolated from wt hearts. We conclude that mitochondria function at a higher level of resting calcium in the intact mdx myocyte and activation of the L-type Ca(2+) channel contributes to alterations in calcium handling by the mitochondria. This perturbation may contribute to the development of cardiomyopathy.
Collapse
Affiliation(s)
- Helena M Viola
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Crawley, Western Australia, Australia
| | | | | | | |
Collapse
|
20
|
Kim HK, Park WS, Warda M, Park SY, Ko EA, Kim MH, Jeong SH, Heo HJ, Choi TH, Hwang YW, Lee SI, Ko KS, Rhee BD, Kim N, Han J. Beta adrenergic overstimulation impaired vascular contractility via actin-cytoskeleton disorganization in rabbit cerebral artery. PLoS One 2012; 7:e43884. [PMID: 22916309 PMCID: PMC3423383 DOI: 10.1371/journal.pone.0043884] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 07/26/2012] [Indexed: 11/22/2022] Open
Abstract
Background and Purpose Beta adrenergic overstimulation may increase the vascular damage and stroke. However, the underlying mechanisms of beta adrenergic overstimulation in cerebrovascular dysfunctions are not well known. We investigated the possible cerebrovascular dysfunction response to isoproterenol induced beta-adrenergic overstimulation (ISO) in rabbit cerebral arteries (CAs). Methods ISO was induced in six weeks aged male New Zealand white rabbit (0.8–1.0 kg) by 7-days isoproterenol injection (300 μg/kg/day). We investigated the alteration of protein expression in ISO treated CAs using 2DE proteomics and western blot analysis. Systemic properties of 2DE proteomics result were analyzed using bioinformatics software. ROS generation and following DNA damage were assessed to evaluate deteriorative effect of ISO on CAs. Intracellular Ca2+ level change and vascular contractile response to vasoactive drug, angiotensin II (Ang II), were assessed to evaluate functional alteration of ISO treated CAs. Ang II-induced ROS generation was assessed to evaluated involvement of ROS generation in CA contractility. Results Proteomic analysis revealed remarkably decreased expression of cytoskeleton organizing proteins (e.g. actin related protein 1A and 2, α-actin, capping protein Z beta, and vimentin) and anti-oxidative stress proteins (e.g. heat shock protein 9A and stress-induced-phosphoprotein 1) in ISO-CAs. As a cause of dysregulation of actin-cytoskeleton organization, we found decreased level of RhoA and ROCK1, which are major regulators of actin-cytoskeleton organization. As functional consequences of proteomic alteration, we found the decreased transient Ca2+ efflux and constriction response to angiotensin II and high K+ in ISO-CAs. ISO also increased basal ROS generation and induced oxidative damage in CA; however, it decreased the Ang II-induced ROS generation rate. These results indicate that ISO disrupted actin cytoskeleton proteome network through down-regulation of RhoA/ROCK1 proteins and increased oxidative damage, which consequently led to contractile dysfunction in CA.
Collapse
Affiliation(s)
- Hyoung Kyu Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Mohamad Warda
- Department of Biochemistry, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - So Youn Park
- Department of Pharmacology, College of Medicine and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Busan, Korea
| | - Eun A. Ko
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Min Hee Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Seung Hun Jeong
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Hye-Jin Heo
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Tae-Hoon Choi
- Department of Physical Education, Andong Science College, Andong, Korea
| | - Young-Won Hwang
- Department of Neurosurgery, College of Medicine, Inje University, Busan Paik Hospital, Busan, Korea
| | - Sun-Il Lee
- Department of Neurosurgery, College of Medicine, Inje University, Busan Paik Hospital, Busan, Korea
| | - Kyung Soo Ko
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Byoung Doo Rhee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Nari Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
- * E-mail:
| |
Collapse
|
21
|
Müller AL, Hryshko LV, Dhalla NS. Extracellular and intracellular proteases in cardiac dysfunction due to ischemia-reperfusion injury. Int J Cardiol 2012; 164:39-47. [PMID: 22357424 DOI: 10.1016/j.ijcard.2012.01.103] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 10/19/2011] [Accepted: 01/28/2012] [Indexed: 12/20/2022]
Abstract
Various procedures such as angioplasty, thrombolytic therapy, coronary bypass surgery, and cardiac transplantation are invariably associated with ischemia-reperfusion (I/R) injury. Impaired recovery of cardiac function due to I/R injury is considered to be a consequence of the occurrence of both oxidative stress and intracellular Ca(2+)-overload in the myocardium. These changes in the ischemic myocardium appear to activate both extracellular and intracellular proteases which are responsible for the cleavage of extracellular matrix and subcellular structures involved in the maintenance of cardiac function. It is thus intended to discuss the actions of I/R injury on several proteases, with a focus on calpain, matrix metalloproteinases, and cathepsins as well as their role in inducing alterations both inside and outside the cardiomyocytes. In addition, modifications of subcellular organelles such as myofibrils, sarcoplasmic reticulum and sarcolemma as well as extracellular matrix, and the potential regulatory effects of endogenous inhibitors on protease activities are identified. Both extracellular and intracellular proteolytic activities appear to be imperative in determining the true extent of I/R injury and their inhibition seems to be of critical importance for improving the recovery of cardiac function. Thus, both extracellular and intracellular proteases may serve as potential targets for the development of cardioprotective interventions for reducing damage to the heart and retarding the development of contractile dysfunction caused by I/R injury.
Collapse
Affiliation(s)
- Alison L Müller
- Institute of Cardiovascular Sciences, St Boniface Hospital Research Centre, and Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | |
Collapse
|
22
|
Lou Q, Li W, Efimov IR. The role of dynamic instability and wavelength in arrhythmia maintenance as revealed by panoramic imaging with blebbistatin vs. 2,3-butanedione monoxime. Am J Physiol Heart Circ Physiol 2011; 302:H262-9. [PMID: 22037192 DOI: 10.1152/ajpheart.00711.2011] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Unlike other excitation-contraction uncouplers, blebbistatin has few electrophysiological side effects and has gained increasing acceptance as an excitation-contraction uncoupler in optical mapping experiments. However, the possible role of blebbistatin in ventricular arrhythmia has hitherto been unknown. Furthermore, experiments with blebbistatin and 2,3-butanedione monoxime (BDM) offer an opportunity to assess the contribution of dynamic instability and wavelength of impulse propagation to the induction and maintenance of ventricular arrhythmia. Recordings of monophasic action potentials were used to assess effects of blebbistatin in Langendorff-perfused rabbit hearts (n = 5). Additionally, panoramic optical mapping experiments were conducted in rabbit hearts (n = 7) that were sequentially perfused with BDM, then washed out, and subsequently perfused with blebbistatin. The susceptibility to arrhythmia was investigated using a shock-on-T protocol. We found that 1) application of blebbistatin did not change action potential duration (APD) restitution; 2) in contrast to blebbistatin, BDM flattened APD restitution curve and reduced the wavelength; and 3) incidence of sustained arrhythmia was much lower under blebbistatin than under BDM (2/123 vs. 23/99). While arrhythmias under BDM were able to stabilize, the arrhythmias under blebbistatin were unstable and terminated spontaneously. In conclusion, the lower susceptibility to arrhythmia under blebbistatin than under BDM indicates that blebbistatin has less effects on arrhythmia dynamics. A steep restitution slope under blebbistatin is associated with higher dynamic instability, manifested by the higher incidence of not only wave breaks but also wave extinctions. This relatively high dynamic instability leads to the self-termination of arrhythmia because of the sufficiently long wavelength under blebbistatin.
Collapse
Affiliation(s)
- Qing Lou
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri 63130-4899, USA
| | | | | |
Collapse
|
23
|
Rosker C, Salvarani N, Schmutz S, Grand T, Rohr S. Abolishing myofibroblast arrhythmogeneicity by pharmacological ablation of α-smooth muscle actin containing stress fibers. Circ Res 2011; 109:1120-31. [PMID: 21921266 DOI: 10.1161/circresaha.111.244798] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Myofibroblasts typically appear in the myocardium after insults to the heart like mechanical overload and infarction. Apart from contributing to fibrotic remodeling, myofibroblasts induce arrhythmogenic slow conduction and ectopic activity in cardiomyocytes after establishment of heterocellular electrotonic coupling in vitro. So far, it is not known whether α-smooth muscle actin (α-SMA) containing stress fibers, the cytoskeletal components that set myofibroblasts apart from resident fibroblasts, are essential for myofibroblasts to develop arrhythmogenic interactions with cardiomyocytes. OBJECTIVE We investigated whether pharmacological ablation of α-SMA containing stress fibers by actin-targeting drugs affects arrhythmogenic myofibroblast-cardiomyocyte cross-talk. METHODS AND RESULTS Experiments were performed with patterned growth cell cultures of neonatal rat ventricular cardiomyocytes coated with cardiac myofibroblasts. The preparations exhibited slow conduction and ectopic activity under control conditions. Exposure to actin-targeting drugs (Cytochalasin D, Latrunculin B, Jasplakinolide) for 24 hours led to disruption of α-SMA containing stress fibers. In parallel, conduction velocities increased dose-dependently to values indistinguishable from cardiomyocyte-only preparations and ectopic activity measured continuously over 24 hours was completely suppressed. Mechanistically, antiarrhythmic effects were due to myofibroblast hyperpolarization (Cytochalasin D, Latrunculin B) and disruption of heterocellular gap junctional coupling (Jasplakinolide), which caused normalization of membrane polarization of adjacent cardiomyocytes. CONCLUSIONS The results suggest that α-SMA containing stress fibers importantly contribute to myofibroblast arrhythmogeneicity. After ablation of this cytoskeletal component, cells lose their arrhythmic effects on cardiomyocytes, even if heterocellular electrotonic coupling is sustained. The findings identify α-SMA containing stress fibers as a potential future target of antiarrhythmic therapy in hearts undergoing structural remodeling.
Collapse
Affiliation(s)
- Christian Rosker
- Dept. of Physiology, University of Bern, Bühlplatz 5, CH-3012 Bern, Switzerland
| | | | | | | | | |
Collapse
|
24
|
Li F, Wang W, Gu M, Gyoneva S, Zhang J, Huang S, Traynelis SF, Cai H, Guggino SE, Zhang X. L-type calcium channel activity in osteoblast cells is regulated by the actin cytoskeleton independent of protein trafficking. J Bone Miner Metab 2011; 29:515-25. [PMID: 21246227 DOI: 10.1007/s00774-010-0252-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 11/14/2010] [Indexed: 11/27/2022]
Abstract
Voltage-dependent L-type calcium channels (VDCC) play important roles in many cellular processes. The interaction of the actin cytoskeleton with the channel in nonexcitable cells is less well understood. We performed whole-cell patch-clamp surface biotinylation and calcium imaging on different osteoblast cells to determine channel kinetics, amplitude, surface abundance, and intracellular calcium, respectively. Patch-clamp studies showed that actin polymerization by phalloidin increased the peak current density of I (Ca), whereas actin depolymerization by cytochalasin D (CD) significantly decreased the current amplitude. This result is consistent with calcium imaging, which showed that CD significantly decreased Bay K8644-induced intracellular calcium increase. Surface biotinylation studies showed that CD is not able to affect the surface expression of the pore-forming subunit α(1C). Interestingly, application of CD caused a significantly negative shift in the steady-state inactivation kinetics of I (Ca). There were decreases in the voltage at half-maximal inactivation that changed in a dose-dependent manner. CD also reduced the effect of activated vitamin D(3) (1α,25-D3) on VDCC and intracellular calcium. We conclude that in osteoblasts the actin cytoskeleton affects α(1C) by altering the channel kinetic properties, instead of changing the surface expression, and it is able to regulate 1α,25-D3 signaling through VDCC. Our study provides a new insight into calcium regulation in osteoblasts, which are essential in many physiological functions of this cell.
Collapse
Affiliation(s)
- Fangping Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Pudong New District, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Yoshikawa Y, Zhang GX, Obata K, Matsuyoshi H, Asada K, Taniguchi S, Takaki M. A cardioprotective agent of a novel calpain inhibitor, SNJ-1945, exerts beta1 actions on left ventricular mechanical work and energetics. Am J Physiol Heart Circ Physiol 2010; 299:H396-401. [PMID: 20511411 DOI: 10.1152/ajpheart.00153.2010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously shown that a newly developed calpain inhibitor, SNJ-1945 (SNJ), with good aqueous solubility prevents the heart from KCl arrest-reperfusion injury associated with the impairment of total Ca(2+) handling by inhibiting the proteolysis of alpha-fodrin as a cardioplegia. The aim of the present study was to investigate certain actions of this calpain inhibitor, SNJ, on left ventricular (LV) mechanical work and energetics in cross-circulated excised rat hearts undergoing blood perfusion with 40 microM SNJ. Mean end-systolic pressure at midrange LV volume and systolic pressure-volume area (PVA) at mLVV (a total mechanical energy/beat) were significantly increased by SNJ perfusion (P < 0.01). Mean myocardial oxygen consumption per beat (Vo(2)) intercepts (Vo(2) for the total Ca(2+) handling in excitation-contraction coupling and basal metabolism) of Vo(2)-PVA linear relations were significantly increased (P < 0.01) with unchanged mean slopes of Vo(2)-PVA linear relations. Pretreatment with the selective beta(1)-blocker landiolol (10 microM) blocked these effects of SNJ perfusion. There were no significant differences in mean basal metabolic oxygen consumption among normal, 40 microM SNJ, and 10 microM landiolol + 40 microM SNJ groups. Our results indicate that water-soluble SNJ exerted positive actions on mechanical work and energetics mediated via beta(1)-adrenergic receptors associated with the enhancement of total Ca(2+) handling in excitation-contraction coupling and with unchanged contractile efficiency. In clinical settings, this pharmacological action of SNJ is beneficial as an additive agent for cardioplegia.
Collapse
Affiliation(s)
- Yoshiro Yoshikawa
- Dept. of Physiology II, Nara Medical Univ. School of Medicine, 840 Shijo-cho, Kashihara, Nara 634.8521, Japan
| | | | | | | | | | | | | |
Collapse
|
26
|
Aromolaran KA, Benzow KA, Cribbs LL, Koob MD, Piedras-Rentería ES. T-type current modulation by the actin-binding protein Kelch-like 1. Am J Physiol Cell Physiol 2010; 298:C1353-62. [PMID: 20147652 DOI: 10.1152/ajpcell.00235.2009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We report a novel form of modulation of T-type calcium currents carried out by the neuronal actin-binding protein (ABP) Kelch-like 1 (KLHL1). KLHL1 is a constitutive neuronal ABP localized to the soma and dendritic arbors; its genetic elimination in Purkinje neurons leads to dendritic atrophy and motor insufficiency. KLHL1 participates in neurite outgrowth and upregulates voltage-gated P/Q-type calcium channel function; here we investigated KLHL1's role as a modulator of low-voltage-gated calcium channels and determined the molecular mechanism of this modulation with electrophysiology and biochemistry. Coexpression of KLHL1 with Ca(V)3.1 or Ca(V)3.2 (alpha(1G) or alpha(1H) subunits) caused increases in T-type current density (35%) and calcium influx (75-83%) when carried out by alpha(1H) but not by alpha(1G). The association between KLHL1 and alpha(1H) was determined by immunoprecipitation and immunolocalization in brain membrane fractions and in vitro in HEK-293 cells. Noise analysis showed that neither alpha(1H) single-channel conductance nor open probability was altered by KLHL1, yet a significant increase in channel number was detected and further corroborated by Western blot analysis. KLHL1 also induced an increase in alpha(1H) current deactivation time (tau(deactivation)). Interestingly, the majority of KLHL1's effects were eliminated when the actin-binding motif (kelch) was removed, with the exception of the calcium influx increase during action potentials, indicating that KLHL1 interacts with alpha(1H) and actin and selectively regulates alpha(1H) function by increasing the number of alpha(1H) channels. This constitutes a novel regulatory mechanism of T-type calcium currents and supports the role of KLHL1 in the modulation of cellular excitability.
Collapse
|
27
|
The multiple roles of myelin protein genes during the development of the oligodendrocyte. ASN Neuro 2010; 2:e00027. [PMID: 20017732 PMCID: PMC2814326 DOI: 10.1042/an20090051] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Revised: 12/14/2009] [Accepted: 12/17/2009] [Indexed: 11/22/2022] Open
Abstract
It has become clear that the products of several of the earliest identified myelin protein genes perform functions that extend beyond the myelin sheath. Interestingly, these myelin proteins, which comprise proteolipid protein, 2′,3′-cyclic nucleotide 3′-phosphodiesterase and the classic and golli MBPs (myelin basic proteins), play important roles during different stages of oligodendroglial development. These non-myelin-related functions are varied and include roles in the regulation of process outgrowth, migration, RNA transport, oligodendrocyte survival and ion channel modulation. However, despite the wide variety of cellular functions performed by the different myelin genes, the route by which they achieve these many functions seems to converge upon a common mechanism involving Ca2+ regulation, cytoskeletal rearrangements and signal transduction. In the present review, the newly emerging functions of these myelin proteins will be described, and these will then be discussed in the context of their contribution to oligodendroglial development.
Collapse
|
28
|
Yoshikawa Y, Zhang GX, Obata K, Ohga Y, Matsuyoshi H, Taniguchi S, Takaki M. Cardioprotective effects of a novel calpain inhibitor SNJ-1945 for reperfusion injury after cardioplegic cardiac arrest. Am J Physiol Heart Circ Physiol 2009; 298:H643-51. [PMID: 19966051 DOI: 10.1152/ajpheart.00849.2009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We have previously indicated that calpain inhibitor-1 prevents the heart from ischemia- reperfusion injury associated with the impairment of total Ca(2+) handling by inhibiting the proteolysis of alpha-fodrin. However, this inhibitor is insoluble with water and inappropriate for clinical application. The aim of the present study was to investigate the protective effect of a newly developed calpain inhibitor, SNJ-1945 (SNJ), with good aqueous solubility on left ventricular (LV) mechanical work and energetics in the cross-circulated rat hearts. SNJ (150 microM) was added to KCl (30 meq) cardioplegia (CP). Mean end-systolic pressure at midrange LV volume (ESP(mLVV)) and systolic pressure-volume area (PVA) at mLVV (PVA(mLVV); a total mechanical energy per beat) were hardly changed after CP plus SNJ arrest-reperfusion (post-CP + SNJ), whereas ESP(mLVV) and PVA(mLVV) in post-CP group were significantly (P < 0.01) decreased. Mean myocardial oxygen consumption for the total Ca(2+) handling in excitation-contraction coupling did not significantly decrease in post-CP + SNJ group, whereas it was significantly (P < 0.01) decreased in post-CP group. The mean amounts of 145- and 150-kDa fragments of alpha-fodrin in the post-CP group were significantly larger than those in normal and post-CP + SNJ groups. In contrast, the mean amounts of L-type Ca(2+) channel and sarcoplasmic reticulum Ca(2+)-ATPase were not significantly different among normal, post-CP, and post-CP + SNJ groups. Our results indicate that soluble SNJ attenuates cardiac dysfunction due to CP arrest-reperfusion injury associated with the impairment of the total Ca(2+) handling in excitation-contraction coupling by inhibiting the proteolysis of alpha-fodrin.
Collapse
Affiliation(s)
- Yoshiro Yoshikawa
- Department of Physiology II, Nara Medical University School of Medicine, Kashihara, Nara, Japan
| | | | | | | | | | | | | |
Collapse
|
29
|
Shao Y, Czymmek KJ, Jones PA, Fomin VP, Akanbi K, Duncan RL, Farach-Carson MC. Dynamic interactions between L-type voltage-sensitive calcium channel Cav1.2 subunits and ahnak in osteoblastic cells. Am J Physiol Cell Physiol 2009; 296:C1067-78. [PMID: 19261907 PMCID: PMC2681378 DOI: 10.1152/ajpcell.00427.2008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Accepted: 03/03/2009] [Indexed: 11/22/2022]
Abstract
Voltage-sensitive Ca(2+) channels (VSCCs) mediate Ca(2+) permeability in osteoblasts. Association between VSCC alpha(1)- and beta-subunits targets channel complexes to the plasma membrane and modulates function. In mechanosensitive tissues, a 700-kDa ahnak protein anchors VSCCs to the actin cytoskeleton via the beta(2)-subunit of the L-type Ca(v)1.2 (alpha(1C)) VSCC complex. Ca(v)1.2 is the major alpha(1)-subunit in osteoblasts, but the cytoskeletal complex and subunit composition are unknown. Among the four beta-subtypes, the beta(2)-subunit and, to a lesser extent, the beta(3)-subunit coimmunoprecipitated with the Ca(v)1.2 subunit in MC3T3-E1 preosteoblasts. Fluorescence resonance energy transfer revealed a complex between Ca(v)1.2 and beta(2)-subunits and demonstrated their association in the plasma membrane and secretory pathway. Western blot and immunohistochemistry showed ahnak association with the channel complex in the plasma membrane via the beta(2)-subunit. Cytochalasin D exposure disrupted the actin cytoskeleton but did not disassemble or disrupt the function of the complex of L-type VSCC Ca(v)1.2 and beta(2)-subunits and ahnak. Similarly, small interfering RNA knockdown of ahnak did not disrupt the actin cytoskeleton but significantly impaired Ca(2+) influx. Collectively, we showed that Ca(v)1.2 and beta(2)-subunits and ahnak form a stable complex in osteoblastic cells that permits Ca(2+) signaling independently of association with the actin cytoskeleton.
Collapse
Affiliation(s)
- Ying Shao
- Dept. of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Tsuji T, del Monte F, Yoshikawa Y, Abe T, Shimizu J, Nakajima-Takenaka C, Taniguchi S, Hajjar RJ, Takaki M. Rescue of Ca2+ overload-induced left ventriclur dysfunction by targeted ablation of phospholamban. Am J Physiol Heart Circ Physiol 2009; 296:H310-7. [DOI: 10.1152/ajpheart.00975.2008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In failing hearts, a deficiency in sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)2a results in abnormal Ca2+ handling and diminished contraction. In addition, a decrease in the phosphorylation of phospholamban (PLB) has been reported. Gene transfer of antisense PLB (asPLB) can improve contractile function in the failing human myocardium. Gene transfer of SERCA2a improves survival and the energy potential in failing hearts. The aim of present study was to evaluate whether enhancement of SERCA2a function prevents acute Ca2+ overload-induced left ventricular (LV) dysfunction in rat hearts. We ablated PLB using adenoviral gene transfer of asPLB by a new and less invasive gene delivery method, which involved a percutaneous technique. Experiments were performed on 13 excised cross-circulated rat hearts: 5 rats underwent sham operations, 4 rats underwent gene transfer of the reporter gene β-galactosidase (Ad.β-gal), and 4 rats underwent gene transfer of asPLB (Ad.asPLB). After clearance of high Ca2+ infused into the coronary, there was LV contractile dysfunction associated with the decreased myocardial O2 consumption per beat (Vo2) intercept (equal to decreased Vo2 for Ca2+ handling in excitation-contraction coupling) of the Vo2-systolic pressure-volume area (PVA; total mechanical energy per beat) linear relation in the hearts that underwent sham operation and had been infected with Ad.β-gal. Hearts that had been infected with Ad.asPLB were rescued from LV contractile dysfunction associated with an unchanged Vo2 intercept of the Vo2-PVA linear relation. We conclude that SERCA2a function enhanced by adenoviral gene transfer of asPLB prevents Ca2+ overload-induced LV contractile dysfunction in terms of mechanical work and especially energetics.
Collapse
|
31
|
Viola HM, Arthur PG, Hool LC. Evidence for regulation of mitochondrial function by the L-type Ca2+ channel in ventricular myocytes. J Mol Cell Cardiol 2009; 46:1016-26. [PMID: 19166857 DOI: 10.1016/j.yjmcc.2008.12.015] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Revised: 12/12/2008] [Accepted: 12/16/2008] [Indexed: 10/21/2022]
Abstract
The L-type Ca(2+) channel is responsible for initiating contraction in the heart. Mitochondria are responsible for meeting the cellular energy demands and calcium is required for the activity of metabolic intermediates. We examined whether activation of the L-type Ca(2+) channel alone is sufficient to alter mitochondrial function. The channel was activated directly with the dihydropyridine agonist BayK(-) or voltage-clamp of the plasma membrane and indirectly by depolarization of the membrane with high KCl. Activation of the channel increased superoxide production (assessed as changes in dihydroethidium fluorescence), NADH production and metabolic activity (assessed as formation of formazan from tetrazolium) in a calcium-dependent manner. Activation of the channel also increased mitochondrial membrane potential assessed as changes in JC-1 fluorescence. The response was reversible upon inactivation of the channel during voltage-clamp of the plasma membrane and did not appear to require calcium. We examined whether the response may be mediated through movement of cytoskeletal proteins. Depolymerization of actin or exposing cells to a peptide directed against the alpha-interacting domain of the alpha(1C)-subunit of the channel (thereby preventing movement of the beta-subunit) attenuated the increase in mitochondrial membrane potential. We conclude that activation of the L-type Ca(2+) channel can regulate mitochondrial function and the response appears to be modulated by movement through the cytoskeleton.
Collapse
Affiliation(s)
- Helena M Viola
- School of Biomedical, Biomolecular and Chemical Sciences, The University of Western Australia, Crawley, Australia
| | | | | |
Collapse
|
32
|
Pica-Mattoccia L, Orsini T, Basso A, Festucci A, Liberti P, Guidi A, Marcatto-Maggi AL, Nobre-Santana S, Troiani AR, Cioli D, Valle C. Schistosoma mansoni: Lack of correlation between praziquantel-induced intra-worm calcium influx and parasite death. Exp Parasitol 2008; 119:332-5. [DOI: 10.1016/j.exppara.2008.03.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Revised: 03/10/2008] [Accepted: 03/10/2008] [Indexed: 11/16/2022]
|
33
|
Koltsova SV, Gusakova SV, Anfinogenova YJ, Baskakov MB, Orlov SN. Vascular Smooth Muscle Contraction Evoked by Cell Volume Modulation: Role of the Cytoskeleton Network. Cell Physiol Biochem 2008; 21:29-36. [DOI: 10.1159/000113744] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2007] [Indexed: 01/23/2023] Open
|
34
|
Wang J, Weigand L, Foxson J, Shimoda LA, Sylvester JT. Ca2+ signaling in hypoxic pulmonary vasoconstriction: effects of myosin light chain and Rho kinase antagonists. Am J Physiol Lung Cell Mol Physiol 2007; 293:L674-85. [PMID: 17575009 DOI: 10.1152/ajplung.00141.2007] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Antagonists of myosin light chain (MLC) kinase (MLCK) and Rho kinase (ROK) are thought to inhibit hypoxic pulmonary vasoconstriction (HPV) by decreasing the concentration of phosphorylated MLC at any intracellular Ca(2+) concentration ([Ca(2+)](i)) in pulmonary arterial smooth muscle cells (PASMC); however, these antagonists can also decrease [Ca(2+)](i). To determine whether MLCK and ROK antagonists alter Ca(2+) signaling in HPV, we measured the effects of ML-9, ML-7, Y-27632, and HA-1077 on [Ca(2+)](i), Ca(2+) entry, and Ca(2+) release in rat distal PASMC exposed to hypoxia or depolarizing concentrations of KCl. We performed parallel experiments in isolated rat lungs to confirm the inhibitory effects of these agents on pulmonary vasoconstriction. Our results demonstrate that MLCK and ROK antagonists caused concentration-dependent inhibition of hypoxia-induced increases in [Ca(2+)](i) in PASMC and HPV in isolated lungs and suggest that this inhibition was due to blockade of Ca(2+) release from the sarcoplasmic reticulum and Ca(2+) entry through store- and voltage-operated Ca(2+) channels in PASMC. Thus MLCK and ROK antagonists might block HPV by inhibiting Ca(2+) signaling, as well as the actin-myosin interaction, in PASMC. If effects on Ca(2+) signaling were due to decreased phosphorylated myosin light chain concentration, their diversity suggests that MLCK and ROK antagonists may have acted by inhibiting myosin motors and/or altering the cytoskeleton in a manner that prevented achievement of required spatial relationships among the cellular components of the response.
Collapse
Affiliation(s)
- Jian Wang
- Division of Pulmonary & Critical Care Medicine, The Johns Hopkins Asthma and Allergy Center, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
35
|
Shimada S, Sunagawa M, Hanashiro K, Nakamura M, Kosugi T. RNA interference targeting embryonic myosin heavy chain isoform inhibited mRNA expressions of phenotype markers in rabbit cultured vascular smooth muscle cells. Heart Vessels 2007; 22:41-7. [PMID: 17285445 DOI: 10.1007/s00380-006-0929-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2005] [Accepted: 05/27/2006] [Indexed: 12/01/2022]
Abstract
To investigate whether the knockdown of SMemb gene expression induces phenotypic modulation of vascular smooth muscle (VSM) cells toward a contractile type, we constructed a siRNA targeting the 3' untranslated region (UTR) of SMemb gene (SMemb-siRNA). The SMemb-siRNA was introduced into cultured rabbit VSM cells for 48 h at 37 degrees C by the lipofection method. The mRNA expressions were estimated by comparative reverse transcription-polymerase chain reaction (RT-PCR). SMemb-siRNA significantly decreased the ratio of SMemb to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA expression in a dose-dependent manner (P < 0.01): 0 nM, 0.90 +/- 0.08; 100 nM, 0.43 +/- 0.07. Immunofluorescence and immunoblot analyses demonstrated that SMemb-siRNA markedly decreased SMemb protein expression to 56% +/- 7.8% (P < 0.01). Other MHC isoform (SM1 and SM2) mRNA expressions were not changed. The relative mRNA expressions of other phenotype markers (plasminogen activator inhibitor (PAI)-1 and beta-actin) were significantly decreased by SMemb-siRNA to 71% +/- 7.5% and 61% +/- 7.5%, respectively (P < 0.01). Expression of smooth muscle (SM) alpha-actin protein and cell proliferation was not changed by SMemb-siRNA. Thus, SMemb gene might be involved in the transcription of PAI-1 and beta-actin, but not involved in SM alpha-actin and cell proliferation in cultured VSM.
Collapse
Affiliation(s)
- Seiji Shimada
- First Department of Physiology, Unit of Physiological Science, School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
| | | | | | | | | |
Collapse
|
36
|
Kim JH, Rhee PL, Kang TM. Actin cytoskeletons regulate the stretch-induced increase of Ca2+ current in human gastric myocytes. Biochem Biophys Res Commun 2006; 352:503-8. [PMID: 17126300 DOI: 10.1016/j.bbrc.2006.11.051] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2006] [Accepted: 11/10/2006] [Indexed: 10/23/2022]
Abstract
Using the whole-cell and single channel recording techniques, the influence of actin cytoskeletons on L-type Ca2+ current was investigated in human gastric smooth muscle cells. In isotonic condition, an actin depolymerizer cytochalasin D (Cyt-D) markedly decreased the whole-cell current (I(Ba)) without changing steady-state voltage dependency and single channel conductance. Intracellular dialysis of phalloidin, an actin polymerizer, significantly increased the I(Ba). Hypotonic stretch (222 mOsm/L) of the myocytes increased the I(Ba), and Cyt-D significantly inhibited the I(Ba) increase by the stretch. Phalloidin was without effect on the I(Ba) increase by the stretch. Phalloidin antagonized the Cyt-D inhibition of the stretch-induced I(Ba) increase. Neither heterotrimeric G protein modifiers (GTPgammaS and GDPbetaS) nor rho GTPase inhibitor (C3 exoenzyme) influenced the stretch-induced responses. These results reveal that the integrity of the actin cytoskeleton is an important factor which determines the activity of L-type Ca2+ channels and a response to stretch.
Collapse
Affiliation(s)
- Jun Hee Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon 440-746, Republic of Korea
| | | | | |
Collapse
|
37
|
Pica-Mattoccia L, Valle C, Basso A, Troiani AR, Vigorosi F, Liberti P, Festucci A, Cioli D. Cytochalasin D abolishes the schistosomicidal activity of praziquantel. Exp Parasitol 2006; 115:344-51. [PMID: 17083933 DOI: 10.1016/j.exppara.2006.09.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2006] [Revised: 09/22/2006] [Accepted: 09/23/2006] [Indexed: 11/22/2022]
Abstract
To test the hypothesis that calcium channels of schistosomes are the targets for the action of praziquantel, we subjected schistosomes in vitro to pharmacological agents capable of interfering with the functioning of calcium channels. After 1-h exposure to these agents, praziquantel was added and incubation continued overnight. Worms were then washed, resuspended in drug-free medium and observed during the following 7-10 days. About 50% of schistosomes pre-exposed to the calcium channel blockers nicardipine and nifedipine were able to survive a praziquantel concentration (3 microM) that normally killed the majority of adult male worms. Since the organization of the actin cytoskeleton controls the activity of calcium channels in a number of different systems, we also pre-exposed schistosomes to the actin depolymerizing agent cytochalasin D. This treatment rendered the parasites completely refractory to the effects of very high praziquantel levels (up to 36 microM). These results are consistent with the hypothesis that schistosome calcium channels are involved in the mechanism of action of praziquantel.
Collapse
|
38
|
Zhang J, Ryder KD, Bethel JA, Ramirez R, Duncan RL. PTH-induced actin depolymerization increases mechanosensitive channel activity to enhance mechanically stimulated Ca2+ signaling in osteoblasts. J Bone Miner Res 2006; 21:1729-37. [PMID: 17002579 DOI: 10.1359/jbmr.060722] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
UNLABELLED Disruption of the actin cytoskeleton with cytochalasin D enhanced the mechanically induced increase in intracellular Ca(2+) ([Ca(2+)](i)) in osteoblasts in a manner similar to that of PTH. Stabilization of actin with phalloidin prevented the PTH enhanced [Ca(2+)](i) response to shear. Patch-clamp analyses show that the MSCC is directly influenced by alterations in actin integrity. INTRODUCTION PTH significantly enhances the fluid shear-induced increase in [Ca(2+)](i) in osteoblasts, in part, through increased activation of both the mechanosensitive, cation-selective channel (MSCC) and L-type voltage-sensitive Ca(2+) channel (L-VSCC). Both stimuli have been shown to produce dynamic changes in the organization of the actin cytoskeleton. In this study, we examined the effects of alterations in actin polymerization on [Ca(2+)](i) and MSCC activity in MC3T3-E1 and UMR-106.01 osteoblasts in response to shear +/- PTH pretreatment. MATERIALS AND METHODS MC3T3-E1 or UMR-106.01 cells were plated onto type I collagen-coated quartz slides, allowed to proliferate to 60% confluency, and mounted on a modified parallel plate chamber and subjected to 12 dynes/cm(2). For patch-clamp studies, cells were plated on collagen-coated glass coverslips, mounted on the patch chamber, and subjected to pipette suction. Modulators of actin cytoskeleton polymerization were added 30 minutes before the experiments, whereas channel inhibitors were added 10 minutes before mechanical stimulation. All drugs were maintained in the flow medium for the duration of the experiment. RESULTS AND CONCLUSIONS Depolymerization of actin with 1-5 microM cytochalasin D (cyto D) augmented the peak [Ca(2+)](i) response and increased the number of cells responding to shear, similar to the increased responses induced by pretreatment with 50 nM PTH. Stabilization of actin with phalloidin prevented the PTH enhanced [Ca(2+)](i) response to shear. Inhibition of the MSCC with Gd(3+) significantly blocked both the peak Ca(2+) response and the number of cells responding to shear in cells pretreated with either PTH or cyto D. Inhibition of the L-VSCC reduced the peak [Ca(2+)](i) response to shear in cells pretreated with PTH, but not with cyto D. Patch-clamp analyses found that addition of PTH or cyto D significantly increased the MSCC open probability in response to mechanical stimulation, whereas phalloidin significantly attenuated the PTH-enhanced MSCC activation. These data indicate that actin reorganization increases MSCC activity in a manner similar to PTH and may be one mechanism through which PTH may reduce the mechanical threshold of osteoblasts.
Collapse
Affiliation(s)
- Jinsong Zhang
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | | | |
Collapse
|
39
|
Ben-Tabou De-Leon S, Ben-Zeev G, Nussinovitch I. Effects of osmotic shrinkage on voltage-gated Ca2+ channel currents in rat anterior pituitary cells. Am J Physiol Cell Physiol 2006; 290:C222-32. [PMID: 16148035 DOI: 10.1152/ajpcell.00118.2005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Increased extracellular osmolarity ([Os]e) suppresses stimulated hormone secretion from anterior pituitary cells. Ca2+ influx may mediate this effect. We show that increase in [Os]e (by 18–125%) differentially suppresses L-type and T-type Ca2+ channel currents ( IL and IT, respectively); IL was more sensitive than IT. Hyperosmotic suppression of IL depended on the magnitude of increase in [Os]e and was correlated with the percent decrease in pituitary cell volume, suggesting that pituitary cell shrinkage can modulate L-type currents. The hyperosmotic suppression of IL and IT persisted after incubation of pituitary cells either with the actin-disrupter cytochalasin D or with the actin stabilizer phalloidin, suggesting that the actin cytoskeleton is not involved in this modulation. The hyperosmotic suppression of Ca2+ influx was not correlated with changes in reversal potential, membrane capacitance, and access resistance. Together, these results suggest that the hyperosmotic suppression of Ca2+ influx involves Ca2+ channel proteins. We therefore recorded the activity of L-type Ca2+ channels from cell-attached patches while exposing the cell outside the patch pipette to hyperosmotic media. Increased [Os]e reduced the activity of Ca2+ channels but did not change single-channel conductance. This hyperosmotic suppression of Ca2+ currents may therefore contribute to the previously reported hyperosmotic suppression of hormone secretion.
Collapse
Affiliation(s)
- Shlomo Ben-Tabou De-Leon
- Dept. of Anatomy and Cell Biology, Hebrew Univ. Medical School, PO Box 12272, Jerusalem 91120, Israel
| | | | | |
Collapse
|
40
|
Fatima S, Yaghini FA, Pavicevic Z, Kalyankrishna S, Jafari N, Luong E, Estes A, Malik KU. Intact actin filaments are required for cytosolic phospholipase A2 translocation but not for its activation by norepinephrine in vascular smooth muscle cells. J Pharmacol Exp Ther 2005; 313:1017-26. [PMID: 15705737 DOI: 10.1124/jpet.104.081992] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cytosolic phospholipase A(2) (cPLA(2)) is activated and translocated to the nuclear envelope by various vasoactive agents, including norepinephrine (NE), and releases arachidonic acid (AA) from tissue phospholipids. We previously demonstrated that NE-induced cPLA(2) translocation to the nuclear envelope is mediated via its phosphorylation by calcium/calmodulin-dependent kinase-II in rabbit vascular smooth muscle cells (VSMCs). Cytoskeletal structures actin and microtubule filaments have been implicated in the trafficking of proteins to various cellular sites. This study was conducted to investigate the contribution of actin and microtubule filaments to cPLA(2) translocation to the nuclear envelope and its activation by NE in rabbit VSMCs. NE (10 microM) caused cPLA(2) translocation to the nuclear envelope as determined by immunofluorescence. Cytochalasin D (CD; 0.5 microM) and latrunculin A (LA; 0.5 microM) that disrupted actin filaments, blocked cPLA(2) translocation elicited by NE. On the other hand, disruption of microtubule filaments by 10 microM colchicine did not block NE-induced cPLA(2) translocation to the nuclear envelope. CD and LA did not inhibit NE-induced increase in cytosolic calcium and cPLA(2) activity, determined from the hydrolysis of l-1-[(14)C]arachidonyl phosphatidylcholine and release of AA. Coimmunoprecipitation studies showed an association of actin with cPLA(2), which was not altered by CD or LA. Far-Western analysis showed that cPLA(2) interacts directly with actin. Our data suggest that NE-induced cPLA(2) translocation to the nuclear envelope requires an intact actin but not microtubule filaments and that cPLA(2) phosphorylation and activation and AA release are independent of its translocation to the nuclear envelope in rabbit VSMCs.
Collapse
Affiliation(s)
- Soghra Fatima
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, 38163, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Morales S, Camello PJ, Rosado JA, Mawe GM, Pozo MJ. Disruption of the filamentous actin cytoskeleton is necessary for the activation of capacitative calcium entry in naive smooth muscle cells. Cell Signal 2005; 17:635-45. [PMID: 15683738 DOI: 10.1016/j.cellsig.2004.10.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2004] [Accepted: 10/11/2004] [Indexed: 11/23/2022]
Abstract
It has been proposed that cytoskeleton plays a key positive role in the activation of capacitative calcium entry (CCE), which supported the secretion-like hypothesis for the mechanisms underlying this process. However, its role on CCE in native smooth muscle is unknown. Here we demonstrate that CCE in isolated gallbladder myocytes was enhanced by cytochalasin D or latrunculin A treatments (agents that cause actin disassembly) whereas it was reduced by jasplakinolide treatment (which causes actin polymerization), suggesting that actin cytoskeleton acts as a barrier in CCE. In addition, we show for the first time that depletion of intracellular Ca2+ stores by thapsigargin and cholecystokinin in BAPTA-loaded cells induced a decrease in F-actin content that was consistent with a link between CCE and actin reorganization. In conclusion, these data suggest an active participation of actin reorganization in the implementation of CCE and support a conformational coupling model for this process in naive smooth muscle cells.
Collapse
Affiliation(s)
- Sara Morales
- Department of Physiology, University of Extremadura, 10071 Cáceres, Spain
| | | | | | | | | |
Collapse
|
42
|
Kempson SA, Montrose MH. Osmotic regulation of renal betaine transport: transcription and beyond. Pflugers Arch 2005; 449:227-34. [PMID: 15452713 DOI: 10.1007/s00424-004-1338-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cells in the kidney inner medulla are routinely exposed to high extracellular osmolarity during normal operation of the urinary concentrating mechanism. One adaptation critical for survival in this environment is the intracellular accumulation of organic osmolytes to balance the osmotic stress. Betaine is an important osmolyte that is accumulated via the betaine/gamma-aminobutyric acid transporter (BGT1) in the basolateral plasma membrane of medullary epithelial cells. In response to hypertonic stress, there is transcriptional activation of the BGT1 gene, followed by trafficking and membrane insertion of BGT1 protein. Transcriptional activation, triggered by changes in ionic strength and water content, is an early response that is a key regulatory step and has been studied in detail. Recent studies suggest there are additional post-transcriptional regulatory steps in the pathway leading to upregulation of BGT1 transport, and that additional proteins are required for membrane insertion. Reversal of this adaptive process, upon removal of hypertonic stress, involves a rapid efflux of betaine through specific release pathways, a reduction in betaine influx, and a slower downregulation of BGT1 protein abundance. There is much more to be learned about many of these steps in BGT1 regulation.
Collapse
Affiliation(s)
- Stephen A Kempson
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Medical Sciences Bldg., Room 309, 635 Barnhill Drive, Indianapolis, IN 46202-5120, USA.
| | | |
Collapse
|
43
|
Hagihara H, Yoshikawa Y, Ohga Y, Takenaka C, Murata KY, Taniguchi S, Takaki M. Na+/Ca2+ exchange inhibition protects the rat heart from ischemia-reperfusion injury by blocking energy-wasting processes. Am J Physiol Heart Circ Physiol 2005; 288:H1699-707. [PMID: 15626686 DOI: 10.1152/ajpheart.01033.2004] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have recently reported that exposure of rat hearts to high Ca2+ produces a Ca2+ overload-induced contractile failure in rat hearts, which was associated with proteolysis of α-fodrin. We hypothesized that contractile failure after ischemia-reperfusion (I/R) is similar to that after high Ca2+ infusion. To test this hypothesis, we investigated left ventricular (LV) mechanical work and energetics in the cross-circulated rat hearts, which were subjected to 15 min global ischemia and 60 min reperfusion. Sixty minutes after I/R, mean systolic pressure-volume area (PVA; a total mechanical energy per beat) at midrange LV volume (mLVV) (PVAmLVV) was significantly decreased from 5.89 ± 1.55 to 3.83 ± 1.16 mmHg·ml·beat−1·g−1 ( n = 6). Mean myocardial oxygen consumption per beat (Vo2) intercept of (Vo2-PVA linear relation was significantly decreased from 0.21 ± 0.05 to 0.15 ± 0.03 μl O2·beat−1·g−1 without change in its slope. Initial 30-min reperfusion with a Na+/Ca2+ exchanger (NCX) inhibitor KB-R7943 (KBR; 10 μmol/l) significantly reduced the decrease in mean PVAmLVV and Vo2 intercept ( n = 6). Although Vo2 for the Ca2+ handling was finally decreased, it transiently but significantly increased from the control for 10–15 min after I/R. This increase in Vo2 for the Ca2+ handling was completely blocked by KBR, suggesting an inhibition of reverse-mode NCX by KBR. α-Fodrin proteolysis, which was significantly increased after I/R, was also significantly reduced by KBR. Our study shows that the contractile failure after I/R is similar to that after high Ca2+ infusion, although the contribution of reverse-mode NCX to the contractile failure is different. An inhibition of reverse-mode NCX during initial reperfusion protects the heart against reperfusion injury.
Collapse
Affiliation(s)
- Hiroji Hagihara
- Dept. of Physiology II, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
Yoshikawa Y, Hagihara H, Ohga Y, Nakajima-Takenaka C, Murata KY, Taniguchi S, Takaki M. Calpain inhibitor-1 protects the rat heart from ischemia-reperfusion injury: analysis by mechanical work and energetics. Am J Physiol Heart Circ Physiol 2005; 288:H1690-8. [PMID: 15528229 DOI: 10.1152/ajpheart.00666.2004] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We hypothesized that calpain inhibitor-1 protected left ventricular (LV) function from ischemia-reperfusion injury by inhibiting the proteolysis of α-fodrin. To test this hypothesis, we investigated the effect of calpain inhibitor-1 on LV mechanical work and energetics in the cross-circulated rat hearts that underwent 15-min global ischemia and 60-min reperfusion ( n = 9). After ischemia-reperfusion with calpain inhibitor-1, mean end-systolic pressure at midrange LV volume and systolic pressure-volume area (PVA) at midrange LV volume (total mechanical energy per beat) were hardly changed, although they were significantly ( P < 0.01) decreased after ischemia-reperfusion without calpain inhibitor-1. Mean myocardial oxygen consumption per beat (Vo2) intercepts (PVA-independent Vo2; Vo2 for the total Ca2+ handling in excitation-contraction coupling and basal metabolism) of Vo2-PVA linear relations were also unchanged after ischemia-reperfusion with calpain inhibitor-1, although they were significantly ( P < 0.01) decreased after ischemia-reperfusion without calpain inhibitor-1. There were no significant differences in O2 costs of LV PVA and contractility among the hearts in control (or normal) postischemia-reperfusion and postischemia-reperfusion with calpain inhibitor-1. Western blot analysis of α-fodrin and the immunostaining of 150-kDa products of α-fodrin confirmed that calpain inhibitor-1 almost completely protected the proteolysis of α-fodrin. Our results indicate that calpain inhibitor-1 prevents the heart from ischemia-reperfusion injury associated with the impairment of total Ca2+ handling by directly inhibiting the proteolysis of α-fodrin.
Collapse
Affiliation(s)
- Yoshiro Yoshikawa
- Dept. of Physiology II, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Zhao Y, Xiao H, Long W, Pearce WJ, Longo LD. Expression of several cytoskeletal proteins in ovine cerebral arteries: developmental and functional considerations. J Physiol 2004; 558:623-32. [PMID: 15181158 PMCID: PMC1664981 DOI: 10.1113/jphysiol.2004.064220] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cytoskeleton proteins play important roles in regulating vascular smooth muscle (VSM) contraction and relaxation. We tested the hypotheses that the expression levels of several of these proteins change significantly during the course of development, and that these changes contribute to age-related changes in contractile responses. In cerebral arteries from 95-day (d) gestation and 140-d fetus, newborn lambs, and adult sheep, by Western immunoblot (n= 5 for each age) we quantified the relative expression of alpha-actin, alpha-tubulin, cyclophilin A, and proliferating cell nuclear antigen (PCNA). In addition, we examined middle cerebral artery tension responses to phenylephrine (PHE) stimulation in the absence or presence of cytochalasin D (3 x 10(-7)m) and nocodazole (3 x 10(-6)m), inhibitors of alpha-actin and alpha-tubulin polymerization, respectively. The expression levels of alpha-actin and cyclophilin A varied little during the course of development. In contrast, alpha-tubulin expression was approximately 2.5-fold greater in both fetal age groups as compared to adult. Also, as compared to adult and as expected, expression of PCNA was several-fold greater in cerebral arteries of the 95-d fetus (x8), 140-d fetus (x 5), and newborn (x 3). In both adult and fetal middle cerebral artery, cytochalasin D-induced inhibition of actin polymerization decreased PHE-induced contraction, to approximately 60 and approximately 40% of control, respectively (despite no significant change in expression level). In contrast, alpha-tubulin inhibition by nocodazole showed little effect on PHE-induced tension (in spite of the age-related decrease in expression). In conclusion, expression levels of alpha-actin, a thin filament protein involved in contraction, remained relatively constant during the course of development, as did the effects of inhibition of its polymerization on contractility. In contrast, alpha-tubulin, important in intracellular protein trafficking, showed a significant age-related decrease in expression and played a relatively minor role in contractility. The present studies suggest that other cytoskeletal structural proteins and/or elements of pharmaco-mechanical coupling are important to developmental differences in cerebrovascular contractility. In addition, the relatively constant expression levels of alpha-actin and cyclophilin A with development, suggest that these are useful internal standards for studies of cytosolic protein expression.
Collapse
Affiliation(s)
- Yu Zhao
- Center for Perinatal Biology, Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | | | | | | | | |
Collapse
|
46
|
Alvarez J, Hamplova J, Hohaus A, Morano I, Haase H, Vassort G. Calcium Current in Rat Cardiomyocytes Is Modulated by the Carboxyl-terminal Ahnak Domain. J Biol Chem 2004; 279:12456-61. [PMID: 14722071 DOI: 10.1074/jbc.m312177200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ahnak, a protein of 5643 amino acids, interacts with the regulatory beta-subunit of cardiac calcium channels and with F-actin. Recently, we defined the binding sites among the protein partners in the carboxyl-terminal domain of ahnak. Here we further narrowed down the beta(2)-interaction sites to the carboxyl-terminal 188 amino acids of ahnak by the recombinant ahnak protein fragments P3 (amino acids 5456-5556) and P4 (amino acids 5556-5643). The effects of these P3 and P4 fragments on the calcium current were investigated under whole-cell patch clamp conditions on rat ventricular cardiomyocytes. P4 but not P3 increased significantly the current amplitude by 22.7 +/- 5% without affecting its voltage dependence. The slow component of calcium current inactivation was slowed down by both P3 and P4, whereas only P3 slowed significantly the fast one. The composite recombinant protein fragment P3-P4 induced similar modifications to the ones induced by each of the ahnak fragments. In the presence of carboxyl-terminal ahnak protein fragments, isoprenaline induced a similar relative increase in current amplitude and shift in current kinetics. The actin-stabilizing agents, phalloidin and jasplakinolide, did not modify the effects of these ahnak protein fragments on calcium current in control conditions nor in the presence of isoprenaline. Hence, our results suggest that the functional effects of P3, P4, and P3-P4 on calcium current are mediated by targeting the ahnak-beta(2)-subunit interaction rather than by targeting the ahnak-F-actin interaction. More specifically they suggest that binding of the beta(2)-subunit to the endogenous subsarcolemmal giant ahnak protein re-primes the alpha(1C)/beta(2)-subunit interaction and that the ahnak-derived proteins relieve the beta(2)-subunit from this inhibition.
Collapse
Affiliation(s)
- Julio Alvarez
- Physiopathologie Cardiovasculaire, INSERM U-390, CHU Arnaud de Villeneuve, F-34295 Montpellier Cedex 5, France
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Studies on left ventricular mechanical work and energetics in rat and mouse hearts are reviewed. First, left ventricular linear end-systolic pressure-volume relation (ESPVR) and curved end-diastolic pressure-volume relation (EDPVR) in canine hearts and left ventricular curved ESPVR and curved EDPVR in rat hearts are reviewed. Second, as an index for total mechanical energy per beat in rat hearts as in canine hearts, a systolic pressure-volume area (PVA) is proposed. By the use of our original system for measuring continuous oxygen consumption for rat left ventricular mechanical work, the linear left ventricular myocardial oxygen consumption per beat (VO2)-PVA relation is obtained as in canine hearts. The slope of VO2-PVA relation (oxygen cost of PVA) indicates a ratio of chemomechanical energy transduction. VO2 intercept (PVA-independent VO2) indicates the summation of oxygen consumption for Ca2+ handling in excitation-contraction coupling and for basal metabolism. An equivalent maximal elastance (eEmax) is proposed as a new left ventricular contractility index based on PVA at the midrange left ventricular volume. The slope of the linear relation between PVA-independent VO2 and eEmax (oxygen cost of eEmax) indicates changes in oxygen consumption for Ca2+ handling in excitation-contraction coupling per unit changes in left ventricular contractility. The key framework of VO2-PVA-eEmax can give us a better understanding for the biology and mechanisms of physiological and various failing rat heart models in terms of mechanical work and energetics.
Collapse
Affiliation(s)
- M Takaki
- Department of Physiology II, Nara Medical University, Kashihara, Nara, 634-8521 Japan.
| |
Collapse
|
48
|
Schubert T, Akopian A. Actin filaments regulate voltage-gated ion channels in salamander retinal ganglion cells. Neuroscience 2004; 125:583-90. [PMID: 15099672 DOI: 10.1016/j.neuroscience.2004.02.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2004] [Indexed: 11/28/2022]
Abstract
The regulation of voltage-activated K(+), and Ca(2+) currents by actin filaments was studied in salamander retinal ganglion cells, using the whole-cell patch clamp technique and Ca(2+) imaging. Disruption of F-actin by cytochalasin B or latrunculin B resulted in a reduction of L-type Ca(2+) current by 55+/-4%, and a sustained outward K(+) current (I(k)) by 41+/-3%. The effect was diminished when the F-actin stabilizing agent phalloidin was present in the patch pipette. In a group of cells where I(K) exhibited a small degree of inactivation, the effect of F-actin disruption on current was dual; it increased it by 89+/-16%, at -10 mV, and reduced it by 37+/-5% at +50 mV voltage step from the same holding potential of -70 mV. This was accompanied by a shift in a voltage of half-maximal activation toward negative potentials by approximately 20 mV. In Ca(2+) imaging experiments, 30 min incubation of isolated neurons with latrunculin A reduced a depolarization-induced Ca(2+) accumulation by 45+/-5%. These results suggest a role for the actin cytoskeleton in regulating voltage-gated ion channels in retinal ganglion cells.
Collapse
Affiliation(s)
- T Schubert
- Department of Neurobiology, University of Oldenburg, Oldenburg, Germany
| | | |
Collapse
|
49
|
Geeraert V, Dupont JL, Grant NJ, Huvet C, Chasserot-Golaz S, Janoshazi A, Procksch O, de Barry J. F-actin does not modulate the initial steps of the protein kinase C activation process in living nerve cells. Exp Cell Res 2003; 289:222-36. [PMID: 14499623 DOI: 10.1016/s0014-4827(03)00267-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Actin is a major substrate for protein kinase C (PKC) and PKC is considered a modulator of the actin network. In addition in vitro studies (Biochemistry 39 (2000) 271) have suggested that all PKC isoforms bind to actin during the process of activation of the enzyme. To test the physiological significance of such a coupling we used living PC12 cells and primary cultures of cerebellar granule cells. When PC12 cells were treated with either latrunculin B, which impairs actin polymerization, or phalloidin, which stabilizes actin filaments, we observed a significant reduction of the [Ca2+]i response revealed by Fura-2 fluorescence, while the PKC conformational changes followed by Fim-1 fluorescence were unaffected. The responses induced either by cell depolarization or muscarinic receptor activation were similarly affected by the toxin treatment of PC12 cells. In cerebellar granule cells the [Ca2+]i response induced by KCl depolarization was increased by latrunculin treatment, whereas no effect was observed on the PKC response. Latrunculin had no effect on the NMDA-induced responses in these cells. Finally we also show that the response induced by a long-lasting depolarization, which mimics stimulation leading to neuronal plasticity, was not significantly altered by latrunculin or phalloidin treatment of the cells. These results suggest that the actin network is not involved in the initial steps of the PKC activation process in living nerve cells.
Collapse
Affiliation(s)
- Virginie Geeraert
- Neurotransmission et Secretion Neuroendocrine, UPR 2356 CNRS, 5 rue B Pascal, F-67084 Strasbourg Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Ben-Tabou De-Leon S, Blotnick E, Nussinovitch I. Effects of osmotic swelling on voltage-gated calcium channel currents in rat anterior pituitary cells. Am J Physiol Cell Physiol 2003; 285:C840-52. [PMID: 12773317 DOI: 10.1152/ajpcell.00101.2003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Decrease in extracellular osmolarity ([Os]e) results in stimulation of hormone secretion from pituitary cells. Different mechanisms can account for this stimulation of hormone secretion. In this study we examined the possibility that hyposmolarity directly modulates voltage-gated calcium influx in pituitary cells. The effects of hyposmolarity on L-type (IL) and T-type (IT) calcium currents in pituitary cells were investigated by using two hyposmotic stimuli, moderate (18-22% decrease in [Os]e) and strong (31-32% decrease in [Os]e). Exposure to moderate hyposmotic stimuli resulted in three response types in IL (a decrease, a biphasic effect, and an increase in IL) and in increase in IT. Exposure to strong hyposmotic stimuli resulted only in increases in both IL and IT. Similarly, in intact pituitary cells (perforated patch method), exposure to either moderate or strong hyposmotic stimuli resulted only in increases in both IL and IT. Thus it appears that the main effect of decrease in [Os]e is increase in calcium channel currents. This increase was differential (IL were more sensitive than IT) and voltage independent. In addition, we show that these hyposmotic effects cannot be explained by activation of an anionic conductance or by an increase in cell membrane surface area. In conclusion, this study shows that hyposmotic swelling of pituitary cells can directly modulate voltage-gated calcium influx. This hyposmotic modulation of IL and IT may contribute to the previously reported hyposmotic stimulation of hormone secretion. The mechanisms underlying these hyposmotic effects and their possible physiological relevance are discussed.
Collapse
Affiliation(s)
- Shlomo Ben-Tabou De-Leon
- Department of Anatomy and Cell Biology, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel
| | | | | |
Collapse
|