1
|
Akin EJ, Aoun J, Jimenez C, Mayne K, Baeck J, Young MD, Sullivan B, Sanders KM, Ward SM, Bulley S, Jaggar JH, Earley S, Greenwood IA, Leblanc N. ANO1, CaV1.2, and IP3R form a localized unit of EC-coupling in mouse pulmonary arterial smooth muscle. J Gen Physiol 2023; 155:e202213217. [PMID: 37702787 PMCID: PMC10499037 DOI: 10.1085/jgp.202213217] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/01/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Pulmonary arterial (PA) smooth muscle cells (PASMC) generate vascular tone in response to agonists coupled to Gq-protein receptor signaling. Such agonists stimulate oscillating calcium waves, the frequency of which drives the strength of contraction. These Ca2+ events are modulated by a variety of ion channels including voltage-gated calcium channels (CaV1.2), the Tmem16a or Anoctamin-1 (ANO1)-encoded calcium-activated chloride (CaCC) channel, and Ca2+ release from the sarcoplasmic reticulum through inositol-trisphosphate receptors (IP3R). Although these calcium events have been characterized, it is unclear how these calcium oscillations underly a sustained contraction in these muscle cells. We used smooth muscle-specific ablation of ANO1 and pharmacological tools to establish the role of ANO1, CaV1.2, and IP3R in the contractile and intracellular Ca2+ signaling properties of mouse PA smooth muscle expressing the Ca2+ biosensor GCaMP3 or GCaMP6. Pharmacological block or genetic ablation of ANO1 or inhibition of CaV1.2 or IP3R, or Ca2+ store depletion equally inhibited 5-HT-induced tone and intracellular Ca2+ waves. Coimmunoprecipitation experiments showed that an anti-ANO1 antibody was able to pull down both CaV1.2 and IP3R. Confocal and superresolution nanomicroscopy showed that ANO1 coassembles with both CaV1.2 and IP3R at or near the plasma membrane of PASMC from wild-type mice. We conclude that the stable 5-HT-induced PA contraction results from the integration of stochastic and localized Ca2+ events supported by a microenvironment comprising ANO1, CaV1.2, and IP3R. In this model, ANO1 and CaV1.2 would indirectly support cyclical Ca2+ release events from IP3R and propagation of intracellular Ca2+ waves.
Collapse
Affiliation(s)
- Elizabeth J. Akin
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Joydeep Aoun
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Connor Jimenez
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Katie Mayne
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Julius Baeck
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Michael D. Young
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Brennan Sullivan
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Sean M. Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Simon Bulley
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jonathan H. Jaggar
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Scott Earley
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Iain A. Greenwood
- Department of Vascular Pharmacology, Molecular and Clinical Science Research Institute, St. George’s University of London, London, UK
| | - Normand Leblanc
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| |
Collapse
|
2
|
Sanchez-Alonso JL, Fedele L, Copier JS, Lucarelli C, Mansfield C, Judina A, Houser SR, Brand T, Gorelik J. Functional LTCC-β 2AR Complex Needs Caveolin-3 and Is Disrupted in Heart Failure. Circ Res 2023; 133:120-137. [PMID: 37313722 PMCID: PMC10321517 DOI: 10.1161/circresaha.123.322508] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND Beta-2 adrenergic receptors (β2ARs) but not beta-2 adrenergic receptors (β1ARs) form a functional complex with L-type Ca2+ channels (LTCCs) on the cardiomyocyte membrane. However, how microdomain localization in the plasma membrane affects the function of these complexes is unknown. We aim to study the coupling between LTCC and β adrenergic receptors in different cardiomyocyte microdomains, the distinct involvement of PKA and CAMKII (Ca2+/calmodulin-dependent protein kinase II) and explore how this functional complex is disrupted in heart failure. METHODS Global signaling between LTCCs and β adrenergic receptors was assessed with whole-cell current recordings and western blot analysis. Super-resolution scanning patch-clamp was used to explore the local coupling between single LTCCs and β1AR or β2AR in different membrane microdomains in control and failing cardiomyocytes. RESULTS LTCC open probability (Po) showed an increase from 0.054±0.003 to 0.092±0.008 when β2AR was locally stimulated in the proximity of the channel (<350 nm) in the transverse tubule microdomain. In failing cardiomyocytes, from both rodents and humans, this transverse tubule coupling between LTCC and β2AR was lost. Interestingly, local stimulation of β1AR did not elicit any change in the Po of LTCCs, indicating a lack of proximal functional interaction between the two, but we confirmed a general activation of LTCC via β1AR. By using blockers of PKA and CaMKII and a Caveolin-3-knockout mouse model, we conclude that the β2AR-LTCC regulation requires the presence of caveolin-3 and the activation of the CaMKII pathway. By contrast, at a cellular "global" level PKA plays a major role downstream β1AR and results in an increase in LTCC current. CONCLUSIONS Regulation of the LTCC activity by proximity coupling mechanisms occurs only via β2AR, but not β1AR. This may explain how β2ARs tune the response of LTCCs to adrenergic stimulation in healthy conditions. This coupling is lost in heart failure; restoring it could improve the adrenergic response of failing cardiomyocytes.
Collapse
Affiliation(s)
- Jose L. Sanchez-Alonso
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Laura Fedele
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Jaël S. Copier
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Carla Lucarelli
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Catherine Mansfield
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Aleksandra Judina
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Steven R. Houser
- Department of Physiology, Cardiovascular Research Center, Lewis Katz Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Thomas Brand
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| |
Collapse
|
3
|
Anderson JR, Morin EE, Brayer KJ, Salbato S, Gonzalez Bosc LV, Kanagy NL, Naik JS. Single-cell transcriptomic heterogeneity between conduit and resistance mesenteric arteries in rats. Physiol Genomics 2023; 55:179-193. [PMID: 36912534 PMCID: PMC10085562 DOI: 10.1152/physiolgenomics.00126.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/03/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
The endothelium contains morphologically similar cells throughout the vasculature, but individual cells along the length of a single vascular tree or in different regional circulations function dissimilarly. When observations made in large arteries are extrapolated to explain the function of endothelial cells (ECs) in the resistance vasculature, only a fraction of these observations are consistent between artery sizes. To what extent endothelial (EC) and vascular smooth muscle cells (VSMCs) from different arteriolar segments of the same tissue differ phenotypically at the single-cell level remains unknown. Therefore, single-cell RNA-seq (10x Genomics) was performed using a 10X Genomics Chromium system. Cells were enzymatically digested from large (>300 µm) and small (<150 µm) mesenteric arteries from nine adult male Sprague-Dawley rats, pooled to create six samples (3 rats/sample, 3 samples/group). After normalized integration, the dataset was scaled before unsupervised cell clustering and cluster visualization using UMAP plots. Differential gene expression analysis allowed us to infer the biological identity of different clusters. Our analysis revealed 630 and 641 differentially expressed genes (DEGs) between conduit and resistance arteries for ECs and VSMCs, respectively. Gene ontology analysis (GO-Biological Processes, GOBP) of scRNA-seq data discovered 562 and 270 pathways for ECs and VSMCs, respectively, that differed between large and small arteries. We identified eight and seven unique ECs and VSMCs subpopulations, respectively, with DEGs and pathways identified for each cluster. These results and this dataset allow the discovery and support of novel hypotheses needed to identify mechanisms that determine the phenotypic heterogeneity between conduit and resistance arteries.
Collapse
Affiliation(s)
- Jacob R Anderson
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Emily E Morin
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Kathryn J Brayer
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Sophia Salbato
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Laura V Gonzalez Bosc
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Nancy L Kanagy
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Jay S Naik
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| |
Collapse
|
4
|
van der Horst J, Rognant S, Abbott GW, Ozhathil LC, Hägglund P, Barrese V, Chuang CY, Jespersen T, Davies MJ, Greenwood IA, Gourdon P, Aalkjær C, Jepps TA. Dynein regulates Kv7.4 channel trafficking from the cell membrane. J Gen Physiol 2021; 153:211752. [PMID: 33533890 PMCID: PMC7863719 DOI: 10.1085/jgp.202012760] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/21/2020] [Accepted: 01/08/2021] [Indexed: 12/15/2022] Open
Abstract
The dynein motor protein transports proteins away from the cell membrane along the microtubule network. Recently, we found the microtubule network was important for regulating the membrane abundance of voltage-gated Kv7.4 potassium channels in vascular smooth muscle. Here, we aimed to investigate the influence of dynein on the microtubule-dependent internalization of the Kv7.4 channel. Patch-clamp recordings from HEK293B cells showed Kv7.4 currents were increased after inhibiting dynein function with ciliobrevin D or by coexpressing p50/dynamitin, which specifically interferes with dynein motor function. Mutation of a dynein-binding site in the Kv7.4 C terminus increased the Kv7.4 current and prevented p50 interference. Structured illumination microscopy, proximity ligation assays, and coimmunoprecipitation showed colocalization of Kv7.4 and dynein in mesenteric artery myocytes. Ciliobrevin D enhanced mesenteric artery relaxation to activators of Kv7.2–Kv7.5 channels and increased membrane abundance of Kv7.4 protein in isolated smooth muscle cells and HEK293B cells. Ciliobrevin D failed to enhance the negligible S-1–mediated relaxations after morpholino-mediated knockdown of Kv7.4. Mass spectrometry revealed an interaction of dynein with caveolin-1, confirmed using proximity ligation and coimmunoprecipitation assays, which also provided evidence for interaction of caveolin-1 with Kv7.4, confirming that Kv7.4 channels are localized to caveolae in mesenteric artery myocytes. Lastly, cholesterol depletion reduced the interaction of Kv7.4 with caveolin-1 and dynein while increasing the overall membrane expression of Kv7.4, although it attenuated the Kv7.4 current in oocytes and interfered with the action of ciliobrevin D and channel activators in arterial segments. Overall, this study shows that dynein can traffic Kv7.4 channels in vascular smooth muscle in a mechanism dependent on cholesterol-rich caveolae.
Collapse
Affiliation(s)
| | - Salomé Rognant
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA
| | | | - Per Hägglund
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vincenzo Barrese
- St. George's, University of London, London, UK.,Department of Neuroscience, Reproductive Science and Dentistry, University of Naples "Federico II," Naples, Italy
| | - Christine Y Chuang
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Jespersen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Pontus Gourdon
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Medical Sciences, Lund University, Lund, Sweden
| | - Christian Aalkjær
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Thomas A Jepps
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Sarkar J, Chakraborti T, Pramanik PK, Ghosh P, Mandal A, Chakraborti S. PKCζ-NADPH Oxidase-PKCα Dependent Kv1.5 Phosphorylation by Endothelin-1 Modulates Nav1.5-NCX1-Cav1.2 Axis in Stimulating Ca 2+ Level in Caveolae of Pulmonary Artery Smooth Muscle Cells. Cell Biochem Biophys 2020; 79:57-71. [PMID: 33095400 DOI: 10.1007/s12013-020-00954-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2020] [Indexed: 11/29/2022]
Abstract
Endothelin-1 (ET-1) is a potent endogenously derived vasoconstrictor, which increases pulmonary hypertension via stimulation of [Ca2+]i level in pulmonary artery smooth muscle cells (PASMCs). In this communication, we sought to investigate the mechanism by which ET-1 causes stimulation of Ca2+ concentration in caveolae vesicles of bovine PASMCs (BPASMCs). ET-1 activates PKC-α in the caveolae vesicles by O2.- derived from PKCζ-NADPH oxidase dependent pathway. PKC-α phosphorylates Kv1.5 channels leading to a marked stimulation of Na+ and Ca2+ concentration in the caveolae vesicles. The stimulation of Ca2+ concentration in the caveolae vesicles by ET-1 occurs predominantly via Cav1.2 channels. Additionally, an increase in Na+ concentration by ET-1 due to stimulation of Nav1.5 channels marginally increases Ca2+ level in the caveolae vesicles via reverse-mode Na+/Ca2+ exchanger (NCX-1) and also through "slip-mode conductance" Nav1.5 channels. 4-AP, a well-known inhibitor of Kv channels, also increases Ca2+ concentration in the caveolae vesicles via Cav1.2 channels, reverse-mode NCX-1 and Nav1.5 channels by phosphorylation independent modulation of Kv1.5 channels without the involvement of PKCζ-NADPH oxidase-PKCα signaling axis. Overall, PKCζ-NADPH oxidase-PKCα dependent phosphorylation of Kv1.5 by ET-1 modulates Nav1.5-NCX1-Cav1.2 axis for stimulation of Ca2+ concentration in caveolae vesicles of BPASMCs, which provides a crucial mechanism for better understanding of ET-1-mediated modulation of pulmonary vascular tone.
Collapse
Affiliation(s)
- Jaganmay Sarkar
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, West Bengal, 741235, India
| | - Tapati Chakraborti
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, West Bengal, 741235, India
| | - Pijush Kanti Pramanik
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, West Bengal, 741235, India
| | - Priyanka Ghosh
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, West Bengal, 741235, India
| | - Amritlal Mandal
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, West Bengal, 741235, India
| | - Sajal Chakraborti
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, West Bengal, 741235, India.
| |
Collapse
|
6
|
Abstract
Vascular smooth muscle cells (VSMCs) of small peripheral arteries contribute to blood pressure control by adapting their contractile state. These adaptations depend on the VSMC cytosolic Ca2+ concentration, regulated by complex local elementary Ca2+ signaling pathways. Ca2+ sparks represent local, transient, rapid calcium release events from a cluster of ryanodine receptors (RyRs) in the sarcoplasmic reticulum. In arterial SMCs, Ca2+ sparks activate nearby calcium-dependent potassium channels, cause membrane hyperpolarization and thus decrease the global intracellular [Ca2+] to oppose vasoconstriction. Arterial SMC Cav1.2 L-type channels regulate intracellular calcium stores content, which in turn modulates calcium efflux through RyRs. Cav3.2 T-type channels contribute to a minor extend to Ca2+ spark generation in certain types of arteries. Their localization within cell membrane caveolae is essential. We summarize present data on local elementary calcium signaling (Ca2+ sparks) in arterial SMCs with focus on RyR isoforms, large-conductance calcium-dependent potassium (BKCa) channels, and cell membrane-bound calcium channels (Cav1.2 and Cav3.2), particularly in caveolar microdomains.
Collapse
Affiliation(s)
- Gang Fan
- Charité - Universitätsmedizin Berlin, Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Yingqiu Cui
- Charité - Universitätsmedizin Berlin, Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Maik Gollasch
- Charité - Universitätsmedizin Berlin, Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Mario Kassmann
- Charité - Universitätsmedizin Berlin, Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| |
Collapse
|
7
|
Fan G, Kaßmann M, Cui Y, Matthaeus C, Kunz S, Zhong C, Zhu S, Xie Y, Tsvetkov D, Daumke O, Huang Y, Gollasch M. Age attenuates the T-type Ca V 3.2-RyR axis in vascular smooth muscle. Aging Cell 2020; 19:e13134. [PMID: 32187825 PMCID: PMC7189999 DOI: 10.1111/acel.13134] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 01/28/2020] [Accepted: 02/16/2020] [Indexed: 12/26/2022] Open
Abstract
Caveolae position CaV3.2 (T‐type Ca2+ channel encoded by the α‐3.2 subunit) sufficiently close to RyR (ryanodine receptors) for extracellular Ca2+ influx to trigger Ca2+ sparks and large‐conductance Ca2+‐activated K+ channel feedback in vascular smooth muscle. We hypothesize that this mechanism of Ca2+ spark generation is affected by age. Using smooth muscle cells (VSMCs) from mouse mesenteric arteries, we found that both Cav3.2 channel inhibition by Ni2+ (50 µM) and caveolae disruption by methyl‐ß‐cyclodextrin or genetic abolition of Eps15 homology domain‐containing protein (EHD2) inhibited Ca2+ sparks in cells from young (4 months) but not old (12 months) mice. In accordance, expression of Cav3.2 channel was higher in mesenteric arteries from young than old mice. Similar effects were observed for caveolae density. Using SMAKO Cav1.2−/− mice, caffeine (RyR activator) and thapsigargin (Ca2+ transport ATPase inhibitor), we found that sufficient SR Ca2+ load is a prerequisite for the CaV3.2‐RyR axis to generate Ca2+ sparks. We identified a fraction of Ca2+ sparks in aged VSMCs, which is sensitive to the TRP channel blocker Gd3+ (100 µM), but insensitive to CaV1.2 and CaV3.2 channel blockade. Our data demonstrate that the VSMC CaV3.2‐RyR axis is down‐regulated by aging. This defective CaV3.2‐RyR coupling is counterbalanced by a Gd3+ sensitive Ca2+ pathway providing compensatory Ca2+ influx for triggering Ca2+ sparks in aged VSMCs.
Collapse
Affiliation(s)
- Gang Fan
- Experimental and Clinical Research Center (ECRC) a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC) Charité – Universitätsmedizin Berlin Berlin Germany
- Hunan Cancer Hospital The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha China
| | - Mario Kaßmann
- Experimental and Clinical Research Center (ECRC) a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC) Charité – Universitätsmedizin Berlin Berlin Germany
| | - Yingqiu Cui
- Experimental and Clinical Research Center (ECRC) a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC) Charité – Universitätsmedizin Berlin Berlin Germany
| | - Claudia Matthaeus
- Crystallography Max‐Delbrück‐Center for Molecular Medicine Berlin Germany
| | - Séverine Kunz
- Electron Microscopy Facility Max Delbrück Center for Molecular Medicine (MDC) Berlin Germany
| | - Cheng Zhong
- Experimental and Clinical Research Center (ECRC) a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC) Charité – Universitätsmedizin Berlin Berlin Germany
| | - Shuai Zhu
- Hunan Cancer Hospital The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha China
| | - Yu Xie
- Hunan Cancer Hospital The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha China
| | - Dmitry Tsvetkov
- Experimental and Clinical Research Center (ECRC) a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC) Charité – Universitätsmedizin Berlin Berlin Germany
| | - Oliver Daumke
- Crystallography Max‐Delbrück‐Center for Molecular Medicine Berlin Germany
- Institute of Chemistry and Biochemistry Freie Universität Berlin Berlin Germany
| | - Yu Huang
- Institute of Vascular Medicine and School of Biomedical Sciences Chinese University of Hong Kong Hong Kong China
| | - Maik Gollasch
- Experimental and Clinical Research Center (ECRC) a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC) Charité – Universitätsmedizin Berlin Berlin Germany
- Medical Clinic for Nephrology and Internal Intensive Care Charité – Universitätsmedizin Berlin Berlin Germany
- Department of Geriatrics University Medicine Greifswald Greifswald Germany
| |
Collapse
|
8
|
Lian X, Matthaeus C, Kaßmann M, Daumke O, Gollasch M. Pathophysiological Role of Caveolae in Hypertension. Front Med (Lausanne) 2019; 6:153. [PMID: 31355199 PMCID: PMC6635557 DOI: 10.3389/fmed.2019.00153] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/20/2019] [Indexed: 12/02/2022] Open
Abstract
Caveolae, flask-shaped cholesterol-, and glycosphingolipid-rich membrane microdomains, contain caveolin 1, 2, 3 and several structural proteins, in particular Cavin 1-4, EHD2, pacsin2, and dynamin 2. Caveolae participate in several physiological processes like lipid uptake, mechanosensitivity, or signaling events and are involved in pathophysiological changes in the cardiovascular system. They serve as a specific membrane platform for a diverse set of signaling molecules like endothelial nitric oxide synthase (eNOS), and further maintain vascular homeostasis. Lack of caveolins causes the complete loss of caveolae; induces vascular disorders, endothelial dysfunction, and impaired myogenic tone; and alters numerous cellular processes, which all contribute to an increased risk for hypertension. This brief review describes our current knowledge on caveolae in vasculature, with special focus on their pathophysiological role in hypertension.
Collapse
Affiliation(s)
- Xiaoming Lian
- Experimental and Clinical Research Center—A Joint Cooperation Between the Charité–University Medicine Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Claudia Matthaeus
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Mario Kaßmann
- Experimental and Clinical Research Center—A Joint Cooperation Between the Charité–University Medicine Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Oliver Daumke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Maik Gollasch
- Experimental and Clinical Research Center—A Joint Cooperation Between the Charité–University Medicine Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Medical Clinic for Nephrology and Internal Intensive Care, Berlin, Germany
| |
Collapse
|
9
|
Fan G, Kaßmann M, Hashad AM, Welsh DG, Gollasch M. Differential targeting and signalling of voltage-gated T-type Ca v 3.2 and L-type Ca v 1.2 channels to ryanodine receptors in mesenteric arteries. J Physiol 2018; 596:4863-4877. [PMID: 30146760 PMCID: PMC6187032 DOI: 10.1113/jp276923] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/24/2018] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS In arterial smooth muscle, Ca2+ sparks are elementary Ca2+ -release events generated by ryanodine receptors (RyRs) to cause vasodilatation by opening maxi Ca2+ -sensitive K+ (BKCa ) channels. This study elucidated the contribution of T-type Cav 3.2 channels in caveolae and their functional interaction with L-type Cav 1.2 channels to trigger Ca2+ sparks in vascular smooth muscle cells (VSMCs). Our data demonstrate that L-type Cav 1.2 channels provide the predominant Ca2+ pathway for the generation of Ca2+ sparks in murine arterial VSMCs. T-type Cav 3.2 channels represent an additional source for generation of VSMC Ca2+ sparks. They are located in pit structures of caveolae to provide locally restricted, tight coupling between T-type Cav 3.2 channels and RyRs to ignite Ca2+ sparks. ABSTRACT Recent data suggest that T-type Cav 3.2 channels in arterial vascular smooth muscle cells (VSMCs) and pits structure of caveolae could contribute to elementary Ca2+ signalling (Ca2+ sparks) via ryanodine receptors (RyRs) to cause vasodilatation. While plausible, their precise involvement in igniting Ca2+ sparks remains largely unexplored. The goal of this study was to elucidate the contribution of caveolar Cav 3.2 channels and their functional interaction with Cav 1.2 channels to trigger Ca2+ sparks in VSMCs from mesenteric, tibial and cerebral arteries. We used tamoxifen-inducible smooth muscle-specific Cav 1.2-/- (SMAKO) mice and laser scanning confocal microscopy to assess Ca2+ spark generation in VSMCs. Ni2+ , Cd2+ and methyl-β-cyclodextrin were used to inhibit Cav 3.2 channels, Cav 1.2 channels and caveolae, respectively. Ni2+ (50 μmol L-1 ) and methyl-β-cyclodextrin (10 mmol L-1 ) decreased Ca2+ spark frequency by ∼20-30% in mesenteric VSMCs in a non-additive manner, but failed to inhibit Ca2+ sparks in tibial and cerebral artery VSMCs. Cd2+ (200 μmol L-1 ) suppressed Ca2+ sparks in mesenteric arteries by ∼70-80%. A similar suppression of Ca2+ sparks was seen in mesenteric artery VSMCs of SMAKO mice. The remaining Ca2+ sparks were fully abolished by Ni2+ or methyl-β-cyclodextrin. Our data demonstrate that Ca2+ influx through CaV 1.2 channels is the primary means of triggering Ca2+ sparks in murine arterial VSMCs. CaV 3.2 channels, localized to caveolae and tightly coupled to RyR, provide an additional Ca2+ source for Ca2+ spark generation in mesenteric, but not tibial and cerebral, arteries.
Collapse
Affiliation(s)
- Gang Fan
- Charité – Universitätsmedizin BerlinExperimental and Clinical Research Center (ECRC)Campus BuchBerlinGermany
| | - Mario Kaßmann
- Charité – Universitätsmedizin BerlinExperimental and Clinical Research Center (ECRC)Campus BuchBerlinGermany
- DZHK (German Centre for Cardiovascular Research)BerlinGermany
| | - Ahmed M. Hashad
- Department of Physiology and PharmacologyHotchkiss Brain and Libin Cardiovascular InstitutesUniversity of CalgaryAlbertaCanada
| | - Donald G. Welsh
- Department of Physiology and PharmacologyWestern UniversityLondonONCanada
| | - Maik Gollasch
- Charité – Universitätsmedizin BerlinExperimental and Clinical Research Center (ECRC)Campus BuchBerlinGermany
- DZHK (German Centre for Cardiovascular Research)BerlinGermany
- Charité – Universitätsmedizin BerlinMedical Clinic for Nephrology and Internal Intensive CareCampus VirchowBerlinGermany
| |
Collapse
|
10
|
He D, Pan Q, Chen Z, Sun C, Zhang P, Mao A, Zhu Y, Li H, Lu C, Xie M, Zhou Y, Shen D, Tang C, Yang Z, Jin J, Yao X, Nilius B, Ma X. Treatment of hypertension by increasing impaired endothelial TRPV4-KCa2.3 interaction. EMBO Mol Med 2018; 9:1491-1503. [PMID: 28899928 PMCID: PMC5666316 DOI: 10.15252/emmm.201707725] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The currently available antihypertensive agents have undesirable adverse effects due to systemically altering target activity including receptors, channels, and enzymes. These effects, such as loss of potassium ions induced by diuretics, bronchospasm by beta‐blockers, constipation by Ca2+ channel blockers, and dry cough by ACEI, lead to non‐compliance with therapies (Moser, 1990). Here, based on new hypertension mechanisms, we explored a new antihypertensive approach. We report that transient receptor potential vanilloid 4 (TRPV4) interacts with Ca2+‐activated potassium channel 3 (KCa2.3) in endothelial cells (ECs) from small resistance arteries of normotensive humans, while ECs from hypertensive patients show a reduced interaction between TRPV4 and KCa2.3. Murine hypertension models, induced by high‐salt diet, N(G)‐nitro‐l‐arginine intake, or angiotensin II delivery, showed decreased TRPV4‐KCa2.3 interaction in ECs. Perturbation of the TRPV4‐KCa2.3 interaction in mouse ECs by overexpressing full‐length KCa2.3 or defective KCa2.3 had hypotensive or hypertensive effects, respectively. Next, we developed a small‐molecule drug, JNc‐440, which showed affinity for both TRPV4 and KCa2.3. JNc‐440 significantly strengthened the TRPV4‐KCa2.3 interaction in ECs, enhanced vasodilation, and exerted antihypertensive effects in mice. Importantly, JNc‐440 specifically targeted the impaired TRPV4‐KCa2.3 interaction in ECs but did not systemically activate TRPV4 and KCa2.3. Together, our data highlight the importance of impaired endothelial TRPV4‐KCa2.3 coupling in the progression of hypertension and suggest a novel approach for antihypertensive drug development.
Collapse
Affiliation(s)
- Dongxu He
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Qiongxi Pan
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Zhen Chen
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Chunyuan Sun
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Peng Zhang
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Aiqin Mao
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Yaodan Zhu
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Hongjuan Li
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Chunxiao Lu
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Mingxu Xie
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Yin Zhou
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Daoming Shen
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Chunlei Tang
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Zhenyu Yang
- Heart Centre, Wuxi People's Hospital, Wuxi, China
| | - Jian Jin
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Bernd Nilius
- Department Cell Mol Medicine Laboratory Ion Channel Research Campus Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Xin Ma
- School of Medicine, Jiangnan University, Wuxi, China .,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
11
|
Effects of caveolae depletion and urothelial denudation on purinergic and cholinergic signaling in healthy and cyclophosphamide-induced cystitis in the rat bladder. Auton Neurosci 2018; 213:60-70. [PMID: 30005741 DOI: 10.1016/j.autneu.2018.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/29/2018] [Accepted: 06/05/2018] [Indexed: 01/18/2023]
Abstract
Cholesterol rich membrane invaginations, caveolae, have important roles in various cellular activities, one of them being signal transduction. This signaling pathway seems to be affected during various bladder disorders and the current study aimed to elucidate the plausible involvement of caveolae mediated signal transduction during cyclophosphamide induced cystitis. Furthermore, the urothelial cholinergic part of ATP-evoked contractions and its possible link to caveolae were investigated. Cholinergic, as well as purinergic, contractile responses in rat urinary bladders were examined using a classic organ bath set-up with full-thickness strip preparations or a whole bladder model that enabled luminal administration of substances. Furthermore, sub groups with and without urothelium were examined. The expression of caveolin-1 was also tested using western blot and immunofluorescence. Caveolae cholesterol depletion by methyl-β-cyclodextrin entailed a significant decrease of ATP-evoked bladder contractility. Interestingly, after muscarinic blockade the ATP induced contractions were significantly reduced in the same manner. Furthermore, this atropine-sensitive part of ATP-evoked responses was absent in denuded as well as inflamed bladders. A tendency towards a reduced expression of caveolin-1 was observed in rats with experimental cystitis. The cholinergic part of ATP-induced contractile responses seemed to be affected by urothelium denudation as well as caveolae depletion. Removing one of these structures nullifies the effect of the other, suggesting an important interaction between the urothelium and the caveolar structures. These effects are absent in inflamed animals and might be one pathophysiological aspect behind BPS/IC.
Collapse
|
12
|
Burton RAB, Rog-Zielinska EA, Corbett AD, Peyronnet R, Bodi I, Fink M, Sheldon J, Hoenger A, Calaghan SC, Bub G, Kohl P. Caveolae in Rabbit Ventricular Myocytes: Distribution and Dynamic Diminution after Cell Isolation. Biophys J 2017; 113:1047-1059. [PMID: 28877488 PMCID: PMC5653872 DOI: 10.1016/j.bpj.2017.07.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 05/16/2017] [Accepted: 07/06/2017] [Indexed: 12/12/2022] Open
Abstract
Caveolae are signal transduction centers, yet their subcellular distribution and preservation in cardiac myocytes after cell isolation are not well documented. Here, we quantify caveolae located within 100 nm of the outer cell surface membrane in rabbit single-ventricular cardiomyocytes over 8 h post-isolation and relate this to the presence of caveolae in intact tissue. Hearts from New Zealand white rabbits were either chemically fixed by coronary perfusion or enzymatically digested to isolate ventricular myocytes, which were subsequently fixed at 0, 3, and 8 h post-isolation. In live cells, the patch-clamp technique was used to measure whole-cell plasma membrane capacitance, and in fixed cells, caveolae were quantified by transmission electron microscopy. Changes in cell-surface topology were assessed using scanning electron microscopy. In fixed ventricular myocardium, dual-axis electron tomography was used for three-dimensional reconstruction and analysis of caveolae in situ. The presence and distribution of surface-sarcolemmal caveolae in freshly isolated cells matches that of intact myocardium. With time, the number of surface-sarcolemmal caveolae decreases in isolated cardiomyocytes. This is associated with a gradual increase in whole-cell membrane capacitance. Concurrently, there is a significant increase in area, diameter, and circularity of sub-sarcolemmal mitochondria, indicative of swelling. In addition, electron tomography data from intact heart illustrate the regular presence of caveolae not only at the surface sarcolemma, but also on transverse-tubular membranes in ventricular myocardium. Thus, caveolae are dynamic structures, present both at surface-sarcolemmal and transverse-tubular membranes. After cell isolation, the number of surface-sarcolemmal caveolae decreases significantly within a time frame relevant for single-cell research. The concurrent increase in cell capacitance suggests that membrane incorporation of surface-sarcolemmal caveolae underlies this, but internalization and/or micro-vesicle loss to the extracellular space may also contribute. Given that much of the research into cardiac caveolae-dependent signaling utilizes isolated cells, and since caveolae-dependent pathways matter for a wide range of other study targets, analysis of isolated cell data should take the time post-isolation into account.
Collapse
Affiliation(s)
- Rebecca A B Burton
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Eva A Rog-Zielinska
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | | | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ilona Bodi
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Fink
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Judith Sheldon
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Andreas Hoenger
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado
| | - Sarah C Calaghan
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Gil Bub
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
13
|
Gadeberg HC, Kong CHT, Bryant SM, James AF, Orchard CH. Cholesterol depletion does not alter the capacitance or Ca handling of the surface or t-tubule membranes in mouse ventricular myocytes. Physiol Rep 2017; 5:5/22/e13500. [PMID: 29150591 PMCID: PMC5704078 DOI: 10.14814/phy2.13500] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/28/2017] [Accepted: 10/20/2017] [Indexed: 01/17/2023] Open
Abstract
Cholesterol is a key component of the cell plasma membrane. It has been suggested that the t‐tubule membrane of cardiac ventricular myocytes is enriched in cholesterol and that this plays a role in determining t‐tubule structure and function. We have used methyl‐β‐cyclodextrin (MβCD) to deplete cholesterol in intact and detubulated mouse ventricular myocytes to investigate the contribution of cholesterol to t‐tubule structure, membrane capacitance, and the distribution of Ca flux pathways. Depletion of membrane cholesterol was confirmed using filipin; however, di‐8‐ANEPPS staining showed no differences in t‐tubule structure following MβCD treatment. MβCD treatment had no significant effect on the capacitance:volume relationship of intact myocytes or on the decrease in capacitance:volume caused by detubulation. Similarly, Ca influx and efflux were not altered by MβCD treatment and were reduced by a similar amount following detubulation in untreated and MβCD‐treated cells. These data show that cholesterol depletion has similar effects on the surface and t‐tubule membranes and suggest that cholesterol plays no acute role in determining t‐tubule structure and function.
Collapse
Affiliation(s)
- Hanne C Gadeberg
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Cherrie H T Kong
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Simon M Bryant
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Andrew F James
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Clive H Orchard
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
14
|
Wang Y, Wilson C, Cartwright EJ, Lei M. Plasma membrane Ca 2+ -ATPase 1 is required for maintaining atrial Ca 2+ homeostasis and electrophysiological stability in the mouse. J Physiol 2017; 595:7383-7398. [PMID: 29023784 PMCID: PMC5730856 DOI: 10.1113/jp274110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 10/06/2017] [Indexed: 11/22/2022] Open
Abstract
Key points The role of plasma membrane Ca2+‐ATPase 1 (PMCA1) in Ca2+ homeostasis and electrical stability in atrial tissue has been investigated at both organ and cellular levels in mice with cardiomyocyte‐specific deletion of PMCA1 (PMCA1cko) The PMCA1cko hearts became more susceptible to atrial arrhythmic stress conditions than PMCA1loxP/loxP hearts. PMCA1 deficiency alters cellular Ca2+ homeostasis under both baseline and stress conditions. PMCA1 is required for maintaining cellular Ca2+ homeostasis and electrical stability in murine atria under stress conditions.
Abstract To determine the role of plasma membrane Ca2+‐ATPase 1 (PMCA1) in maintaining Ca2+ homeostasis and electrical stability in the atrium under physiological and stress conditions, mice with a cardiomyocyte‐specific deletion of PMCA1 (PMCA1cko) and their control littermates (PMCA1loxP/loxP) were studied at the organ and cellular levels. At the organ level, the PMCA1cko hearts became more susceptible to atrial arrhythmias under rapid programmed electrical stimulation compared with the PMCA1loxP/loxP hearts, and such arrhythmic events became more severe under Ca2+ overload conditions. At the cellular level, the occurrence of irregular‐type action potentials of PMCA1cko atrial myocytes increased significantly under Ca2+ overload conditions and/or at higher frequency of stimulation. The decay of Na+/Ca2+ exchanger current that followed a stimulation protocol was significantly prolonged in PMCA1cko atrial myocytes under basal conditions, with Ca2+ overload leading to even greater prolongation. In conclusion, PMCA1 is required for maintaining Ca2+ homeostasis and electrical stability in the atrium. This is particularly critical during fast removal of Ca2+ from the cytosol, which is required under stress conditions. The role of plasma membrane Ca2+‐ATPase 1 (PMCA1) in Ca2+ homeostasis and electrical stability in atrial tissue has been investigated at both organ and cellular levels in mice with cardiomyocyte‐specific deletion of PMCA1 (PMCA1cko) The PMCA1cko hearts became more susceptible to atrial arrhythmic stress conditions than PMCA1loxP/loxP hearts. PMCA1 deficiency alters cellular Ca2+ homeostasis under both baseline and stress conditions. PMCA1 is required for maintaining cellular Ca2+ homeostasis and electrical stability in murine atria under stress conditions.
Collapse
Affiliation(s)
- Yanwen Wang
- Department of Pharmacology, University of Oxford, Oxford, UK.,Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Claire Wilson
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Dopico AM, Bukiya AN. Regulation of Ca 2+-Sensitive K + Channels by Cholesterol and Bile Acids via Distinct Channel Subunits and Sites. CURRENT TOPICS IN MEMBRANES 2017; 80:53-93. [PMID: 28863822 DOI: 10.1016/bs.ctm.2017.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cholesterol (CLR) conversion into bile acids (BAs) in the liver constitutes the major pathway for CLR elimination from the body. Moreover, these steroids regulate each other's metabolism. While the roles of CLR and BAs in regulating metabolism and tissue function are well known, research of the last two decades revealed the existence of specific protein receptors for CLR or BAs in tissues with minor contribution to lipid metabolism, raising the possibility that these lipids serve as signaling molecules throughout the body. Among other lipids, CLR and BAs regulate ionic current mediated by the activity of voltage- and Ca2+-gated, K+ channels of large conductance (BK channels) and, thus, modulate cell physiology and participate in tissue pathophysiology. Initial work attributed modification of BK channel function by CLR or BAs to the capability of these steroids to directly interact with bilayer lipids and thus alter the physicochemical properties of the bilayer with eventual modification of BK channel function. Based on our own work and that of others, we now review evidence that supports direct interactions between CLR or BA and specific BK protein subunits, and the consequence of such interactions on channel activity and organ function, with a particular emphasis on arterial smooth muscle. For each steroid type, we will also briefly discuss several mechanisms that may underlie modification of channel steady-state activity. Finally, we will present novel computational data that provide a chemical basis for differential recognition of CLR vs lithocholic acid by distinct BK channel subunits and recognition sites.
Collapse
Affiliation(s)
- Alex M Dopico
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States.
| | - Anna N Bukiya
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
16
|
Busija AR, Patel HH, Insel PA. Caveolins and cavins in the trafficking, maturation, and degradation of caveolae: implications for cell physiology. Am J Physiol Cell Physiol 2017; 312:C459-C477. [PMID: 28122734 DOI: 10.1152/ajpcell.00355.2016] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 01/09/2023]
Abstract
Caveolins (Cavs) are ~20 kDa scaffolding proteins that assemble as oligomeric complexes in lipid raft domains to form caveolae, flask-shaped plasma membrane (PM) invaginations. Caveolae ("little caves") require lipid-lipid, protein-lipid, and protein-protein interactions that can modulate the localization, conformational stability, ligand affinity, effector specificity, and other functions of proteins that are partners of Cavs. Cavs are assembled into small oligomers in the endoplasmic reticulum (ER), transported to the Golgi for assembly with cholesterol and other oligomers, and then exported to the PM as an intact coat complex. At the PM, cavins, ~50 kDa adapter proteins, oligomerize into an outer coat complex that remodels the membrane into caveolae. The structure of caveolae protects their contents (i.e., lipids and proteins) from degradation. Cellular changes, including signal transduction effects, can destabilize caveolae and produce cavin dissociation, restructuring of Cav oligomers, ubiquitination, internalization, and degradation. In this review, we provide a perspective of the life cycle (biogenesis, degradation), composition, and physiologic roles of Cavs and caveolae and identify unanswered questions regarding the roles of Cavs and cavins in caveolae and in regulating cell physiology.1.
Collapse
Affiliation(s)
- Anna R Busija
- Department of Anesthesiology, University of California, San Diego, La Jolla, California.,Department of Pharmacology, University of California, San Diego, La Jolla, California
| | - Hemal H Patel
- Department of Anesthesiology, University of California, San Diego, La Jolla, California
| | - Paul A Insel
- Department of Medicine, University of California, San Diego, La Jolla, California; and .,Department of Pharmacology, University of California, San Diego, La Jolla, California
| |
Collapse
|
17
|
Niazmand S, Fereidouni E, Mahmoudabady M, Hosseini M. Teucrium polium-induced Vasorelaxation Mediated by Endothelium-dependent and Endothelium-independent Mechanisms in Isolated Rat Thoracic Aorta. Pharmacognosy Res 2017; 9:372-377. [PMID: 29263631 PMCID: PMC5717790 DOI: 10.4103/pr.pr_140_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Objective: There are some reports on hypotensive and antispasmodic effects of Teucrium polium L. (Lamiaceae) (TP). Subjects and Methods: The activity of different concentrations of TP extract (1, 2, 4 and 8 mg/ml) was evaluated on contractile responses of isolated aorta to potassium chloride (KCl) and phenylephrine (PE). Results: The cumulative concentrations of the extract induced a concentration-dependent relaxation in the aorta precontracted by PE and KCl. Extract-induced vasorelaxations in denuded aortic rings precontracted by PE and KCl at lower concentrations were considerably less than intact aortic rings, but this effect was significantly more at concentrations of 4 mg/ml for PE-, 4 and 8 mg/ml for KCl-induced contractions. All the extract concentrations (except 1 mg/ml) significantly relaxed PE-induced contraction in the presence of NG-nitro-L-arginine methyl ester. Indomethacin reduced effectively extract-induced vasorelaxation at 1 and 2 mg/ml. The extract reduced PE- and KCl-induced contractions in the presence of cumulative calcium concentrations and after incubation with diltiazem; this vasorelaxant effect of TP was decreased. TP-induced relaxation was inhibited by heparin, ruthenium red, glibenclamide, and tetraethylammonium, but 4-aminopyridine had no effect on TP-induced relaxation. Conclusion: TP extract has vasorelaxant effect on isolated rat thoracic aorta which mediated by endothelium-dependent and endothelium-independent mechanisms. The relaxation mainly was mediated by inhibition of calcium influx in vascular smooth muscle cells. It seems that the vasorelaxant effect of extract at lower concentrations was mediated by nitric oxide and prostacyclin. SUMMARY The vasodilatory effect of Teucrium polium L. was mediated by several mechanisms. First: Teucrium polium L. inhibited receptor operated ROCC and VDCC. Second: Teucrium polium L. also inhibited KATP and KCa channels. Third: Teucrium polium L. blocked IP3 receptor and reduced the release of calcium from intracellular source. Forth: Teucrium polium L. increased the release on NO and PGI2 from endothelial cells.
Abbreviations Used: ROCC: Receptor operated calcium channels, VDCC: Voltage dependent calcium channels, PLC: Phospholipase C, IP3: 1,4,5 triphosphate inositol, IP3R: IP3 receptors, SR: sarcoplasmic reticulum, RYR: ryanodine receptors, K+ATP: ATP-sensitive potassium channel, K+Ca: Calcium-activated potassium channel, cAMP: Cyclic adenosine monophosphate, cGMP: Cyclic guanosine monophosphate, PGI2: Prostaglandin I2, NO: Nitric oxide
Collapse
Affiliation(s)
- Saeed Niazmand
- Cardiovascular Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elahe Fereidouni
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Mahmoudabady
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurocognitive Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Dai R, Wang T, Si X, Jia Y, Wang L, Yuan Y, Lin Q, Yang C. Vasodilatory effects and underlying mechanisms of the ethyl acetate extracts from Gastrodia elata. Can J Physiol Pharmacol 2016; 95:564-571. [PMID: 28177685 DOI: 10.1139/cjpp-2016-0407] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The objective of this study was to assess the ethyl acetate extracts of Gastrodia elata Blume (GEB) on vascular tone and the mechanisms involved. GEB was extracted with 95% EtOH followed by a further extraction with ethyl acetate. The effects of GEB and its ingredients on the isometric tensions of the aortic rings from rats were measured. The ethyl acetate extract of GEB induced a vasodilatory effect on rat aorta, which was partially dependent on endothelium. Four chemical compounds isolated from GEB were identified as 3,4-dihydroxybenzaldehyde (DB), 4-hydroxybenzaldehyde (HB), 4-methoxybenzyl alcohol (MA), and 4,4'-dihydroxydiphenyl methane (DM), respectively. All of these compounds induced vasodilatations, which were dependent on the endothelium to different degrees. After pretreatment with Nω-nitro-l-arginine methyl ester, indomethacin, or methylene blue, the vasodilatations induced by DB, HB, and MA were significantly decreased. In addition, the contractions of the rat aortic rings due to Ca2+ influx and intracellular Ca2+ release were also inhibited by DM. Furthermore, the administration of DB significantly enhanced the productions of nitric oxide (NO) and the activities of the endothelial NO synthase in aorta and in endothelial cells. Thus, GEB may play an important role in the amelioration of hypertension by modulating vascular tones.
Collapse
Affiliation(s)
- Rong Dai
- a Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming 650500, P.R. China
| | - Ting Wang
- b Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, P.R. China
| | - Xiaoqin Si
- a Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming 650500, P.R. China
| | - Yuanyuan Jia
- a Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming 650500, P.R. China
| | - Lili Wang
- a Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming 650500, P.R. China
| | - Yan Yuan
- b Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, P.R. China
| | - Qing Lin
- a Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming 650500, P.R. China
| | - Cui Yang
- b Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, P.R. China
| |
Collapse
|
19
|
Fu ZJ, Zhong XZ, Ma WH, Zhang WD, Shi CY. Lipophilic but not hydrophilic statin functionally inhibit volume-activated chloride channels by inhibiting NADPH oxidase in monocytes. Biochem Biophys Res Commun 2016; 481:117-124. [PMID: 27818195 DOI: 10.1016/j.bbrc.2016.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 11/02/2016] [Indexed: 11/27/2022]
Abstract
Volume-activated Cl- channels (VACCs) can be activated by hypotonic solutions and have been identified in many cell types. Here, we investigated the effects of different statins on VACCs in monocytes. Whole-cell patch clamp recordings demonstrated that a hypotonic solution induced 5-nitro-2- (3-phenylpropylamino) benzoic acid (NPPB)- and 4,4'-diisothiocyanatostilbene-2, 2'-disulfonic acid (DIDS)-sensitive VACC currents in human peripheral monocytes and RAW 264.7 cells. The VACC currents were inhibited by the lipophilic statin (simvastatin) but not by the hydrophilic simvastatin acid and pravastatin. A low-molecular-weight superoxide anion scavenger (tiron, 1 mM) and inhibitor of NADPH oxidase (DPI 10 μM) was able to abolish the VACC currents. A hypotonic solution increased the reactive oxygen species (ROS) detected by the fluorescence of dichlorodihydrofluorescein (DCF), which was abolished by tiron and DPI. NPPB, DIDS, and simvastatin but not pravastatin decreased the fluorescence of DCF. Simvastatin could not further decrease VACC currents when pretreated with tiron or DPI, whereas exogenous H2O2 (100 μM), increased the VACC currents and overcame the blockade of VACC currents by simvastatin. Functionally, hypotonic solution increased the TNF-α mRNA expression, which could be decreased by tiron, DPI, NPPB, DIDS and simvastatin but not pravastatin. However, simvastatin could not decrease the TNF-α expression further when pretreatment with tiron, DPI, NPPB or DIDS. We conclude that lipophilic (simvastatin) rather than hydrophilic statin inhibit VACCs and decrease hyposmolality induced inflammation in monocytes by inhibiting NADPH oxidase.
Collapse
Affiliation(s)
- Zhi-Jie Fu
- Department of Otorhinolaryngology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, China
| | - Xue-Zhen Zhong
- Department of Cardiovascular Disease, Jinan Central Hospital Affiliated to Shandong University, Shandong, Jinan 250013, China
| | - Wei-Hong Ma
- Department of Cardiology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Wen-Dong Zhang
- Department of Pharmacy, QiLu Hospital, Shandong University, Jinan 250012, China.
| | - Cheng-Yao Shi
- Department of Pharmacy, QiLu Hospital, Shandong University, Jinan 250012, China
| |
Collapse
|
20
|
Zhu Y, Zhang C, Chen B, Chen R, Guo A, Hong J, Song LS. Cholesterol is required for maintaining T-tubule integrity and intercellular connections at intercalated discs in cardiomyocytes. J Mol Cell Cardiol 2016; 97:204-12. [PMID: 27255730 PMCID: PMC5002380 DOI: 10.1016/j.yjmcc.2016.05.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/25/2016] [Accepted: 05/25/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUNDS Low serum cholesterol levels are associated with cardiac arrhythmias and poor prognosis in patients with chronic heart failure. However, the underlying mechanisms by which decreases in cholesterol content lead to cardiac dysfunction remain unclear. Multiple studies have implicated damage to cardiac transverse (T)-tubules as a key mediator of excitation-contraction (E-C) coupling dysfunction and heart failure. Since the T-tubule membrane system is enriched in cholesterol, we hypothesized that depletion of membrane cholesterol promotes T-tubule remodeling and E-C coupling dysfunction. METHODS AND RESULTS We first examined the impact of membrane cholesterol depletion on T-tubule architecture by treating isolated C57BL/6 murine cardiomyocytes with methyl-β-cyclodextrin (MβCD). T-tubule structural integrity was progressively decreased by MβCD in a concentration- and time-dependent manner. Membrane cholesterol depletion also promoted a severe decrease in the amplitude of Ca(2+) transients and an increase in Ca(2+) release dyssynchrony as well as a significant increase in the frequency of spontaneous Ca(2+) sparks. Reintroduction of cholesterol restored T-tubule integrity and partially restored Ca(2+) handling properties in acutely-treated myocytes and slowed T-tubule deterioration in response to chronic MβCD exposure. Studies were extended to determine the impact of membrane cholesterol depletion on T-tubule structure in intact hearts. In addition to T-tubule remodeling, Langendorff perfusion of MβCD resulted in rapid and severe disruption of the intercellular connections between cardiomyocytes, in particular at intercalated disc regions in intact hearts. CONCLUSIONS These data provide the first evidence that cholesterol plays a critical role in maintaining cardiac T-tubule structure as well as the integrity of intercalated discs.
Collapse
Affiliation(s)
- Yanqi Zhu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China; Division of Cardiovascular Medicine, Department of Internal Medicine, François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Caimei Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Rong Chen
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China; Department of Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Ang Guo
- Division of Cardiovascular Medicine, Department of Internal Medicine, François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Jiang Hong
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China; Department of Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA.
| |
Collapse
|
21
|
Akinci G, Topaloglu H, Akinci B, Onay H, Karadeniz C, Ergul Y, Demir T, Ozcan EE, Altay C, Atik T, Garg A. Spectrum of clinical manifestations in two young Turkish patients with congenital generalized lipodystrophy type 4. Eur J Med Genet 2016; 59:320-4. [PMID: 27167729 DOI: 10.1016/j.ejmg.2016.05.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/19/2016] [Accepted: 05/04/2016] [Indexed: 12/31/2022]
Abstract
Congenital generalized lipodystrophy type 4 is an extremely rare autosomal recessive disorder. We report our clinical experience on two unrelated Turkish patients with congenital generalized lipodystrophy type 4. A 13-year-old girl (patient-1) presented with generalized lipodystrophy and myopathy. Further tests revealed ventricular and supraventricular arrhythmias, gastrointestinal dysmotility, atlantoaxial instability, lumbosacral scoliosis, and metabolic abnormalities associated with insulin resistance. A 16-year-old girl (patient-2) with congenital generalized lipodystrophy type 4 was previously reported. Here, we report on her long term clinical follow-up. She received several course of anti-arrhythmic treatments for catecholaminergic polymorphic ventricular tachycardia and rapid atrial fibrillation. An implantable cardioverter defibrillator was also placed. A homozygous PTRF mutation, c.259C > T (p.Gln87*), was identified in patient-1. Congenital generalized lipodystrophy type 4 was caused by homozygous PTRF c.481-482insGTGA (p.Lys161Serfs*41) mutation in patient-2. Our data indicate that patients with congenital generalized lipodystrophy type 4 should be meticulously evaluated for cardiac, neuromuscular, gastrointestinal and skeletal diseases, as well as metabolic abnormalities associated with insulin resistance.
Collapse
Affiliation(s)
- Gulcin Akinci
- Division of Pediatric Neurology, Dr. Behcet Uz Children's Hospital, Izmir, Turkey.
| | - Haluk Topaloglu
- Division of Pediatric Neurology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Baris Akinci
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Huseyin Onay
- Department of Medical Genetics, Ege University, Izmir, Turkey
| | - Cem Karadeniz
- Division of Pediatric Cardiology, Dr. Behcet Uz Children's Hospital, Izmir, Turkey
| | - Yakup Ergul
- Department of Pediatric Cardiology, Istanbul Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Center and Research Hospital, Istanbul, Turkey
| | - Tevfik Demir
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Dokuz Eylul University, Izmir, Turkey
| | | | - Canan Altay
- Department of Radiology, Dokuz Eylul University, Izmir, Turkey
| | - Tahir Atik
- Division of Pediatric Genetics, Department of Pediatrics, Ege University, Izmir, Turkey
| | - Abhimanyu Garg
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, UT Southwestern Medical Center at Dallas, Dallas, TX, USA
| |
Collapse
|
22
|
Schilling JM, Horikawa YT, Zemljic-Harpf AE, Vincent KP, Tyan L, Yu JK, McCulloch AD, Balijepalli RC, Patel HH, Roth DM. Electrophysiology and metabolism of caveolin-3-overexpressing mice. Basic Res Cardiol 2016; 111:28. [PMID: 27023865 DOI: 10.1007/s00395-016-0542-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/17/2016] [Indexed: 10/22/2022]
Abstract
Caveolin-3 (Cav-3) plays a critical role in organizing signaling molecules and ion channels involved in cardiac conduction and metabolism. Mutations in Cav-3 are implicated in cardiac conduction abnormalities and myopathies. Additionally, cardiac-specific overexpression of Cav-3 (Cav-3 OE) is protective against ischemic and hypertensive injury, suggesting a potential role for Cav-3 in basal cardiac electrophysiology and metabolism involved in stress adaptation. We hypothesized that overexpression of Cav-3 may alter baseline cardiac conduction and metabolism. We examined: (1) ECG telemetry recordings at baseline and during pharmacological interventions, (2) ion channels involved in cardiac conduction with immunoblotting and computational modeling, and (3) baseline metabolism in Cav-3 OE and transgene-negative littermate control mice. Cav-3 OE mice had decreased heart rates, prolonged PR intervals, and shortened QTc intervals with no difference in activity compared to control mice. Dobutamine or propranolol did not cause significant changes between experimental groups in maximal (dobutamine) or minimal (propranolol) heart rate. Cav-3 OE mice had an overall lower chronotropic response to atropine. The expression of Kv1.4 and Kv4.3 channels, Nav1.5 channels, and connexin 43 were increased in Cav-3 OE mice. A computational model integrating the immunoblotting results indicated shortened action potential duration in Cav-3 OE mice linking the change in channel expression to the observed electrophysiology phenotype. Metabolic profiling showed no gross differences in VO2, VCO2, respiratory exchange ratio, heat generation, and feeding or drinking. In conclusion, Cav-3 OE mice have changes in ECG intervals, heart rates, and cardiac ion channel expression. These findings give novel mechanistic insights into previously reported Cav-3 dependent cardioprotection.
Collapse
Affiliation(s)
- Jan M Schilling
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, University of California San Diego, La Jolla, CA, USA
| | - Yousuke T Horikawa
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.,Department of Pediatrics, Sharp Rees-Stealy Medical Group, San Diego, CA, USA
| | - Alice E Zemljic-Harpf
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, University of California San Diego, La Jolla, CA, USA
| | - Kevin P Vincent
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Leonid Tyan
- Department of Medicine, Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI, USA
| | - Judith K Yu
- Department of Anesthesiology, University of California San Diego, La Jolla, CA, USA
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.,Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ravi C Balijepalli
- Department of Medicine, Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI, USA
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, University of California San Diego, La Jolla, CA, USA
| | - David M Roth
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA. .,Department of Anesthesiology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
23
|
Jiang M, Wan F, Wang F, Wu Q. Irisin relaxes mouse mesenteric arteries through endothelium-dependent and endothelium-independent mechanisms. Biochem Biophys Res Commun 2015; 468:832-6. [PMID: 26582714 DOI: 10.1016/j.bbrc.2015.11.040] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 11/08/2015] [Indexed: 02/07/2023]
Abstract
Irisin, a newly discovered myokine, has been shown to produce modest weight loss and improve glucose intolerance in mice. The purpose of this study was to investigate the effects of irisin on vascular activity and the mechanisms involved. Experiments were performed on mouse mesenteric arteries. We demonstrated that irisin induced relaxation in mesenteric arteries with or without endothelium in a concentration-dependent manner. It was further demonstrated that the irisin-induced vasorelaxation effects on endothelium-intact mesenteric arteries were reduced by pretreatment with Nω-nitro-L-arginine methyl ester (L-NAME) or 1H-[1, 2, 4] oxadizolo [4, 3-a] quinoxalin-1-one (ODQ). However, pretreatment with indomethacin (INDO), a nonselective cyclooxygenase inhibitor did not modulate irisin-induced relaxation. In addition, the contraction due to extracellular Ca(2+) influx and intracellular Ca(2+) release was also inhibited by irisin. In summary, these results suggested that the endothelium-dependent relaxation of irisin is mediated by the nitric oxide (NO)-guanosine 3', 5'-cyclic phosphate (cGMP)-dependent pathway but not the prostaglandin I2 (PGI2)-cyclic adenosine monophosphate (cAMP)-dependent mechanism. Endothelium-independent relaxation may be depend on inhibiting Ca(2+) influx through blocking VDCCs and intracellular Ca(2+) release through both IP3R and RyR channels.
Collapse
Affiliation(s)
- Miao Jiang
- Experimental Animal Center, The Second Hospital of Shandong University, Jinan 250012, PR China
| | - Fangzhu Wan
- Department of Clinical Medicine, School of Medicine, Shandong University, Jinan 250012, PR China
| | - Fang Wang
- Experimental Animal Center, The Second Hospital of Shandong University, Jinan 250012, PR China
| | - Qi Wu
- Department of Clinical Medicine, School of Medicine, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
24
|
Glukhov AV, Balycheva M, Sanchez-Alonso JL, Ilkan Z, Alvarez-Laviada A, Bhogal N, Diakonov I, Schobesberger S, Sikkel MB, Bhargava A, Faggian G, Punjabi PP, Houser SR, Gorelik J. Direct Evidence for Microdomain-Specific Localization and Remodeling of Functional L-Type Calcium Channels in Rat and Human Atrial Myocytes. Circulation 2015; 132:2372-84. [PMID: 26450916 PMCID: PMC4689179 DOI: 10.1161/circulationaha.115.018131] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 10/02/2015] [Indexed: 12/27/2022]
Abstract
Supplemental Digital Content is available in the text. Distinct subpopulations of L-type calcium channels (LTCCs) with different functional properties exist in cardiomyocytes. Disruption of cellular structure may affect LTCC in a microdomain-specific manner and contribute to the pathophysiology of cardiac diseases, especially in cells lacking organized transverse tubules (T-tubules) such as atrial myocytes (AMs).
Collapse
Affiliation(s)
- Alexey V Glukhov
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Marina Balycheva
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Jose L Sanchez-Alonso
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Zeki Ilkan
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Anita Alvarez-Laviada
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Navneet Bhogal
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Ivan Diakonov
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Sophie Schobesberger
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Markus B Sikkel
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Anamika Bhargava
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Giuseppe Faggian
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Prakash P Punjabi
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Steven R Houser
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Julia Gorelik
- From Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, United Kingdom (A.V.G., M.B., J.L.S.-A., Z.I., A.A.-L., N.B., I.D., S.S., M.B.S., A.B., P.P.P., J.G.); University of Verona, School of Medicine, Verona, Italy (M.B., G.F.); Department of Cardiothoracic Surgery, Hammersmith Hospital, National Heart and Lung Institute, Imperial College London, United Kingdom (P.P.P.); and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.).
| |
Collapse
|
25
|
The molecular interaction of heart LIM protein (HLP) with RyR2 and caveolin-3 is essential for Ca 2+ -induced Ca 2+ release in the heart. Biochem Biophys Res Commun 2015; 463:975-81. [DOI: 10.1016/j.bbrc.2015.06.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 06/06/2015] [Indexed: 11/20/2022]
|
26
|
Al-Brakati AY, Kamishima T, Dart C, Quayle JM. Caveolar disruption causes contraction of rat femoral arteries via reduced basal NO release and subsequent closure of BKCa channels. PeerJ 2015; 3:e966. [PMID: 26038721 PMCID: PMC4451037 DOI: 10.7717/peerj.966] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/29/2015] [Indexed: 12/22/2022] Open
Abstract
Background and Purpose. Caveolae act as signalling hubs in endothelial and smooth muscle cells. Caveolar disruption by the membrane cholesterol depleting agent methyl-β-cyclodextrin (M-β-CD) has various functional effects on arteries including (i) impairment of endothelium-dependent relaxation, and (ii) alteration of smooth muscle cell (SMC) contraction independently of the endothelium. The aim of this study was to explore the effects of M-β-CD on rat femoral arteries. Methods. Isometric force was measured in rat femoral arteries stimulated to contract with a solution containing 20 mM K(+) and 200 nM Bay K 8644 (20 K/Bay K) or with one containing 80 mM K(+)(80 K). Results. Incubation of arteries with M-β-CD (5 mM, 60 min) increased force in response to 20 K/Bay K but not that induced by 80 K. Application of cholesterol saturated M-β-CD (Ch-MCD, 5 mM, 50 min) reversed the effects of M-β-CD. After mechanical removal of endothelial cells M-β-CD caused only a small enhancement of contractions to 20 K/Bay K. This result suggests M-β-CD acts via altering release of an endothelial-derived vasodilator or vasoconstrictor. When nitric oxide synthase was blocked by pre-incubation of arteries with L-NAME (250 µM) the contraction of arteries to 20 K/Bay K was enhanced, and this effect was abolished by pre-treatment with M-β-CD. This suggests M-β-CD is inhibiting endothelial NO release. Inhibition of large conductance voltage- and Ca(2+)-activated (BKCa) channels with 2 mM TEA(+) or 100 nM Iberiotoxin (IbTX) enhanced 20 K/Bay K contractions. L-NAME attenuated the contractile effect of IbTX, as did endothelial removal. Conclusions. Our results suggest caveolar disruption results in decreased release of endothelial-derived nitric oxide in rat femoral artery, resulting in a reduced contribution of BKCa channels to the smooth muscle cell membrane potential, causing depolarisation and contraction.
Collapse
Affiliation(s)
- AY Al-Brakati
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - T Kamishima
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - C Dart
- Department of Biochemistry and Cell Biology, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - JM Quayle
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
27
|
Vincze J, Jenes Á, Füzi M, Almássy J, Németh R, Szigeti G, Dienes B, Gaál Z, Szentesi P, Jóna I, Kertai P, Paragh G, Csernoch L. Effects of fluvastatin and coenzyme Q10 on skeletal muscle in normo- and hypercholesterolaemic rats. J Muscle Res Cell Motil 2015; 36:263-74. [PMID: 25920381 DOI: 10.1007/s10974-015-9413-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 04/21/2015] [Indexed: 10/23/2022]
Abstract
Myalgia and muscle weakness may appreciably contribute to the poor adherence to statin therapy. Although the pathomechanism of statin-induced myopathy is not completely understood, changes in calcium homeostasis and reduced coenzyme Q10 levels are hypothesized to play important roles. In our experiments, fluvastatin and/or coenzyme Q10 was administered chronically to normocholesterolaemic or hypercholaestherolaemic rats, and the modifications of the calcium homeostasis and the strength of their muscles were investigated. While hypercholesterolaemia did not change the frequency of sparks, fluvastatin increased it on muscles both from normocholesterolaemic and from hypercholesterolaemic rats. This effect, however, was not mediated by a chronic modification of the ryanodine receptor as shown by the unchanged ryanodine binding in the latter group. While coenzyme Q10 supplementation significantly reduced the frequency of the spontaneous calcium release events, it did not affect their amplitude and spatial spread in muscles from fluvastatin-treated rats. This indicates that coenzyme Q10 supplementation prevented the spark frequency increasing effect of fluvastatin without having a major effect on the amount of calcium released during individual sparks. In conclusion, we have found that fluvastatin, independently of the cholesterol level in the blood, consistently and specifically increased the frequency of calcium sparks in skeletal muscle cells, an effect which could be prevented by the addition of coenzyme Q10 to the diet. These results support theories favouring the role of calcium handling in the pathophysiology of statin-induced myopathy and provide a possible pathway for the protective effect of coenzyme Q10 in statin treated patients symptomatic of this condition.
Collapse
Affiliation(s)
- J Vincze
- Department of Physiology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, P.O. Box 22, Debrecen, 4012, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Trafficking of β-Adrenergic Receptors: Implications in Intracellular Receptor Signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:151-88. [PMID: 26055058 DOI: 10.1016/bs.pmbts.2015.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
β-Adrenergic receptors (βARs), prototypical G-protein-coupled receptors, play a pivotal role in regulating neuronal and cardiovascular responses to catecholamines during stress. Agonist-induced receptor endocytosis is traditionally considered as a primary mechanism to turn off the receptor signaling (or receptor desensitization). However, recent progress suggests that intracellular trafficking of βAR presents a mean to translocate receptor signaling machinery to intracellular organelles/compartments while terminating the signaling at the cell surface. Moreover, the apparent multidimensionality of ligand efficacy in space and time in a cell has forecasted exciting pathophysiological implications, which are just beginning to be explored. As we begin to understand how these pathways impact downstream cellular programs, this will have significant implications for a number of pathophysiological conditions in heart and other systems, that in turn open up new therapeutic opportunities.
Collapse
|
29
|
Balycheva M, Faggian G, Glukhov AV, Gorelik J. Microdomain-specific localization of functional ion channels in cardiomyocytes: an emerging concept of local regulation and remodelling. Biophys Rev 2015; 7:43-62. [PMID: 28509981 PMCID: PMC5425752 DOI: 10.1007/s12551-014-0159-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/18/2014] [Indexed: 12/26/2022] Open
Abstract
Cardiac excitation involves the generation of action potential by individual cells and the subsequent conduction of the action potential from cell to cell through intercellular gap junctions. Excitation of the cellular membrane results in opening of the voltage-gated L-type calcium ion (Ca2+) channels, thereby allowing a small amount of Ca2+ to enter the cell, which in turn triggers the release of a much greater amount of Ca2+ from the sarcoplasmic reticulum, the intracellular Ca2+ store, and gives rise to the systolic Ca2+ transient and contraction. These processes are highly regulated by the autonomic nervous system, which ensures the acute and reliable contractile function of the heart and the short-term modulation of this function upon changes in heart rate or workload. It has recently become evident that discrete clusters of different ion channels and regulatory receptors are present in the sarcolemma, where they form an interacting network and work together as a part of a macro-molecular signalling complex which in turn allows the specificity, reliability and accuracy of the autonomic modulation of the excitation-contraction processes by a variety of neurohormonal pathways. Disruption in subcellular targeting of ion channels and associated signalling proteins may contribute to the pathophysiology of a variety of cardiac diseases, including heart failure and certain arrhythmias. Recent methodological advances have made it possible to routinely image the topography of live cardiomyocytes, allowing the study of clustering functional ion channels and receptors as well as their coupling within a specific microdomain. In this review we highlight the emerging understanding of the functionality of distinct subcellular microdomains in cardiac myocytes (e.g. T-tubules, lipid rafts/caveolae, costameres and intercalated discs) and their functional role in the accumulation and regulation of different subcellular populations of sodium, Ca2+ and potassium ion channels and their contributions to cellular signalling and cardiac pathology.
Collapse
Affiliation(s)
- Marina Balycheva
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, 4th Floor National Heart and Lung Institute, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK
- Cardiosurgery Department, University of Verona School of Medicine, Verona, Italy
| | - Giuseppe Faggian
- Cardiosurgery Department, University of Verona School of Medicine, Verona, Italy
| | - Alexey V Glukhov
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, 4th Floor National Heart and Lung Institute, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| | - Julia Gorelik
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, 4th Floor National Heart and Lung Institute, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
30
|
Chen H, Li S, Wang P, Yan S, Hu L, Pan X, Yang C, Leung GP. Endothelium-dependent and -independent relaxation of rat aorta induced by extract of Schizophyllum commune. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2014; 21:1230-1236. [PMID: 25172784 DOI: 10.1016/j.phymed.2014.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 05/03/2014] [Accepted: 06/19/2014] [Indexed: 06/03/2023]
Abstract
Schizophyllum commune (SC) is widely consumed by Chinese, especially in southern part of China. The aim of the present study was to assess the extract of SC on vascular tone and the mechanisms involved. Experiments were performed on aorta of 18-week-old male Sprague-Dawley rats. Dried SC was extracted with 50% ethanol, 90% ethanol and deionized water, respectively. The effects of SC on the isometric tension of rat aortic rings were measured. Protein expression for the endothelial nitric oxide synthase (eNOS) was also determined in the primarily cultured rat aortic arterial endothelial cells (RAECs). The results showed that the water extract of SC induced a marked relaxation in aortic rings with or without endothelium. After the pretreatments of N(ω)-nitro-l-arginine methyl ester, indomethacin, RP-cAMP, and methylene blue, the SC-induced relaxation was significantly decreased. In addition, the contraction due to Ca(2+) influx and intracellular Ca(2+) release was also inhibited by SC. Furthermore, expression of the eNOS protein was significantly elevated in RAECs after treatment of SC. In conclusion, the water extract of SC induces an endothelium-dependent and -independent relaxation in rat aorta. The relaxing effect of SC involves the modulation of NO-cGMP-dependent pathways, PGI2-cAMP-depedent pathways, Ca(2+) influx though calcium channels and intracellular Ca(2+) release.
Collapse
Affiliation(s)
- Haiyun Chen
- Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, PR China
| | - Sujuan Li
- Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, PR China
| | - Peng Wang
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, PR China
| | - Saimei Yan
- Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, PR China
| | - Lin Hu
- Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, PR China
| | - Xiaoxia Pan
- Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, PR China
| | - Cui Yang
- Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, PR China; Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, PR China.
| | - George Pakheng Leung
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, PR China.
| |
Collapse
|
31
|
Endothelium-independent vasorelaxant effects of hydroalcoholic extract from Nigella sativa seed in rat aorta: the roles of Ca2+ and K+ channels. BIOMED RESEARCH INTERNATIONAL 2014; 2014:247054. [PMID: 24900958 PMCID: PMC4036417 DOI: 10.1155/2014/247054] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/13/2014] [Accepted: 04/21/2014] [Indexed: 12/20/2022]
Abstract
Objective. The aim of this study was to elucidate the mechanism(s) responsible for the vasorelaxant effect of Nigella sativa (N. sativa). Methods. The activity of different concentrations of N. sativa extract was evaluated on contractile responses of isolated aorta to KCl and phenylephrine (PE). Results. The extract (2–14 mg/mL) induced a concentration dependent relaxation both in endothelium-intact and endothelium-denuded aortic rings precontracted by PE (10−6 M) and KCl (6 × 10−2 M). Extract reduced PE- and KCl-induced contractions in presence of cumulative concentrations of calcium (10−5–10−2 M) significantly. L-NAME and indomethacin had no effect on vasorelaxation effect of extract in PE-induced contraction. Diltiazem and heparin reduced significantly this vasorelaxation at a concentration of 14 mg/mL of extract; however, N. sativa-induced relaxation was not affected by ruthenium red. Tetraethylammonium chloride reduced the extract-induced relaxation in concentrations of 2–6 mg/mL of extract significantly but glibenclamide reduced this relaxative effect in all concentrations of extract. Conclusions. The inhibitory effect of N. sativa seed extract on the contraction induced by PE and KCl was endothelium-independent. This relaxation was mediated mainly through the inhibition of Ca2+ and KATP channels and also intracellular calcium release.
Collapse
|
32
|
Niazmand S, Harandizadeh F, Mahmoudabady M, Hosseini M, Hasanzadeh M, Fereidouni E. Mechanism of vasorelaxation induced by Achillea wilhelmsii in rat isolated thoracic aorta. Adv Biomed Res 2014; 3:91. [PMID: 24761399 PMCID: PMC3988604 DOI: 10.4103/2277-9175.128470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 08/21/2013] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Achillea wilhelmsii (A. wilhelmsii) is used in Iraninan folk medicine for the treatment of hypertension; also, in previous reports, the hypotensive and antihypertensive effects of this plant have been indicated. The aim of the present study is to investigate the vasorelaxant effect of the hydroalcholic extract of A. wilhelmsii and its underlying mechanisms in isolated rat aorta. MATERIALS AND METHODS The effect of the hydroalcholic A. wilhelmsii extract was tested on the contractile response of Wistar rat aorta induced by potassium chloride (KCl) and phenylephrine (PE) using a pressure transducer that is connected to the PowerLab. RESULTS The cumulative concentrations of A. wilhelmsii (0.5-8 mg/ml) induced a vasorelaxation both in endothelium-intact and endothelium-denuded aortas precontracted by high K(+) (6 × 10(-2) M) or 10(-6) M PE. A. wilhelmsii, at a concentration of 4 mg/ml, reduced Ca(2+)-induced contraction (P < 0.001 vs. control) after PE or KCl had generated a stable contraction in the Ca(2+)-free solution. Furthermore, after incubation with diltiazem, the vasorelaxant effect of A. wilhelmsii reduced in the endothelium-denuded aortas precontracted by PE or KCl (P < 0.001 vs. control). In contrast, A. wilhelmsii-induced relaxation was not affected by glibenclamide, BaCl2, ruthenium red, methylene blue, or heparin. CONCLUSIONS The results showed that A. wilhelmsii had a vasorelaxation effect, which was not endothelium-dependent. The relaxation was mediated by inhibition of extracellular Ca(2+) influx through voltage- and receptor-operated Ca(2+) channels (VDDCs and ROCCs) in vascular smooth muscle cells.
Collapse
Affiliation(s)
- Saeed Niazmand
- Cardiovascular Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran ; Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Harandizadeh
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Mahmoudabady
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran ; Neurogenic Inflammation Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran ; Neurocognitive Research Center, School of Medicine, Mashhad University of Medical Sciences, Iran
| | - Mehdi Hasanzadeh
- Cardiovascular Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elahe Fereidouni
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
33
|
Sommer B, Montano LM, Chávez J, Carbajal V, García-Hernandez LM, Irles C, Jiménez-Garduno AM, Ortega A. ROCK1 translocates from non-caveolar to caveolar regions upon KCl stimulation in airway smooth muscle. Physiol Res 2014; 63:179-87. [PMID: 24397803 DOI: 10.33549/physiolres.932545] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Airway smooth muscle (ASM) membrane depolarization through KCl opens L-type voltage dependent Ca2+ channels (Ca(v)1.2); its opening was considered the cause of KCl contraction. This substance is used to bypass intracellular second messenger pathways. It is now clear that KCl also activates RhoA/Rho kinase (ROCK) pathway. ROCK isoforms are characterized as ROCK1 and ROCK2. Because ROCK1 seems the most abundant isotype in lung, we studied its participation in KCl stimulated bovine ASM. With methyl-beta-cyclodextrin (MbetaCD) we disrupted caveolae, a membrane compartment considered as the RhoA/ROCK assembly site, and found that KCl contraction was reduced to the same extent (~26%) as Y-27632 (ROCK inhibitor) treated tissues. We confirmed that KCl induces ROCK activation and this effect was annulled by Y-27632 or MbetaCD. In isolated plasmalemma, ROCK1 was localized in non-caveolar membrane fractions in Western blots from control tissues, but it transferred to caveolae in samples from tissues stimulated with KCl. Ca(v)1.2 was found at the non-caveolar membrane fractions in control and MbetaCD treated tissues. In MbetaCD treated tissues stimulated with KCl, contraction was abolished by nifedipine; only the response to Ca(v)1.2 opening remained as the ROCK component disappeared. Our results show that, in ASM, the KCl contraction involves the translocation of ROCK1 from non-caveolar to caveolar regions and that the proper physiological response depends on this translocation.
Collapse
Affiliation(s)
- B Sommer
- Department of Bronchial Hyperreactivity Research, National Institute of Respiratory Diseases, Mexico City, Mexico.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Suzuki Y, Yamamura H, Ohya S, Imaizumi Y. Caveolin-1 facilitates the direct coupling between large conductance Ca2+-activated K+ (BKCa) and Cav1.2 Ca2+ channels and their clustering to regulate membrane excitability in vascular myocytes. J Biol Chem 2013; 288:36750-61. [PMID: 24202214 DOI: 10.1074/jbc.m113.511485] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
L-type voltage-dependent Ca(2+) channels (LVDCC) and large conductance Ca(2+)-activated K(+) channels (BKCa) are the major factors defining membrane excitability in vascular smooth muscle cells (VSMCs). The Ca(2+) release from sarcoplasmic reticulum through ryanodine receptor significantly contributes to BKCa activation in VSMCs. In this study direct coupling between LVDCC (Cav1.2) and BKCa and the role of caveoline-1 on their interaction in mouse mesenteric artery SMCs were examined. The direct activation of BKCa by Ca(2+) influx through coupling LVDCC was demonstrated by patch clamp recordings in freshly isolated VSMCs. Using total internal reflection fluorescence microscopy, it was found that a large part of yellow fluorescent protein-tagged BKCa co-localized with the cyan fluorescent protein-tagged Cav1.2 expressed in the plasma membrane of primary cultured mouse VSMCs and that the two molecules often exhibited FRET. It is notable that each BKα subunit of a tetramer in BKCa can directly interact with Cav1.2 and promotes Cav1.2 cluster in the molecular complex. Furthermore, caveolin-1 deficiency in knock-out (KO) mice significantly reduced not only the direct coupling between BKCa and Cav1.2 but also the functional coupling between BKCa and ryanodine receptor in VSMCs. The measurement of single cell shortening by 40 mm K(+) revealed enhanced contractility in VSMCs from KO mice than wild type. Taken together, caveolin-1 facilitates the accumulation/clustering of BKCa-LVDCC complex in caveolae, which effectively regulates spatiotemporal Ca(2+) dynamics including the negative feedback, to control the arterial excitability and contractility.
Collapse
Affiliation(s)
- Yoshiaki Suzuki
- From the Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan and
| | | | | | | |
Collapse
|
35
|
Alcalay Y, Hochhauser E, Kliminski V, Dick J, Zahalka MA, Parnes D, Schlesinger H, Abassi Z, Shainberg A, Schindler RFR, Brand T, Kessler-Icekson G. Popeye domain containing 1 (Popdc1/Bves) is a caveolae-associated protein involved in ischemia tolerance. PLoS One 2013; 8:e71100. [PMID: 24066022 PMCID: PMC3774711 DOI: 10.1371/journal.pone.0071100] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 06/24/2013] [Indexed: 11/18/2022] Open
Abstract
Popeye domain containing1 (Popdc1), also named Bves, is an evolutionary conserved membrane protein. Despite its high expression level in the heart little is known about its membrane localization and cardiac functions. The study examined the hypothesis that Popdc1 might be associated with the caveolae and play a role in myocardial ischemia tolerance. To address these issues, we analyzed hearts and cardiomyocytes of wild type and Popdc1-null mice. Immunoconfocal microscopy revealed co-localization of Popdc1 with caveolin3 in the sarcolemma, intercalated discs and T-tubules and with costameric vinculin. Popdc1 was co-immunoprecipitated with caveolin3 from cardiomyocytes and from transfected COS7 cells and was co-sedimented with caveolin3 in equilibrium density gradients. Caveolae disruption by methyl-β-cyclodextrin or by ischemia/reperfusion (I/R) abolished the cellular co-localization of Popdc1 with caveolin3 and modified their density co-sedimentation. The caveolin3-rich fractions of Popdc1-null hearts redistributed to fractions of lower buoyant density. Electron microscopy showed a statistically significant 70% reduction in caveolae number and a 12% increase in the average diameter of the remaining caveolae in the mutant hearts. In accordance with these changes, Popdc1-null cardiomyocytes displayed impaired [Ca+2]i transients, increased vulnerability to oxidative stress and no pharmacologic preconditioning. In addition, induction of I/R injury to Langendorff-perfused hearts indicated a significantly lower functional recovery in the mutant compared with wild type hearts while their infarct size was larger. No improvement in functional recovery was observed in Popdc1-null hearts following ischemic preconditioning. The results indicate that Popdc1 is a caveolae-associated protein important for the preservation of caveolae structural and functional integrity and for heart protection.
Collapse
Affiliation(s)
- Yifat Alcalay
- The Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Edith Hochhauser
- The Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Vitaly Kliminski
- The Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Julia Dick
- The Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Muayad A. Zahalka
- The Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Doris Parnes
- The Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Hadassa Schlesinger
- The Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Zaid Abassi
- Department of Physiology, Rappaport Faculty of Medicine, Israel Institute of Technology, Haifa, Israel
| | - Asher Shainberg
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | - Thomas Brand
- Harefield Heart Science Centre, Imperial College, London, United Kingdom
| | - Gania Kessler-Icekson
- The Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- * E-mail:
| |
Collapse
|
36
|
Sun YH, Zhao J, Jin HT, Cao Y, Ming T, Zhang LL, Hu MY, Hamlati H, Pang SB, Ma XP. Vasorelaxant effects of the extracts and some flavonoids from the buds of Coreopsis tinctoria. PHARMACEUTICAL BIOLOGY 2013; 51:1158-64. [PMID: 23763258 DOI: 10.3109/13880209.2013.782320] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
CONTEXT The buds of Coreopsis tinctoria Nutt (Compositae) are used in the treatment of hypertension in the Uyghur folk medicine in China. OBJECTIVE To investigate vasorelaxant properties of extracts and some flavonoids from C. tinctoria (CT) and their underlying mechanisms in isolated rat thoracic aortic rings. MATERIALS AND METHODS Vasorelaxant effects of ethanol extracts of CT (CTA) and its flavonoids as well as water-ethanol eluates from CTA by AB-8 resins (CTAA∼CTAF) were evaluated on rat aortic rings pre-contracted with phenylephrine (PE, 1 µM) or high KCl (60 µM). We evaluated the effect of CTA, CTAD and CTAE on PE-induced contraction in a Ca²⁺-free medium and a dose-effect curve of Ca²⁺ in pre-contracted ring with high KCl. RESULTS Endothelial removal did not modify the effect of CTAD and CTAE (3.00 g/L) neither on PE-pre-contracted rings (164.78 ± 21.44 and 191.47 ± 16.75%) nor on KCl-pre-contracted rings (75.68 ± 10.76 and 125.14 ± 17.41%) compared with intact-endothelium rings pre-contracted with high KCl (100.49 ± 17.30 and 110.81 ± 16.33%). CTAD and CTAE (3.00 g/L) down-regulated the dose-effect curve of Ca²⁺ in pre-contraction with high KCl, and inhibited the pre-contraction with PE in a Ca²⁺-free medium (p < 0.05). Seven flavonoids were obtained from CTAD, of which luteolin (5) and quercetin (6) were found to be the most effective relaxation in rings precontracted with PE (EC₅₀: 0.006 and 0.039 g/L, p < 0.05) or high KCl (EC₅₀: 0.023 and 0.045 g/L, p < 0.05). DISCUSSION AND CONCLUSION These data demonstrated the vasorelaxant effect of CT, and its mechanism is likely due to an inhibitory effect on calcium movements through cell membranes.
Collapse
Affiliation(s)
- Yu-Hua Sun
- Xinjiang Key Laboratory of Xinjiang Uygur Medicine, Xinjiang Institute of Materia Medica, Urumqi, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Zhang GQ, Wei H, Lu J, Wong P, Shim W. Identification and characterization of calcium sparks in cardiomyocytes derived from human induced pluripotent stem cells. PLoS One 2013; 8:e55266. [PMID: 23408964 PMCID: PMC3567046 DOI: 10.1371/journal.pone.0055266] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 12/27/2012] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Ca2+ spark constitutes the elementary units of cardiac excitation-contraction (E-C) coupling in mature cardiomyocytes. Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes are known to have electrophysiological properties similar to mature adult cardiomyocytes. However, it is unclear if they share similar calcium handling property. We hypothesized that Ca2+ sparks in human induced pluripotent stem cell (hiPSCs)-derived cardiomyocytes (hiPSC-CMs) may display unique structural and functional properties than mature adult cardiomyocytes. METHODS AND RESULTS Ca2+ sparks in hiPSC-CMs were recorded with Ca2+ imaging assay with confocal laser scanning microscopy. Those sparks were stochastic with a tendency of repetitive occurrence at the same site. Nevertheless, the spatial-temporal properties of Ca2+ spark were analogous to that of adult CMs. Inhibition of L-type Ca2+ channels by nifedipine caused a 61% reduction in calcium spark frequency without affecting amplitude of those sparks and magnitude of caffeine releasable sarcoplasmic reticulum (SR) Ca2+ content. In contrast, high extracellular Ca2+ and ryanodine increased the frequency, full width at half maximum (FWHM) and full duration at half maximum (FDHM) of spontaneous Ca2+ sparks. CONCLUSIONS For the first time, spontaneous Ca2+ sparks were detected in hiPSC-CMs. The Ca2+ sparks are predominately triggered by L-type Ca2+ channels mediated Ca2+ influx, which is comparable to sparks detected in adult ventricular myocytes in which cardiac E-C coupling was governed by a Ca2+-induced Ca2+ release (CICR) mechanism. However, focal repetitive sparks originated from the same intracellular organelle could reflect an immature status of the hiPSC-CMs.
Collapse
Affiliation(s)
- Guang Qin Zhang
- Research and Development Unit, National Heart Centre Singapore, Singapore, Republic of Singapore
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Heming Wei
- Research and Development Unit, National Heart Centre Singapore, Singapore, Republic of Singapore
- Cardiovascular and Metabolic Disorders Program, DUKE-NUS Graduate Medical School Singapore, Singapore, Republic of Singapore
| | - Jun Lu
- Research and Development Unit, National Heart Centre Singapore, Singapore, Republic of Singapore
| | - Philip Wong
- Research and Development Unit, National Heart Centre Singapore, Singapore, Republic of Singapore
- Cardiovascular and Metabolic Disorders Program, DUKE-NUS Graduate Medical School Singapore, Singapore, Republic of Singapore
| | - Winston Shim
- Research and Development Unit, National Heart Centre Singapore, Singapore, Republic of Singapore
- Cardiovascular and Metabolic Disorders Program, DUKE-NUS Graduate Medical School Singapore, Singapore, Republic of Singapore
| |
Collapse
|
38
|
Stary CM, Tsutsumi YM, Patel PM, Head BP, Patel HH, Roth DM. Caveolins: targeting pro-survival signaling in the heart and brain. Front Physiol 2012; 3:393. [PMID: 23060817 PMCID: PMC3464704 DOI: 10.3389/fphys.2012.00393] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 09/14/2012] [Indexed: 12/20/2022] Open
Abstract
The present review discusses intracellular signaling moieties specific to membrane lipid rafts (MLRs) and the scaffolding proteins caveolin and introduces current data promoting their potential role in the treatment of pathologies of the heart and brain. MLRs are discreet microdomains of the plasma membrane enriched in gylcosphingolipids and cholesterol that concentrate and localize signaling molecules. Caveolin proteins are necessary for the formation of MLRs, and are responsible for coordinating signaling events by scaffolding and enriching numerous signaling moieties in close proximity. Specifically in the heart and brain, caveolins are necessary for the cytoprotective phenomenon termed ischemic and anesthetic preconditioning. Targeted overexpression of caveolin in the heart and brain leads to induction of multiple pro-survival and pro-growth signaling pathways; thus, caveolins represent a potential novel therapeutic target for cardiac and neurological pathologies.
Collapse
Affiliation(s)
- Creed M Stary
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, University of California San Diego, La Jolla, CA, USA
| | | | | | | | | | | |
Collapse
|
39
|
Lai Y, Cheng K, Kisaalita W. Three dimensional neuronal cell cultures more accurately model voltage gated calcium channel functionality in freshly dissected nerve tissue. PLoS One 2012; 7:e45074. [PMID: 23049767 PMCID: PMC3458113 DOI: 10.1371/journal.pone.0045074] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 08/16/2012] [Indexed: 12/01/2022] Open
Abstract
It has been demonstrated that neuronal cells cultured on traditional flat surfaces may exhibit exaggerated voltage gated calcium channel (VGCC) functionality. To gain a better understanding of this phenomenon, primary neuronal cells harvested from mice superior cervical ganglion (SCG) were cultured on two dimensional (2D) flat surfaces and in three dimensional (3D) synthetic poly-L-lactic acid (PLLA) and polystyrene (PS) polymer scaffolds. These 2D- and 3D-cultured cells were compared to cells in freshly dissected SCG tissues, with respect to intracellular calcium increase in response to high K+ depolarization. The calcium increases were identical for 3D-cultured and freshly dissected, but significantly higher for 2D-cultured cells. This finding established the physiological relevance of 3D-cultured cells. To shed light on the mechanism behind the exaggerated 2D-cultured cells’ functionality, transcriptase expression and related membrane protein distributions (caveolin-1) were obtained. Our results support the view that exaggerated VGCC functionality from 2D cultured SCG cells is possibly due to differences in membrane architecture, characterized by uniquely organized caveolar lipid rafts. The practical implication of use of 3D-cultured cells in preclinical drug discovery studies is that such platforms would be more effective in eliminating false positive hits and as such improve the overall yield from screening campaigns.
Collapse
Affiliation(s)
- Yinzhi Lai
- Cellular Bioengineering Laboratory, College of Engineering, University of Georgia, Athens, Georgia, United States of America
| | - Ke Cheng
- Cellular Bioengineering Laboratory, College of Engineering, University of Georgia, Athens, Georgia, United States of America
| | - William Kisaalita
- Cellular Bioengineering Laboratory, College of Engineering, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
40
|
Abstract
Caveolae are omega-shaped membrane invaginations present in essentially all cell types of the cardiovascular system, including endothelial cells, smooth muscle cells, macrophages, cardiac myocytes, and fibroblasts. Numerous functions have been ascribed to this omega-shaped structure. Caveolae are enriched with different signaling molecules and ion channel regulatory proteins and function both in protein trafficking and signal transduction in these cell types. Caveolins are the structural proteins that are necessary for the formation of caveola membrane domains. Mechanistically, caveolins interact with a variety of downstream signaling molecules, as, for example, Src-family tyrosine kinase, p42/44 mitogen-activated protein (MAP) kinase, and endothelial nitric oxide synthase (eNOS) and hold the signal transducers in the inactive condition until activated with proper stimulus. Caveolae are gradually acquiring increasing attention as cellular organelles contributing to the pathogenesis of several structural and functional processes including cardiac hypertrophy, atherosclerosis, and heart failure. At present, very little is known about the role of caveolae in cardiac function and dysfunction, although recent studies with caveolin knock-out mouse have shown that caveolae and caveolins play a pivotal role in various human pathobiological conditions. This review will discuss the possible role and mechanism of action of caveolae and caveolins in different cardiac diseases.
Collapse
Affiliation(s)
- Manika Das
- Cardiovascular Research Center, University of Connecticut School of Medicine, Farmington, CT 06030-1110, USA
| | | |
Collapse
|
41
|
Yamamura H, Ikeda C, Suzuki Y, Ohya S, Imaizumi Y. Molecular assembly and dynamics of fluorescent protein-tagged single KCa1.1 channel in expression system and vascular smooth muscle cells. Am J Physiol Cell Physiol 2012; 302:C1257-68. [PMID: 22301058 DOI: 10.1152/ajpcell.00191.2011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The large-conductance Ca(2+)-activated K(+) (K(Ca)1.1, BK) channel has pivotal roles in the regulation of vascular tone. To clarify the molecular dynamics of BK channels and their functionally coupled protein on the membrane surface, we examined single-molecule imaging of fluorescent-labeled BK subunits in the plasma membrane using total internal reflection fluorescence (TIRF) microscopy. The dynamic mobility of yellow fluorescent protein (YFP)-tagged BKα subunit (BKα-YFP) expressed in human embryo kidney 293 (HEK) cells was detected in TIRF regions at the level of individual channels and their clusters on the plasma membrane with a diffusion coefficient of 6.7 × 10(3) nm(2)/s. When BKα-YFP was coexpressed with cyan fluorescent protein (CFP)-tagged BKβ1 subunit (BKβ1-CFP) in HEK cells, the mobility was reduced by ∼50%. Fluorescent image analyses suggest that green fluorescent protein (GFP)-tagged BKα subunit (BKα-GFP) expressed in vascular smooth muscle cells (VSMCs), at low density, preferentially formed a heterotetrameric molecular assembly with native BKα subunits, rather than homotetrameric BKα-GFP. Movement of BKα-YFP in VSMCs (0.29 × 10(3) nm(2)/s) was far more restricted than BKα-YFP/BKβ1-CFP in HEK cells (2.5 × 10(3) nm(2)/s). Actin disruption by pretreatment with cytochalasin D in VSMCs appeared to increase the mobile behavior of BKα-YFP, which was then significantly reduced by addition of jasplakinolide. Most BKα-YFP colocalized with caveolin 1 (Cav1)-CFP in VSMCs, but unexpectedly not frequently in HEK cells. Fluorescence resonance energy transfer analyses showed the direct interaction between BKα-YFP and Cav1-CFP, particularly in VSMCs. These results, obtained by single molecule imaging in living cells, indicate that the dynamics of BKα molecules on the membrane surface are strongly restricted or regulated by its auxiliary β-subunit, cytoskeleton, and direct interaction with Cav1 in VSMCs.
Collapse
Affiliation(s)
- Hisao Yamamura
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan
| | | | | | | | | |
Collapse
|
42
|
Senejoux F, Girard-Thernier C, Berthelot A, Bévalot F, Demougeot C. New insights into the mechanisms of the vasorelaxant effects of apocynin in rat thoracic aorta. Fundam Clin Pharmacol 2012; 27:262-70. [PMID: 22233502 DOI: 10.1111/j.1472-8206.2011.01025.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Apocynin is a naturally occurring acetophenone widely used as an inhibitor of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Recent data suggested that apocynin might exert NADPH oxidase-independent pharmacological properties. Among them, vasorelaxant properties have been described, but the mechanisms still give rise to debates. The present study investigated the mechanisms involved in the vasorelaxant effect of apocynin on the in vitro model of rat isolated thoracic aortic rings. Apocynin (30 μM to 10 mM) induced a dose-dependent relaxation in both endothelium-intact and endothelium-denuded aortic rings with respective EC50 values of 0.78 ± 0.08 and 1.91 ± 0.21 mM. Endothelium removal or inhibition of nitric oxide (NO) synthase with N(ω)-nitro-L-arginine-methyl ester (L-NAME) significantly decreased but did not abolish the effect of apocynin. By contrast, apocynin-induced relaxation was unchanged after incubation with indomethacin or charybdotoxin plus apamin. In endothelium-denuded aortas, the vasorelaxant effect of apocynin was significantly reduced by glibenclamide but not by 4-aminopyridine nor by iberiotoxin. Apocynin significantly decreased Ca(2+)-induced contraction and inhibited intracellular Ca(2+) mobilization after contraction with phenylephrine. Finally, the acute intravenous injection of apocynin led to an immediate and transient hypotensive effect in spontaneously hypertensive rats (SHR). In conclusion, our data demonstrated that apocynin induces both endothelium-independent relaxant effects involving inhibition of Ca(2+) mobilization and activation of KATP channels in vascular smooth muscle cells and endothelium-dependent effects mediated by NO. These results should provide a basis for caution when interpreting results on the vascular effects of apocynin.
Collapse
Affiliation(s)
- François Senejoux
- EA 3185 Fonctions et Dysfonctions Epithéliales, UFR des Sciences Médicales et Pharmaceutiques, 25030 Besançon, France
| | | | | | | | | |
Collapse
|
43
|
Abstract
Cardiomyocytes have multiple Ca(2+) fluxes of varying duration that work together to optimize function (1,2). Changes in Ca(2+) activity in response to extracellular agents is predominantly regulated by the phospholipase Cβ- Gα(q;) pathway localized on the plasma membrane which is stimulated by agents such as acetylcholine (3,4). We have recently found that plasma membrane protein domains called caveolae(5,6) can entrap activated Gα(q;)(7). This entrapment has the effect of stabilizing the activated state of Gα(q;) and resulting in prolonged Ca(2+) signals in cardiomyocytes and other cell types(8). We uncovered this surprising result by measuring dynamic calcium responses on a fast scale in living cardiomyocytes. Briefly, cells are loaded with a fluorescent Ca(2+) indicator. In our studies, we used Ca(2+) Green (Invitrogen, Inc.) which exhibits an increase in fluorescence emission intensity upon binding of calcium ions. The fluorescence intensity is then recorded for using a line-scan mode of a laser scanning confocal microscope. This method allows rapid acquisition of the time course of fluorescence intensity in pixels along a selected line, producing several hundreds of time traces on the microsecond time scale. These very fast traces are transferred into excel and then into Sigmaplot for analysis, and are compared to traces obtained for electronic noise, free dye, and other controls. To dissect Ca(2+) responses of different flux rates, we performed a histogram analysis that binned pixel intensities with time. Binning allows us to group over 500 traces of scans and visualize the compiled results spatially and temporally on a single plot. Thus, the slow Ca(2+) waves that are difficult to discern when the scans are overlaid due to different peak placement and noise, can be readily seen in the binned histograms. Very fast fluxes in the time scale of the measurement show a narrow distribution of intensities in the very short time bins whereas longer Ca(2+) waves show binned data with a broad distribution over longer time bins. These different time distributions allow us to dissect the timing of Ca(2+)fluxes in the cells, and to determine their impact on various cellular events.
Collapse
Affiliation(s)
- Urszula Golebiewska
- Department of Biological Sciences and Geology, Queensborough Community College
| | | |
Collapse
|
44
|
Harvey RD, Calaghan SC. Caveolae create local signalling domains through their distinct protein content, lipid profile and morphology. J Mol Cell Cardiol 2011; 52:366-75. [PMID: 21782827 DOI: 10.1016/j.yjmcc.2011.07.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 06/21/2011] [Accepted: 07/07/2011] [Indexed: 01/02/2023]
Abstract
Compartmentation of signalling allows multiple stimuli to achieve diverse cellular responses with only a limited pool of second messengers. This spatial control of signalling is achieved, in part, by cellular structures which bring together elements of a particular cascade. One such structure is the caveola, a flask-shaped lipid raft. Caveolae are well-recognised as signalosomes, platforms for assembly of signalling complexes of receptors, effectors and their targets, which can facilitate efficient and specific cellular responses. Here we extend this simple model and present evidence to show how the protein and lipid profiles of caveolae, as well as their characteristic morphology, define their roles in creating local signalling domains in the cardiac myocyte. This article is part of a Special Issue entitled "Local Signaling in Myocytes."
Collapse
Affiliation(s)
- Robert D Harvey
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | |
Collapse
|
45
|
Das M, Das DK. Lipid raft in cardiac health and disease. Curr Cardiol Rev 2011; 5:105-11. [PMID: 20436850 PMCID: PMC2805812 DOI: 10.2174/157340309788166660] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2008] [Revised: 08/25/2008] [Accepted: 08/25/2008] [Indexed: 01/01/2023] Open
Abstract
Lipid rafts are sphingolipid and cholesterol rich micro-domains of the plasma membrane that coordinate and regulate varieties of signaling processes. Lipid rafts are also present in cardiac myocytes and are enriched in signaling molecules and ion channel regulatory proteins. Lipid rafts are receiving increasing attention as cellular organelles contributing to the pathogenesis of several structural and functional processes including cardiac hypertrophy and heart failure. At present, very little is known about the role of lipid rafts in cardiac function and dysfunction. This review will discuss the possible role of lipid rafts in cardiac health and disease.
Collapse
Affiliation(s)
- Manika Das
- Cardiovascular Research Center, University of Connecticut School of Medicine, Farmington, CT 06030-110, USA
| | | |
Collapse
|
46
|
Winslow RL, Greenstein JL. Cardiac myocytes and local signaling in nano-domains. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2011; 107:48-59. [PMID: 21718716 DOI: 10.1016/j.pbiomolbio.2011.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 06/14/2011] [Indexed: 10/18/2022]
Abstract
It is well known that calcium-induced calcium-release in cardiac myocytes takes place in spatially restricted regions known as dyads, where discrete patches of junctional sarcoplasmic reticulum tightly associate with the t-tubule membrane. The dimensions of a dyad are so small that it contains only a few Ca²⁺ ions at any given time. Ca²⁺ signaling in the dyad is therefore noisy, and dominated by the Brownian motion of Ca²⁺ ions in a potential field. Remarkably, from this complexity emerges the integrated behavior of the myocyte in which, under normal conditions, precise control of Ca²⁺ release and muscle contraction is maintained over the life of the cell. This is but one example of how signal processing within the cardiac myocyte and other cells often occurs in small "nano-domains" where proteins and protein complexes interact at spatial dimensions on the order of ∼1-10 nm and at time-scales on the order of nanoseconds to perform the functions of the cell. In this article, we will review several examples of local signaling in nano-domains, how it contributes to the integrative behavior of the cardiac myocyte, and present computational methods for modeling signal processing within these domains across differing spatio-temporal scales.
Collapse
Affiliation(s)
- Raimond L Winslow
- The Institute for Computational Medicine & Department of Biomedical Engineering, The Johns Hopkins University, School of Medicine & Whiting School of Engineering, Baltimore, MD 21218, USA.
| | | |
Collapse
|
47
|
Somara S, Bashllari D, Gilmont RR, Bitar KN. Real-time dynamic movement of caveolin-1 during smooth muscle contraction of human colon and aged rat colon transfected with caveolin-1 cDNA. Am J Physiol Gastrointest Liver Physiol 2011; 300:G1022-32. [PMID: 21372166 PMCID: PMC3119117 DOI: 10.1152/ajpgi.00301.2010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Caveolin-1 (cav-1) plays a key role in PKC-α and RhoA signaling pathways during acetylcholine (ACh)-induced contraction of colonic smooth muscle cells (CSMC). Aged rat CSMC showed sluggish contractility, concomitant with reduced expression of cav-1 with an associated reduction in activation of PKC-α and RhoA signaling pathway. Real-time monitoring of live human CSMC transfected with yellow fluorescent protein-tagged wild-type caveolin 1 cDNA (YFP-wt-cav-1) cDNA in the present study suggests that cav-1 cycles within and along the membrane in a synchronized, highly organized cytoskeletal path. These studies provide, for the first time, the advantages of real-time monitoring of the dynamic movement of caveolin in living cells. Rapid movement of cav-1 in response to ACh suggests its dynamic role in CSMC contraction. Human CSMC transfected with YFP-ΔTFT-cav-1 dominant negative cDNA show fluorescence in the cytosol of the CSMC and no movement of fluorescent cav-1 in response to ACh mimicking the response shown by aged rat CSMC. Transfection of CSMC from aged rat with YFP-wt-cav-1 cDNA restored the physiological contractile response to ACh as well as the dynamic movement of cav-1 along the organized cytoskeletal path observed in normal adult CSMC. To study the force generation by CSMC, three-dimensional colonic rings were bioengineered. Colonic bioengineered rings from aged CSMC showed reduced force generation compared with colonic bioengineered rings from adult CSMC. Colonic bioengineered rings from aged CSMC transfected with wt-cav-1 cDNA showed force generation similar to colonic bioengineered rings from adult rat CSMC. The data suggest that contraction in CSMC is dependent on cav-1 reorganization dynamics, which restores the physiological contractile response in aged CSMC. We hypothesize that dynamic movement of cav-1 is essential for physiological contractile response of colonic smooth muscle.
Collapse
Affiliation(s)
- Sita Somara
- Gastrointestinal Molecular Motors Laboratory, Department of Pediatrics, Gastroenterology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Daniela Bashllari
- Gastrointestinal Molecular Motors Laboratory, Department of Pediatrics, Gastroenterology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Robert R. Gilmont
- Gastrointestinal Molecular Motors Laboratory, Department of Pediatrics, Gastroenterology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Khalil N. Bitar
- Gastrointestinal Molecular Motors Laboratory, Department of Pediatrics, Gastroenterology, University of Michigan Medical Center, Ann Arbor, Michigan
| |
Collapse
|
48
|
Sathish V, Yang B, Meuchel LW, VanOosten SK, Ryu AJ, Thompson MA, Prakash YS, Pabelick CM. Caveolin-1 and force regulation in porcine airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2011; 300:L920-9. [PMID: 21421751 DOI: 10.1152/ajplung.00322.2010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Caveolae are specialized membrane microdomains expressing the scaffolding protein caveolin-1. We recently demonstrated the presence of caveolae in human airway smooth muscle (ASM) and the contribution of caveolin-1 to intracellular calcium ([Ca(2+)](i)) regulation. In the present study, we tested the hypothesis that caveolin-1 regulates ASM contractility. We examined the role of caveolins in force regulation of porcine ASM under control conditions as well as TNF-α-induced airway inflammation. In porcine ASM strips, exposure to 10 mM methyl-β-cyclodextrin (CD) or 5 μM of the caveolin-1 specific scaffolding domain inhibitor peptide (CSD) resulted in time-dependent decrease in force responses to 1 μM ACh. Overnight exposure to the cytokine TNF-α (50 ng/ml) accelerated and increased caveolin-1 expression and enhanced force responses to ACh. Suppression of caveolin-1 with small interfering RNA mimicked the effects of CD or CSD. Regarding mechanisms by which caveolae contribute to contractile changes, inhibition of MAP kinase with 10 μM PD98059 did not alter control or TNF-α-induced increases in force responses to ACh. However, inhibiting RhoA with 100 μM fasudil or 10 μM Y27632 resulted in significant decreases in force responses, with lesser effects in TNF-α exposed samples. Furthermore, Ca(2+) sensitivity for force generation was substantially reduced by fasudil or Y27632, an effect even more enhanced in the absence of caveolin-1 signaling. Overall, these results indicate that caveolin-1 is a critical player in enhanced ASM contractility with airway inflammation.
Collapse
Affiliation(s)
- Venkatachalem Sathish
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Schulson MN, Scriven DRL, Fletcher P, Moore EDW. Couplons in rat atria form distinct subgroups defined by their molecular partners. J Cell Sci 2011; 124:1167-74. [PMID: 21385843 DOI: 10.1242/jcs.080929] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Standard local control theory, which describes Ca(2+) release during excitation-contraction coupling (ECC), assumes that all ryanodine receptor 2 (RyR2) complexes are equivalent. Findings from our laboratory have called this assumption into question. Specifically, we have shown that the RyR2 complexes in ventricular myocytes are different, depending on their location within the cell. This has led us to hypothesize that similar differences occur within the rat atrial cell. To test this hypothesis, we have triple-labelled enzymatically isolated fixed myocytes to examine the distribution and colocalization of RyR2, calsequestrin (Casq), voltage-gated Ca(2+) channels (Ca(v)1.2), the sodium-calcium exchanger (Ncx) and caveolin-3 (Cav3). A number of different surface RyR2 populations were identified, and one of these groups, in which RyR2, Ca(v)1.2 and Ncx colocalized, might provide the structural basis for 'eager' sites of Ca(2+) release in atria. A small percentage of the dyads containing RyR2 and Ca(v)1.2 were colocalized with Cav3, and therefore could be influenced by the signalling molecules it anchors. The majority of the RyR2 clusters were tightly linked to Ca(v)1.2, and, whereas some were coupled to both Ca 1.2 and Ncx, none were with Ncx alone. This suggests that Ca(v)1.2-mediated Ca(2+) -induced Ca(2+) release is the primary method of ECC. The two molecules studied that were found in the interior of atrial cells, RyR2 and Casq, showed significantly less colocalization and a reduced nearest-neighbour distance in the interior, compared with the surface of the cell. These differences might result in a higher excitability for RyR2 in the interior of the cells, facilitating the spread of excitation from the periphery to the centre. We also present morphometric data for all of the molecules studied, as well as for those colocalizations found to be significant.
Collapse
Affiliation(s)
- Meredith N Schulson
- Department of Cellular and Physiological Sciences, University of British Columbia, Life Sciences Institute, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | | | | | | |
Collapse
|
50
|
Westcott EB, Jackson WF. Heterogeneous function of ryanodine receptors, but not IP3 receptors, in hamster cremaster muscle feed arteries and arterioles. Am J Physiol Heart Circ Physiol 2011; 300:H1616-30. [PMID: 21357503 DOI: 10.1152/ajpheart.00728.2010] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The roles played by ryanodine receptors (RyRs) and inositol 1,4,5-trisphosphate receptors (IP₃Rs) in vascular smooth muscle in the microcirculation remain unclear. Therefore, the function of both RyRs and IP₃Rs in Ca(²+) signals and myogenic tone in hamster cremaster muscle feed arteries and downstream arterioles were assessed using confocal imaging and pressure myography. Feed artery vascular smooth muscle displayed Ca(²+) sparks and Ca(²+) waves, which were inhibited by the RyR antagonists ryanodine (10 μM) or tetracaine (100 μM). Despite the inhibition of sparks and waves, ryanodine or tetracaine increased global intracellular Ca(²+) and constricted the arteries. The blockade of IP₃Rs with xestospongin D (5 μM) or 2-aminoethoxydiphenyl borate (100 μM) or the inhibition of phospholipase C using U-73122 (10 μM) also attenuated Ca(2+) waves without affecting Ca(²+) sparks. Importantly, the IP₃Rs and phospholipase C antagonists decreased global intracellular Ca(2+) and dilated the arteries. In contrast, cremaster arterioles displayed only Ca(²+) waves: Ca(²+) sparks were not observed, and neither ryanodine (10-50 μM) nor tetracaine (100 μM) affected either Ca(²+) signals or arteriolar tone despite the presence of functional RyRs as assessed by responses to the RyR agonist caffeine (10 mM). As in feed arteries, arteriolar Ca(²+) waves were attenuated by xestospongin D (5 μM), 2-aminoethoxydiphenyl borate (100 μM), and U-73122 (10 μM), accompanied by decreased global intracellular Ca(²+) and vasodilation. These findings highlight the contrasting roles played by RyRs and IP₃Rs in Ca(²+) signals and myogenic tone in feed arteries and demonstrate important differences in the function of RyRs between feed arteries and downstream arterioles.
Collapse
Affiliation(s)
- Erika B Westcott
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA.
| | | |
Collapse
|