1
|
Takahashi H, Nishitani K, Kawarasaki S, Martin-Morales A, Nagai H, Kuwata H, Tokura M, Okaze H, Mohri S, Ara T, Ito T, Nomura W, Jheng HF, Kawada T, Inoue K, Goto T. Metabolome analysis reveals that cyclic adenosine diphosphate ribose contributes to the regulation of differentiation in mice adipocyte. FASEB J 2024; 38:e23391. [PMID: 38145327 DOI: 10.1096/fj.202300850rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023]
Abstract
Adipocytes play a key role in energy storage and homeostasis. Although the role of transcription factors in adipocyte differentiation is known, the effect of endogenous metabolites of low molecular weight remains unclear. Here, we analyzed time-dependent changes in the levels of these metabolites throughout adipocyte differentiation, using metabolome analysis, and demonstrated that there is a positive correlation between cyclic adenosine diphosphate ribose (cADPR) and Pparγ mRNA expression used as a marker of differentiation. We also found that the treatment of C3H10T1/2 adipocytes with cADPR increased the mRNA expression of those marker genes and the accumulation of triglycerides. Furthermore, inhibition of ryanodine receptors (RyR), which are activated by cADPR, caused a significant reduction in mRNA expression levels of the marker genes and triglyceride accumulation in adipocytes. Our findings show that cADPR accelerates adipocytic differentiation via RyR pathway.
Collapse
Affiliation(s)
- Haruya Takahashi
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Kento Nishitani
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Satoko Kawarasaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Agustin Martin-Morales
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Hiroyuki Nagai
- Gifu Prefectural Research Institute for Health and Environmental Science, Gifu, Japan
| | - Hidetoshi Kuwata
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Motohiro Tokura
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Haruka Okaze
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Shinsuke Mohri
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Takeshi Ara
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tetsuro Ito
- Gifu Prefectural Research Institute for Health and Environmental Science, Gifu, Japan
- Laboratory of Pharmacognosy, Department of Pharmacy, Faculty of Pharmacy, Gifu University of Medical Science, Gifu, Japan
| | - Wataru Nomura
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
- Research Unit for Physiological Chemistry, Kyoto University, Kyoto, Japan
| | - Huei-Fen Jheng
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Teruo Kawada
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
- Research Unit for Physiological Chemistry, Kyoto University, Kyoto, Japan
| | - Kazuo Inoue
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
- Research Unit for Physiological Chemistry, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
- Research Unit for Physiological Chemistry, Kyoto University, Kyoto, Japan
| |
Collapse
|
2
|
Badin J, Rodenbeck S, McKenney-Drake ML, Sturek M. Multiphasic changes in smooth muscle Ca 2+ transporters during the progression of coronary atherosclerosis. CURRENT TOPICS IN MEMBRANES 2022; 90:95-121. [PMID: 36368876 DOI: 10.1016/bs.ctm.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Ischemic heart disease due to macrovascular atherosclerosis and microvascular dysfunction is the major cause of death worldwide and the unabated increase in metabolic syndrome is a major reason why this will continue. Intracellular free Ca2+ ([Ca2+]i) regulates a variety of cellular functions including contraction, proliferation, migration, and transcription. It follows that studies of vascular Ca2+ regulation in reductionist models and translational animal models are vital to understanding vascular health and disease. Swine with metabolic syndrome (MetS) develop the full range of coronary atherosclerosis from mild to severe disease. Intravascular imaging enables quantitative measurement of atherosclerosis in vivo, so viable coronary smooth muscle (CSM) cells can be dispersed from the arteries to enable Ca2+ transport studies in native cells. Transition of CSM from the contractile phenotype in the healthy swine to the proliferative phenotype in mild atherosclerosis was associated with increases in SERCA activity, sarcoplasmic reticulum Ca2+, and voltage-gated Ca2+ channel function. In vitro organ culture confirmed that SERCA activation induces CSM proliferation. Transition from the proliferative to a more osteogenic phenotype was associated with decreases in all three Ca2+ transporters. Overall, there was a biphasic change in Ca2+ transporters over the progression of atherosclerosis in the swine model and this was confirmed in CSM from failing explanted hearts of humans. A major determinant of endolysosome content in human CSM is the severity of atherosclerosis. In swine CSM endolysosome Ca2+ release occurred through the TPC2 channel. We propose a multiphasic change in Ca2+ transporters over the progression of coronary atherosclerosis.
Collapse
Affiliation(s)
- Jill Badin
- ZOLL Medical Corporation, Chelmsford, MA, United States
| | - Stacey Rodenbeck
- Department of Biology, Harding University, Searcy, AR, United States
| | - Mikaela L McKenney-Drake
- Butler University, Health Sciences Department, Pharmacy and Health Sciences, Indianapolis, IN, United States
| | - Michael Sturek
- Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
3
|
Faris P, Casali C, Negri S, Iengo L, Biggiogera M, Maione AS, Moccia F. Nicotinic Acid Adenine Dinucleotide Phosphate Induces Intracellular Ca2+ Signalling and Stimulates Proliferation in Human Cardiac Mesenchymal Stromal Cells. Front Cell Dev Biol 2022; 10:874043. [PMID: 35392169 PMCID: PMC8980055 DOI: 10.3389/fcell.2022.874043] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 02/24/2022] [Indexed: 12/18/2022] Open
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is a newly discovered second messenger that gates two pore channels 1 (TPC1) and 2 (TPC2) to elicit endo-lysosomal (EL) Ca2+ release. NAADP-induced lysosomal Ca2+ release may be amplified by the endoplasmic reticulum (ER) through the Ca2+-induced Ca2+ release (CICR) mechanism. NAADP-induced intracellular Ca2+ signals were shown to modulate a growing number of functions in the cardiovascular system, but their occurrence and role in cardiac mesenchymal stromal cells (C-MSCs) is still unknown. Herein, we found that exogenous delivery of NAADP-AM induced a robust Ca2+ signal that was abolished by disrupting the lysosomal Ca2+ store with Gly-Phe β-naphthylamide, nigericin, and bafilomycin A1, and blocking TPC1 and TPC2, that are both expressed at protein level in C-MSCs. Furthermore, NAADP-induced EL Ca2+ release resulted in the Ca2+-dependent recruitment of ER-embedded InsP3Rs and SOCE activation. Transmission electron microscopy revealed clearly visible membrane contact sites between lysosome and ER membranes, which are predicted to provide the sub-cellular framework for lysosomal Ca2+ to recruit ER-embedded InsP3Rs through CICR. NAADP-induced EL Ca2+ mobilization via EL TPC was found to trigger the intracellular Ca2+ signals whereby Fetal Bovine Serum (FBS) induces C-MSC proliferation. Furthermore, NAADP-evoked Ca2+ release was required to mediate FBS-induced extracellular signal-regulated kinase (ERK), but not Akt, phosphorylation in C-MSCs. These finding support the notion that NAADP-induced TPC activation could be targeted to boost proliferation in C-MSCs and pave the way for future studies assessing whether aberrant NAADP signaling in C-MSCs could be involved in cardiac disorders.
Collapse
Affiliation(s)
- Pawan Faris
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Claudio Casali
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Sharon Negri
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Lara Iengo
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Marco Biggiogera
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Angela Serena Maione
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
- *Correspondence: Angela Serena Maione, ; Francesco Moccia,
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
- *Correspondence: Angela Serena Maione, ; Francesco Moccia,
| |
Collapse
|
4
|
Hu W, Zhao F, Chen L, Ni J, Jiang Y. NAADP-induced intracellular calcium ion is mediated by the TPCs (two-pore channels) in hypoxia-induced pulmonary arterial hypertension. J Cell Mol Med 2021; 25:7485-7499. [PMID: 34263977 PMCID: PMC8335677 DOI: 10.1111/jcmm.16783] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 06/21/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a form of obstructive vascular disease. Chronic hypoxic exposure leads to excessive proliferation of pulmonary arterial smooth muscle cells and pulmonary arterial endothelial cells. This condition can potentially be aggravated by [Ca2+] i mobilization. In the present study, hypoxia exposure of rat's model was established. Two‐pore segment channels (TPCs) silencing was achieved in rats' models by injecting Lsh‐TPC1 or Lsh‐TPC2. The effects of TPC1/2 silencing on PAH were evaluated by H&E staining detecting pulmonary artery wall thickness and ELISA assay kit detecting NAADP concentrations in lung tissues. TPC1/2 silencing was achieved in PASMCs and PAECs, and cell proliferation was detected by MTT and BrdU incorporation assays. As the results shown, NAADP‐activated [Ca2+]i shows to be mediated via two‐pore segment channels (TPCs) in PASMCs, with TPC1 being the dominant subtype. NAADP generation and TPC1/2 mRNA and protein levels were elevated in the hypoxia‐induced rat PAH model; NAADP was positively correlated with TPC1 and TPC2 expression, respectively. In vivo, Lsh‐TPC1 or Lsh‐TPC2 infection significantly improved the mean pulmonary artery pressure and PAH morphology. In vitro, TPC1 silencing inhibited NAADP‐AM‐induced PASMC proliferation and [Ca2+]i in PASMCs, whereas TPC2 silencing had minor effects during this process; TPC2 silencing attenuated NAADP‐AM‐ induced [Ca2+]i and ECM in endothelial cells, whereas TPC1 silencing barely ensued any physiological changes. In conclusion, TPC1/2 might provide a unifying mechanism within pulmonary arterial hypertension, which can potentially be regarded as a therapeutic target.
Collapse
Affiliation(s)
- Wen Hu
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, China
| | - Fei Zhao
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, China
| | - Ling Chen
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, China
| | - Jiamin Ni
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, China
| | - Yongliang Jiang
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, China
| |
Collapse
|
5
|
Negri S, Faris P, Moccia F. Endolysosomal Ca 2+ signaling in cardiovascular health and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 363:203-269. [PMID: 34392930 DOI: 10.1016/bs.ircmb.2021.03.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
An increase in intracellular Ca2+ concentration ([Ca2+]i) regulates a plethora of functions in the cardiovascular (CV) system, including contraction in cardiomyocytes and vascular smooth muscle cells (VSMCs), and angiogenesis in vascular endothelial cells and endothelial colony forming cells. The sarco/endoplasmic reticulum (SR/ER) represents the largest endogenous Ca2+ store, which releases Ca2+ through ryanodine receptors (RyRs) and/or inositol-1,4,5-trisphosphate receptors (InsP3Rs) upon extracellular stimulation. The acidic vesicles of the endolysosomal (EL) compartment represent an additional endogenous Ca2+ store, which is targeted by several second messengers, including nicotinic acid adenine dinucleotide phosphate (NAADP) and phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2], and may release intraluminal Ca2+ through multiple Ca2+ permeable channels, including two-pore channels 1 and 2 (TPC1-2) and Transient Receptor Potential Mucolipin 1 (TRPML1). Herein, we discuss the emerging, pathophysiological role of EL Ca2+ signaling in the CV system. We describe the role of cardiac TPCs in β-adrenoceptor stimulation, arrhythmia, hypertrophy, and ischemia-reperfusion injury. We then illustrate the role of EL Ca2+ signaling in VSMCs, where TPCs promote vasoconstriction and contribute to pulmonary artery hypertension and atherosclerosis, whereas TRPML1 sustains vasodilation and is also involved in atherosclerosis. Subsequently, we describe the mechanisms whereby endothelial TPCs promote vasodilation, contribute to neurovascular coupling in the brain and stimulate angiogenesis and vasculogenesis. Finally, we discuss about the possibility to target TPCs, which are likely to mediate CV cell infection by the Severe Acute Respiratory Disease-Coronavirus-2, with Food and Drug Administration-approved drugs to alleviate the detrimental effects of Coronavirus Disease-19 on the CV system.
Collapse
Affiliation(s)
- Sharon Negri
- Laboratory of Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Pawan Faris
- Laboratory of Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Francesco Moccia
- Laboratory of Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy.
| |
Collapse
|
6
|
Moccia F, Negri S, Faris P, Perna A, De Luca A, Soda T, Berra-Romani R, Guerra G. Targeting Endolysosomal Two-Pore Channels to Treat Cardiovascular Disorders in the Novel COronaVIrus Disease 2019. Front Physiol 2021; 12:629119. [PMID: 33574769 PMCID: PMC7870486 DOI: 10.3389/fphys.2021.629119] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
Emerging evidence hints in favor of a life-threatening link between severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) and the cardiovascular system. SARS-CoV-2 may result in dramatic cardiovascular complications, whereas the severity of COronaVIrus Disease 2019 (COVID-19) and the incidence of fatalities tend to increase in patients with pre-existing cardiovascular complications. SARS-CoV-2 is internalized into the host cells by endocytosis and may then escape the endolysosomal system via endosomes. Two-pore channels drive endolysosomal trafficking through the release of endolysosomal Ca2+. Recent evidence suggested that the pharmacological inhibition of TPCs prevents Ebola virus and Middle East Respiratory Syndrome COronaVirus (MERS-CoV) entry into host cells. In this perspective, we briefly summarize the biophysical and pharmacological features of TPCs, illustrate their emerging role in the cardiovascular system, and finally present them as a reliable target to treat cardiovascular complications in COVID-19 patients.
Collapse
Affiliation(s)
- Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Sharon Negri
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Pawan Faris
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Angelica Perna
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Antonio De Luca
- Section of Human Anatomy, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Teresa Soda
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Roberto Berra-Romani
- School of Medicine, Department of Biomedicine, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Germano Guerra
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| |
Collapse
|
7
|
The Role of Two-Pore Channels in Norepinephrine-Induced [Ca 2+] i Rise in Rat Aortic Smooth Muscle Cells and Aorta Contraction. Cells 2019; 8:cells8101144. [PMID: 31557916 PMCID: PMC6829401 DOI: 10.3390/cells8101144] [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: 07/31/2019] [Revised: 09/21/2019] [Accepted: 09/23/2019] [Indexed: 12/17/2022] Open
Abstract
Second messenger nicotinic acid adenine dinucleotide phosphate (NAADP) triggers Ca2+ release via two-pore channels (TPCs) localized in endolysosomal vesicles. The aim of the present work is to evaluate the role of TPCs in the action of norepinephrine (NE), angiotensin II (AngII), vasopressin (AVP), and 5-hydroxytriptamine (5-HT) on free cytoplasmic calcium concentration ([Ca2+]i) in smooth muscle cells (SMCs) isolated from rat aorta and on aorta contraction. To address this issue, the NAADP structural analogue and inhibitor of TPCs, NED 19, was applied. We have demonstrated a high degree of colocalization of the fluorescent signals of cis-NED 19 and endolysosmal probe LysoTracker in SMCs. Both cis- or trans-NED 19 inhibited the rise of [Ca2+]i in SMCs induced by 100 μM NE by 50–60%. IC50 for cis- and trans-NED 19 were 2.7 and 8.9 μM, respectively. The inhibition by NED 19 stereoisomers of the effects of AngII, AVP, and 5-HT was much weaker. Both forms of NED 19 caused relaxation of aortic rings preconstricted by NE, with relative potency of cis-NED 19 several times higher than that of trans-NED 19. Inhibition by cis-NED 19 of NE-induced contraction was maintained after intensive washing and slowly reversed within an hour of incubation. Cis- and trans-NED 19 did not cause decrease in the force of aorta contraction in response to Ang II and AVP, and only slightly relaxed aorta preconstricted by 5-HT and by KCl. Suppression of TPC1 in SMCs with siRNA caused a 40% decrease in [Ca2+]i in response to NE, whereas siRNA against TPC2 did not change NE calcium signaling. These data suggest that TPC1 is involved in the NE-stimulated [Ca2+]i rise in SMCs. Inhibition of TPC1 activity by NED 19 could be the reason for partial inhibition of aortic rings contraction in response to NE.
Collapse
|
8
|
Faris P, Pellavio G, Ferulli F, Di Nezza F, Shekha M, Lim D, Maestri M, Guerra G, Ambrosone L, Pedrazzoli P, Laforenza U, Montagna D, Moccia F. Nicotinic Acid Adenine Dinucleotide Phosphate (NAADP) Induces Intracellular Ca 2+ Release through the Two-Pore Channel TPC1 in Metastatic Colorectal Cancer Cells. Cancers (Basel) 2019; 11:cancers11040542. [PMID: 30991693 PMCID: PMC6521149 DOI: 10.3390/cancers11040542] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 04/09/2019] [Indexed: 12/12/2022] Open
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) gates two-pore channels 1 and 2 (TPC1 and TPC2) to elicit endo-lysosomal (EL) Ca2+ release. NAADP-induced EL Ca2+ signals may be amplified by the endoplasmic reticulum (ER) through the Ca2+-induced Ca2+ release mechanism (CICR). Herein, we aimed at assessing for the first time the role of EL Ca2+ signaling in primary cultures of human metastatic colorectal carcinoma (mCRC) by exploiting Ca2+ imaging and molecular biology techniques. The lysosomotropic agent, Gly-Phe β-naphthylamide (GPN), and nigericin, which dissipates the ΔpH which drives Ca2+ refilling of acidic organelles, caused massive Ca2+ release in the presence of a functional inositol-1,4,5-trisphosphate (InsP3)-sensitive ER Ca2+ store. Liposomal delivery of NAADP induced a transient Ca2+ release that was reduced by GPN and NED-19, a selective TPC antagonist. Pharmacological and genetic manipulations revealed that the Ca2+ response to NAADP was triggered by TPC1, the most expressed TPC isoform in mCRC cells, and required ER-embedded InsP3 receptors. Finally, NED-19 and genetic silencing of TPC1 reduced fetal calf serum-induced Ca2+ signals, proliferation, and extracellular signal-regulated kinase and Akt phoshorylation in mCRC cells. These data demonstrate that NAADP-gated TPC1 could be regarded as a novel target for alternative therapies to treat mCRC.
Collapse
Affiliation(s)
- Pawan Faris
- Laboratory of General Physiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy.
- Research Centre, Salahaddin University-Erbil, 44001 Erbil, Kurdistan-Region of Iraq, Iraq.
| | - Giorgia Pellavio
- Human Physiology Unit, via Forlanini 6, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy.
| | - Federica Ferulli
- Laboratory of Immunology Transplantation, Foundation IRCCS Policlinico San Matteo, 27100 Pavia, Italy.
| | - Francesca Di Nezza
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, 86100 Campobasso, Italy.
| | - Mudhir Shekha
- Research Centre, Salahaddin University-Erbil, 44001 Erbil, Kurdistan-Region of Iraq, Iraq.
- Department of Pathological Analysis, College of Science, Knowledge University, 074016 Erbil, Kurdistan-Region of Iraq, Iraq.
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, 28100 Novara, Italy.
| | - Marcello Maestri
- Unit of General Surgery, Foundation IRCCS Policlinico San Matteo, 27100 Pavia, Italy.
- Department of Sciences Clinic-Surgical, Diagnostic and Pediatric, University of Pavia, 27100 Pavia, Italy.
| | - Germano Guerra
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, 86100 Campobasso, Italy.
| | - Luigi Ambrosone
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, 86100 Campobasso, Italy.
| | - Paolo Pedrazzoli
- Medical Oncology, oundation IRCCS Policlinico San Matteo, 27100 Pavia, Italy.
| | - Umberto Laforenza
- Human Physiology Unit, via Forlanini 6, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy.
| | - Daniela Montagna
- Laboratory of Immunology Transplantation, Foundation IRCCS Policlinico San Matteo, 27100 Pavia, Italy.
- Department of Sciences Clinic-Surgical, Diagnostic and Pediatric, University of Pavia, 27100 Pavia, Italy.
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy.
| |
Collapse
|
9
|
Endolysosomal Ca 2+ Signalling and Cancer Hallmarks: Two-Pore Channels on the Move, TRPML1 Lags Behind! Cancers (Basel) 2018; 11:cancers11010027. [PMID: 30591696 PMCID: PMC6356888 DOI: 10.3390/cancers11010027] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 12/22/2022] Open
Abstract
The acidic vesicles of the endolysosomal (EL) system are emerging as an intracellular Ca2+ store implicated in the regulation of multiple cellular functions. The EL Ca2+ store releases Ca2+ through a variety of Ca2+-permeable channels, including Transient Receptor Potential (TRP) Mucolipin 1-3 (TRPML1-3) and two-pore channels 1-2 (TPC1-2), whereas EL Ca2+ refilling is sustained by the proton gradient across the EL membrane and/or by the endoplasmic reticulum (ER). EL Ca2+ signals may be either spatially restricted to control vesicle trafficking, autophagy and membrane repair or may be amplified into a global Ca2+ signal through the Ca2+-dependent recruitment of ER-embedded channels. Emerging evidence suggested that nicotinic acid adenine dinucleotide phosphate (NAADP)-gated TPCs sustain multiple cancer hallmarks, such as migration, invasiveness and angiogenesis. Herein, we first survey the EL Ca2+ refilling and release mechanisms and then focus on the oncogenic role of EL Ca2+ signaling. While the evidence in favor of TRPML1 involvement in neoplastic transformation is yet to be clearly provided, TPCs are emerging as an alternative target for anticancer therapies.
Collapse
|
10
|
Yip KP, Balasubramanian L, Kan C, Wang L, Liu R, Ribeiro-Silva L, Sham JSK. Intraluminal pressure triggers myogenic response via activation of calcium spark and calcium-activated chloride channel in rat renal afferent arteriole. Am J Physiol Renal Physiol 2018; 315:F1592-F1600. [PMID: 30089032 DOI: 10.1152/ajprenal.00239.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Myogenic contraction of renal arterioles is an important regulatory mechanism for renal blood flow autoregulation. We have previously demonstrated that integrin-mediated mechanical force increases the occurrence of Ca2+ sparks in freshly isolated renal vascular smooth muscle cells (VSMCs). To further test whether the generation of Ca2+ sparks is a downstream signal of mechanotransduction in pressure-induced myogenic constriction, the relationship between Ca2+ sparks and transmural perfusion pressure was investigated in intact VSMCs of pressurized rat afferent arterioles. Spontaneous Ca2+ sparks were found in VSMCs when afferent arterioles were perfused at 80 mmHg. The spark frequency was significantly increased when perfusion pressure was increased to 120 mmHg. A similar increase of spark frequency was also observed in arterioles stimulated with β1-integrin-activating antibody. Moreover, spark frequency was significantly higher in arterioles of spontaneous hypertensive rats at 80 and 120 mmHg. Spontaneous membrane current recorded using whole cell perforated patch in renal VSMCs showed predominant activity of spontaneous transient inward currents instead of spontaneous transient outward currents when holding potential was set close to physiological resting membrane potential. Real-time PCR and immunohistochemistry confirmed the expression of Ca2+-activated Cl- channel (ClCa) TMEM16A in renal VSMCs. Inhibition of TMEM16A with T16Ainh-A01 impaired the pressure-induced myogenic contraction in perfused afferent arterioles. Our study, for the first time to our knowledge, detected Ca2+ sparks in VSMCs of intact afferent arterioles, and their frequencies were positively modulated by the perfusion pressure. Our results suggest that Ca2+ sparks may couple to ClCa channels and trigger pressure-induced myogenic constriction via membrane depolarization.
Collapse
Affiliation(s)
- Kay-Pong Yip
- Department of Molecular Pharmacology and Physiology, University of South Florida , Tampa, Florida
| | - Lavanya Balasubramanian
- Department of Molecular Pharmacology and Physiology, University of South Florida , Tampa, Florida
| | - Chen Kan
- Department of Industrial, Manufacturing, and System Engineering, University of Texas at Arlington , Arlington, Texas
| | - Lei Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida , Tampa, Florida
| | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology, University of South Florida , Tampa, Florida
| | - Luisa Ribeiro-Silva
- Department of Molecular Pharmacology and Physiology, University of South Florida , Tampa, Florida
| | - James S K Sham
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland
| |
Collapse
|
11
|
Jiang Y, Zhou Y, Peng G, Tian H, Pan D, Liu L, Yang X, Li C, Li W, Chen L, Ran P, Dai A. Two-pore channels mediated receptor-operated Ca 2+ entry in pulmonary artery smooth muscle cells in response to hypoxia. Int J Biochem Cell Biol 2018; 97:28-35. [PMID: 29355755 DOI: 10.1016/j.biocel.2018.01.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/12/2018] [Accepted: 01/15/2018] [Indexed: 01/26/2023]
Abstract
The aim of this study was to investigate the influence of two-pore channels mediated receptor-operated Ca2+ entry on pulmonary arterial smooth muscle cell (PASMC) under hypoxia conditions. PASMCs were separated using the direct adherent culture method. The cultured cells were observed under optic microscope and the phenotypes of cells were identified by immunohistochemistry. The expression of NAADP was examined by ELISA. CaN, TPC1, TPC2 and NFATc3 protein levels were examined using Western blotting. Real-time PCR was utilized to detect the level of TPC1 and TPC2 mRNA. Fluorescent probe technique was used to explore the [Ca2+]i in PASMCs. Proliferation and migration of PASMCs were examined by MTT assay and Transwell, respectively. The results showed that cells displayed a typical "peak-valley" growth pattern and positive for α-actin staining. Expression of NAADP, CaN, NFATc3, TPC1 and TPC2 under PASMCs exposed to hypoxia after 24 h and 48 h were higher than control, however, cells treated with Ned-19 were significantly decreased compared with control. Levels of CaN and NFATc3 protein collected from RPASMCs transfected with TPCs siRNA were observably decreased than scrambled siRNA. Under hypoxia condition for 12 h, 24 h and 48 h, TPC1 and TPC2 mRNA levels were higher in PASMCs compared as control. The [Ca2+]i evoked by hypoxia significantly increased than normoxia group. Nevertheless, the [Ca2+]i of the groups treated with Ned-19 and transfected with TPCs siRNA were markedly lower compared with control. In conclusion, the TPCs influence on function of pulmonary artery smooth muscle cells by mediated Ca2+ Signals under hypoxia condition.
Collapse
Affiliation(s)
- Yongliang Jiang
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410219, PR China
| | - Yumin Zhou
- State Key Lab of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, PR China
| | - Gongyong Peng
- State Key Lab of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, PR China
| | - Heshen Tian
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410219, PR China
| | - Dan Pan
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410219, PR China
| | - Lei Liu
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410219, PR China
| | - Xing Yang
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410219, PR China
| | - Chao Li
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410219, PR China
| | - Wen Li
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410219, PR China
| | - Ling Chen
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410219, PR China
| | - Pixin Ran
- State Key Lab of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, PR China.
| | - Aiguo Dai
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410219, PR China; Institute of Respiratory Medicine, Changsha Medical College, Changsha 410219, PR China.
| |
Collapse
|
12
|
Lin AHY, Shang Y, Mitzner W, Sham JSK, Tang WY. Aberrant DNA Methylation of Phosphodiesterase [corrected] 4D Alters Airway Smooth Muscle Cell Phenotypes. Am J Respir Cell Mol Biol 2016; 54:241-9. [PMID: 26181301 DOI: 10.1165/rcmb.2015-0079oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Airway hyperresponsiveness (AHR) is a hallmark feature in asthma characterized by exaggerated airway contractile response to stimuli due to increased airway sensitivity and chronic airway remodeling. We have previously shown that allergen-induced AHR in mice is associated with aberrant DNA methylation in the lung genome, suggesting that AHR could be epigenetically regulated, and these changes might predispose the animals to asthma. Previous studies demonstrated that overexpression of phosphodiesterase 4D (PDE4D) is associated with increased AHR. However, epigenetic regulation of this gene in asthmatic airway smooth muscle cells (ASMCs) has not been examined. In this study, we aimed to examine the relationship between epigenetic regulation of PDE4D and ASMC phenotypes. We identified CpG site-specific hypomethylation at PDE4D promoter in human asthmatic ASMCs. We next used methylated oligonucleotides to introduce CpG site-specific methylation at PDE4D promoter and examined its effect on ASMCs. We showed that PDE4D methylation decreased cell proliferation and migration of asthmatic ASMCs. We further elucidated that methylated PDE4D decreased PDE4D expression in asthmatic ASMCs, increased cAMP level, and inhibited the aberrant increase in Ca(2+) level. Moreover, PDE4D methylation reduced the phosphorylation level of downstream effectors of Ca(2+) signaling, including myosin light chain kinase and p38. Taken together, our findings demonstrate that gene-specific epigenetic changes may predispose ASMCs to asthma through alterations in cell phenotypes. Modulation of ASMC phenotypes by methylated PDE4D oligonucleotides can reverse the aberrant ASMC functions to normal phenotypes. This has provided new insight to the development of novel therapeutic options for this debilitative disease.
Collapse
Affiliation(s)
- Amanda H Y Lin
- 1 Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland; and
| | - Yan Shang
- 2 Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Wayne Mitzner
- 2 Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - James S K Sham
- 1 Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland; and.,2 Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Wan-yee Tang
- 2 Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
13
|
Abstract
Extracellular stimuli evoke the synthesis of intracellular second messengers, several of which couple to the release of Ca2+ from Ca2+-storing organelles via activation of cognate organellar Ca2+-channel complexes. The archetype is the inositol 1,4,5-trisphosphate (IP3) and IP3 receptor (IP3R) on the endoplasmic reticulum (ER). A less understood, parallel Ca2+ signalling cascade is that involving the messenger nicotinic acid adenine dinucleotide phosphate (NAADP) that couples to Ca2+ release from acidic Ca2+ stores [e.g. endo-lysosomes, secretory vesicles, lysosome-related organelles (LROs)]. NAADP-induced Ca2+ release absolutely requires organellar TPCs (two-pore channels). This review discusses how ER and acidic Ca2+ stores physically and functionally interact to generate and shape global and local Ca2+ signals, with particular emphasis on the two-way dialogue between these two organelles.
Collapse
|
14
|
Zhang DX, Zhang JP, Hu JY, Huang YS. The potential regulatory roles of NAD(+) and its metabolism in autophagy. Metabolism 2016; 65:454-62. [PMID: 26975537 DOI: 10.1016/j.metabol.2015.11.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 10/29/2015] [Accepted: 11/25/2015] [Indexed: 02/02/2023]
Abstract
(Macro)autophagy mediates the bulk degradation of defective organelles, long-lived proteins and protein aggregates in lysosomes and plays a critical role in cellular and tissue homeostasis. Defective autophagy processes have been found to contribute to a variety of metabolic diseases. However, the regulatory mechanisms of autophagy are not fully understood. Increasing data indicate that nicotinamide adenine nucleotide (NAD(+)) homeostasis correlates intimately with autophagy. NAD(+) is a ubiquitous coenzyme that functions primarily as an electron carrier of oxidoreductase in multiple redox reactions. Both NAD(+) homeostasis and its metabolism are thought to play critical roles in regulating autophagy. In this review, we discuss how the regulation of NAD(+) and its metabolism can influence autophagy. We focus on the regulation of NAD(+)/NADH homeostasis and the effects of NAD(+) consumption by poly(ADP-ribose) (PAR) polymerase-1 (PARP-1), NAD(+)-dependent deacetylation by sirtuins and NAD(+) metabolites on autophagy processes and the underlying mechanisms. Future studies should provide more direct evidence for the regulation of autophagy processes by NAD(+). A better understanding of the critical roles of NAD(+) and its metabolites on autophagy will shed light on the complexity of autophagy regulation, which is essential for the discovery of new therapeutic tools for autophagy-related diseases.
Collapse
Affiliation(s)
- Dong-Xia Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, PR China, 400038
| | - Jia-Ping Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, PR China, 400038
| | - Jiong-Yu Hu
- Endocrinology Department, Southwest Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, PR China, 400038
| | - Yue-Sheng Huang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, PR China, 400038.
| |
Collapse
|
15
|
Lin AHY, Sun H, Paudel O, Lin MJ, Sham JSK. Conformation of ryanodine receptor-2 gates store-operated calcium entry in rat pulmonary arterial myocytes. Cardiovasc Res 2016; 111:94-104. [PMID: 27013634 DOI: 10.1093/cvr/cvw067] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 03/18/2016] [Indexed: 12/31/2022] Open
Abstract
AIMS Store-operated Ca(2+) entry (SOCE) contributes to a multitude of physiological and pathophysiological functions in pulmonary vasculatures. SOCE attributable to inositol 1,4,5-trisphosphate receptor (InsP3R)-gated Ca(2+) store has been studied extensively, but the role of ryanodine receptor (RyR)-gated store in SOCE remains unclear. The present study aims to delineate the relationship between RyR-gated Ca(2+) stores and SOCE, and characterize the properties of RyR-gated Ca(2+) entry in pulmonary artery smooth muscle cells (PASMCs). METHODS AND RESULTS PASMCs were isolated from intralobar pulmonary arteries of male Wister rats. Application of the RyR1/2 agonist 4-chloro-m-cresol (4-CmC) activated robust Ca(2+) entry in PASMCs. It was blocked by Gd(3+) and the RyR2 modulator K201 but was unaffected by the RyR1/3 antagonist dantrolene and the InsP3R inhibitor xestospongin C, suggesting RyR2 is mainly involved in the process. siRNA knockdown of STIM1, TRPC1, and Orai1, or interruption of STIM1 translocation with ML-9 significantly attenuated the 4-CmC-induced SOCE, similar to SOCE induced by thapsigargin. However, depletion of RyR-gated store with caffeine failed to activate Ca(2+) entry. Inclusion of ryanodine, which itself did not cause Ca(2+) entry, uncovered caffeine-induced SOCE in a concentration-dependent manner, suggesting binding of ryanodine to RyR is permissive for the process. This Ca(2+) entry had the same molecular and pharmacological properties of 4-CmC-induced SOCE, and it persisted once activated even after caffeine washout. Measurement of Ca(2+) in sarcoplasmic reticulum (SR) showed that 4-CmC and caffeine application with or without ryanodine reduced SR Ca(2+) to similar extent, suggesting store-depletion was not the cause of the discrepancy. Moreover, caffeine/ryanodine and 4-CmC failed to initiate SOCE in cells transfected with the ryanodine-binding deficient mutant RyR2-I4827T. CONCLUSIONS RyR2-gated Ca(2+) store contributes to SOCE in PASMCs; however, store-depletion alone is insufficient but requires a specific RyR conformation modifiable by ryanodine binding to activate Ca(2+) entry.
Collapse
Affiliation(s)
- Amanda H Y Lin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| | - Hui Sun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| | - Omkar Paudel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| | - Mo-Jun Lin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - James S K Sham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| |
Collapse
|
16
|
Wolf IMA, Diercks BP, Gattkowski E, Czarniak F, Kempski J, Werner R, Schetelig D, Mittrücker HW, Schumacher V, von Osten M, Lodygin D, Flügel A, Fliegert R, Guse AH. Frontrunners of T cell activation: Initial, localized Ca2+ signals mediated by NAADP and the type 1 ryanodine receptor. Sci Signal 2015; 8:ra102. [PMID: 26462735 DOI: 10.1126/scisignal.aab0863] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The activation of T cells is the fundamental on switch for the adaptive immune system. Ca(2+) signaling is essential for T cell activation and starts as initial, short-lived, localized Ca(2+) signals. The second messenger nicotinic acid adenine dinucleotide phosphate (NAADP) forms rapidly upon T cell activation and stimulates early Ca(2+) signaling. We developed a high-resolution imaging technique using multiple fluorescent Ca(2+) indicator dyes to characterize these early signaling events and investigate the channels involved in NAADP-dependent Ca(2+) signals. In the first seconds of activation of either primary murine T cells or human Jurkat cells with beads coated with an antibody against CD3, we detected Ca(2+) signals with diameters close to the limit of detection and that were close to the activation site at the plasma membrane. In Jurkat cells in which the ryanodine receptor (RyR) was knocked down or in primary T cells from RyR1(-/-) mice, either these early Ca(2+) signals were not detected or the number of signals was markedly reduced. Local Ca(2+) signals observed within 20 ms upon microinjection of Jurkat cells with NAADP were also sensitive to RyR knockdown. In contrast, TRPM2 (transient receptor potential channel, subtype melastatin 2), a potential NAADP target channel, was not required for the formation of initial Ca(2+) signals in primary T cells. Thus, through our high-resolution imaging method, we characterized early Ca(2+) release events in T cells and obtained evidence for the involvement of RyR and NAADP in such signals.
Collapse
Affiliation(s)
- Insa M A Wolf
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ellen Gattkowski
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Frederik Czarniak
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jan Kempski
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - René Werner
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Daniel Schetelig
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Hans-Willi Mittrücker
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Valéa Schumacher
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Manuel von Osten
- Institute for Multiple Sclerosis Research, Department of Neuroimmunology, Gemeinnützige Hertie-Stiftung and University Medical Center Göttingen, Waldweg 33, 37073 Göttingen, Germany. Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Dimitri Lodygin
- Institute for Multiple Sclerosis Research, Department of Neuroimmunology, Gemeinnützige Hertie-Stiftung and University Medical Center Göttingen, Waldweg 33, 37073 Göttingen, Germany. Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Alexander Flügel
- Institute for Multiple Sclerosis Research, Department of Neuroimmunology, Gemeinnützige Hertie-Stiftung and University Medical Center Göttingen, Waldweg 33, 37073 Göttingen, Germany. Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Ralf Fliegert
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Andreas H Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
17
|
Davidson SM, Foote K, Kunuthur S, Gosain R, Tan N, Tyser R, Zhao YJ, Graeff R, Ganesan A, Duchen MR, Patel S, Yellon DM. Inhibition of NAADP signalling on reperfusion protects the heart by preventing lethal calcium oscillations via two-pore channel 1 and opening of the mitochondrial permeability transition pore. Cardiovasc Res 2015; 108:357-66. [PMID: 26395965 PMCID: PMC4648198 DOI: 10.1093/cvr/cvv226] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 08/06/2015] [Indexed: 12/21/2022] Open
Abstract
Aims In the heart, a period of ischaemia followed by reperfusion evokes powerful cytosolic Ca2+ oscillations that can cause lethal cell injury. These signals represent attractive cardioprotective targets, but the underlying mechanisms of genesis are ill-defined. Here, we investigated the role of the second messenger nicotinic acid adenine dinucleotide phosphate (NAADP), which is known in several cell types to induce Ca2+ oscillations that initiate from acidic stores such as lysosomes, likely via two-pore channels (TPCs, TPC1 and 2). Methods and results An NAADP antagonist called Ned-K was developed by rational design based on a previously existing scaffold. Ned-K suppressed Ca2+ oscillations and dramatically protected cardiomyocytes from cell death in vitro after ischaemia and reoxygenation, preventing opening of the mitochondrial permeability transition pore. Ned-K profoundly decreased infarct size in mice in vivo. Transgenic mice lacking the endo-lysosomal TPC1 were also protected from injury. Conclusion NAADP signalling plays a major role in reperfusion-induced cell death and represents a potent pathway for protection against reperfusion injury.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| | - Kirsty Foote
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| | - Suma Kunuthur
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| | - Raj Gosain
- School of Chemistry, University of Southampton, Highfield, Southampton, UK
| | - Noah Tan
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| | - Richard Tyser
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| | - Yong Juan Zhao
- Department of Physiology, Li Ka Shing School of Medicine, The University of Hong Kong, Hong Kong, China
| | - Richard Graeff
- Department of Physiology, Li Ka Shing School of Medicine, The University of Hong Kong, Hong Kong, China
| | - A Ganesan
- School of Pharmacy, University of East Anglia, Norwich, UK
| | - Michael R Duchen
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| |
Collapse
|
18
|
Lee S, Paudel O, Jiang Y, Yang XR, Sham JSK. CD38 mediates angiotensin II-induced intracellular Ca(2+) release in rat pulmonary arterial smooth muscle cells. Am J Respir Cell Mol Biol 2015; 52:332-41. [PMID: 25078456 DOI: 10.1165/rcmb.2014-0141oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
CD38 is a multifunctional enzyme that catalyzes the formation of the endogenous Ca(2+)-mobilizing messengers cyclic ADP-ribose (cADPR) and nicotinic acid adenosine dinucleotide phosphate (NAADP) for the activation of ryanodine receptors (RyRs) of sarcoplasmic reticulum and NAADP-sensitive Ca(2+) release channels in endolysosomes, respectively. It plays important roles in systemic vascular functions, but there is little information on CD38 in pulmonary arterial smooth muscle cells (PASMCs). Earlier studies suggested a redox-sensing role of CD38 in hypoxic pulmonary vasoconstriction. This study sought to characterize its roles in angiotensin II (Ang II)-induced Ca(2+) release (AICR) in PASMCs. Examination of CD38 expression in various rat arteries found high levels of CD38 mRNA and protein in pulmonary arteries. The Ang II-elicited Ca(2+) response consisted of extracellular Ca(2+) influx and intracellular Ca(2+) release in PASMCs. AICR activated in the absence of extracellular Ca(2+) was reduced by pharmacological or siRNA inhibition of CD38, by the cADPR antagonist 8-bromo-cADPR or ryanodine, and by the NAADP antagonist Ned-19 or disruption of endolysosomal Ca(2+) stores with the vacuolar H(+)-ATPase inhibitor bafilomycin A1. Suppression of AICR by the inhibitions of cADPR- and NAADP-dependent pathways were nonadditive, indicating interdependence of RyR- and NAADP-gated Ca(2+) release. Furthermore, AICR was inhibited by the protein kinase C inhibitor staurosporine, the nonspecific NADPH oxidase (NOX) inhibitors apocynin and diphenyleneiodonium, the NOX2-specific inhibitor gp91ds-tat, and the scavenger of reactive oxygen species (ROS) tempol. These results provide the first evidence that Ang II activates CD38-dependent Ca(2+) release via the NOX2-ROS pathway in PASMCs.
Collapse
Affiliation(s)
- Suengwon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | | | | |
Collapse
|
19
|
Galione A. A primer of NAADP-mediated Ca(2+) signalling: From sea urchin eggs to mammalian cells. Cell Calcium 2014; 58:27-47. [PMID: 25449298 DOI: 10.1016/j.ceca.2014.09.010] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 09/28/2014] [Accepted: 09/29/2014] [Indexed: 02/04/2023]
Abstract
Since the discovery of the Ca(2+) mobilizing effects of the pyridine nucleotide metabolite, nicotinic acid adenine dinucleotide phosphate (NAADP), this molecule has been demonstrated to function as a Ca(2+) mobilizing intracellular messenger in a wide range of cell types. In this review, I will briefly summarize the distinct principles behind NAADP-mediated Ca(2+) signalling before going on to outline the role of this messenger in the physiology of specific cell types. Central to the discussion here is the finding that NAADP principally mobilizes Ca(2+) from acidic organelles such as lysosomes and it is this property that allows NAADP to play a unique role in intracellular Ca(2+) signalling. Lysosomes and related organelles are small Ca(2+) stores but importantly may also initiate a two-way dialogue with other Ca(2+) storage organelles to amplify Ca(2+) release, and may be strategically localized to influence localized Ca(2+) signalling microdomains. The study of NAADP signalling has created a new and fruitful focus on the lysosome and endolysosomal system as major players in calcium signalling and pathophysiology.
Collapse
Affiliation(s)
- Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| |
Collapse
|
20
|
Pitt SJ, Lam AKM, Rietdorf K, Galione A, Sitsapesan R. Reconstituted human TPC1 is a proton-permeable ion channel and is activated by NAADP or Ca2+. Sci Signal 2014; 7:ra46. [PMID: 24847115 DOI: 10.1126/scisignal.2004854] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
NAADP potently triggers Ca2+ release from acidic lysosomal and endolysosomal Ca2+ stores. Human two-pore channels (TPC1 and TPC2), which are located on these stores, are involved in this process, but there is controversy over whether TPC1 and TPC2 constitute the Ca2+ release channels. We therefore examined the single-channel properties of human TPC1 after reconstitution into bilayers of controlled composition. We found that TPC1 was permeable not only to Ca2+ but also to monovalent cations and that permeability to protons was the highest (relative permeability sequence: H+ >> K+ > Na(+) ≥ Ca2+). NAADP or Ca2+ activated TPC1, and the presence of one of these ligands was required for channel activation. The endolysosome-located lipid phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2] had no effect on TPC1 open probability but significantly increased the relative permeability of Na+ to Ca2+ and of H+ to Ca2+. Furthermore, our data showed that, although both TPC1 and TPC2 are stimulated by NAADP, these channels differ in ion selectivity and modulation by Ca2+ and pH. We propose that NAADP triggers H+ release from lysosomes and endolysomes through activation of TPC1, but that the Ca2+ -releasing ability of TPC1 will depend on the ionic composition of the acidic stores and may be influenced by other regulators that affect TPC1 ion permeation.
Collapse
Affiliation(s)
- Samantha J Pitt
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
| | - Andy K M Lam
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Katja Rietdorf
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Rebecca Sitsapesan
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| |
Collapse
|
21
|
Pereira GJS, Hirata H, do Carmo LG, Stilhano RS, Ureshino RP, Medaglia NC, Han SW, Churchill G, Bincoletto C, Patel S, Smaili SS. NAADP-sensitive two-pore channels are present and functional in gastric smooth muscle cells. Cell Calcium 2014; 56:51-8. [PMID: 24882212 DOI: 10.1016/j.ceca.2014.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 04/11/2014] [Accepted: 04/21/2014] [Indexed: 01/06/2023]
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) has been identified as an important modulator of Ca(2+) release from the endo-lysosomal system in a variety of cells by a new and ubiquitous class of endo-lysosomal ion channels known as the two-pore channels (TPCs). However, the role of TPCs in NAADP action in smooth muscle is not known. In the present work, we investigated the effects of NAADP in gastric smooth muscle cells and its ability to release Ca(2+) by TPCs. We show that Ca(2+) signals mediated by NAADP were inhibited by disrupting Ca(2+) handling by either acidic organelles (using bafilomycin A1) or the Endoplasmic Reticulum (using thapsigargin, ryanodine or 2-APB). Transcripts for endogenous TPC1 and TPC2 were readily detected and recombinant TPCs localized to the endosomes and/or lysosomes. Overexpression of wild-type TPCs but not pore mutants enhanced NAADP-mediated cytosolic Ca(2+) signals. Desensitizing the NAADP pathway inhibited Ca(2+)-responses to extracellular stimulation with carbachol but not ATP. Taken together, these results indicate that NAADP likely induces Ca(2+) release from the endolysosomal system through TPCs which is subsequently amplified via the ER in an agonist-specific manner. Thus, we suggest a second messenger role for NAADP in smooth muscle cells.
Collapse
Affiliation(s)
- Gustavo J S Pereira
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Hanako Hirata
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Lúcia G do Carmo
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Roberta S Stilhano
- Interdisciplinary Center for Gene Therapy, Federal University of São Paulo, São Paulo, Brazil
| | - Rodrigo P Ureshino
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Natalia C Medaglia
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Sang W Han
- Interdisciplinary Center for Gene Therapy, Federal University of São Paulo, São Paulo, Brazil
| | - Grant Churchill
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Claudia Bincoletto
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Soraya S Smaili
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil.
| |
Collapse
|
22
|
Gilbert G, Ducret T, Marthan R, Savineau JP, Quignard JF. Stretch-induced Ca2+ signalling in vascular smooth muscle cells depends on Ca2+ store segregation. Cardiovasc Res 2014; 103:313-23. [PMID: 24692174 DOI: 10.1093/cvr/cvu069] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AIM Calcium is a key second messenger that can be mobilized from both the extracellular medium and intracellular calcium stores. Pulmonary arterial smooth muscle cells (PASMCs) respond to stretch by a calcium increase, a mechanism enhanced during pulmonary hypertension (PH). We investigated the role of the spatial organization between plasma membrane stretch-activated channels (SACs) and intracellular calcium stores [sarcoplasmic reticulum (SR), mitochondria, and lysosomes) in response to stretch. METHODS AND RESULTS Studies were performed in freshly isolated PASMCs from both control and two different rat models of PH (chronically hypoxic and monocrotaline-treated rats). Co-immunolabellings revealed that the subcellular segregation between each subtype of SR ryanodine receptors (RyR1, RyR2, and RyR3), SERCA2 pumps (SERCA2a and SERCA2b), mitochondria, or lysosomes in freshly isolated PASMCs differs from control and PH PASMCs. In control PASMCs, stretching the membrane activates a Ca(2+) influx through SACs. This influx is amplified by cell hyperpolarization, a calcium release by subplasmalemmal RyR1 and is then buffered by mitochondria. In two different PH rat models, the calcium response to stretch is enhanced due to hyper-reactivity of SACs and a greater calcium amplification by all RyR subtypes. CONCLUSION The spatial organization of RyR and calcium stores in PASMCs is important for cell signalling and plays a causal role in PH.
Collapse
Affiliation(s)
- Guillaume Gilbert
- Centre de Recherche Cardio-Thoracique de Bordeaux, Universite de Bordeaux, Bordeaux F-33000, France Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, U 1045, Bordeaux F-33000, France
| | - Thomas Ducret
- Centre de Recherche Cardio-Thoracique de Bordeaux, Universite de Bordeaux, Bordeaux F-33000, France Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, U 1045, Bordeaux F-33000, France
| | - Roger Marthan
- Centre de Recherche Cardio-Thoracique de Bordeaux, Universite de Bordeaux, Bordeaux F-33000, France Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, U 1045, Bordeaux F-33000, France CHU Bordeaux, Exploration Fonctionnelle Respiratoire, Bordeaux F-33000, France
| | - Jean-Pierre Savineau
- Centre de Recherche Cardio-Thoracique de Bordeaux, Universite de Bordeaux, Bordeaux F-33000, France Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, U 1045, Bordeaux F-33000, France
| | - Jean-François Quignard
- Centre de Recherche Cardio-Thoracique de Bordeaux, Universite de Bordeaux, Bordeaux F-33000, France Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, U 1045, Bordeaux F-33000, France Centre de Recherche Cardio-Thoracique de Bordeaux, 146 rue Léo Saignat, Bat TP - 2ème étage, 33076 Bordeaux Cedex, France
| |
Collapse
|
23
|
Park KH, Weisleder N, Zhou J, Gumpper K, Zhou X, Duann P, Ma J, Lin PH. Assessment of calcium sparks in intact skeletal muscle fibers. J Vis Exp 2014:e50898. [PMID: 24638093 DOI: 10.3791/50898] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Maintaining homeostatic Ca(2+) signaling is a fundamental physiological process in living cells. Ca(2+) sparks are the elementary units of Ca(2+) signaling in the striated muscle fibers that appear as highly localized Ca(2+) release events mediated by ryanodine receptor (RyR) Ca(2+) release channels on the sarcoplasmic reticulum (SR) membrane. Proper assessment of muscle Ca(2+) sparks could provide information on the intracellular Ca(2+) handling properties of healthy and diseased striated muscles. Although Ca(2+) sparks events are commonly seen in resting cardiomyocytes, they are rarely observed in resting skeletal muscle fibers; thus there is a need for methods to generate and analyze sparks in skeletal muscle fibers. Detailed here is an experimental protocol for measuring Ca(2+) sparks in isolated flexor digitorm brevis (FDB) muscle fibers using fluorescent Ca(2+) indictors and laser scanning confocal microscopy. In this approach, isolated FDB fibers are exposed to transient hypoosmotic stress followed by a return to isotonic physiological solution. Under these conditions, a robust Ca(2+) sparks response is detected adjacent to the sarcolemmal membrane in young healthy FDB muscle fibers. Altered Ca(2+) sparks response is detected in dystrophic or aged skeletal muscle fibers. This approach has recently demonstrated that membrane-delimited signaling involving cross-talk between inositol (1,4,5)-triphosphate receptor (IP3R) and RyR contributes to Ca(2+) spark activation in skeletal muscle. In summary, our studies using osmotic stress induced Ca(2+) sparks showed that this intracellular response reflects a muscle signaling mechanism in physiology and aging/disease states, including mouse models of muscle dystrophy (mdx mice) or amyotrophic lateral sclerosis (ALS model).
Collapse
Affiliation(s)
- Ki Ho Park
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center
| | - Noah Weisleder
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center
| | - Jingsong Zhou
- Department of Molecular Biophysics and Physiology, Rush University Medical Center
| | - Kristyn Gumpper
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center
| | - Xinyu Zhou
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center
| | - Pu Duann
- Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center
| | - Jianjie Ma
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center
| | - Pei-Hui Lin
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center;
| |
Collapse
|
24
|
Subedi KP, Paudel O, Sham JSK. Detection of differentially regulated subsarcolemmal calcium signals activated by vasoactive agonists in rat pulmonary artery smooth muscle cells. Am J Physiol Cell Physiol 2013; 306:C659-69. [PMID: 24352334 DOI: 10.1152/ajpcell.00341.2013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intracellular calcium (Ca(2+)) plays pivotal roles in distinct cellular functions through global and local signaling in various subcellular compartments, and subcellular Ca(2+) signal is the key factor for independent regulation of different cellular functions. In vascular smooth muscle cells, subsarcolemmal Ca(2+) is an important regulator of excitation-contraction coupling, and nucleoplasmic Ca(2+) is crucial for excitation-transcription coupling. However, information on Ca(2+) signals in these subcellular compartments is limited. To study the regulation of the subcellular Ca(2+) signals, genetically encoded Ca(2+) indicators (cameleon), D3cpv, targeting the plasma membrane (PM), cytoplasm, and nucleoplasm were transfected into rat pulmonary arterial smooth muscle cells (PASMCs) and Ca(2+) signals were monitored using laser scanning confocal microscopy. In situ calibration showed that the Kd for Ca(2+) of D3cpv was comparable in the cytoplasm and nucleoplasm, but it was slightly higher in the PM. Stimulation of digitonin-permeabilized cells with 1,4,5-trisphosphate (IP3) elicited a transient elevation of Ca(2+) concentration with similar amplitude and kinetics in the nucleoplasm and cytoplasm. Activation of G protein-coupled receptors by endothelin-1 and angiotensin II preferentially elevated the subsarcolemmal Ca(2+) signal with higher amplitude in the PM region than the nucleoplasm and cytoplasm. In contrast, the receptor tyrosine kinase activator, platelet-derived growth factor, elicited Ca(2+) signals with similar amplitudes in all three regions, except that the rise-time and decay-time were slightly slower in the PM region. These data clearly revealed compartmentalization of Ca(2+) signals in the subsarcolemmal regions and provide the basis for further investigations of differential regulation of subcellular Ca(2+) signals in PASMCs.
Collapse
Affiliation(s)
- Krishna P Subedi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | |
Collapse
|