1
|
Timour G, Fréderic V, Olivier S, Shango DN. Nicardipine-induced acute respiratory failure: Case report and literature review. Clin Case Rep 2023; 11:e7186. [PMID: 37143457 PMCID: PMC10151601 DOI: 10.1002/ccr3.7186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/07/2022] [Accepted: 03/22/2023] [Indexed: 05/06/2023] Open
Abstract
Hypoxic pulmonary vasoconstriction (HPV) is a major physiological mechanism that prevents the development of hypoxemia secondary to a regional decrease in the ventilation-perfusion ratio (the intrapulmonary shunt effect). Calcium plays a critical role in the cellular response to hypoxia and the regulation of the pulmonary vascular tone. Therefore, calcium channel antagonists such as nicardipine have the potential to interfere with the pulmonary response to hypoxia, increasing intrapulmonary blood shunt and thus worsening underlying hypoxemia. This article reports the case of a 40-year-old man suffering from lobar pneumonia, who developed a rapidly progressing hypoxemia after starting nicardipine infusion for blood pressure control. After ruling out all major causes of hypoxemic respiratory failure, the involvement of the calcium channel antagonist was strongly suspected. Hypoxemia caused by HPV release is an underreported side effect of calcium channel blockers. There are few clinical reports that describe the occurrence of this adverse event, and to our knowledge, only one other publication describes a patient suffering from infectious pneumopathy. In this article, we discuss the cellular mechanisms behind the HPV, as well as the pharmacology of calcium channel antagonists and their involvement in the development of acute respiratory failure. The purpose of this report is to remind clinicians dealing with patients affected by acute hypoxemia that pharmacologic HPV inhibition should be considered as part of the differential diagnosis, thus avoiding unnecessary costly and time-consuming assessments.
Collapse
Affiliation(s)
- Gizzatullin Timour
- Department of Intensive Care CentreHospitalier de Wallonie Picarde (CHwapi)TournaiBelgium
| | - Vallot Fréderic
- Department of Intensive Care CentreHospitalier de Wallonie Picarde (CHwapi)TournaiBelgium
| | - Simonet Olivier
- Department of Intensive Care CentreHospitalier de Wallonie Picarde (CHwapi)TournaiBelgium
| | | |
Collapse
|
2
|
Lade JM, Andrade MR, Undem C, Walker J, Jiang H, Yun X, Shimoda LA. Hypoxia enhances interactions between Na +/H + exchanger isoform 1 and actin filaments via ezrin in pulmonary vascular smooth muscle. Front Physiol 2023; 14:1108304. [PMID: 36926194 PMCID: PMC10011449 DOI: 10.3389/fphys.2023.1108304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/24/2023] [Indexed: 03/08/2023] Open
Abstract
Exposure to hypoxia, due to high altitude or chronic lung disease, leads to structural changes in the pulmonary vascular wall, including hyperplasia and migration of pulmonary arterial smooth muscle cells (PASMCs). Previous studies showed that hypoxia upregulates the expression of Na+/H+ exchanger isoform 1 (NHE1) and that inhibition or loss of NHE1 prevents hypoxia-induced PASMC migration and proliferation. The exact mechanism by which NHE1 controls PASMC function has not been fully delineated. In fibroblasts, NHE1 has been shown to act as a membrane anchor for actin filaments, via binding of the adaptor protein, ezrin. Thus, in this study, we tested the role of ezrin and NHE1/actin interactions in controlling PASMC function. Using rat PASMCs exposed to in vitro hypoxia (4% O2, 24 h) we found that hypoxic exposure increased phosphorylation (activation) of ezrin, and promoted interactions between NHE1, phosphorylated ezrin and smooth muscle specific α-actin (SMA) as measured via immunoprecipitation and co-localization. Overexpression of wild-type human NHE1 in the absence of hypoxia was sufficient to induce PASMC migration and proliferation, whereas inhibiting ezrin phosphorylation with NSC668394 suppressed NHE1/SMA co-localization and migration in hypoxic PASMCs. Finally, overexpressing a version of human NHE1 in which amino acids were mutated to prevent NHE1/ezrin/SMA interactions was unable to increase PASMC migration and proliferation despite exhibiting normal Na+/H+ exchange activity. From these results, we conclude that hypoxic exposure increases ezrin phosphorylation in PASMCs, leading to enhanced ezrin/NHE1/SMA interaction. We further speculate that these interactions promote anchoring of the actin cytoskeleton to the membrane to facilitate the changes in cell movement and shape required for migration and proliferation.
Collapse
Affiliation(s)
- Julie M Lade
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Baltimore, MD, United States
| | - Manuella R Andrade
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Clark Undem
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Baltimore, MD, United States
| | - Jasmine Walker
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Baltimore, MD, United States
| | - Haiyang Jiang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Baltimore, MD, United States
| | - Xin Yun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Baltimore, MD, United States
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Baltimore, MD, United States
| |
Collapse
|
3
|
Mechanism of Hypoxia-Mediated Smooth Muscle Cell Proliferation Leading to Vascular Remodeling. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3959845. [PMID: 36593773 PMCID: PMC9805398 DOI: 10.1155/2022/3959845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/25/2022] [Accepted: 12/07/2022] [Indexed: 12/25/2022]
Abstract
Vascular remodeling refers to changes in the size, contraction, distribution, and flow rate of blood vessels and even changes in vascular function. Vascular remodeling can cause cardiovascular and cerebrovascular diseases. It can also lead to other systemic diseases, such as pulmonary hypertension, pulmonary atherosclerosis, chronic obstructive pulmonary disease, stroke, and ascites of broilers. Hypoxia is one of the main causes of vascular remodeling. Prolonged hypoxia or intermittent hypoxia can lead to loss of lung ventilation, causing respiratory depression, irregular respiratory rhythms, and central respiratory failure. Animals that are unable to adapt to the highland environment are also prone to sustained constriction of the small pulmonary arteries, increased resistance to pulmonary circulation, and impaired blood circulation, leading to pulmonary hypertension and right heart failure if they live in a highland environment for long periods of time. However, limited studies have been found on the relationship between hypoxia and vascular remodeling. Therefore, this review will explore the relationship between hypoxia and vascular remodeling from the aspects of endoplasmic reticulum stress, mitochondrial dysfunction, abnormal calcium channel, disordered cellular metabolism, abnormal expression of miRNA, and other factors. This will help to understand the detailed mechanism of hypoxia-mediated smooth muscle cell proliferation and vascular remodeling for the better treatment and management of diseases due to vascular remodeling.
Collapse
|
4
|
Lee MH, Sanders L, Kumar R, Hernandez-Saavedra D, Yun X, Ford JA, Perez MJ, Mickael C, Gandjeva A, Koyanagi DE, Harral JW, Irwin DC, Kassa B, Eckel RH, Shimoda LA, Graham BB, Tuder RM. Contribution of fatty acid oxidation to the pathogenesis of pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2022; 323:L355-L371. [PMID: 35763400 PMCID: PMC9448289 DOI: 10.1152/ajplung.00039.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/05/2022] [Accepted: 06/25/2022] [Indexed: 11/22/2022] Open
Abstract
Dysregulated metabolism characterizes both animal and human forms of pulmonary hypertension (PH). Enzymes involved in fatty acid metabolism have previously not been assessed in human pulmonary arteries affected by pulmonary arterial hypertension (PAH), and how inhibition of fatty acid oxidation (FAO) may attenuate PH remains unclear. Fatty acid metabolism gene transcription was quantified in laser-dissected pulmonary arteries from 10 explanted lungs with advanced PAH (5 idiopathic, 5 associated with systemic sclerosis), and 5 donors without lung diseases. Effects of oxfenicine, a FAO inhibitor, on female Sugen 5416-chronic hypoxia (SuHx) rats were studied in vivo using right heart catheterization, and ex vivo using perfused lungs and pulmonary artery ring segments. The impact of pharmacologic (oxfenicine) and genetic (carnitine palmitoyltransferase 1a heterozygosity) FAO suppression was additionally probed in mouse models of Schistosoma and hypoxia-induced PH. Potential mechanisms underlying FAO-induced PH pathogenesis were examined by quantifying ATP and mitochondrial mass in oxfenicine-treated SuHx pulmonary arterial cells, and by assessing pulmonary arterial macrophage infiltration with immunohistochemistry. We found upregulated pulmonary arterial transcription of 26 and 13 FAO genes in idiopathic and systemic sclerosis-associated PAH, respectively. In addition to promoting de-remodeling of pulmonary arteries in SuHx rats, oxfenicine attenuated endothelin-1-induced vasoconstriction. FAO inhibition also conferred modest benefit in the two mouse models of PH. Oxfenicine increased mitochondrial mass in cultured rat pulmonary arterial cells, and decreased the density of perivascular macrophage infiltration in pulmonary arteries of treated SuHx rats. In summary, FAO inhibition attenuated experimental PH, and may be beneficial in human PAH.
Collapse
Affiliation(s)
- Michael H Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Linda Sanders
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Rahul Kumar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Daniel Hernandez-Saavedra
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Xin Yun
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Joshay A Ford
- University of Colorado School of Medicine, Aurora, Colorado
| | - Mario J Perez
- Department of Psychiatry, University of Colorado, Aurora, Colorado
| | - Claudia Mickael
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Aneta Gandjeva
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Daniel E Koyanagi
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Julie W Harral
- Cardiovascular Pulmonary Research Laboratory, Department of Pediatrics and Medicine, University of Colorado, Aurora, Colorado
| | - David C Irwin
- Cardiovascular Pulmonary Research Laboratory, Department of Pediatrics and Medicine, University of Colorado, Aurora, Colorado
| | - Biruk Kassa
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Robert H Eckel
- Division of Endocrinology, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Brian B Graham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Rubin M Tuder
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| |
Collapse
|
5
|
Castillo-Galán S, Riquelme B, Iturriaga R. Crucial Role of Stromal Interaction Molecule-Activated TRPC-ORAI Channels in Vascular Remodeling and Pulmonary Hypertension Induced by Intermittent Hypoxia. Front Physiol 2022; 13:841828. [PMID: 35370769 PMCID: PMC8969100 DOI: 10.3389/fphys.2022.841828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Obstructive sleep apnea (OSA), a sleep breathing disorder featured by chronic intermittent hypoxia (CIH), is associate with pulmonary hypertension. Rats exposed to CIH develop lung vascular remodeling and pulmonary hypertension, which paralleled the upregulation of stromal interaction molecule (STIM)-activated TRPC-ORAI Ca2+ channels (STOC) in the lung, suggesting that STOC participate in the pulmonary vascular alterations. Accordingly, to evaluate the role played by STOC in pulmonary hypertension we studied whether the STOC blocker 2-aminoethoxydiphenyl borate (2-APB) may prevent the vascular remodeling and the pulmonary hypertension induced by CIH in a rat model of OSA. We assessed the effects of 2-APB on right ventricular systolic pressure (RVSP), pulmonary vascular remodeling, α-actin and proliferation marker Ki-67 levels in pulmonary arterial smooth muscle cells (PASMC), mRNA levels of STOC subunits, and systemic and pulmonary oxidative stress (TBARS) in male Sprague-Dawley (200 g) rats exposed to CIH (5% O2, 12 times/h for 8h) for 28 days. At 14 days of CIH, osmotic pumps containing 2-APB (10 mg/kg/day) or its vehicle were implanted and rats were kept for 2 more weeks in CIH. Exposure to CIH for 28 days raised RVSP > 35 mm Hg, increased the medial layer thickness and the levels of α-actin and Ki-67 in PASMC, and increased the gene expression of TRPC1, TRPC4, TRPC6 and ORAI1 subunits. Treatment with 2-APB prevented the raise in RVSP and the increment of the medial layer thickness, as well as the increased levels of α-actin and Ki-67 in PASMC, and the increased gene expression of STOC subunits. In addition, 2-APB did not reduced the lung and systemic oxidative stress, suggesting that the effects of 2-APB on vascular remodeling and pulmonary hypertension are independent on the reduction of the oxidative stress. Thus, our results supported that STIM-activated TRPC-ORAI Ca2+ channels contributes to the lung vascular remodeling and pulmonary hypertension induced by CIH.
Collapse
Affiliation(s)
- Sebastián Castillo-Galán
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bárbara Riquelme
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Iturriaga
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
- *Correspondence: Rodrigo Iturriaga,
| |
Collapse
|
6
|
Masson B, Montani D, Humbert M, Capuano V, Antigny F. Role of Store-Operated Ca 2+ Entry in the Pulmonary Vascular Remodeling Occurring in Pulmonary Arterial Hypertension. Biomolecules 2021; 11:1781. [PMID: 34944425 PMCID: PMC8698435 DOI: 10.3390/biom11121781] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 12/31/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe and multifactorial disease. PAH pathogenesis mostly involves pulmonary arterial endothelial and pulmonary arterial smooth muscle cell (PASMC) dysfunction, leading to alterations in pulmonary arterial tone and distal pulmonary vessel obstruction and remodeling. Unfortunately, current PAH therapies are not curative, and therapeutic approaches mostly target endothelial dysfunction, while PASMC dysfunction is under investigation. In PAH, modifications in intracellular Ca2+ homoeostasis could partly explain PASMC dysfunction. One of the most crucial actors regulating Ca2+ homeostasis is store-operated Ca2+ channels, which mediate store-operated Ca2+ entry (SOCE). This review focuses on the main actors of SOCE in human and experimental PASMC, their contribution to PAH pathogenesis, and their therapeutic potential in PAH.
Collapse
Affiliation(s)
- Bastien Masson
- Faculté de Médecine, School of Medicine, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France; (B.M.); (D.M.); (M.H.); (V.C.)
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Groupe Hospitalier Paris Saint-Joseph, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - David Montani
- Faculté de Médecine, School of Medicine, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France; (B.M.); (D.M.); (M.H.); (V.C.)
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Groupe Hospitalier Paris Saint-Joseph, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, 94276 Le Kremlin-Bicêtre, France
| | - Marc Humbert
- Faculté de Médecine, School of Medicine, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France; (B.M.); (D.M.); (M.H.); (V.C.)
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Groupe Hospitalier Paris Saint-Joseph, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, 94276 Le Kremlin-Bicêtre, France
| | - Véronique Capuano
- Faculté de Médecine, School of Medicine, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France; (B.M.); (D.M.); (M.H.); (V.C.)
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Groupe Hospitalier Paris Saint-Joseph, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Research and Innovation Unit, Groupe Hospitalier Paris Saint-Joseph, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Fabrice Antigny
- Faculté de Médecine, School of Medicine, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France; (B.M.); (D.M.); (M.H.); (V.C.)
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Groupe Hospitalier Paris Saint-Joseph, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| |
Collapse
|
7
|
Shimoda LA, Suresh K, Undem C, Jiang H, Yun X, Sylvester JT, Swenson ER. Acetazolamide prevents hypoxia-induced reactive oxygen species generation and calcium release in pulmonary arterial smooth muscle. Pulm Circ 2021; 11:20458940211049948. [PMID: 34646499 PMCID: PMC8504243 DOI: 10.1177/20458940211049948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Abstract
Upon sensing a reduction in local oxygen partial pressure, pulmonary vessels constrict, a phenomenon known as hypoxic pulmonary vasoconstriction. Excessive hypoxic pulmonary vasoconstriction can occur with ascent to high altitude and is a contributing factor to the development of high-altitude pulmonary edema. The carbonic anhydrase inhibitor, acetazolamide, attenuates hypoxic pulmonary vasoconstriction through stimulation of alveolar ventilation via modulation of acid-base homeostasis and by direct effects on pulmonary vascular smooth muscle. In pulmonary arterial smooth muscle cells (PASMCs), acetazolamide prevents hypoxia-induced increases in intracellular calcium concentration ([Ca2+]i), although the exact mechanism by which this occurs is unknown. In this study, we explored the effect of acetazolamide on various calcium-handling pathways in PASMCs. Using fluorescent microscopy, we tested whether acetazolamide directly inhibited store-operated calcium entry or calcium release from the sarcoplasmic reticulum, two well-documented sources of hypoxia-induced increases in [Ca2+]i in PASMCs. Acetazolamide had no effect on calcium entry stimulated by store-depletion, nor on calcium release from the sarcoplasmic reticulum induced by either phenylephrine to activate inositol triphosphate receptors or caffeine to activate ryanodine receptors. In contrast, acetazolamide completely prevented Ca2+-release from the sarcoplasmic reticulum induced by hypoxia (4% O2). Since these results suggest the acetazolamide interferes with a mechanism upstream of the inositol triphosphate and ryanodine receptors, we also determined whether acetazolamide might prevent hypoxia-induced changes in reactive oxygen species production. Using roGFP, a ratiometric reactive oxygen species-sensitive fluorescent probe, we found that hypoxia caused a significant increase in reactive oxygen species in PASMCs that was prevented by 100 μM acetazolamide. Together, these results suggest that acetazolamide prevents hypoxia-induced changes in [Ca2+]i by attenuating reactive oxygen species production and subsequent activation of Ca2+-release from sarcoplasmic reticulum stores.
Collapse
Affiliation(s)
- Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Clark Undem
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Haiyang Jiang
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Xin Yun
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - J T Sylvester
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Erik R Swenson
- Division of Pulmonary and Critical Care Medicine, VA Puget Sound Health Care System and University of Washington School of Medicine, St. Louis, MO, USA
| |
Collapse
|
8
|
Potential role of diacylglycerol kinases in immune-mediated diseases. Clin Sci (Lond) 2021; 134:1637-1658. [PMID: 32608491 DOI: 10.1042/cs20200389] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/08/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023]
Abstract
The mechanism promoting exacerbated immune responses in allergy and autoimmunity as well as those blunting the immune control of cancer cells are of primary interest in medicine. Diacylglycerol kinases (DGKs) are key modulators of signal transduction, which blunt diacylglycerol (DAG) signals and produce phosphatidic acid (PA). By modulating lipid second messengers, DGK modulate the activity of downstream signaling proteins, vesicle trafficking and membrane shape. The biological role of the DGK α and ζ isoforms in immune cells differentiation and effector function was subjected to in deep investigations. DGK α and ζ resulted in negatively regulating synergistic way basal and receptor induced DAG signals in T cells as well as leukocytes. In this way, they contributed to keep under control the immune response but also downmodulate immune response against tumors. Alteration in DGKα activity is also implicated in the pathogenesis of genetic perturbations of the immune function such as the X-linked lymphoproliferative disease 1 and localized juvenile periodontitis. These findings suggested a participation of DGK to the pathogenetic mechanisms underlying several immune-mediated diseases and prompted several researches aiming to target DGK with pharmacologic and molecular strategies. Those findings are discussed inhere together with experimental applications in tumors as well as in other immune-mediated diseases such as asthma.
Collapse
|
9
|
Liu G, Fu D, Tian H, Dai A. The mechanism of ions in pulmonary hypertension. Pulm Circ 2021; 11:2045894020987948. [PMID: 33614016 PMCID: PMC7869166 DOI: 10.1177/2045894020987948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
Pulmonary hypertension(PH)is a kind of hemodynamic and pathophysiological state, in which the pulmonary artery pressure (PAP) rises above a certain threshold. The main pathological manifestation is pulmonary vasoconstriction and remodelling progressively. More and more studies have found that ions play a major role in the pathogenesis of PH. Many vasoactive substances, inflammatory mediators, transcription-inducing factors, apoptosis mediators, redox substances and translation modifiers can control the concentration of ions inside and outside the cell by regulating the activity of ion channels, which can regulate vascular contraction, cell proliferation, migration, apoptosis, inflammation and other functions. We all know that there are no effective drugs to treat PH. Ions are involved in the occurrence and development of PH, so it is necessary to clarify the mechanism of ions in PH as a therapeutic target for PH. The main ions involved in PH are calcium ion (Ca2+), potassium ion (K+), sodium ion (Na+) and chloride ion (Cl-). Here, we mainly discuss the distribution of these ions and their channels in pulmonary arteries and their role in the pathogenesis of PH.
Collapse
Affiliation(s)
- Guogu Liu
- Department of Graduate School, University of South China,
Hengyang, China
- Department of Respiratory Medicine, Hunan Provincial People’s
Hospital, Changsha, China
| | - Daiyan Fu
- Department of Respiratory Medicine, Hunan Provincial People’s
Hospital, Changsha, China
| | - Heshen Tian
- Department of Graduate School, University of South China,
Hengyang, China
- Department of Respiratory Medicine, Hunan Provincial People’s
Hospital, Changsha, China
| | - Aiguo Dai
- Department of Respiratory Diseases, Hunan University of Chinese
Medicine, Changsha, China
| |
Collapse
|
10
|
Castillo-Galán S, Arenas GA, Reyes RV, Krause BJ, Iturriaga R. Stim-activated TRPC-ORAI channels in pulmonary hypertension induced by chronic intermittent hypoxia. Pulm Circ 2020; 10:13-22. [PMID: 33110495 PMCID: PMC7557718 DOI: 10.1177/2045894020941484] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/18/2020] [Indexed: 12/30/2022] Open
Abstract
Obstructive sleep apnea (OSA), a breathing disorder featured by chronic intermittent
hypoxia (CIH) is associated with pulmonary hypertension (PH). Rodents exposed to CIH
develop pulmonary vascular remodeling and PH, but the pathogenic mechanisms are not well
known. Overexpression of Stim-activated Transient Receptor Potential Channels (TRPC) and
Calcium Release-Activated Calcium Channel Protein (ORAI) TRPC-ORAI Ca2+
channels (STOC) has been involved in pulmonary vascular remodeling and PH in sustained
hypoxia. However, it is not known if CIH may change STOC levels. Accordingly, we studied
the effects of CIH on the expression of STOC subunits in the lung and if these changes
paralleled the progression of the vascular pulmonary remodeling and PH in a preclinical
model of OSA. Male Sprague-Dawley rats (∼200 g) were exposed to CIH (5%O2, 12
times/h for 8 h) for 14, 21, and 28 days. We measured right ventricular systolic pressure
(RVSP), cardiac morphometry with MRI, pulmonary vascular remodeling, and wire-myographic
arterial responses to KCl and endothelin-1 (ET-1). Pulmonary RNA and protein STOC levels
of TRPC1, TRPC4, TRPC6, ORAI 1, ORAI 2, and STIM1 subunits were measured by qPCR and
western blot, and results were compared with age-matched controls. CIH elicited a
progressive increase of RVSP and vascular contractile responses to KCl and ET-1, leading
to vascular remodeling and augmented right ventricular ejection fraction, which was
significant at 28 days of CIH. The levels of TRPC1, TRPC4, TRPC 6, ORAI 1, and STIM 1
channels increased following CIH, and some of them paralleled morphologic and functional
changes. Our findings show that CIH increased pulmonary STOC expression, paralleling
vascular remodeling and PH.
Collapse
Affiliation(s)
- Sebastian Castillo-Galán
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - German A Arenas
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roberto V Reyes
- Laboratorio de Bioquímica y Biología Molecular de la Hipoxia, Universidad de Chile, Santiago, Chile
| | - Bernardo J Krause
- Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua, Chile
| | - Rodrigo Iturriaga
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
11
|
Jain PP, Hosokawa S, Xiong M, Babicheva A, Zhao T, Rodriguez M, Rahimi S, Pourhashemi K, Balistrieri F, Lai N, Malhotra A, Shyy JYJ, Valdez-Jasso D, Thistlethwaite PA, Makino A, Yuan JXJ. Revisiting the mechanism of hypoxic pulmonary vasoconstriction using isolated perfused/ventilated mouse lung. Pulm Circ 2020; 10:2045894020956592. [PMID: 33282184 PMCID: PMC7691930 DOI: 10.1177/2045894020956592] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/16/2020] [Indexed: 12/13/2022] Open
Abstract
Hypoxic Pulmonary Vasoconstriction (HPV) is an important physiological mechanism of the lungs that matches perfusion to ventilation thus maximizing O2 saturation of the venous blood within the lungs. This study emphasizes on principal pathways in the initiation and modulation of hypoxic pulmonary vasoconstriction with a primary focus on the role of Ca2+ signaling and Ca2+ influx pathways in hypoxic pulmonary vasoconstriction. We used an ex vivo model, isolated perfused/ventilated mouse lung to evaluate hypoxic pulmonary vasoconstriction. Alveolar hypoxia (utilizing a mini ventilator) rapidly and reversibly increased pulmonary arterial pressure due to hypoxic pulmonary vasoconstriction in the isolated perfused/ventilated lung. By applying specific inhibitors for different membrane receptors and ion channels through intrapulmonary perfusion solution in isolated lung, we were able to define the targeted receptors and channels that regulate hypoxic pulmonary vasoconstriction. We show that extracellular Ca2+ or Ca2+ influx through various Ca2+-permeable channels in the plasma membrane is required for hypoxic pulmonary vasoconstriction. Removal of extracellular Ca2+ abolished hypoxic pulmonary vasoconstriction, while blockade of L-type voltage-dependent Ca2+ channels (with nifedipine), non-selective cation channels (with 30 µM SKF-96365), and TRPC6/TRPV1 channels (with 1 µM SAR-7334 and 30 µM capsazepine, respectively) significantly and reversibly inhibited hypoxic pulmonary vasoconstriction. Furthermore, blockers of Ca2+-sensing receptors (by 30 µM NPS2143, an allosteric Ca2+-sensing receptors inhibitor) and Notch (by 30 µM DAPT, a γ-secretase inhibitor) also attenuated hypoxic pulmonary vasoconstriction. These data indicate that Ca2+ influx in pulmonary arterial smooth muscle cells through voltage-dependent, receptor-operated, and store-operated Ca2+ entry pathways all contribute to initiation of hypoxic pulmonary vasoconstriction. The extracellular Ca2+-mediated activation of Ca2+-sensing receptors and the cell-cell interaction via Notch ligands and receptors contribute to the regulation of hypoxic pulmonary vasoconstriction.
Collapse
Affiliation(s)
- Pritesh P. Jain
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Susumu Hosokawa
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
- Department of Pediatrics, Tokyo Medical
and Dental University, Tokyo, Japan
| | - Mingmei Xiong
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
- Department of Critical Medicine, The
Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aleksandra Babicheva
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Tengteng Zhao
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Marisela Rodriguez
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Shamin Rahimi
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Kiana Pourhashemi
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Francesca Balistrieri
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Ning Lai
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Atul Malhotra
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - John Y.-J. Shyy
- Division of Cardiovascular Medicine,
Department of Medicine, University of California, San Diego, USA
| | | | | | - Ayako Makino
- Division of Endocrinology and
Metabolism, University of California, San Diego, CA, USA
| | - Jason X.-J. Yuan
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| |
Collapse
|
12
|
Kaymak E, Akin AT, Tufan E, Başaran KE, Taheri S, Özdamar S, Yakan B. The effect of chloroquine on the TRPC1, TRPC6, and CaSR in the pulmonary artery smooth muscle cells in hypoxia-induced experimental pulmonary artery hypertension. J Biochem Mol Toxicol 2020; 35:e22636. [PMID: 32956540 DOI: 10.1002/jbt.22636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/30/2020] [Accepted: 09/09/2020] [Indexed: 01/10/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by a constant high pulmonary artery pressure and the remodeling of the vessel. Chloroquine (CLQ) has been observed to inhibit calcium influx. The aim of this study is to investigate the effect of CLQ on transient receptor cationic proteins (TRPC1 and TRPC6) and extracellular calcium-sensitive receptor (CaSR) in a hypoxic PAH model. In this study, 8- to 12-week-old 32 male Wistar albino rats, weighing 200 to 300 g, were used. The rats were studied in four groups, including normoxy control, n = 8; normoxy CLQ (50 mg/kg/28 d), n = 8; hypoxia (HX; 10% oxygen/28 d) control, n = 8; and HX (10% oxygen/28 d) + CLQ (50 mg/kg), N = 8. Pulmonary arterial medial wall thickness, pulmonary arteriole wall, TRPC1, TRPC6, and CaSR expressions were evaluated by immunohistochemistry, polymerase chain reaction, and enzyme-linked immunosorbent assay methods. At the end of the experiment, a statistically significant increase in the medial wall thickness was observed in the hypoxic group as compared with the control group. However, in the HX + CLQ group, there was a statistically significant decrease in the vessel medial wall as compared with the HX group. In the TRPC1-, TRPC6-, and CaSR-immunopositive cell numbers, messenger RNA expressions and biochemical results showed an increase in the HX group, whereas they were decreased in the HX + CLQ group. The inhibitory effect of CLQ on calcium receptors in arterioles was observed in PAH.
Collapse
Affiliation(s)
- Emin Kaymak
- Department of Histology and Embryology, Yozgat Bozok University, Yozgat, Turkey
| | | | - Esra Tufan
- Department of Physiology, Erciyes University, Kayseri, Turkey
| | | | - Serpil Taheri
- Department of Medical Biology, Erciyes University, Kayseri, Turkey
| | - Saim Özdamar
- Department of Histology and Embryology, Pamukkale University, Denizli, Turkey
| | - Birkan Yakan
- Department of Histology and Embryology, Erciyes University, Kayseri, Turkey
| |
Collapse
|
13
|
Chen Y, Lu W, Yang K, Duan X, Li M, Chen X, Zhang J, Kuang M, Liu S, Wu X, Zou G, Liu C, Hong C, He W, Liao J, Hou C, Zhang Z, Zheng Q, Chen J, Zhang N, Tang H, Vanderpool RR, Desai AA, Rischard F, Black SM, Garcia JGN, Makino A, Yuan JXJ, Zhong N, Wang J. Tetramethylpyrazine: A promising drug for the treatment of pulmonary hypertension. Br J Pharmacol 2020; 177:2743-2764. [PMID: 31976548 DOI: 10.1111/bph.15000] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/28/2019] [Accepted: 01/07/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Tetramethylpyrazine (TMP) was originally isolated from the traditional Chinese herb ligusticum and the fermented Japanese food natto and has since been synthesized. TMP has a long history of beneficial effects in the treatment of many cardiovascular diseases. Here we have evaluated the therapeutic effects of TMP on pulmonary hypertension (PH) in animal models and in patients with pulmonary arterial hypertension (PAH) or chronic thromboembolic pulmonary hypertension (CTEPH). EXPERIMENTAL APPROACH Three well-defined models of PH -chronic hypoxia (10% O2 )-induced PH (HPH), monocrotaline-induced PH (MCT-PH) and Sugen 5416/hypoxia-induced PH (SuHx-PH) - were used in Sprague-Dawley rats, and assessed by echocardiography, along with haemodynamic and histological techniques. Primary cultures of rat distal pulmonary arterial smooth muscle cells (PASMCs) were used to study intracellular calcium levels. Western blots and RT-qPCR assays were also used. In the clinical cohort, patients with PAH or CTEPH were recruited. The effects of TMP were evaluated in all systems. KEY RESULTS TMP (100 mg·kg-1 ·day-1 ) prevented rats from developing experimental PH and ameliorated three models of established PH: HPH, MCT-PH and SuHx-PH. The therapeutic effects of TMP were accompanied by inhibition of intracellular calcium homeostasis in PASMCs. In a small cohort of patients with PAH or CTEPH, oral administration of TMP (100 mg, t.i.d. for 16 weeks) increased the 6-min walk distance and improved the 1-min heart rate recovery. CONCLUSION AND IMPLICATIONS Our results suggest that TMP is a novel and inexpensive medication for treatment of PH. Clinical trial is registered with www.chictr.org.cn (ChiCTR-IPR-14005379).
Collapse
Affiliation(s)
- Yuqin Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kai Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xin Duan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengxi Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiuqing Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jie Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Meidan Kuang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shiyun Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiongting Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guofa Zou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chunli Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cheng Hong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenjun He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Liao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chi Hou
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Zhe Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiuyu Zheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiyuan Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nuofu Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Rebecca R Vanderpool
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Ankit A Desai
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Franz Rischard
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Stephen M Black
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Joe G N Garcia
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Ayako Makino
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Jason X-J Yuan
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Division of Pulmonary and Critical Care Medicine, The People's Hospital of Inner Mongolia, Huhhot, China.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
14
|
Reyes RV, Castillo-Galán S, Hernandez I, Herrera EA, Ebensperger G, Llanos AJ. Revisiting the Role of TRP, Orai, and ASIC Channels in the Pulmonary Arterial Response to Hypoxia. Front Physiol 2018; 9:486. [PMID: 29867539 PMCID: PMC5949889 DOI: 10.3389/fphys.2018.00486] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/16/2018] [Indexed: 12/11/2022] Open
Abstract
The pulmonary arteries are exquisitely responsive to oxygen changes. They rapidly and proportionally contract as arterial PO2 decrease, and they relax as arterial PO2 is re-established. The hypoxic pulmonary vasoconstriction (HPV) is intrinsic since it does not require neural or endocrine factors, as evidenced in isolated vessels. On the other hand, pulmonary arteries also respond to sustained hypoxia with structural and functional remodeling, involving growth of smooth muscle medial layer and later recruitment of adventitial fibroblasts, secreted mitogens from endothelium and changes in the response to vasoconstrictor and vasodilator stimuli. Hypoxic pulmonary arterial vasoconstriction and remodeling are relevant biological responses both under physiological and pathological conditions, to explain matching between ventilation and perfusion, fetal to neonatal transition of pulmonary circulation and pulmonary artery over-constriction and thickening in pulmonary hypertension. Store operated channels (SOC) and receptor operated channels (ROC) are plasma membrane cationic channels that mediate calcium influx in response to depletion of internal calcium stores or receptor activation, respectively. They are involved in both HPV and pathological remodeling since their pharmacological blockade or genetic suppression of several of the Stim, Orai, TRP, or ASIC proteins in SOC or ROC complexes attenuate the calcium increase, the tension development, the pulmonary artery smooth muscle proliferation, and pulmonary arterial hypertension. In this Mini Review, we discussed the evidence obtained in in vivo animal models, at the level of isolated organ or cells of pulmonary arteries, and we identified and discussed the questions for future research needed to validate these signaling complexes as targets against pulmonary hypertension.
Collapse
Affiliation(s)
- Roberto V Reyes
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| | - Sebastián Castillo-Galán
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ismael Hernandez
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Emilio A Herrera
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| | - Germán Ebensperger
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| | - Aníbal J Llanos
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| |
Collapse
|
15
|
Abstract
Hypoxic pulmonary vasoconstriction (HPV) in combination with hypercapnic pulmonary vasoconstriction redistributes pulmonary blood flow from poorly aerated to better ventilated lung regions by an active process of local vasoconstriction. Impairment of HPV results in ventilation-perfusion mismatch and is commonly associated with various lung diseases including pneumonia, sepsis, or cystic fibrosis. Although several regulatory pathways have been identified, considerable knowledge gaps persist, and a unifying concept of the signaling pathways that underlie HPV and their impairment in lung diseases has not yet emerged. In the past, conceptual models of HPV have focused on pulmonary arterial smooth muscle cells (PASMC) acting as sensor and effector of hypoxia in the pulmonary vasculature. In contrast, the endothelium was considered a modulating bystander in this scenario. For an ideal design, however, the oxygen sensor in HPV should be located in the region of gas exchange, i.e., in the alveolar capillary network. This concept requires the retrograde propagation of the hypoxic signal along the endothelial layer of the vascular wall and subsequent contraction of PASMC in upstream arterioles that is elicited via temporospatially tightly controlled endothelial-smooth muscle cell crosstalk. The present review summarizes recent work that provides proof-of-principle for the existence and functional relevance of such signaling pathway in HPV that involves important roles for connexin 40, epoxyeicosatrienoic acids, sphingolipids, and cystic fibrosis transmembrane conductance regulator. Of translational relevance, implication of these molecules provides for novel mechanistic explanations for impaired ventilation/perfusion matching in patients with pneumonia, sepsis, cystic fibrosis, and presumably various other lung diseases.
Collapse
Affiliation(s)
- Benjamin Grimmer
- Institute of Physiology, Charité Universitätsmedizin Berlin, Berlin , Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité Universitätsmedizin Berlin, Berlin , Germany
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital , Toronto, Ontario , Canada
- Departments of Surgery and Physiology, University of Toronto , Toronto, Ontario , Canada
| |
Collapse
|
16
|
Abstract
This chapter offers a brief introduction of the functions of TRPC channels in non-neuronal systems. We focus on three major organs of which the research on TRPC channels have been most focused on: kidney, heart, and lung. The chapter highlights on cellular functions and signaling pathways mediated by TRPC channels. It also summarizes several inherited diseases in humans that are related to or caused by TRPC channel mutations and malfunction. A better understanding of TRPC channels functions and the importance of TRPC channels in health and disease should lead to new insights and discovery of new therapeutic approaches for intractable disease.
Collapse
|
17
|
Barreto Ortiz S, Hori D, Nomura Y, Yun X, Jiang H, Yong H, Chen J, Paek S, Pandey D, Sikka G, Bhatta A, Gillard A, Steppan J, Kim JH, Adachi H, Barodka VM, Romer L, An SS, Shimoda LA, Santhanam L, Berkowitz DE. Opsin 3 and 4 mediate light-induced pulmonary vasorelaxation that is potentiated by G protein-coupled receptor kinase 2 inhibition. Am J Physiol Lung Cell Mol Physiol 2017; 314:L93-L106. [PMID: 28882814 DOI: 10.1152/ajplung.00091.2017] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We recently demonstrated that blue light induces vasorelaxation in the systemic mouse circulation, a phenomenon mediated by the nonvisual G protein-coupled receptor melanopsin (Opsin 4; Opn4). Here we tested the hypothesis that nonvisual opsins mediate photorelaxation in the pulmonary circulation. We discovered Opsin 3 (Opn3), Opn4, and G protein-coupled receptor kinase 2 (GRK2) in rat pulmonary arteries (PAs) and in pulmonary arterial smooth muscle cells (PASMCs), where the opsins interact directly with GRK2, as demonstrated with a proximity ligation assay. Light elicited an intensity-dependent relaxation of PAs preconstricted with phenylephrine (PE), with a maximum response between 400 and 460 nm (blue light). Wavelength-specific photorelaxation was attenuated in PAs from Opn4-/- mice and further reduced following shRNA-mediated knockdown of Opn3. Inhibition of GRK2 amplified the response and prevented physiological desensitization to repeated light exposure. Blue light also prevented PE-induced constriction in isolated PAs, decreased basal tone, ablated PE-induced single-cell contraction of PASMCs, and reversed PE-induced depolarization in PASMCs when GRK2 was inhibited. The photorelaxation response was modulated by soluble guanylyl cyclase but not by protein kinase G or nitric oxide. Most importantly, blue light induced significant vasorelaxation of PAs from rats with chronic pulmonary hypertension and effectively lowered pulmonary arterial pressure in isolated intact perfused rat lungs subjected to acute hypoxia. These findings show that functional Opn3 and Opn4 in PAs represent an endogenous "optogenetic system" that mediates photorelaxation in the pulmonary vasculature. Phototherapy in conjunction with GRK2 inhibition could therefore provide an alternative treatment strategy for pulmonary vasoconstrictive disorders.
Collapse
Affiliation(s)
- Sebastian Barreto Ortiz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Daijiro Hori
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland.,Division of Cardiac Surgery, Johns Hopkins University , Baltimore, Maryland
| | - Yohei Nomura
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland.,Division of Cardiac Surgery, Johns Hopkins University , Baltimore, Maryland
| | - Xin Yun
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Asthma and Allergy Center, Johns Hopkins University , Baltimore, Maryland
| | - Haiyang Jiang
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Asthma and Allergy Center, Johns Hopkins University , Baltimore, Maryland
| | - Hwanmee Yong
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| | - James Chen
- Department of Biomedical Engineering, Johns Hopkins University , Baltimore, Maryland
| | - Sam Paek
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| | - Deepesh Pandey
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Gautam Sikka
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Anil Bhatta
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Andrew Gillard
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Jochen Steppan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Jae Hyung Kim
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Hideo Adachi
- Department of Cardiovascular Surgery, Saitama Medical Center, Jichi Medical University, Shimotsuke, Japan
| | - Viachaslau M Barodka
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Lewis Romer
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland.,Department of Biomedical Engineering, Johns Hopkins University , Baltimore, Maryland.,Departments of Cell Biology, Pediatrics, and the Center for Cell Dynamics, Johns Hopkins University , Baltimore, Maryland
| | - Steven S An
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Asthma and Allergy Center, Johns Hopkins University , Baltimore, Maryland
| | - Lakshmi Santhanam
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland.,Department of Biomedical Engineering, Johns Hopkins University , Baltimore, Maryland
| | - Dan E Berkowitz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland.,Department of Biomedical Engineering, Johns Hopkins University , Baltimore, Maryland
| |
Collapse
|
18
|
Strielkov I, Pak O, Sommer N, Weissmann N. Recent advances in oxygen sensing and signal transduction in hypoxic pulmonary vasoconstriction. J Appl Physiol (1985) 2017; 123:1647-1656. [PMID: 28751366 DOI: 10.1152/japplphysiol.00103.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypoxic pulmonary vasoconstriction (HPV) is a physiological reaction, which adapts lung perfusion to regional ventilation and optimizes gas exchange. Impaired HPV may cause systemic hypoxemia, while generalized HPV contributes to the development of pulmonary hypertension. The triggering mechanisms underlying HPV are still not fully elucidated. Several hypotheses are currently under debate, including a possible decrease as well as an increase in reactive oxygen species as a triggering event. Recent findings suggest an increase in the production of reactive oxygen species in pulmonary artery smooth muscle cells by complex III of the mitochondrial electron transport chain and occurrence of oxygen sensing at complex IV. Other essential components are voltage-dependent potassium and possibly L-type, transient receptor potential channel 6, and transient receptor potential vanilloid 4 channels. The release of arachidonic acid metabolites appears also to be involved in HPV regulation. Further investigation of the HPV mechanisms will facilitate the development of novel therapeutic strategies for the treatment of HPV-related disorders.
Collapse
Affiliation(s)
- Ievgen Strielkov
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen , Germany
| | - Oleg Pak
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen , Germany
| | - Natasha Sommer
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen , Germany
| | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen , Germany
| |
Collapse
|
19
|
Malczyk M, Erb A, Veith C, Ghofrani HA, Schermuly RT, Gudermann T, Dietrich A, Weissmann N, Sydykov A. The Role of Transient Receptor Potential Channel 6 Channels in the Pulmonary Vasculature. Front Immunol 2017; 8:707. [PMID: 28670316 PMCID: PMC5472666 DOI: 10.3389/fimmu.2017.00707] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/31/2017] [Indexed: 01/21/2023] Open
Abstract
Canonical or classical transient receptor potential channel 6 (TRPC6) is a Ca2+-permeable non-selective cation channel that is widely expressed in the heart, lung, and vascular tissues. The use of TRPC6-deficient (“knockout”) mice has provided important insights into the role of TRPC6 in normal physiology and disease states of the pulmonary vasculature. Evidence indicates that TRPC6 is a key regulator of acute hypoxic pulmonary vasoconstriction. Moreover, several studies implicated TRPC6 in the pathogenesis of pulmonary hypertension. Furthermore, a unique genetic variation in the TRPC6 gene promoter has been identified, which might link the inflammatory response to the upregulation of TRPC6 expression and ultimate development of pulmonary vascular abnormalities in idiopathic pulmonary arterial hypertension. Additionally, TRPC6 is critically involved in the regulation of pulmonary vascular permeability and lung edema formation during endotoxin or ischemia/reperfusion-induced acute lung injury. In this review, we will summarize latest findings on the role of TRPC6 in the pulmonary vasculature.
Collapse
Affiliation(s)
- Monika Malczyk
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Alexandra Erb
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Christine Veith
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Hossein Ardeschir Ghofrani
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Thomas Gudermann
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig Maximilian University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Alexander Dietrich
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig Maximilian University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Akylbek Sydykov
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| |
Collapse
|
20
|
Chen TX, Xu XY, Zhao Z, Zhao FY, Gao YM, Yan XH, Wan Y. Hydrogen peroxide is a critical regulator of the hypoxia-induced alterations of store-operated Ca2+ entry into rat pulmonary arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2017; 312:L477-L487. [DOI: 10.1152/ajplung.00138.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 01/23/2017] [Accepted: 01/23/2017] [Indexed: 12/28/2022] Open
Abstract
To investigate the association between store-operated Ca2+ entry (SOCE) and reactive oxygen species (ROS) during hypoxia, this study determined the changes of transient receptor potential canonical 1 (TRPC1) and Orai1, two candidate proteins for store-operated Ca2+ (SOC) channels and their gate regulator, stromal interaction molecule 1 (STIM1), in a hypoxic environment and their relationship with ROS in pulmonary arterial smooth muscle cells (PASMCs). Exposure to hypoxia caused a transient Ca2+ spike and subsequent Ca2+ plateau of SOCE to be intensified in PASMCs when TRPC1, STIM1, and Orai1 were upregulated. SOCE in cells transfected with specific short hairpin RNA (shRNA) constructs was almost completely eliminated by the knockdown of TRPC1, STIM1, or Orai1 alone and was no longer affected by hypoxia exposure. Hypoxia-induced SOCE enhancement was further strengthened by PEG-SOD but was attenuated by PEG-catalase, with correlated changes to intracellular hydrogen peroxide (H2O2) levels and protein levels of TRPC1, STIM1, and Orai1. Exogenous H2O2 could mimic alterations of the interactions of STIM1 with TRPC1 and Orai1 in hypoxic cells. These findings suggest that TRPC1, STIM1, and Orai1 are essential for the initiation of SOCE in PASMCs. Hypoxia-induced ROS promoted the expression and interaction of the SOC channel molecules and their gate regulator via their converted product, H2O2.
Collapse
Affiliation(s)
- Tao-Xiang Chen
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xiao-Ya Xu
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Zhao Zhao
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Fang-Yu Zhao
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yi-Mei Gao
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xiao-Hong Yan
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yu Wan
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
21
|
Brinks L, Moonen RMJ, Moral-Sanz J, Barreira B, Kessels L, Perez-Vizcaino F, Cogolludo A, Villamor E. Hypoxia-induced contraction of chicken embryo mesenteric arteries: mechanisms and developmental changes. Am J Physiol Regul Integr Comp Physiol 2016; 311:R858-R869. [DOI: 10.1152/ajpregu.00461.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 08/10/2016] [Indexed: 11/22/2022]
Abstract
The fetal cardiovascular responses to acute hypoxia include a redistribution of the cardiac output toward the heart and the brain at the expense of other organs, such as the intestine. We hypothesized that hypoxia exerts a direct effect on the mesenteric artery (MA) that may contribute to this response. Using wire myography, we investigated the response to hypoxia (Po2 ~2.5 kPa for 20 min) of isolated MAs from 15- to 21-day chicken embryos (E15, E19, E21), and 1- to 45-day-old chickens (P1, P3, P14, P45). Agonist-induced pretone or an intact endothelium were not required to obtain a consistent and reproducible response to hypoxia, which showed a pattern of initial rapid phasic contraction followed by a sustained tonic contraction. Phasic contraction was reduced by elimination of extracellular Ca2+ or by presence of the neurotoxin tetrodotoxin, the α1-adrenoceptor antagonist prazosin, or inhibitors of L-type voltage-gated Ca2+ channels (nifedipine), mitochondrial electron transport chain (rotenone and antimycin A), and NADPH oxidase (VAS2870). The Rho-kinase inhibitor Y27632 impaired both phasic and tonic contraction and, when combined with elimination of extracellular Ca2+, hypoxia-induced contraction was virtually abolished. Hypoxic MA contraction was absent at E15 but present from E19 and increased toward the first days posthatching. It then decreased during the first weeks of life and P45 MAs were unable to sustain hypoxia-induced contraction over time. In conclusion, the results of the present study demonstrate that hypoxic vasoconstriction is an intrinsic feature of chicken MA vascular smooth muscle cells during late embryogenesis and the perinatal period.
Collapse
Affiliation(s)
- Leonie Brinks
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| | - Rob M. J. Moonen
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
- Department of Pediatrics, Zuyderland Medical Center, Heerlen, The Netherlands; and
| | - Javier Moral-Sanz
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Bianca Barreira
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Lilian Kessels
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| | - Francisco Perez-Vizcaino
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| |
Collapse
|
22
|
Castillo-Galán S, Quezada S, Moraga FA, Ebensperger G, Herrera EA, Beñaldo F, Hernandez I, Ebensperger R, Ramirez S, Llanos AJ, Reyes RV. 2-AMINOETHYLDIPHENYLBORINATE MODIFIES THE PULMONARY CIRCULATION IN PULMONARY HYPERTENSIVE NEWBORN LAMBS WITH PARTIAL GESTATION AT HIGH ALTITUDE. Am J Physiol Lung Cell Mol Physiol 2016; 311:L788-L799. [PMID: 27542806 DOI: 10.1152/ajplung.00230.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/14/2016] [Indexed: 12/22/2022] Open
Abstract
Calcium signaling through store operated channels (SOC) is involved in hypoxic pulmonary hypertension. We determined whether a treatment with 2-aminoethyldiphenylborinate (2-APB), a compound with SOC blocker activity, reduces pulmonary hypertension and vascular remodeling. Twelve newborn lambs exposed to perinatal chronic hypoxia were studied, 6 of them received a 2-APB treatment and the other 6 received vehicle treatment, for 10 days in both cases. Throughout this period, we recorded cardiopulmonary variables and on day 11 we evaluated the response to an acute hypoxic challenge. Additionally, we assessed the vasoconstrictor and vasodilator function in isolated pulmonary arteries as well as their remodeling in lung slices. 2-APB reduced pulmonary arterial pressure at the third and tenth days, cardiac output between the fourth and eighth days, and pulmonary vascular resistance at the tenth day of treatment. The pulmonary vasoconstrictor response to acute hypoxia was reduced by the end of treatment. 2-APB also decreased maximal vasoconstrictor response to the thromboxane mimetic U46619 and endothelin-1 and increased maximal relaxation to 8-Br-cGMP. The maximal relaxation and potency to phosphodiesterase-5 and Rho-kinase inhibition with sildenafil and fasudil respectively, were also increased. Finally, 2-APB reduced the medial and adventitial layers' thickness, the expression of α-actin and the percentage of Ki67+ nuclei of small pulmonary arteries. Taken together, our results indicate that 2-APB reduces pulmonary hypertension, vasoconstrictor responses and pathological remodeling in pulmonary hypertensive lambs. We conclude that SOC targeting may be a useful strategy for the treatment of neonatal pulmonary hypertension, however, further testing of specific blockers is needed.
Collapse
Affiliation(s)
| | - Sebastián Quezada
- Universidad de Chile, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM)
| | | | - Germán Ebensperger
- Facultad de Medicina, Universidad de Chile, Instituto de Ciencias Biomédicas (ICBM), Santiago, Chile
| | | | | | - Ismael Hernandez
- Facultad de Medicina, Universidad de Chile, Instituto de Ciencias Biomédicas (ICBM), Santiago, Chile
| | - Renato Ebensperger
- Facultad de Medicina, Universidad de Chile, Instituto de Ciencias Biomédicas (ICBM), Santiago, Chile
| | - Santiago Ramirez
- Facultad de Medicina, Universidad de Chile, Instituto de Ciencias Biomédicas (ICBM), Santiago, Chile
| | | | - Roberto V Reyes
- Universidad de Chile, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM)
| |
Collapse
|
23
|
From contraction to gene expression: nanojunctions of the sarco/endoplasmic reticulum deliver site- and function-specific calcium signals. SCIENCE CHINA-LIFE SCIENCES 2016; 59:749-63. [PMID: 27376531 DOI: 10.1007/s11427-016-5071-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 04/07/2016] [Indexed: 10/21/2022]
Abstract
Calcium signals determine, for example, smooth muscle contraction and changes in gene expression. How calcium signals select for these processes is enigmatic. We build on the "panjunctional sarcoplasmic reticulum" hypothesis, describing our view that different calcium pumps and release channels, with different kinetics and affinities for calcium, are strategically positioned within nanojunctions of the SR and help demarcate their respective cytoplasmic nanodomains. SERCA2b and RyR1 are preferentially targeted to the sarcoplasmic reticulum (SR) proximal to the plasma membrane (PM), i.e., to the superficial buffer barrier formed by PM-SR nanojunctions, and support vasodilation. In marked contrast, SERCA2a may be entirely restricted to the deep, perinuclear SR and may supply calcium to this sub-compartment in support of vasoconstriction. RyR3 is also preferentially targeted to the perinuclear SR, where its clusters associate with lysosome-SR nanojunctions. The distribution of RyR2 is more widespread and extends from this region to the wider cell. Therefore, perinuclear RyR3s most likely support the initiation of global calcium waves at L-SR junctions, which subsequently propagate by calcium-induced calcium release via RyR2 in order to elicit contraction. Data also suggest that unique SERCA and RyR are preferentially targeted to invaginations of the nuclear membrane. Site- and function-specific calcium signals may thus arise to modulate stimulus-response coupling and transcriptional cascades.
Collapse
|
24
|
Jiang Q, Lu W, Yang K, Hadadi C, Fu X, Chen Y, Yun X, Zhang J, Li M, Xu L, Tang H, Yuan JXJ, Wang J, Sun D. Sodium tanshinone IIA sulfonate inhibits hypoxia-induced enhancement of SOCE in pulmonary arterial smooth muscle cells via the PKG-PPAR-γ signaling axis. Am J Physiol Cell Physiol 2016; 311:C136-49. [PMID: 27194472 PMCID: PMC4967135 DOI: 10.1152/ajpcell.00252.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 05/02/2016] [Indexed: 11/22/2022]
Abstract
Our laboratory previously showed that sodium tanshinone IIA sulfonate (STS) inhibited store-operated Ca(2+) entry (SOCE) through store-operated Ca(2+) channels (SOCC) via downregulating the expression of transient receptor potential canonical proteins (TRPC), which contribute to the formation of SOCC (Wang J, Jiang Q, Wan L, Yang K, Zhang Y, Chen Y, Wang E, Lai N, Zhao L, Jiang H, Sun Y, Zhong N, Ran P, Lu W. Am J Respir Cell Mol Biol 48: 125-134, 2013). The detailed molecular mechanisms by which STS inhibits SOCE and downregulates TRPC, however, remain largely unknown. We have previously shown that, under hypoxic conditions, inhibition of protein kinase G (PKG) and peroxisome proliferator-activated receptor-γ (PPAR-γ) signaling axis results in the upregulation of TRPC (Wang J, Yang K, Xu L, Zhang Y, Lai N, Jiang H, Zhang Y, Zhong N, Ran P, Lu W. Am J Respir Cell Mol Biol 49: 231-240, 2013). This suggests that strategies targeting the restoration of this signaling pathway may be an effective treatment strategy for pulmonary hypertension. In this study, our results demonstrated that STS treatment can effectively prevent the hypoxia-mediated inhibition of the PKG-PPAR-γ signaling axis in rat distal pulmonary arterial smooth muscle cells (PASMCs) and distal pulmonary arteries. These effects of STS treatment were blocked by pharmacological inhibition or specific small interfering RNA knockdown of either PKG or PPAR-γ. Moreover, targeted PPAR-γ agonist markedly enhanced the beneficial effects of STS. These results comprehensively suggest that STS treatment can prevent hypoxia-mediated increases in intracellular calcium homeostasis and cell proliferation, by targeting and restoring the hypoxia-inhibited PKG-PPAR-γ signaling pathway in PASMCs.
Collapse
Affiliation(s)
- Qian Jiang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kai Yang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cyrus Hadadi
- Department of Cardiology, Geisinger Medical Center, Danville, Pennsylvania
| | - Xin Fu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xin Yun
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jie Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Meichan Li
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lei Xu
- Department of Respiratory Diseases, The Affiliated Hospital of Inner Mongolia Medical University Hohhot, Inner Mongolia, China; and
| | - Haiyang Tang
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona, Tucson, Arizona
| | - Jason X-J Yuan
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona, Tucson, Arizona
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona, Tucson, Arizona; Division of Pulmonary Medicine, The People's Hospital of Inner Mongolia, Hohhot, Inner Mongolia, China;
| | - Dejun Sun
- Division of Pulmonary Medicine, The People's Hospital of Inner Mongolia, Hohhot, Inner Mongolia, China
| |
Collapse
|
25
|
Abstract
The circulation of the lung is unique both in volume and function. For example, it is the only organ with two circulations: the pulmonary circulation, the main function of which is gas exchange, and the bronchial circulation, a systemic vascular supply that provides oxygenated blood to the walls of the conducting airways, pulmonary arteries and veins. The pulmonary circulation accommodates the entire cardiac output, maintaining high blood flow at low intravascular arterial pressure. As compared with the systemic circulation, pulmonary arteries have thinner walls with much less vascular smooth muscle and a relative lack of basal tone. Factors controlling pulmonary blood flow include vascular structure, gravity, mechanical effects of breathing, and the influence of neural and humoral factors. Pulmonary vascular tone is also altered by hypoxia, which causes pulmonary vasoconstriction. If the hypoxic stimulus persists for a prolonged period, contraction is accompanied by remodeling of the vasculature, resulting in pulmonary hypertension. In addition, genetic and environmental factors can also confer susceptibility to development of pulmonary hypertension. Under normal conditions, the endothelium forms a tight barrier, actively regulating interstitial fluid homeostasis. Infection and inflammation compromise normal barrier homeostasis, resulting in increased permeability and edema formation. This article focuses on reviewing the basics of the lung circulation (pulmonary and bronchial), normal development and transition at birth and vasoregulation. Mechanisms contributing to pathological conditions in the pulmonary circulation, in particular when barrier function is disrupted and during development of pulmonary hypertension, will also be discussed.
Collapse
Affiliation(s)
- Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Larissa A. Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
26
|
Sommer N, Strielkov I, Pak O, Weissmann N. Oxygen sensing and signal transduction in hypoxic pulmonary vasoconstriction. Eur Respir J 2015; 47:288-303. [PMID: 26493804 DOI: 10.1183/13993003.00945-2015] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 08/24/2015] [Indexed: 01/17/2023]
Abstract
Hypoxic pulmonary vasoconstriction (HPV), also known as the von Euler-Liljestrand mechanism, is an essential response of the pulmonary vasculature to acute and sustained alveolar hypoxia. During local alveolar hypoxia, HPV matches perfusion to ventilation to maintain optimal arterial oxygenation. In contrast, during global alveolar hypoxia, HPV leads to pulmonary hypertension. The oxygen sensing and signal transduction machinery is located in the pulmonary arterial smooth muscle cells (PASMCs) of the pre-capillary vessels, albeit the physiological response may be modulated in vivo by the endothelium. While factors such as nitric oxide modulate HPV, reactive oxygen species (ROS) have been suggested to act as essential mediators in HPV. ROS may originate from mitochondria and/or NADPH oxidases but the exact oxygen sensing mechanisms, as well as the question of whether increased or decreased ROS cause HPV, are under debate. ROS may induce intracellular calcium increase and subsequent contraction of PASMCs via direct or indirect interactions with protein kinases, phospholipases, sarcoplasmic calcium channels, transient receptor potential channels, voltage-dependent potassium channels and L-type calcium channels, whose relevance may vary under different experimental conditions. Successful identification of factors regulating HPV may allow development of novel therapeutic approaches for conditions of disturbed HPV.
Collapse
Affiliation(s)
- Natascha Sommer
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Ievgen Strielkov
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Oleg Pak
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
27
|
NADPH oxidases—do they play a role in TRPC regulation under hypoxia? Pflugers Arch 2015; 468:23-41. [DOI: 10.1007/s00424-015-1731-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/23/2015] [Accepted: 08/25/2015] [Indexed: 12/25/2022]
|
28
|
Li Y, Guo B, Xie Q, Ye D, Zhang D, Zhu Y, Chen H, Zhu B. STIM1 Mediates Hypoxia-Driven Hepatocarcinogenesis via Interaction with HIF-1. Cell Rep 2015; 12:388-95. [DOI: 10.1016/j.celrep.2015.06.033] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 04/30/2015] [Accepted: 06/08/2015] [Indexed: 01/06/2023] Open
|
29
|
Veit F, Pak O, Brandes RP, Weissmann N. Hypoxia-dependent reactive oxygen species signaling in the pulmonary circulation: focus on ion channels. Antioxid Redox Signal 2015; 22:537-52. [PMID: 25545236 PMCID: PMC4322788 DOI: 10.1089/ars.2014.6234] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE An acute lack of oxygen in the lung causes hypoxic pulmonary vasoconstriction, which optimizes gas exchange. In contrast, chronic hypoxia triggers a pathological vascular remodeling causing pulmonary hypertension, and ischemia can cause vascular damage culminating in lung edema. RECENT ADVANCES Regulation of ion channel expression and gating by cellular redox state is a widely accepted mechanism; however, it remains a matter of debate whether an increase or a decrease in reactive oxygen species (ROS) occurs under hypoxic conditions. Ion channel redox regulation has been described in detail for some ion channels, such as Kv channels or TRPC6. However, in general, information on ion channel redox regulation remains scant. CRITICAL ISSUES AND FUTURE DIRECTIONS In addition to the debate of increased versus decreased ROS production during hypoxia, we aim here at describing and deciphering why different oxidants, under different conditions, can cause both activation and inhibition of channel activity. While the upstream pathways affecting channel gating are often well described, we need a better understanding of redox protein modifications to be able to determine the complexity of ion channel redox regulation. Against this background, we summarize the current knowledge on hypoxia-induced ROS-mediated ion channel signaling in the pulmonary circulation.
Collapse
Affiliation(s)
- Florian Veit
- 1 Excellence Cluster Cardiopulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL) , Giessen, Germany
| | | | | | | |
Collapse
|
30
|
Yoo HY, Park SJ, Kim HJ, Kim WK, Kim SJ. Integrative understanding of hypoxic pulmonary vasoconstriction using in vitro models: from ventilated/perfused lung to single arterial myocyte. Integr Med Res 2014; 3:180-188. [PMID: 28664095 PMCID: PMC5481745 DOI: 10.1016/j.imr.2014.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/27/2014] [Accepted: 08/27/2014] [Indexed: 10/25/2022] Open
Abstract
Contractile response of a pulmonary artery (PA) to hypoxia (hypoxic pulmonary vasoconstriction; HPV) is a unique physiological reaction. HPV is beneficial for the optimal distribution of blood flow to differentially ventilated alveolar regions in the lung, thereby preventing systemic hypoxemia. Numerous in vitro studies have been conducted to elucidate the mechanisms underlying HPV. These studies indicate that PA smooth muscle cells (PASMCs) sense lowers the oxygen partial pressure (PO2) and contract under hypoxia. As for the PO2-sensing molecules, a variety of ion channels in PASMCs had been suggested. Nonetheless, the modulator(s) of the ion channels alone cannot mimic HPV in the experiments using PA segments and/or isolated organs. We compared the hypoxic responses of PASMCs, PAs, lung slices, and total lungs using a variety of methods (e.g., patch-clamp technique, isometric contraction measurement, video analysis of precision-cut lung slices, and PA pressure measurement in ventilated/perfused lungs). In this review, the relevant results are compared to provide a comprehensive understanding of HPV. Integration of the influences from surrounding tissues including blood cells as well as the hypoxic regulation of ion channels in PASMCs are indispensable for insights into HPV and other related clinical conditions.
Collapse
Affiliation(s)
- Hae Young Yoo
- Red Cross College of Nursing, Chung-Ang University, Seoul, Korea
| | - Su Jung Park
- Department of Physiology, College of Medicine, Seoul National University, Seoul, Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Hae Jin Kim
- Department of Physiology, College of Medicine, Seoul National University, Seoul, Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Woo Kyung Kim
- Department of Internal Medicine and Channelopathy Research Institute (CRC), College of Medicine, Dongguk University, Goyang, Korea
| | - Sung Joon Kim
- Department of Physiology, College of Medicine, Seoul National University, Seoul, Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
31
|
Jiang Q, Fu X, Tian L, Chen Y, Yang K, Chen X, Zhang J, Lu W, Wang J. NOX4 mediates BMP4-induced upregulation of TRPC1 and 6 protein expressions in distal pulmonary arterial smooth muscle cells. PLoS One 2014; 9:e107135. [PMID: 25203114 PMCID: PMC4159322 DOI: 10.1371/journal.pone.0107135] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 08/10/2014] [Indexed: 02/07/2023] Open
Abstract
Rationale Our previous studies demonstrated that bone morphogenetic protein 4 (BMP4) mediated, elevated expression of canonical transient receptor potential (TRPC) largely accounts for the enhanced proliferation in pulmonary arterial smooth muscle cells (PASMCs). In the present study, we sought to determine the signaling pathway through which BMP4 up-regulates TRPC expression. Methods We employed recombinant human BMP4 (rhBMP4) to determine the effects of BMP4 on NADPH oxidase 4 (NOX4) and reactive oxygen species (ROS) production in rat distal PASMCs. We also designed small interfering RNA targeting NOX4 (siNOX4) and detected whether NOX4 knockdown affects rhBMP4-induced ROS, TRPC1 and 6 expression, cell proliferation and intracellular Ca2+ determination in PASMCs. Results In rhBMP4 treated rat distal PASMCs, NOX4 expression was (226.73±11.13) %, and the mean ROS level was (123.65±1.62) % of that in untreated control cell. siNOX4 transfection significantly reduced rhBMP4-induced elevation of the mean ROS level in PASMCs. Moreover, siNOX4 transfection markedly reduced rhBMP4-induced elevation of TRPC1 and 6 proteins, basal [Ca2+]i and SOCE. Furthermore, compared with control group (0.21±0.001), the proliferation of rhBMP4 treated cells was significantly enhanced (0.41±0.001) (P<0.01). However, such increase was attenuated by knockdown of NOX4. Moreover, external ROS (H2O2 100 µM, 24 h) rescued the effects of NOX4 knockdown, which included the declining of TRPC1 and 6 expression, basal intracellular calcium concentration ([Ca2+]i) and store-operated calcium entry (SOCE), suggesting that NOX4 plays as an important mediator in BMP4-induced proliferation and intracellular calcium homeostasis. Conclusion These results suggest that BMP4 may increase ROS level, enhance TRPC1 and 6 expression and proliferation by up-regulating NOX4 expression in PASMCs.
Collapse
Affiliation(s)
- Qian Jiang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xin Fu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lichun Tian
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kai Yang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Xiuqing Chen
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jie Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- * E-mail: (JW); (WL)
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Pulmonary, Inner Mongolia People’s Hospital, Huhhot, Inner Mongolia, China
- * E-mail: (JW); (WL)
| |
Collapse
|
32
|
Papamatheakis DG, Blood AB, Kim JH, Wilson SM. Antenatal hypoxia and pulmonary vascular function and remodeling. Curr Vasc Pharmacol 2014; 11:616-40. [PMID: 24063380 DOI: 10.2174/1570161111311050006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 06/25/2012] [Accepted: 07/12/2012] [Indexed: 01/02/2023]
Abstract
This review provides evidence that antenatal hypoxia, which represents a significant and worldwide problem, causes prenatal programming of the lung. A general overview of lung development is provided along with some background regarding transcriptional and signaling systems of the lung. The review illustrates that antenatal hypoxic stress can induce a continuum of responses depending on the species examined. Fetuses and newborns of certain species and specific human populations are well acclimated to antenatal hypoxia. However, antenatal hypoxia causes pulmonary vascular disease in fetuses and newborns of most mammalian species and humans. Disease can range from mild pulmonary hypertension, to severe vascular remodeling and dangerous elevations in pressure. The timing, length, and magnitude of the intrauterine hypoxic stress are important to disease development, however there is also a genetic-environmental relationship that is not yet completely understood. Determining the origins of pulmonary vascular remodeling and pulmonary hypertension and their associated effects is a challenging task, but is necessary in order to develop targeted therapies for pulmonary hypertension in the newborn due to antenatal hypoxia that can both treat the symptoms and curtail or reverse disease progression.
Collapse
Affiliation(s)
- Demosthenes G Papamatheakis
- Center for Perinatal Biology, Loma Linda University School of Medicine, 11234 Anderson Street, Loma Linda, 92350 CA, USA.
| | | | | | | |
Collapse
|
33
|
Jernigan NL, Resta TC. Calcium Homeostasis and Sensitization in Pulmonary Arterial Smooth Muscle. Microcirculation 2014; 21:259-71. [DOI: 10.1111/micc.12096] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 09/25/2013] [Indexed: 01/10/2023]
Affiliation(s)
- Nikki L. Jernigan
- Vascular Physiology Group; Department of Cell Biology and Physiology; University of New Mexico Health Sciences Center; Albuquerque New Mexico USA
| | - Thomas C. Resta
- Vascular Physiology Group; Department of Cell Biology and Physiology; University of New Mexico Health Sciences Center; Albuquerque New Mexico USA
| |
Collapse
|
34
|
Nitta CH, Osmond DA, Herbert LM, Beasley BF, Resta TC, Walker BR, Jernigan NL. Role of ASIC1 in the development of chronic hypoxia-induced pulmonary hypertension. Am J Physiol Heart Circ Physiol 2014; 306:H41-52. [PMID: 24186095 PMCID: PMC3920158 DOI: 10.1152/ajpheart.00269.2013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 10/30/2013] [Indexed: 11/22/2022]
Abstract
Chronic hypoxia (CH) associated with respiratory disease results in elevated pulmonary vascular intracellular Ca(2+) concentration, which elicits enhanced vasoconstriction and promotes vascular arterial remodeling and thus has important implications in the development of pulmonary hypertension (PH). Store-operated Ca(2+) entry (SOCE) contributes to this elevated intracellular Ca(2+) concentration and has also been linked to acute hypoxic pulmonary vasoconstriction (HPV). Since our laboratory has recently demonstrated an important role for acid-sensing ion channel 1 (ASIC1) in mediating SOCE, we hypothesized that ASIC1 contributes to both HPV and the development of CH-induced PH. To test this hypothesis, we examined responses to acute hypoxia in isolated lungs and assessed the effects of CH on indexes of PH, arterial remodeling, and vasoconstrictor reactivity in wild-type (ASIC1(+/+)) and ASIC1 knockout (ASIC1(-/-)) mice. Restoration of ASIC1 expression in pulmonary arterial smooth muscle cells from ASIC1(-/-) mice rescued SOCE, confirming the requirement for ASIC1 in this response. HPV responses were blunted in lungs from ASIC1(-/-) mice. Both SOCE and receptor-mediated Ca(2+) entry, along with agonist-dependent vasoconstrictor responses, were diminished in small pulmonary arteries from control ASIC(-/-) mice compared with ASIC(+/+) mice. The effects of CH to augment receptor-mediated vasoconstrictor and SOCE responses in vessels from ASIC1(+/+) mice were not observed after CH in ASIC1(-/-) mice. In addition, ASIC1(-/-) mice exhibited diminished right ventricular systolic pressure, right ventricular hypertrophy, and arterial remodeling in response to CH compared with ASIC1(+/+) mice. Taken together, these data demonstrate an important role for ASIC1 in both HPV and the development of CH-induced PH.
Collapse
Affiliation(s)
- Carlos H Nitta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | | | | | | | | | | | | |
Collapse
|
35
|
Wang J, Yang K, Xu L, Zhang Y, Lai N, Jiang H, Zhang Y, Zhong N, Ran P, Lu W. Sildenafil inhibits hypoxia-induced transient receptor potential canonical protein expression in pulmonary arterial smooth muscle via cGMP-PKG-PPARγ axis. Am J Respir Cell Mol Biol 2013; 49:231-40. [PMID: 23526219 DOI: 10.1165/rcmb.2012-0185oc] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Transient receptor potential canonical (TRPC) proteins play important roles in chronically hypoxic pulmonary hypertension (CHPH). Previous results indicated that sildenafil inhibited TRPC1 and TRPC6 expression in rat distal pulmonary arteries (PAs). However, the underlying mechanisms remain unknown. We undertook this study to investigate the downstream signaling of sildenafil's regulation on TRPC1 and TRPC6 expression in pulmonary arterial smooth muscle cells (PASMCs). Hypoxia-exposed rats (10% O2 for 21 d) and rat distal PASMCs (4% O2 for 60 h) were taken as models to mimic CHPH. Real-time PCR, Western blotting, and Fura-2-based fluorescent microscopy were performed for mRNA, protein, and Ca(2+) measurements, respectively. The cellular cyclic guanosine monophosphate (cGMP) analogue 8-(4-chlorophenylthio)-guanosine 3',5'-cyclic monophosphate sodium salt (CPT-cGMP) (100 μM) inhibited TRPC1 and TRPC6 expression, store-operated Ca(2+) entry (SOCE), and the proliferation and migration of PASMCs exposed to prolonged hypoxia. The inhibition of CPT-cGMP on TRPC1 and TRPC6 expression in PASMCs was relieved by either the inhibition or knockdown of cGMP-dependent protein kinase (PKG) and peroxisome proliferator-activated receptor γ (PPARγ) expression. Under hypoxic conditions, CPT-cGMP increased PPARγ expression. This increase was abolished by the PKG antagonists Rp8 or KT5823. PPARγ agonist GW1929 significantly decreased TRPC1 and TRPC6 expression in PASMCs. Moreover, hypoxia exposure decreased, whereas sildenafil treatment increased, PKG and PPARγ expression in PASMCs ex vivo, and in rat distal PAs in vivo. The suppressive effects of sildenafil on TRPC1 and TRPC6 in rat distal PAs and on the hemodynamic parameters of CHPH were inhibited by treatment with the PPARγ antagonist T0070907. We conclude that sildenafil inhibits TRPC1 and TRPC6 expression in PASMCs via cGMP-PKG-PPARγ-dependent signaling during CHPH.
Collapse
Affiliation(s)
- Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kizub IV, Strielkov IV, Shaifta Y, Becker S, Prieto-Lloret J, Snetkov VA, Soloviev AI, Aaronson PI, Ward JP. Gap junctions support the sustained phase of hypoxic pulmonary vasoconstriction by facilitating calcium sensitization. Cardiovasc Res 2013; 99:404-11. [PMID: 23708740 PMCID: PMC3718323 DOI: 10.1093/cvr/cvt129] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 05/09/2013] [Accepted: 05/18/2013] [Indexed: 01/04/2023] Open
Abstract
AIMS To determine the role of gap junctions (GJs) in hypoxic pulmonary vasoconstriction (HPV). METHODS AND RESULTS Studies were performed in rat isolated intrapulmonary arteries (IPAs) mounted on a myograph and in anaesthetized rats. Hypoxia induced a biphasic HPV response in IPAs preconstricted with prostaglandin F2α (PGF2α, 3 µM) or 20 mM K⁺. The GJ inhibitors 18β-glycyrrhetinic acid (18β-GA, 30 µM), heptanol (3.5 mM), or 2-aminoethoxydiphenyl borate (2-APB) (75 µM) had little effect on the transient Phase 1 of HPV, but abolished the sustained Phase 2 which is associated with Ca²⁺ sensitization. The voltage-dependent Ca²⁺ channel blocker diltiazem (10 µM) had no effect on HPV, and did not alter the inhibitory action of 18β-GA. Sustained HPV is enhanced by high glucose (15 mM) via potentiation of Ca²⁺ sensitization, in the presence of high glucose 18β-GA still abolished sustained HPV. Simultaneous measurement of tension and intracellular Ca²⁺ using Fura PE-3 demonstrated that whilst 18β-GA abolished tension development during sustained HPV, it did not affect the elevation of intracellular Ca²⁺. Consistent with this, 18β-GA abolished hypoxia-induced phosphorylation of the Rho kinase target MYPT-1. In anaesthetized rats hypoxia caused a biphasic increase in systolic right ventricular pressure. Treatment with oral 18β-GA (25 mg/kg) abolished the sustained component of the hypoxic pressor response. CONCLUSION These results imply that GJs are critically involved in the signalling pathways leading to Rho kinase-dependent Ca²⁺ sensitization during sustained HPV, but not elevation of intracellular Ca²⁺, and may explain the dependence of the former on an intact endothelium.
Collapse
Affiliation(s)
- Igor V. Kizub
- Department of Experimental Therapeutics, Institute of Pharmacology and Toxicology of National Academy of Medical Sciences of Ukraine, Kiev, Ukraine
- Division of Asthma, Allergy and Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Campus, London SE1 9RT, UK
| | - Ievgen V. Strielkov
- Department of Experimental Therapeutics, Institute of Pharmacology and Toxicology of National Academy of Medical Sciences of Ukraine, Kiev, Ukraine
| | - Yasin Shaifta
- Division of Asthma, Allergy and Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Campus, London SE1 9RT, UK
| | - Silke Becker
- Division of Asthma, Allergy and Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Campus, London SE1 9RT, UK
| | - Jesus Prieto-Lloret
- Division of Asthma, Allergy and Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Campus, London SE1 9RT, UK
| | - Vladimir A. Snetkov
- Division of Asthma, Allergy and Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Campus, London SE1 9RT, UK
| | - Anatoly I. Soloviev
- Department of Experimental Therapeutics, Institute of Pharmacology and Toxicology of National Academy of Medical Sciences of Ukraine, Kiev, Ukraine
| | - Philip I. Aaronson
- Division of Asthma, Allergy and Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Campus, London SE1 9RT, UK
| | - Jeremy P.T. Ward
- Division of Asthma, Allergy and Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Campus, London SE1 9RT, UK
| |
Collapse
|
37
|
Connolly MJ, Prieto-Lloret J, Becker S, Ward JPT, Aaronson PI. Hypoxic pulmonary vasoconstriction in the absence of pretone: essential role for intracellular Ca2+ release. J Physiol 2013; 591:4473-98. [PMID: 23774281 DOI: 10.1113/jphysiol.2013.253682] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hypoxic pulmonary vasoconstriction (HPV) maintains blood oxygenation during acute hypoxia but contributes to pulmonary hypertension during chronic hypoxia. The mechanisms of HPV remain controversial, in part because HPV is usually studied in the presence of agonist-induced preconstriction ('pretone'). This potentiates HPV but may obscure and distort its underlying mechanisms. We therefore carried out an extensive assessment of proposed mechanisms contributing to HPV in isolated intrapulmonary arteries (IPAs) in the absence of pretone by using a conventional small vessel myograph. Hypoxia elicited a biphasic constriction consisting of a small transient (phase 1) superimposed upon a sustained (phase 2) component. Neither phase was affected by the L-type Ca2+ channel antagonists diltiazem (10 and 30 μm) or nifedipine (3 μm). Application of the store-operated Ca2+ entry (SOCE) blockers BTP2 (10 μm) or SKF96365 (50 μm) attenuated phase 2 but not phase 1, whereas a lengthy (30 min) incubation in Ca2+-free physiological saline solution similarly reduced phase 2 but abolished phase 1. No further effect of inhibition of HPV was observed if the sarco/endoplasmic reticulum Ca2+-ATPase inhibitor cyclopiazonic acid (30 μm) was also applied during the 30 min incubation in Ca2+-free physiological saline solution. Pretreatment with 10 μm ryanodine and 15 mm caffeine abolished both phases, whereas treatment with 100 μm ryanodine attenuated both phases. The two-pore channel blocker NED-19 (1 μm) and the nicotinic acid adenine dinucleotide phosphate (NAADP) antagonist BZ194 (200 μm) had no effect on either phase of HPV. The lysosomal Ca2+-depleting agent concanamycin (1 μm) enhanced HPV if applied during hypoxia, but had no effect on HPV during a subsequent hypoxic challenge. The cyclic ADP ribose antagonist 8-bromo-cyclic ADP ribose (30 μm) had no effect on either phase of HPV. Neither the Ca2+-sensing receptor (CaSR) blocker NPS2390 (0.1 and 10 μm) nor FK506 (10 μm), a drug which displaces FKBP12.6 from ryanodine receptor 2 (RyR2), had any effect on HPV. HPV was virtually abolished by the rho kinase blocker Y-27632 (1 μm) and attenuated by the protein kinase C inhibitor Gö6983 (3 μm). Hypoxia for 45 min caused a significant increase in the ratio of oxidised to reduced glutathione (GSSG/GSH). HPV was unaffected by the NADPH oxidase inhibitor VAS2870 (10 μm), whereas phase 2 was inhibited but phase 1 was unaffected by the antioxidants ebselen (100 μm) and TEMPOL (3 mm). We conclude that both phases of HPV in this model are mainly dependent on [Ca2+]i release from the sarcoplasmic reticulum. Neither phase of HPV requires voltage-gated Ca2+ entry, but SOCE contributes to phase 2. We can detect no requirement for cyclic ADP ribose, NAADP-dependent lysosomal Ca2+ release, activation of the CaSR, or displacement of FKBP12.6 from RyR2 for either phase of HPV. Sustained HPV is associated with an oxidising shift in the GSSG/GSH redox potential and is inhibited by the antioxidants ebselen and TEMPOL, consistent with the concept that it requires an oxidising shift in the cell redox state or the generation of reactive oxygen species.
Collapse
Affiliation(s)
- Michelle J Connolly
- P. I. Aaronson: Room 1.19, Henriette Raphael House, Guy's Campus, King's College London, London SE1 9HN, UK.
| | | | | | | | | |
Collapse
|
38
|
Zhang Y, Lu W, Yang K, Xu L, Lai N, Tian L, Jiang Q, Duan X, Chen M, Wang J. Bone morphogenetic protein 2 decreases TRPC expression, store-operated Ca(2+) entry, and basal [Ca(2+)]i in rat distal pulmonary arterial smooth muscle cells. Am J Physiol Cell Physiol 2013; 304:C833-43. [PMID: 23447035 DOI: 10.1152/ajpcell.00036.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent studies indicate that multiple bone morphogenetic protein (BMP) family ligands and receptors are involved in the development of pulmonary arterial hypertension, yet the underlying mechanisms are incompletely understood. Although BMP2 and BMP4 share high homology in amino acid sequence, they appear to exert divergent effects on chronic hypoxic pulmonary hypertension (CHPH). While BMP4 promotes vascular remodeling, BMP2 prevents CHPH. We previously demonstrated that BMP4 upregulates the expression of canonical transient receptor potential channel (TRPC) proteins and, thereby, enhances store-operated Ca(2+) entry (SOCE) and elevates intracellular Ca(2+) concentration ([Ca(2+)]i) in pulmonary arterial smooth muscle cells (PASMCs). In this study, we investigated the effects of BMP2 on these variables in rat distal PASMCs. We found that treatment with BMP2 (50 ng/ml, 60 h) inhibited TRPC1, TRPC4, and TRPC6 mRNA and protein expression. Moreover, BMP2 treatment led to reduced SOCE and decreased basal [Ca(2+)]i in PASMCs. These alterations were associated with decreased PASMC proliferation and migration. Conversely, knockdown of BMP2 with specific small interference RNA resulted in increased cellular levels of TRPC1, TRPC4, and TRPC6 mRNA and protein, enhanced SOCE, elevated basal [Ca(2+)]i, and increased proliferation and migration of PASMCs. Together, these results indicate that BMP2 participates in regulating Ca(2+) signaling in PASMCs by inhibiting TRPC1, TRPC4, and TRPC6 expression, thus leading to reduced SOCE and basal [Ca(2+)]i and inhibition of cell proliferation and migration.
Collapse
Affiliation(s)
- Yi Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Parrau D, Ebensperger G, Herrera EA, Moraga F, Riquelme RA, Ulloa CE, Rojas RT, Silva P, Hernandez I, Ferrada J, Diaz M, Parer JT, Cabello G, Llanos AJ, Reyes RV. Store-operated channels in the pulmonary circulation of high- and low-altitude neonatal lambs. Am J Physiol Lung Cell Mol Physiol 2013; 304:L540-8. [PMID: 23418093 DOI: 10.1152/ajplung.00024.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We determined whether store-operated channels (SOC) are involved in neonatal pulmonary artery function under conditions of acute and chronic hypoxia, using newborn sheep gestated and born either at high altitude (HA, 3,600 m) or low altitude (LA, 520 m). Cardiopulmonary variables were recorded in vivo, with and without SOC blockade by 2-aminoethyldiphenylborinate (2-APB), during basal or acute hypoxic conditions. 2-APB did not have effects on basal mean pulmonary arterial pressure (mPAP), cardiac output, systemic arterial blood pressure, or systemic vascular resistance in both groups of neonates. During acute hypoxia 2-APB reduced mPAP and pulmonary vascular resistance in LA and HA, but this reduction was greater in HA. In addition, isolated pulmonary arteries mounted in a wire myograph were assessed for vascular reactivity. HA arteries showed a greater relaxation and sensitivity to SOC blockers than LA arteries. The pulmonary expression of two SOC-forming subunits, TRPC4 and STIM1, was upregulated in HA. Taken together, our results show that SOC contribute to hypoxic pulmonary vasoconstriction in newborn sheep and that SOC are upregulated by chronic hypoxia. Therefore, SOC may contribute to the development of neonatal pulmonary hypertension. We propose SOC channels could be potential targets to treat neonatal pulmonary hypertension.
Collapse
Affiliation(s)
- Daniela Parrau
- Laboratorios de Fisiología y Fisiopatología del Desarrollo, y de Bioquímica y Biología Molecular de la Hipoxia, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ng LC, O'Neill KG, French D, Airey JA, Singer CA, Tian H, Shen XM, Hume JR. TRPC1 and Orai1 interact with STIM1 and mediate capacitative Ca2+ entry caused by acute hypoxia in mouse pulmonary arterial smooth muscle cells. Am J Physiol Cell Physiol 2012; 303:C1156-72. [DOI: 10.1152/ajpcell.00065.2012] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Previous studies in pulmonary artery smooth muscle cells (PASMCs) showed that acute hypoxia activates capacitative Ca2+ entry (CCE) but the molecular candidate(s) mediating CCE caused by acute hypoxia remain unclear. The present study aimed to determine if transient receptor potential canonical 1 (TRPC1) and Orai1 interact with stromal interacting molecule 1 (STIM1) and mediate CCE caused by acute hypoxia in mouse PASMCs. In primary cultured PASMCs loaded with fura-2, acute hypoxia caused a transient followed by a sustained rise in intracellular Ca2+ concentration ([Ca2+]i). The transient but not sustained rise in [Ca2+]i was partially inhibited by nifedipine. Acute hypoxia also increased the rate of Mn2+ quench of fura-2 fluorescence that was inhibited by SKF 96365, Ni2+, La3+, and Gd3+, exhibiting pharmacological properties characteristic of CCE. The nifedipine-insensitive rise in [Ca2+]i and the increase in Mn2+ quench rate were both inhibited in cells treated with TRPC1 antibody or TRPC1 small interfering (si)RNA, in STIM1 siRNA-transfected cells and in Orai1 siRNA-transfected cells. Moreover, overexpression of STIM1 resulted in a marked increase in [Ca2+]i and Mn2+ quench rate caused by acute hypoxia, and they were reduced in cells treated with TRPC1 antibody and in cells transfected with Orai1 siRNA. Furthermore, TRPC1 and Orai1 coimmunoprecipitated with STIM1 and the precipitation levels of TRPC1 and Orai1 were increased in cells exposed to acute hypoxia. Immunostaining showed colocalizations of TRPC1-STIM1 and Orai1-STIM1, and the colocalizations of these proteins were more apparent in acute hypoxia. These data provide direct evidence that TRPC1 and Orai1 channels mediate CCE through activation of STIM1 in acute hypoxic mouse PASMCs.
Collapse
Affiliation(s)
- Lih Chyuan Ng
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada; and
| | - Kathryn G. O'Neill
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada; and
| | - Dominique French
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada; and
| | - Judith A. Airey
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada; and
| | - Cherie A. Singer
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada; and
| | - Honglin Tian
- Department of Pathology, University of Nevada School of Medicine, Reno, Nevada
| | - Xiao-Ming Shen
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada; and
| | - Joseph R. Hume
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada; and
| |
Collapse
|
41
|
Blood AB, Terry MH, Merritt TA, Papamatheakis DG, Blood Q, Ross JM, Power GG, Longo LD, Wilson SM. Effect of chronic perinatal hypoxia on the role of rho-kinase in pulmonary artery contraction in newborn lambs. Am J Physiol Regul Integr Comp Physiol 2012; 304:R136-46. [PMID: 23152110 DOI: 10.1152/ajpregu.00126.2012] [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]
Abstract
Exposure to chronic hypoxia during gestation predisposes infants to neonatal pulmonary hypertension, but the underlying mechanisms remain unclear. Here, we test the hypothesis that moderate continuous hypoxia during gestation causes changes in the rho-kinase pathway that persist in the newborn period, altering vessel tone and responsiveness. Lambs kept at 3,801 m above sea level during gestation and the first 2 wk of life were compared with those with gestation at low altitude. In vitro studies of isolated pulmonary arterial rings found a more forceful contraction in response to KCl and 5-HT in high-altitude compared with low-altitude lambs. There was no difference between the effects of blockers of various pathways of extracellular Ca(2+) entry in low- and high-altitude arteries. In contrast, inhibition of rho-kinase resulted in significantly greater attenuation of 5-HT constriction in high-altitude compared with low-altitude arteries. High-altitude lambs had higher baseline pulmonary artery pressures and greater elevations in pulmonary artery pressure during 15 min of acute hypoxia compared with low-altitude lambs. Despite evidence for an increased role for rho-kinase in high-altitude arteries, in vivo studies found no significant difference between the effects of rho-kinase inhibition on hypoxic pulmonary vasoconstriction in intact high-altitude and low-altitude lambs. We conclude that chronic hypoxia in utero results in increased vasopressor response to both acute hypoxia and serotonin, but that rho-kinase is involved only in the increased response to serotonin.
Collapse
Affiliation(s)
- Arlin B Blood
- Department of Pediatrics, Division of Neonatology, Loma Linda University School of Medicine, Loma Linda, CA 92373, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Shimoda LA. 55th Bowditch Lecture: Effects of chronic hypoxia on the pulmonary circulation: role of HIF-1. J Appl Physiol (1985) 2012; 113:1343-52. [PMID: 22923506 DOI: 10.1152/japplphysiol.00843.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
When exposed to chronic hypoxia (CH), the pulmonary circulation responds with enhanced contraction and vascular remodeling, resulting in elevated pulmonary arterial pressures. Our work has identified CH-induced alterations in the expression and activity of several ion channels and transporters in pulmonary vascular smooth muscle that contribute to the development of hypoxic pulmonary hypertension and uncovered a critical role for the transcription factor hypoxia-inducible factor-1 (HIF-1) in mediating these responses. Current work is focused on the regulation of HIF in the chronically hypoxic lung and evaluation of the potential for pharmacological inhibitors of HIF to prevent, reverse, or slow the progression of pulmonary hypertension.
Collapse
Affiliation(s)
- Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21224, USA.
| |
Collapse
|
43
|
Zhang J, Zhou J, Cai L, Lu Y, Wang T, Zhu L, Hu Q. Extracellular calcium-sensing receptor is critical in hypoxic pulmonary vasoconstriction. Antioxid Redox Signal 2012; 17:471-84. [PMID: 22098336 DOI: 10.1089/ars.2011.4168] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
AIMS The initiation of hypoxic pulmonary vasoconstriction (HPV) involves an increase in cytosolic calcium ([Ca(2+)](i)) in pulmonary artery (PA) smooth muscle cells (PASMCs). Both the processes depend on extracellular Ca(2+). Extracellular Ca(2+) can be sensed by extracellular calcium-sensing receptor (CaSR). This study aims at determining whether CaSR is pivotal in the initiation of HPV. RESULTS Experiments were performed in cultured PASMCs, isolated PAs, and rats including CaSR knockdown preparations. Both hypoxia and H(2)O(2) equivalent to the level achieved by hypoxia increased [Ca(2+)](i) in an extracellular Ca(2+)-dependent manner in PASMCs, and this was inhibited by CaSR knockdown or its negative allosteric modulator, Calhex231. Hypoxia-increased H(2)O(2) generation was diminished by mitochondria depletion. Mitochondria depletion abolished hypoxia-induced [Ca(2+)](i) increase (HICI), which was reversed by H(2)O(2) repletion. CaSR knockdown or Calhex231, however, prevented the reversible effect of H(2)O(2). HICI was abolished by catalase-polyethylene glycol (PEG-Catalase), not superoxide dismutase-polyethylene glycol (PEG-SOD) pretreatment, attenuated by ryanodine receptor3-knockdown or inhibition of store-operated Ca(2+) entry. HPV in vitro and in vivo was inhibited by Calhex231 and by CaSR knockdown. INNOVATION A novel mechanism underlying HPV is revealed by the role of CaSR in orchestrating reactive oxygen species and [Ca(2+)](i) signaling. CONCLUSIONS The activation of mitochondrial H(2)O(2)-sensitized CaSR by extracellular Ca(2+) mediates HICI in PASMCs and, thus, initiates HPV.
Collapse
Affiliation(s)
- Jiwei Zhang
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
44
|
Wang J, Shimoda LA, Sylvester JT. Ca2+ responses of pulmonary arterial myocytes to acute hypoxia require release from ryanodine and inositol trisphosphate receptors in sarcoplasmic reticulum. Am J Physiol Lung Cell Mol Physiol 2012; 303:L161-8. [PMID: 22582116 DOI: 10.1152/ajplung.00348.2011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In pulmonary arterial smooth muscle cells (PASMC), acute hypoxia increases intracellular Ca(2+) concentration ([Ca(2+)](i)) by inducing Ca(2+) release from the sarcoplasmic reticulum (SR) and Ca(2+) influx through store- and voltage-operated Ca(2+) channels in sarcolemma. To evaluate the mechanisms of hypoxic Ca(2+) release, we measured [Ca(2+)](i) with fluorescent microscopy in primary cultures of rat distal PASMC. In cells perfused with Ca(2+)-free Krebs Ringer bicarbonate solution (KRBS), brief exposures to caffeine (30 mM) and norepinephrine (300 μM), which activate SR ryanodine and inositol trisphosphate receptors (RyR, IP(3)R), respectively, or 4% O(2) caused rapid transient increases in [Ca(2+)](i), indicating intracellular Ca(2+) release. Preexposure of these cells to caffeine, norepinephrine, or the SR Ca(2+)-ATPase inhibitor cyclopiazonic acid (CPA; 10 μM) blocked subsequent Ca(2+) release to caffeine, norepinephrine, and hypoxia. The RyR antagonist ryanodine (10 μM) blocked Ca(2+) release to caffeine and hypoxia but not norepinephrine. The IP(3)R antagonist xestospongin C (XeC, 0.1 μM) blocked Ca(2+) release to norepinephrine and hypoxia but not caffeine. In PASMC perfused with normal KRBS, acute hypoxia caused a sustained increase in [Ca(2+)](i) that was abolished by ryanodine or XeC. These results suggest that in rat distal PASMC 1) the initial increase in [Ca(2+)](i) induced by hypoxia, as well as the subsequent Ca(2+) influx that sustained this increase, required release of Ca(2+) from both RyR and IP(3)R, and 2) the SR Ca(2+) stores accessed by RyR, IP(3)R, and hypoxia functioned as a common store, which was replenished by a CPA-inhibitable Ca(2+)-ATPase.
Collapse
Affiliation(s)
- Jian Wang
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
45
|
Jernigan NL, Herbert LM, Walker BR, Resta TC. Chronic hypoxia upregulates pulmonary arterial ASIC1: a novel mechanism of enhanced store-operated Ca2+ entry and receptor-dependent vasoconstriction. Am J Physiol Cell Physiol 2012; 302:C931-40. [PMID: 22205392 PMCID: PMC3311238 DOI: 10.1152/ajpcell.00332.2011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 12/23/2011] [Indexed: 12/19/2022]
Abstract
Acid-sensing ion channel 1 (ASIC1) is a newly characterized contributor to store-operated Ca(2+) entry (SOCE) in pulmonary vascular smooth muscle (VSM). Since SOCE is implicated in elevated basal VSM intracellular Ca(2+) concentration ([Ca(2+)](i)) and augmented vasoconstriction in chronic hypoxia (CH)-induced pulmonary hypertension, we hypothesized that ASIC1 contributes to these responses. To test this hypothesis, we examined effects of the specific pharmacologic ASIC1a inhibitor, psalmotoxin 1 (PcTX1), on vasoconstrictor and vessel wall [Ca(2+)](i) responses to UTP and KCl (depolarizing stimulus) in fura-2-loaded, pressurized small pulmonary arteries from control and CH (4 wk at 0.5 atm) Wistar rats. PcTX1 had no effect on basal vessel wall [Ca(2+)](i), but attenuated vasoconstriction and increases in vessel wall [Ca(2+)](i) to UTP in arteries from control and CH rats; normalizing responses between groups. In contrast, responses to the depolarizing stimulus, KCl, were unaffected by CH exposure or PcTX1. Upon examining potential Ca(2+) influx mechanisms, we found that PcTX1 prevented augmented SOCE following CH. Exposure to CH resulted in a significant increase in pulmonary arterial ASIC1 protein. This study supports a novel role of ASIC1 in elevated receptor-stimulated vasoconstriction following CH which is likely mediated through increased ASIC1 expression and SOCE.
Collapse
Affiliation(s)
- Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, USA.
| | | | | | | |
Collapse
|
46
|
Abstract
It has been known for more than 60 years, and suspected for over 100, that alveolar hypoxia causes pulmonary vasoconstriction by means of mechanisms local to the lung. For the last 20 years, it has been clear that the essential sensor, transduction, and effector mechanisms responsible for hypoxic pulmonary vasoconstriction (HPV) reside in the pulmonary arterial smooth muscle cell. The main focus of this review is the cellular and molecular work performed to clarify these intrinsic mechanisms and to determine how they are facilitated and inhibited by the extrinsic influences of other cells. Because the interaction of intrinsic and extrinsic mechanisms is likely to shape expression of HPV in vivo, we relate results obtained in cells to HPV in more intact preparations, such as intact and isolated lungs and isolated pulmonary vessels. Finally, we evaluate evidence regarding the contribution of HPV to the physiological and pathophysiological processes involved in the transition from fetal to neonatal life, pulmonary gas exchange, high-altitude pulmonary edema, and pulmonary hypertension. Although understanding of HPV has advanced significantly, major areas of ignorance and uncertainty await resolution.
Collapse
Affiliation(s)
- J T Sylvester
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, The Johns Hopkins University School ofMedicine, Baltimore, Maryland, USA.
| | | | | | | |
Collapse
|
47
|
Madden JA, Ahlf SB, Dantuma MW, Olson KR, Roerig DL. Precursors and inhibitors of hydrogen sulfide synthesis affect acute hypoxic pulmonary vasoconstriction in the intact lung. J Appl Physiol (1985) 2011; 112:411-8. [PMID: 22074719 DOI: 10.1152/japplphysiol.01049.2011] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The effects of hydrogen sulfide (H(2)S) and acute hypoxia are similar in isolated pulmonary arteries from various species. However, the involvement of H(2)S in hypoxic pulmonary vasoconstriction (HPV) has not been studied in the intact lung. The present study used an intact, isolated, perfused rat lung preparation to examine whether adding compounds essential to H(2)S synthesis or to its inhibition would result in a corresponding increase or decrease in the magnitude of HPV. Western blots performed in lung tissue identified the presence of the H(2)S-synthesizing enzymes, cystathionine γ-lyase (CSE) and 3-mercaptopyruvate sulfur transferase (3-MST), but not cystathionine β-synthase (CBS). Adding three H(2)S synthesis precursors, cysteine and oxidized or reduced glutathione, to the perfusate significantly increased peak arterial pressure during hypoxia compared with control (P < 0.05). Adding α-ketoglutarate to enhance the 3-MST enzyme pathway also resulted in an increase (P < 0.05). Both aspartate, which inhibits the 3-MST synthesis pathway, and propargylglycine (PPG), which inhibits the CSE pathway, significantly reduced the increases in arterial pressure during hypoxia. Diethylmaleate (DEM), which conjugates sulfhydryls, also reduced the peak hypoxic arterial pressure at concentrations >2 mM. Finally, H(2)S concentrations as measured with a specially designed polarographic electrode decreased markedly in lung tissue homogenate and in small pulmonary arteries when air was added to the hypoxic environment of the measurement chamber. The results of this study provide evidence that the rate of H(2)S synthesis plays a role in the magnitude of acute HPV in the isolated perfused rat lung.
Collapse
Affiliation(s)
- Jane A Madden
- Department of Neurology, The Medical College of Wisconsin and Research Service, Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin 53295, USA.
| | | | | | | | | |
Collapse
|
48
|
Shimoda LA, Polak J. Hypoxia. 4. Hypoxia and ion channel function. Am J Physiol Cell Physiol 2011; 300:C951-67. [PMID: 21178108 PMCID: PMC3093942 DOI: 10.1152/ajpcell.00512.2010] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 12/16/2010] [Indexed: 12/19/2022]
Abstract
The ability to sense and respond to oxygen deprivation is required for survival; thus, understanding the mechanisms by which changes in oxygen are linked to cell viability and function is of great importance. Ion channels play a critical role in regulating cell function in a wide variety of biological processes, including neuronal transmission, control of ventilation, cardiac contractility, and control of vasomotor tone. Since the 1988 discovery of oxygen-sensitive potassium channels in chemoreceptors, the effect of hypoxia on an assortment of ion channels has been studied in an array of cell types. In this review, we describe the effects of both acute and sustained hypoxia (continuous and intermittent) on mammalian ion channels in several tissues, the mode of action, and their contribution to diverse cellular processes.
Collapse
Affiliation(s)
- Larissa A Shimoda
- Div. of Pulmonary and Critical Care Medicine, Johns Hopkins University, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA.
| | | |
Collapse
|
49
|
Fuchs B, Rupp M, Ghofrani HA, Schermuly RT, Seeger W, Grimminger F, Gudermann T, Dietrich A, Weissmann N. Diacylglycerol regulates acute hypoxic pulmonary vasoconstriction via TRPC6. Respir Res 2011; 12:20. [PMID: 21294865 PMCID: PMC3042943 DOI: 10.1186/1465-9921-12-20] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 02/04/2011] [Indexed: 11/25/2022] Open
Abstract
Background Hypoxic pulmonary vasoconstriction (HPV) is an essential mechanism of the lung that matches blood perfusion to alveolar ventilation to optimize gas exchange. Recently we have demonstrated that acute but not sustained HPV is critically dependent on the classical transient receptor potential 6 (TRPC6) channel. However, the mechanism of TRPC6 activation during acute HPV remains elusive. We hypothesize that a diacylglycerol (DAG)-dependent activation of TRPC6 regulates acute HPV. Methods We investigated the effect of the DAG analog 1-oleoyl-2-acetyl-sn-glycerol (OAG) on normoxic vascular tone in isolated perfused and ventilated mouse lungs from TRPC6-deficient and wild-type mice. Moreover, the effects of OAG, the DAG kinase inhibitor R59949 and the phospholipase C inhibitor U73122 on the strength of HPV were investigated compared to those on non-hypoxia-induced vasoconstriction elicited by the thromboxane mimeticum U46619. Results OAG increased normoxic vascular tone in lungs from wild-type mice, but not in lungs from TRPC6-deficient mice. Under conditions of repetitive hypoxic ventilation, OAG as well as R59949 dose-dependently attenuated the strength of acute HPV whereas U46619-induced vasoconstrictions were not reduced. Like OAG, R59949 mimicked HPV, since it induced a dose-dependent vasoconstriction during normoxic ventilation. In contrast, U73122, a blocker of DAG synthesis, inhibited acute HPV whereas U73343, the inactive form of U73122, had no effect on HPV. Conclusion These findings support the conclusion that the TRPC6-dependency of acute HPV is induced via DAG.
Collapse
Affiliation(s)
- Beate Fuchs
- Excellence Cluster Cardio-Pulmonary System, University of Giessen Lung Center, Department of Internal Medicine II, Justus-Liebig-University Giessen, Giessen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Salomonsson M, Braunstein TH, Holstein-Rathlou NH, Jensen LJ. Na+-independent, nifedipine-resistant rat afferent arteriolar Ca2+ responses to noradrenaline: possible role of TRPC channels. Acta Physiol (Oxf) 2010; 200:265-78. [PMID: 20426773 DOI: 10.1111/j.1748-1716.2010.02141.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIM In rat afferent arterioles we investigated the role of Na(+) entry in noradrenaline (NA)-induced depolarization and voltage-dependent Ca(2+) entry together with the importance of the transient receptor potential channel (TRPC) subfamily for non-voltage-dependent Ca(2+) entry. METHODS R (340/380) Fura-2 fluorescence was used as an index for intracellular free Ca(2+) concentration ([Ca(2+)](i)). Immunofluorescence detected the expression of TRPC channels. RESULTS TRPC 1, 3 and 6 were expressed in afferent arteriolar vascular smooth muscle cells. Under extracellular Na(+)-free (0 Na) conditions, the plateau response to NA was 115% of the baseline R(340/380) (control response 123%). However, as the R(340/380) baseline increased (7%) after 0 Na the plateau reached the same level as during control conditions. Similar responses were obtained after blockade of the Na(+)/Ca(2+) exchanger. The L-type blocker nifedipine reduced the plateau response to NA both under control (from 134% to 116% of baseline) and 0 Na conditions (from 112% to 103% of baseline). In the presence of nifedipine, the putative TRPC channel blockers SKF 96365 (30 μm) and Gd(3+) (100 μm) further reduced the plateau Ca(2+) responses to NA (from 117% to 102% and from 117% to 110% respectively). CONCLUSION We found that Na(+) is not crucial for the NA-induced depolarization that mediates Ca(2+) entry via L-type channels. In addition, the results are consistent with the idea that TRPC1/3/6 Ca(2+) -permeable cation channels expressed in afferent arteriolar smooth muscle cells mediate Ca(2+) entry during NA stimulation.
Collapse
Affiliation(s)
- M Salomonsson
- Division of Renal and Vascular Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | |
Collapse
|