1
|
Ryszkiewicz P, Schlicker E, Malinowska B. Is Inducible Nitric Oxide Synthase (iNOS) Promising as a New Target Against Pulmonary Hypertension? Antioxidants (Basel) 2025; 14:377. [PMID: 40298665 PMCID: PMC12024173 DOI: 10.3390/antiox14040377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Pulmonary hypertension (PH) is a progressive disease characterized by elevated blood pressure in the pulmonary arteries, associated also with inflammation and oxidative stress. Inducible nitric oxide synthase (iNOS) is one of the key mediators of inflammation and immune system activation. Although preclinical studies mostly suggest a detrimental role of iNOS overactivation in PH, there is a lack of exhaustive analyses and summaries. Therefore, this literature overview aims to fill this gap. The involvement of iNOS in the pathogenesis of the four main clinical groups of PH is discussed to assess whether targeting iNOS could be a promising way to treat PH. iNOS expression patterns in the organs primarily affected by PH are analyzed both in animals and in humans. Consequently, the effectiveness of pharmacological iNOS inhibition and/or iNOS gene deletion is discussed and compared, also with reference to the activity of constitutive NOS isoforms, particularly endothelial NOS (eNOS). Overall, our overview suggests that selective iNOS inhibitors could be considered as a novel treatment strategy for PH, as decreases in right ventricular and pulmonary artery pressure, the alleviation of ventricular hypertrophy, and improvements of pulmonary and cardiac function were observed, among others. Nevertheless, further research efforts in this area are needed.
Collapse
Affiliation(s)
- Piotr Ryszkiewicz
- Department of Experimental Physiology and Pathophysiology, Medical University of Bialystok, Mickiewicz Str. 2A, 15-222 Bialystok, Poland
| | - Eberhard Schlicker
- Department of Pharmacology and Toxicology, University of Bonn, Venusberg Campus 1, 53127 Bonn, Germany;
| | - Barbara Malinowska
- Department of Experimental Physiology and Pathophysiology, Medical University of Bialystok, Mickiewicz Str. 2A, 15-222 Bialystok, Poland
| |
Collapse
|
2
|
Tsai J, Malik S, Tjen-A-Looi SC. Pulmonary Hypertension: Pharmacological and Non-Pharmacological Therapies. Life (Basel) 2024; 14:1265. [PMID: 39459565 PMCID: PMC11509317 DOI: 10.3390/life14101265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Pulmonary hypertension (PH) is a severe and chronic disease characterized by increased pulmonary vascular resistance and remodeling, often precipitating right-sided heart dysfunction and death. Although the condition is progressive and incurable, current therapies for the disease focus on multiple different drugs and general supportive therapies to manage symptoms and prolong survival, ranging from medications more specific to pulmonary arterial hypertension (PAH) to exercise training. Moreover, there are multiple studies exploring novel experimental drugs and therapies including unique neurostimulation, to help better manage the disease. Here, we provide a narrative review focusing on current PH treatments that target multiple underlying biochemical mechanisms, including imbalances in vasoconstrictor-vasodilator and autonomic nervous system function, inflammation, and bone morphogenic protein (BMP) signaling. We also focus on the potential of novel therapies for managing PH, focusing on multiple types of neurostimulation including acupuncture. Lastly, we also touch upon the disease's different subgroups, clinical presentations and prognosis, diagnostics, demographics, and cost.
Collapse
Affiliation(s)
- Jason Tsai
- Susan Samueli Integrative Health Institute, College of Health Sciences, University of California-Irvine, Irvine, CA 92617, USA;
| | | | - Stephanie C. Tjen-A-Looi
- Susan Samueli Integrative Health Institute, College of Health Sciences, University of California-Irvine, Irvine, CA 92617, USA;
| |
Collapse
|
3
|
Mallah MA, Soomro T, Ali M, Noreen S, Khatoon N, Kafle A, Feng F, Wang W, Naveed M, Zhang Q. Cigarette smoking and air pollution exposure and their effects on cardiovascular diseases. Front Public Health 2023; 11:967047. [PMID: 38045957 PMCID: PMC10691265 DOI: 10.3389/fpubh.2023.967047] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 06/26/2023] [Indexed: 12/05/2023] Open
Abstract
Cardiovascular disease (CVD) has no socioeconomic, topographical, or sex limitations as reported by the World Health Organization (WHO). The significant drivers of CVD are cardio-metabolic, behavioral, environmental, and social risk factors. However, some significant risk factors for CVD (e.g., a pitiable diet, tobacco smoking, and a lack of physical activities), have also been linked to an elevated risk of cardiovascular disease. Lifestyles and environmental factors are known key variables in cardiovascular disease. The familiarity with smoke goes along with the contact with the environment: air pollution is considered a source of toxins that contribute to the CVD burden. The incidence of myocardial infarction increases in males and females and may lead to fatal coronary artery disease, as confirmed by epidemiological studies. Lipid modification, inflammation, and vasomotor dysfunction are integral components of atherosclerosis development and advancement. These aspects are essential for the identification of atherosclerosis in clinical investigations. This article aims to show the findings on the influence of CVD on the health of individuals and human populations, as well as possible pathology and their involvement in smoking-related cardiovascular diseases. This review also explains lifestyle and environmental factors that are known to contribute to CVD, with indications suggesting an affiliation between cigarette smoking, air pollution, and CVD.
Collapse
Affiliation(s)
| | - Tahmina Soomro
- Department of Sociology, Shah Abdul Latif University, Khairpur, Pakistan
| | - Mukhtiar Ali
- Department of Chemical Engineering, Quaid-e-Awam University of Engineering, Science and Technology, Nawabshah, Sindh, Pakistan
| | - Sobia Noreen
- Department of Pharmaceutics Technology, Institute of Pharmacy, University of Innsbruck, Insbruck, Austria
| | - Nafeesa Khatoon
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Akriti Kafle
- School of Nursing, Zhengzhou University, Zhengzhou, China
| | - Feifei Feng
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Wei Wang
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Muhammad Naveed
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | - Qiao Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
4
|
Morris HE, Neves KB, Nilsen M, Montezano AC, MacLean MR, Touyz RM. Notch3/Hes5 Induces Vascular Dysfunction in Hypoxia-Induced Pulmonary Hypertension Through ER Stress and Redox-Sensitive Pathways. Hypertension 2023; 80:1683-1696. [PMID: 37254738 PMCID: PMC10355806 DOI: 10.1161/hypertensionaha.122.20449] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/24/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND Notch3 (neurogenic locus notch homolog protein 3) is implicated in vascular diseases, including pulmonary hypertension (PH)/pulmonary arterial hypertension. However, molecular mechanisms remain elusive. We hypothesized increased Notch3 activation induces oxidative and endoplasmic reticulum (ER) stress and downstream redox signaling, associated with procontractile pulmonary artery state, pulmonary vascular dysfunction, and PH development. METHODS Studies were performed in TgNotch3R169C mice (harboring gain-of-function [GOF] Notch3 mutation) exposed to chronic hypoxia to induce PH, and examined by hemodynamics. Molecular and cellular studies were performed in pulmonary artery smooth muscle cells from pulmonary arterial hypertension patients and in mouse lung. Notch3-regulated genes/proteins, ER stress, ROCK (Rho-associated kinase) expression/activity, Ca2+ transients and generation of reactive oxygen species, and nitric oxide were measured. Pulmonary vascular reactivity was assessed in the presence of fasudil (ROCK inhibitor) and 4-phenylbutyric acid (ER stress inhibitor). RESULTS Hypoxia induced a more severe PH phenotype in TgNotch3R169C mice versus controls. TgNotch3R169C mice exhibited enhanced Notch3 activation and expression of Notch3 targets Hes Family BHLH Transcription Factor 5 (Hes5), with increased vascular contraction and impaired vasorelaxation that improved with fasudil/4-phenylbutyric acid. Notch3 mutation was associated with increased pulmonary vessel Ca2+ transients, ROCK activation, ER stress, and increased reactive oxygen species generation, with reduced NO generation and blunted sGC (soluble guanylyl cyclase)/cGMP signaling. These effects were ameliorated by N-acetylcysteine. pulmonary artery smooth muscle cells from patients with pulmonary arterial hypertension recapitulated Notch3/Hes5 signaling, ER stress and redox changes observed in PH mice. CONCLUSIONS Notch3 GOF amplifies vascular dysfunction in hypoxic PH. This involves oxidative and ER stress, and ROCK. We highlight a novel role for Notch3/Hes5-redox signaling and important interplay between ER and oxidative stress in PH.
Collapse
Affiliation(s)
- Hannah E Morris
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (H.E.M., K.B.N., A.C.M., R.M.T.)
| | - Karla B Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (H.E.M., K.B.N., A.C.M., R.M.T.)
| | - Margaret Nilsen
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, United Kingdom (M.N., M.R.M.)
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (H.E.M., K.B.N., A.C.M., R.M.T.)
| | - Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, United Kingdom (M.N., M.R.M.)
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (H.E.M., K.B.N., A.C.M., R.M.T.)
- Research Institute of McGill University Health Centre, McGill University, Canada (R.M.T.)
| |
Collapse
|
5
|
Ghosh A, Ghosh B, Parihar N, Ilaweibaphyrnai M, Panda SR, Alexander A, Chella N, Murty U, Naidu V, Kumar G J, Pemmaraju DB. Nutraceutical prospects of Houttuynia cordata against the infectious viruses. FOOD BIOSCI 2022; 50:101977. [PMID: 36059903 PMCID: PMC9423882 DOI: 10.1016/j.fbio.2022.101977] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 08/04/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022]
Abstract
The novel enveloped β-coronavirus SARS-CoV-2 (COVID-19) has offered a surprising health challenge all over the world. It develops severe pneumonia leading to acute respiratory distress syndrome (ARDS). Like SARS-COV-2, other encapsulated viruses like HIV, HSV, and influenza have also offered a similar challenge in the past. In this regard, many antiviral drugs are being explored with varying degrees of success to combat the associated pathological conditions. Therefore, upon scientific validation & development, these antiviral phytochemicals can attain a futuristic nutraceutical prospect in managing different encapsulated viruses. Houttuynia cordata (HC) is widely reported for activities such as antioxidant, anti-inflammatory, and antiviral properties. The major antiviral bioactive components of HC include essential oils (methyl n-nonyl ketone, lauryl aldehyde, capryl aldehyde), flavonoids (quercetin, rutin, hyperin, quercitrin, isoquercitrin), and alkaloids (norcepharadione B) & polysaccharides. HC can further be explored as a potential nutraceutical agent in the therapy of encapsulated viruses like HIV, HSV, and influenza. The review listed various conventional and green technologies that are being employed to extract potent phytochemicals with diverse activities from the HC. It was indicated that HC also inhibited molecular targets like 3C-like protease (3CLPRO) and RNA-dependent RNA polymerase (RdRp) of COVID-19 by blocking viral RNA synthesis and replication. Antioxidant and hepatoprotective effects of HC have been evident in impeding complications from marketed drugs during antiviral therapies. The use of HC as a nutraceutical is localized within some parts of Southeast Asia. Further technological advances can establish it as a nutraceutical-based functional food against pathogenic enveloped viruses like COVID 19.
Collapse
Affiliation(s)
- Aparajita Ghosh
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Assam, 781101, India
| | - Bijoyani Ghosh
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Assam, 781101, India
| | - Nidhi Parihar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Assam, 781101, India
| | - Myrthong Ilaweibaphyrnai
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Assam, 781101, India
| | - Samir R Panda
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Assam, 781101, India
| | - Amit Alexander
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam, 781101, India
| | - Naveen Chella
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam, 781101, India
| | - Usn Murty
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Assam, 781101, India
| | - Vgm Naidu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Assam, 781101, India
| | - Jagadeesh Kumar G
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Assam, 781101, India
| | - Deepak B Pemmaraju
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Assam, 781101, India
| |
Collapse
|
6
|
Wang D, Ji Y, Wang R, Cheng K, Liu L, Wu N, Tang Q, Zheng X, Li J, Zhu Z, Wang Q, Zhang X, Li R, Pan J, Sui Z, Yuan Y. Lycopene Ameliorates Hypoxic Pulmonary Hypertension via Suppression of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9179427. [PMID: 39282152 PMCID: PMC11401662 DOI: 10.1155/2022/9179427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/24/2022] [Accepted: 09/09/2022] [Indexed: 09/18/2024]
Abstract
Hypoxic pulmonary hypertension (HPH) is a progressive cardiopulmonary system disease characterized by pulmonary vascular remodeling. Its occurrence and progression are closely related to oxidative stress. Lycopene, extracted from red vegetables and fruits, exhibits a particularly high antioxidant capacity that is beneficial for cardiovascular diseases. Nevertheless, the role and mechanism of lycopene in HPH remain unknown. Here, we found that lycopene reversed the elevated right ventricular systolic pressure (RVSP), right ventricular hypertrophy, and pulmonary vascular remodeling induced by hypoxia in rats. In vitro, lycopene caused lower proliferation and migration of PASMCs, with higher apoptosis. Consistent with the antiproliferative result of lycopene on hypoxic PASMCs, the hippo signaling pathway associated with cell growth was activated. Furthermore, lycopene reduced malondialdehyde (MDA) levels and enhanced superoxide dismutase (SOD) activity in the lungs and serum of rats under hypoxia conditions. The expression of NOX4 in the lungs was also significantly decreased. Hypoxic PASMCs subjected to lycopene showed decreased reactive oxygen species (ROS) production and NOX4 expression. Importantly, lycopene repressed HIF-1α expression both in the lungs and PASMCs in response to hypoxia in the absence of a significant change of HIF-1α mRNA. Compared with 2ME2 (a HIF-1α inhibitor) alone treatment, lycopene treatment did not significantly change PASMC proliferation, NOX4 expression, and ROS production after 2ME2 blocked HIF-1α, suggesting the inhibitory effect of lycopene on HIF-1α-NOX4-ROS axis and the targeted effect on HIF-1α. After CHX blocked protein synthesis, lycopene promoted the protein degradation of HIF-1α. MG-132, a proteasome inhibitor, notably reversed the decrease in HIF-1α protein level induced by lycopene in response to hypoxia. Therefore, lycopene suppressed hypoxia-induced oxidative stress through HIF-1α-NOX4-ROS axis, thereby alleviating HPH. Our findings will provide a new research direction for clinical HPH therapies.
Collapse
Affiliation(s)
- Dingyou Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Yuke Ji
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Rui Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Ke Cheng
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Liang Liu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Na Wu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Qing Tang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Xu Zheng
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Junxia Li
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Zhilong Zhu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Qinghua Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Xueyan Zhang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Runbo Li
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Jinjin Pan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Zheng Sui
- Department of Vasculocardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Yuhui Yuan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| |
Collapse
|
7
|
Liu R, Xu C, Zhang W, Cao Y, Ye J, Li B, Jia S, Weng L, Liu Y, Liu L, Zheng M. FUNDC1-mediated mitophagy and HIF1α activation drives pulmonary hypertension during hypoxia. Cell Death Dis 2022; 13:634. [PMID: 35864106 PMCID: PMC9304375 DOI: 10.1038/s41419-022-05091-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 01/21/2023]
Abstract
Hypoxic pulmonary hypertension (PH) is a progressive disease characterized by hyper-proliferation of pulmonary vascular cells including pulmonary artery smooth muscle cells (PASMCs) and can lead to right heart failure and early death. Selective degradation of mitochondria by mitophagy during hypoxia regulates mitochondrial functions in many cells, however, it is not clear if mitophagy is involved in the pathogenesis of hypoxic PH. By employing the hypoxic mitophagy receptor Fundc1 knockout (KO) and transgenic (TG) mouse models, combined hypoxic PH models, the current study found that mitophagy is actively involved in hypoxic PH through regulating PASMC proliferation. In the pulmonary artery medium from hypoxic PH mice, mitophagy was upregulated, accompanied with the increased active form of FUNDC1 protein and the enhanced binding affinity of FUNDC1 with LC3B. In PASMCs, overexpression of FUNDC1 increased mitophagy and cell proliferation while knockdown of FUNDC1 inhibited hypoxia-induced mitophagy and PASMC proliferation. Stimulation of mitophagy by FUNDC1 in PASMCs elevated ROS production and inhibited ubiquitination of hypoxia inducible factor 1α (HIF1α), and inhibition of mitophagy by FUNDC1 knockdown or knockout abolished hypoxia-induced ROS-HIF1α upregulation. Moreover, Fundc1 TG mice developed severe hemodynamics changes and pulmonary vascular remodeling, and Fundc1 KO mice were much resistant to hypoxic PH. In addition, intraperitoneal injection of a specific FUNDC1 peptide inhibitor to block mitophagy ameliorated hypoxic PH. Our results reveal that during hypoxic PH, FUNDC1-mediated mitophagy is upregulated which activates ROS-HIF1α pathway and promotes PASMC proliferation, ultimately leads to pulmonary vascular remodeling and PH.
Collapse
Affiliation(s)
- Ruxia Liu
- grid.11135.370000 0001 2256 9319Ministry of Education Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China ,grid.265021.20000 0000 9792 1228Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| | - Chunling Xu
- grid.11135.370000 0001 2256 9319Ministry of Education Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Weilin Zhang
- grid.9227.e0000000119573309State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yangpo Cao
- grid.11135.370000 0001 2256 9319Ministry of Education Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jingjing Ye
- grid.11135.370000 0001 2256 9319Ministry of Education Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Bo Li
- grid.11135.370000 0001 2256 9319Ministry of Education Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Shi Jia
- grid.11135.370000 0001 2256 9319Ministry of Education Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Lin Weng
- grid.11135.370000 0001 2256 9319Ministry of Education Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yingying Liu
- grid.11135.370000 0001 2256 9319Ministry of Education Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Lei Liu
- grid.9227.e0000000119573309State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ming Zheng
- grid.11135.370000 0001 2256 9319Ministry of Education Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|
8
|
Artemisinin and Its Derivate Alleviate Pulmonary Hypertension and Vasoconstriction in Rodent Models. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2782429. [PMID: 35757500 PMCID: PMC9232380 DOI: 10.1155/2022/2782429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 03/20/2022] [Accepted: 05/16/2022] [Indexed: 11/17/2022]
Abstract
Background Pulmonary arterial hypertension (PAH) is a complex pulmonary vasculature disease characterized by progressive obliteration of small pulmonary arteries and persistent increase in pulmonary vascular resistance, resulting in right heart failure and death if left untreated. Artemisinin (ARS) and its derivatives, which are common antimalarial drugs, have been found to possess a broad range of biological effects. Here, we sought to determine the therapeutic benefit and mechanism of ARS and its derivatives treatment in experimental pulmonary hypertension (PH) models. Methods Isolated perfused/ventilated lung and isometric tension measurements in arteries were performed to test pulmonary vasoconstriction and relaxation. Monocrotaline (MCT) and hypoxia+Su5416 (SuHx) were administered to rats to induce severe PH. Evaluation methods of ARS treatment and its derivatives in animal models include echocardiography, hemodynamics measurement, and histological staining. In vitro, the effect of these drugs on proliferation, viability, and hypoxia-inducible factor 1α (HIF1α) was examined in human pulmonary arterial smooth muscle cells (hPASMCs). Results ARS treatment attenuated pulmonary vasoconstriction induced by high K+ solution or alveolar hypoxia, decreased pulmonary artery (PA) basal vascular tension, improved acetylcholine- (ACh-) induced endothelial-dependent relaxation, increased endothelial nitric oxide (NO) synthase (eNOS) activity and NO levels, and decreased levels of NAD(P)H oxidase subunits (NOX2 and NOX4) expression, NAD(P)H oxidase activity, and reactive oxygen species (ROS) levels of pulmonary arteries (PAs) in MCT-PH rats. NOS inhibitor, L-NAME, abrogated the effects of ARS on PA constriction and relaxation. Furthermore, chronic application of both ARS and its derivative dihydroartemisinin (DHA) attenuated right ventricular systolic pressure (RVSP), Fulton index (right ventricular hypertrophy), and vascular remodeling of PAs in the two rat PH models. In addition, DHA inhibited proliferation and migration of hypoxia-induced PASMCs. Conclusions In conclusion, these results indicate that treatment with ARS or DHA can inhibit PA vasoconstriction, PASMC proliferation and migration, and vascular remodeling, as well as improve PA endothelium-dependent relaxation, and eventually attenuate the development and progression of PH. These effects might be achieved by decreasing NAD(P)H oxidase generated ROS production and increasing eNOS activation to release NO in PAs. ARS and its derivatives might have the potential to be novel drugs for the treatment of PH.
Collapse
|
9
|
Truong L, Zheng YM, Wang YX. The Potential Important Role of Mitochondrial Rieske Iron-Sulfur Protein as a Novel Therapeutic Target for Pulmonary Hypertension in Chronic Obstructive Pulmonary Disease. Biomedicines 2022; 10:957. [PMID: 35625694 PMCID: PMC9138741 DOI: 10.3390/biomedicines10050957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/19/2022] [Accepted: 04/19/2022] [Indexed: 02/01/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide, which is often due to pulmonary hypertension (PH). The underlying molecular mechanisms are poorly understood, and current medications are neither specific nor always effective. In this review, we highlight the recent findings on the roles of altered mitochondrial bioenergetics in PH in COPD. We also discuss the central role of mitochondrial reactive oxygen species (ROS) generation mediated by Rieske iron-sulfur protein (RISP) and review the contributions of RISP-dependent DNA damage and NF-κB-associated inflammatory signaling. Finally, the potential importance of mitochondrial RISP and its associated molecules as novel therapeutic targets for PH in COPD are meticulously discussed.
Collapse
Affiliation(s)
| | | | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (L.T.); (Y.-M.Z.)
| |
Collapse
|
10
|
Maladaptive Pulmonary Vascular Responses to Chronic Sustained and Chronic Intermittent Hypoxia in Rat. Antioxidants (Basel) 2021; 11:antiox11010054. [PMID: 35052557 PMCID: PMC8773044 DOI: 10.3390/antiox11010054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic sustained hypoxia (CSH), as found in individuals living at a high altitude or in patients suffering respiratory disorders, initiates physiological adaptations such as carotid body stimulation to maintain oxygen levels, but has deleterious effects such as pulmonary hypertension (PH). Obstructive sleep apnea (OSA), a respiratory disorder of increasing prevalence, is characterized by a situation of chronic intermittent hypoxia (CIH). OSA is associated with the development of systemic hypertension and cardiovascular pathologies, due to carotid body and sympathetic overactivation. There is growing evidence that CIH can also compromise the pulmonary circulation, causing pulmonary hypertension in OSA patients and animal models. The aim of this work was to compare hemodynamics, vascular contractility, and L-arginine-NO metabolism in two models of PH in rats, associated with CSH and CIH exposure. We demonstrate that whereas CSH and CIH cause several common effects such as an increased hematocrit, weight loss, and an increase in pulmonary artery pressure (PAP), compared to CIH, CSH seems to have more of an effect on the pulmonary circulation, whereas the effects of CIH are apparently more targeted on the systemic circulation. The results suggest that the endothelial dysfunction evident in pulmonary arteries with both hypoxia protocols are not due to an increase in methylated arginines in these arteries, although an increase in plasma SDMA could contribute to the apparent loss of basal NO-dependent vasodilation and, therefore, the increase in PAP that results from CIH.
Collapse
|
11
|
Piknova B, Schechter AN, Park JW, Vanhatalo A, Jones AM. Skeletal Muscle Nitrate as a Regulator of Systemic Nitric Oxide Homeostasis. Exerc Sport Sci Rev 2021; 50:2-13. [PMID: 34669624 DOI: 10.1249/jes.0000000000000272] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT Non-enzymatic nitric oxide (NO) generation via the reduction of nitrate and nitrite ions, along with remarkably high levels of nitrate ions in skeletal muscle, have been recently described. Skeletal muscle nitrate storage may be critical for maintenance of NO homeostasis in healthy ageing and nitrate supplementation may be useful for treatment of specific pathophysiologies as well as enhancing normal functions.
Collapse
Affiliation(s)
- Barbora Piknova
- Molecular Medicine Branch, NIDDK, National Institutes of Health,Bethesda, MD 20892, U.S. Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | | | | | | | | |
Collapse
|
12
|
Huynh GT, Kesarwani V, Walker JA, Frith JE, Meagher L, Corrie SR. Review: Nanomaterials for Reactive Oxygen Species Detection and Monitoring in Biological Environments. Front Chem 2021; 9:728717. [PMID: 34568279 PMCID: PMC8461210 DOI: 10.3389/fchem.2021.728717] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/25/2021] [Indexed: 12/19/2022] Open
Abstract
Reactive oxygen species (ROS) and dissolved oxygen play key roles across many biological processes, and fluorescent stains and dyes are the primary tools used to quantify these species in vitro. However, spatio-temporal monitoring of ROS and dissolved oxygen in biological systems are challenging due to issues including poor photostability, lack of reversibility, and rapid off-site diffusion. In particular, ROS monitoring is hindered by the short lifetime of ROS molecules and their low abundance. The combination of nanomaterials and fluorescent detection has led to new opportunities for development of imaging probes, sensors, and theranostic products, because the scaffolds lead to improved optical properties, tuneable interactions with cells and media, and ratiometric sensing robust to environmental drift. In this review, we aim to critically assess and highlight recent development in nanosensors and nanomaterials used for the detection of oxygen and ROS in biological systems, and their future potential use as diagnosis tools.
Collapse
Affiliation(s)
- Gabriel T. Huynh
- Department of Chemical Engineering, Monash University, Clayton, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Node, Clayton, VIC, Australia
| | - Vidhishri Kesarwani
- Department of Chemical Engineering, Monash University, Clayton, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Node, Clayton, VIC, Australia
| | - Julia A. Walker
- Department of Chemical Engineering, Monash University, Clayton, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Node, Clayton, VIC, Australia
| | - Jessica E. Frith
- Monash Institute of Medical Engineering, Monash University, Clayton, VIC, Australia
- Department of Material Science and Engineering, Monash University, Clayton, VIC, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, Australia
| | - Laurence Meagher
- Department of Material Science and Engineering, Monash University, Clayton, VIC, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, Australia
| | - Simon R. Corrie
- Department of Chemical Engineering, Monash University, Clayton, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Node, Clayton, VIC, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, Australia
| |
Collapse
|
13
|
Song JL, Zheng SY, He RL, Gui LX, Lin MJ, Sham JSK. Serotonin and chronic hypoxic pulmonary hypertension activate a NADPH oxidase 4 and TRPM2 dependent pathway for pulmonary arterial smooth muscle cell proliferation and migration. Vascul Pharmacol 2021; 138:106860. [PMID: 33794383 DOI: 10.1016/j.vph.2021.106860] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 03/17/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023]
Abstract
5-Hydroxytryptamine (5-HT)-dependent signaling mediated through its transporters and receptors plays important roles in chronic hypoxic pulmonary hypertension (CHPH), which is associated with aberrant reactive oxygen species (ROS) production. NADPH oxidase 4 (NOX4) is one of the major sources of ROS in pulmonary vasculature, and has been implicated in the development of PH. NOX4 generates H2O2, which can activate the transient receptor potential melastatin 2 (TRPM2) channels, providing Ca2+ signals for cell proliferation and migration. However, the connection between 5-HT, NOX4, ROS and TRPM2 in the context of PH has not been established. Here we examined the level of 5-HT and expression of NOX4 and TRPM2, and their roles in pulmonary arterial smooth muscle cells (PASMCs) proliferation and migration. NOX4 and TRPM2 were upregulated in pulmonary arteries of CHPH rats, which were associated with elevated levels of 5-HT and ROS, and enhanced proliferation and migration in PASMCs. The increase in ROS, and the enhanced proliferation and migration of PASMCs from CHPH rats were mimicked by treating normoxic PASMCs with 5-HT. 5-HT; and CH-induced ROS production were reversed by catalase, the NOX1/NOX4 inhibitor GKT137831, and Nox4 siRNA. 5-HT and H2O2 elicited Ca2+ responses were significantly augmented in CHPH PASMCs; and the augmented Ca2+ responses were obliterated by the 2-Aminoethoxydiphenyl borate (2-APB) and Trpm2-specific siRNA. Moreover, 5-HT and CH-induced proliferation and migration were suppressed by Nox4 or Trpm2 siRNA; and simultaneous transfection of both siRNA did not cause further inhibition. These results suggest that the 5-HT and CH-induced PASMC proliferation and migration were mediated, at least in part, by TRPM2 via activation of NOX4-dependent ROS production; and revealed a novel NOX4-ROS-TRPM2 signaling pathway for the pathogenesis of CHPH.
Collapse
MESH Headings
- Animals
- Calcium Signaling
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Chronic Disease
- Disease Models, Animal
- Hypoxia/complications
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- NADPH Oxidase 4/genetics
- NADPH Oxidase 4/metabolism
- Pulmonary Arterial Hypertension/enzymology
- Pulmonary Arterial Hypertension/etiology
- Pulmonary Arterial Hypertension/pathology
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/enzymology
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Serotonin/metabolism
- Serotonin/pharmacology
- TRPM Cation Channels/genetics
- TRPM Cation Channels/metabolism
- Vascular Remodeling/drug effects
- Rats
Collapse
Affiliation(s)
- Jia-Lin Song
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, PR China
| | - Si-Yi Zheng
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, PR China
| | - Rui-Lan He
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, PR China
| | - Long-Xin Gui
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, PR China
| | - Mo-Jun Lin
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, PR China.
| | - James S K Sham
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
14
|
Wang Y, Li X, Niu W, Chen J, Zhang B, Zhang X, Wang Y, Dang S, Li Z. The alveolar epithelial cells are involved in pulmonary vascular remodeling and constriction of hypoxic pulmonary hypertension. Respir Res 2021; 22:134. [PMID: 33947399 PMCID: PMC8094493 DOI: 10.1186/s12931-021-01708-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/08/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hypoxic pulmonary hypertension (HPH) is a common type of pulmonary hypertension and characterized by pulmonary vascular remodeling and constriction. Alveolar epithelial cells (AECs) primarily sense alveolar hypoxia, but the role of AECs in HPH remains unclear. In this study, we explored whether AECs are involved in pulmonary vascular remodeling and constriction. METHODS In the constructed rat HPH model, hemodynamic and morphological characteristics were measured. By treating AECs with hypoxia, we further detected the levels of superoxide dismutase 2 (SOD2), catalase (CAT), reactive oxygen species (ROS) and hydrogen peroxide (H2O2), respectively. To detect the effects of AECs on pulmonary vascular remodeling and constriction, AECs and pulmonary artery smooth cells (PASMCs) were co-cultured under hypoxia, and PASMCs and isolated pulmonary artery (PA) were treated with AECs hypoxic culture medium. In addition, to explore the mechanism of AECs on pulmonary vascular remodeling and constriction, ROS inhibitor N-acetylcysteine (NAC) was used. RESULTS Hypoxia caused pulmonary vascular remodeling and increased pulmonary artery pressure, but had little effect on non-pulmonary vessels in vivo. Meanwhile, in vitro, hypoxia promoted the imbalance of SOD2 and CAT in AECs, leading to increased ROS and hydrogen peroxide (H2O2) production in the AECs culture medium. In addition, AECs caused the proliferation of co-cultured PASMCs under hypoxia, and the hypoxic culture medium of AECs enhanced the constriction of isolated PA. However, treatment with ROS inhibitor NAC effectively alleviated the above effects. CONCLUSION The findings of present study demonstrated that AECs were involved in pulmonary vascular remodeling and constriction under hypoxia by paracrine H2O2 into the pulmonary vascular microenvironment.
Collapse
Affiliation(s)
- Yanxia Wang
- Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Xiaoming Li
- Department of Pathophysiology, Xi'an Peihua University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Wen Niu
- Department of Pathophysiology, School of Basic Medicine, Fourth Military Medical University, 169 Changle Western Street, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Jian Chen
- Department of Respiratory and Critical Care, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Street, Xi'an, Shaanxi, 710038, People's Republic of China
| | - Bo Zhang
- Department of Pathophysiology, School of Basic Medicine, Fourth Military Medical University, 169 Changle Western Street, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Xiumin Zhang
- Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Yingmei Wang
- Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Shaokang Dang
- Department of Respiratory and Critical Care, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Street, Xi'an, Shaanxi, 710038, People's Republic of China.
| | - Zhichao Li
- Department of Pathophysiology, School of Basic Medicine, Fourth Military Medical University, 169 Changle Western Street, Xi'an, Shaanxi, 710032, People's Republic of China.
- Northwest University School of Medicine, Xi'an, Shaanxi, 710075, People's Republic of China.
| |
Collapse
|
15
|
McCarty MF. Nutraceutical, Dietary, and Lifestyle Options for Prevention and Treatment of Ventricular Hypertrophy and Heart Failure. Int J Mol Sci 2021; 22:ijms22073321. [PMID: 33805039 PMCID: PMC8037104 DOI: 10.3390/ijms22073321] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Although well documented drug therapies are available for the management of ventricular hypertrophy (VH) and heart failure (HF), most patients nonetheless experience a downhill course, and further therapeutic measures are needed. Nutraceutical, dietary, and lifestyle measures may have particular merit in this regard, as they are currently available, relatively safe and inexpensive, and can lend themselves to primary prevention as well. A consideration of the pathogenic mechanisms underlying the VH/HF syndrome suggests that measures which control oxidative and endoplasmic reticulum (ER) stress, that support effective nitric oxide and hydrogen sulfide bioactivity, that prevent a reduction in cardiomyocyte pH, and that boost the production of protective hormones, such as fibroblast growth factor 21 (FGF21), while suppressing fibroblast growth factor 23 (FGF23) and marinobufagenin, may have utility for preventing and controlling this syndrome. Agents considered in this essay include phycocyanobilin, N-acetylcysteine, lipoic acid, ferulic acid, zinc, selenium, ubiquinol, astaxanthin, melatonin, tauroursodeoxycholic acid, berberine, citrulline, high-dose folate, cocoa flavanols, hawthorn extract, dietary nitrate, high-dose biotin, soy isoflavones, taurine, carnitine, magnesium orotate, EPA-rich fish oil, glycine, and copper. The potential advantages of whole-food plant-based diets, moderation in salt intake, avoidance of phosphate additives, and regular exercise training and sauna sessions are also discussed. There should be considerable scope for the development of functional foods and supplements which make it more convenient and affordable for patients to consume complementary combinations of the agents discussed here. Research Strategy: Key word searching of PubMed was employed to locate the research papers whose findings are cited in this essay.
Collapse
Affiliation(s)
- Mark F McCarty
- Catalytic Longevity Foundation, 811 B Nahant Ct., San Diego, CA 92109, USA
| |
Collapse
|
16
|
Lázár Z, Mészáros M, Bikov A. The Nitric Oxide Pathway in Pulmonary Arterial Hypertension: Pathomechanism, Biomarkers and Drug Targets. Curr Med Chem 2021; 27:7168-7188. [PMID: 32442078 DOI: 10.2174/0929867327666200522215047] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/03/2020] [Accepted: 02/20/2020] [Indexed: 11/22/2022]
Abstract
The altered Nitric Oxide (NO) pathway in the pulmonary endothelium leads to increased vascular smooth muscle tone and vascular remodelling, and thus contributes to the development and progression of pulmonary arterial hypertension (PAH). The pulmonary NO signalling is abrogated by the decreased expression and dysfunction of the endothelial NO synthase (eNOS) and the accumulation of factors blocking eNOS functionality. The NO deficiency of the pulmonary vasculature can be assessed by detecting nitric oxide in the exhaled breath or measuring the degradation products of NO (nitrite, nitrate, S-nitrosothiol) in blood or urine. These non-invasive biomarkers might show the potential to correlate with changes in pulmonary haemodynamics and predict response to therapies. Current pharmacological therapies aim to stimulate pulmonary NO signalling by suppressing the degradation of NO (phosphodiesterase- 5 inhibitors) or increasing the formation of the endothelial cyclic guanosine monophosphate, which mediates the downstream effects of the pathway (soluble guanylate cyclase sensitizers). Recent data support that nitrite compounds and dietary supplements rich in nitrate might increase pulmonary NO availability and lessen vascular resistance. This review summarizes current knowledge on the involvement of the NO pathway in the pathomechanism of PAH, explores novel and easy-to-detect biomarkers of the pulmonary NO.
Collapse
Affiliation(s)
- Zsófia Lázár
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Martina Mészáros
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Andras Bikov
- Department of Pulmonology, Semmelweis University, Budapest, Hungary,Manchester University NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
17
|
Bhatia V, Elnagary L, Dakshinamurti S. Tracing the path of inhaled nitric oxide: Biological consequences of protein nitrosylation. Pediatr Pulmonol 2021; 56:525-538. [PMID: 33289321 DOI: 10.1002/ppul.25201] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/28/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO) is a comprehensive regulator of vascular and airway tone. Endogenous NO produced by nitric oxide synthases regulates multiple signaling cascades, including activation of soluble guanylate cyclase to generate cGMP, relaxing smooth muscle cells. Inhaled NO is an established therapy for pulmonary hypertension in neonates, and has been recently proposed for the treatment of hypoxic respiratory failure and acute respiratory distress syndrome due to COVID-19. In this review, we summarize the effects of endogenous and exogenous NO on protein S-nitrosylation, which is the selective and reversible covalent attachment of a nitrogen monoxide group to the thiol side chain of cysteine. This posttranslational modification targets specific cysteines based on the acid/base sequence of surrounding residues, with significant impacts on protein interactions and function. S-nitrosothiol (SNO) formation is tightly compartmentalized and enzymatically controlled, but also propagated by nonenzymatic transnitrosylation of downstream protein targets. Redox-based nitrosylation and denitrosylation pathways dynamically regulate the equilibrium of SNO-proteins. We review the physiological roles of SNO proteins, including nitrosohemoglobin and autoregulation of blood flow through hypoxic vasodilation, and pathological effects of nitrosylation including inhibition of critical vasodilator enzymes; and discuss the intersection of NO source and dose with redox environment, in determining the effects of protein nitrosylation.
Collapse
Affiliation(s)
- Vikram Bhatia
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Lara Elnagary
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Shyamala Dakshinamurti
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada.,Section of Neonatology, Departments of Pediatrics and Physiology, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
18
|
Infante T, Costa D, Napoli C. Novel Insights Regarding Nitric Oxide and Cardiovascular Diseases. Angiology 2021; 72:411-425. [PMID: 33478246 DOI: 10.1177/0003319720979243] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO) is a powerful mediator with biological activities such as vasodilation and prevention of vascular smooth muscle cell proliferation as well as functional regulation of cardiac cells. Thus, impaired production or reduced bioavailability of NO predisposes to the onset of different cardiovascular (CV) diseases. Alterations in the redox balance associated with excitation-contraction coupling have been identified in heart failure (HF), thus contributing to contractile abnormalities and arrhythmias. For its ability to influence cell proliferation and angiogenesis, NO may be considered a therapeutic option for the management of several CV diseases. Several clinical studies and trials investigated therapeutic NO strategies for systemic hypertension, atherosclerosis, and/or prevention of in stent restenosis, coronary heart disease (CHD), pulmonary arterial hypertension (PAH), and HF, although with mixed results in long-term treatment and effective dose administered in selected groups of patients. Tadalafil, sildenafil, and cinaguat were evaluated for the treatment of PAH, whereas vericiguat was investigated in the treatment of HF patients with reduced ejection fraction. Furthermore, supplementation with hydrogen sulfide, tetrahydrobiopterin, and nitrite/nitrate has shown beneficial effects at the vascular level.
Collapse
Affiliation(s)
- Teresa Infante
- Department of Advanced Clinical and Surgical Sciences, 18994University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Dario Costa
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, 18994University of Campania "L. Vanvitelli," Naples, Italy
| | - Claudio Napoli
- Department of Advanced Clinical and Surgical Sciences, 18994University of Campania "Luigi Vanvitelli," Naples, Italy.,IRCCS SDN, Naples, Italy
| |
Collapse
|
19
|
Si R, Zhang Q, Cabrera JTO, Zheng Q, Tsuji-Hosokawa A, Watanabe M, Hosokawa S, Xiong M, Jain PP, Ashton AW, Yuan JXJ, Wang J, Makino A. Chronic Hypoxia Decreases Endothelial Connexin 40, Attenuates Endothelium-Dependent Hyperpolarization-Mediated Relaxation in Small Distal Pulmonary Arteries, and Leads to Pulmonary Hypertension. J Am Heart Assoc 2020; 9:e018327. [PMID: 33307937 PMCID: PMC7955394 DOI: 10.1161/jaha.120.018327] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/02/2020] [Indexed: 11/17/2022]
Abstract
Background Abnormal endothelial function in the lungs is implicated in the development of pulmonary hypertension; however, there is little information about the difference of endothelial function between small distal pulmonary artery (PA) and large proximal PA and their contribution to the development of pulmonary hypertension. Herein, we investigate endothelium-dependent relaxation in different orders of PAs and examine the molecular mechanisms by which chronic hypoxia attenuates endothelium-dependent pulmonary vasodilation, leading to pulmonary hypertension. Methods and Results Endothelium-dependent relaxation in large proximal PAs (second order) was primarily caused by releasing NO from the endothelium, whereas endothelium-dependent hyperpolarization (EDH)-mediated vasodilation was prominent in small distal PAs (fourth-fifth order). Chronic hypoxia abolished EDH-mediated relaxation in small distal PAs without affecting smooth muscle-dependent relaxation. RNA-sequencing data revealed that, among genes related to EDH, the levels of Cx37, Cx40, Cx43, and IK were altered in mouse pulmonary endothelial cells isolated from chronically hypoxic mice in comparison to mouse pulmonary endothelial cells from normoxic control mice. The protein levels were significantly lower for connexin 40 (Cx40) and higher for connexin 37 in mouse pulmonary endothelial cells from hypoxic mice than normoxic mice. Cx40 knockout mice exhibited significant attenuation of EDH-mediated relaxation and marked increase in right ventricular systolic pressure. Interestingly, chronic hypoxia led to a further increase in right ventricular systolic pressure in Cx40 knockout mice without altering EDH-mediated relaxation. Furthermore, overexpression of Cx40 significantly decreased right ventricular systolic pressure in chronically hypoxic mice. Conclusions These data suggest that chronic hypoxia-induced downregulation of endothelial Cx40 results in impaired EDH-mediated relaxation in small distal PAs and contributes to the development of pulmonary hypertension.
Collapse
Affiliation(s)
- Rui Si
- Department of Physiology The University of Arizona Tucson AZ
| | - Qian Zhang
- Department of Physiology The University of Arizona Tucson AZ
- State Key Laboratory of Respiratory Disease The First Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | | | - Qiuyu Zheng
- Department of Medicine University of California San Diego CA
- State Key Laboratory of Respiratory Disease The First Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | | | - Makiko Watanabe
- Department of Physiology The University of Arizona Tucson AZ
| | | | - Mingmei Xiong
- Department of Medicine University of California San Diego CA
- State Key Laboratory of Respiratory Disease The First Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Pritesh P Jain
- Department of Medicine University of California San Diego CA
| | - Anthony W Ashton
- Division of Perinatal Research Kolling Institute of Medical ResearchUniversity of Sydney New South Wales Australia
| | - Jason X-J Yuan
- Department of Medicine The University of Arizona Tucson AZ
- Department of Medicine University of California San Diego CA
| | - Jian Wang
- Department of Medicine The University of Arizona Tucson AZ
- Department of Medicine University of California San Diego CA
- State Key Laboratory of Respiratory Disease The First Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Ayako Makino
- Department of Physiology The University of Arizona Tucson AZ
- Department of Medicine The University of Arizona Tucson AZ
- Department of Medicine University of California San Diego CA
| |
Collapse
|
20
|
Cai ZL, Liu C, Yao Q, Xie QW, Hu TT, Wu QQ, Tang QZ. The pro-migration and anti-apoptosis effects of HMGA2 in HUVECs stimulated by hypoxia. Cell Cycle 2020; 19:3534-3545. [PMID: 33315504 DOI: 10.1080/15384101.2020.1850970] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
High-mobility group AT-hook2 (HMGA2), serving as an architectural transcription factor, participates in plenty of biological processes. Our study is aimed at illustrating the effect of HMGA2 on hypoxia-induced HUVEC injury and the underlying mechanism. To induce hypoxia-related cell injury, HUVECs were exposed to hypoxic condition for 12-24 h. Molecular expression was determined by Western blot analysis, real-time PCR and immunofluorescence staining. Cell migration was monitored by wound healing assay and Transwell chamber assay. Cell proliferation and apoptosis were measured by MTT assay kits and TUNEL staining. In this study, we discovered that HMGA2 was upregulated in hypoxia-induced HUVECs. Overexpression of HMGA2 promoted cell migration, decreased the apoptosis ratio in response to hypoxia stimulation, while HMGA2 knockdown inhibited cell migration and accelerated apoptosis in HUVECs under hypoxic condition. Mechanistically, we found that HMGA2 induced increased expression of HIF-1α,VEGF, eNOS and AKT. eNOS knockdown significantly reduced HMGA2-mediated pro-migration effects, and AKT knockdown strikingly counteracted HMGA2-mediated anti-apoptotic effect. Hence, our data indicated that HMGA2 promoted cell migration by regulating HIF-1α/VGEF/eNOS signaling and prevented cell apoptosis by activating HIF-1α/VGEF/AKT signaling in HUVECs.
Collapse
Affiliation(s)
- Zhu-Lan Cai
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| | - Chen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| | - Qi Yao
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| | - Qing-We Xie
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| | - Tong-Tong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| | - Qing-Qing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| |
Collapse
|
21
|
Systematic analysis of molecular mechanism of resveratrol for treating pulmonary hypertension based on network pharmacology technology. Eur J Pharmacol 2020; 888:173466. [DOI: 10.1016/j.ejphar.2020.173466] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 12/21/2022]
|
22
|
Pu X, Lin X, Duan X, Wang J, Shang J, Yun H, Chen Z. Oxidative and Endoplasmic Reticulum Stress Responses to Chronic High-Altitude Exposure During the Development of High-Altitude Pulmonary Hypertension. High Alt Med Biol 2020; 21:378-387. [DOI: 10.1089/ham.2019.0143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Xiaoyan Pu
- School of Life Science, Qinghai Normal University, Xining, China
- Medical College, Qinghai University, Xining, China
| | - Xue Lin
- Medical College, Qinghai University, Xining, China
| | - Xianglan Duan
- School of Life Science, Qinghai Normal University, Xining, China
| | - Junjie Wang
- School of Life Science, Qinghai Normal University, Xining, China
| | - Jun Shang
- School of Life Science, Qinghai Normal University, Xining, China
| | - Haixia Yun
- School of Life Science, Qinghai Normal University, Xining, China
| | - Zhi Chen
- School of Life Science, Qinghai Normal University, Xining, China
| |
Collapse
|
23
|
Oliveira TC, Gomes MS, Gomes AC. The Crossroads between Infection and Bone Loss. Microorganisms 2020; 8:microorganisms8111765. [PMID: 33182721 PMCID: PMC7698271 DOI: 10.3390/microorganisms8111765] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 01/18/2023] Open
Abstract
Bone homeostasis, based on a tight balance between bone formation and bone degradation, is affected by infection. On one hand, some invading pathogens are capable of directly colonizing the bone, leading to its destruction. On the other hand, immune mediators produced in response to infection may dysregulate the deposition of mineral matrix by osteoblasts and/or the resorption of bone by osteoclasts. Therefore, bone loss pathologies may develop in response to infection, and their detection and treatment are challenging. Possible biomarkers of impaired bone metabolism during chronic infection need to be identified to improve the diagnosis and management of infection-associated osteopenia. Further understanding of the impact of infections on bone metabolism is imperative for the early detection, prevention, and/or reversion of bone loss. Here, we review the mechanisms responsible for bone loss as a direct and/or indirect consequence of infection.
Collapse
Affiliation(s)
- Tiago Carvalho Oliveira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.C.O.); (M.S.G.)
- Faculdade de Ciências da Universidade do Porto, 4169-007 Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar da Universidade do Porto, 4050-313 Porto, Portugal
| | - Maria Salomé Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.C.O.); (M.S.G.)
- Instituto de Ciências Biomédicas de Abel Salazar da Universidade do Porto, 4050-313 Porto, Portugal
| | - Ana Cordeiro Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.C.O.); (M.S.G.)
- Correspondence:
| |
Collapse
|
24
|
Tao H, Ge G, Liang X, Zhang W, Sun H, Li M, Geng D. ROS signaling cascades: dual regulations for osteoclast and osteoblast. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1055-1062. [PMID: 33085739 DOI: 10.1093/abbs/gmaa098] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/16/2020] [Accepted: 07/30/2020] [Indexed: 02/04/2023] Open
Abstract
Accumulating evidence indicates that intracellular reactive oxygen species (ROS) production is highly involved in bone homeostasis by intervening osteoclast or osteoblast differentiation. Interestingly, ROS that are known as oxidizing agents exert dose-dependent biphasic properties in bone remodeling, including preventing osteoblast activity but accelerating osteoclast resorption. ROS mainly composed of superoxide anion radical, hydroxyl radical, nitric oxide, and two-electron reduction product hydrogen peroxide, which are important components to regulate bone cell metabolism and function in mammal skeleton. These free radicals can be partly produced in bone and boosted in an inflammation state. Although numerous researches have emphasized the impacts of ROS on bone cell biology and verified the mechanism of ROS signaling cascades, the recapitulatory commentary is necessary. In this review article, we particularly focus on the regulation of the intracellular ROS and its potential mechanism impacting on cell-signaling transduction in osteoclast and osteoblast differentiation for preferable understanding the pathogenesis and searching for novel therapeutic protocols for human bone diseases.
Collapse
Affiliation(s)
- Huaqiang Tao
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
| | - Gaoran Ge
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
| | - Xiaolong Liang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
| | - Weicheng Zhang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
| | - Houyi Sun
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
| | - Meng Li
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
- Division of Life Sciences and Medicine, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230000, China
| | - Dechun Geng
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
| |
Collapse
|
25
|
Ye W, Guo H, Xu J, Cai S, He Y, Shui X, Huang S, Luo H, Lei W. Heart‑lung crosstalk in pulmonary arterial hypertension following myocardial infarction (Review). Int J Mol Med 2020; 46:913-924. [PMID: 32582962 PMCID: PMC7388838 DOI: 10.3892/ijmm.2020.4650] [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: 01/26/2020] [Accepted: 05/04/2020] [Indexed: 11/20/2022] Open
Abstract
Left heart disease is the main cause of clinical pulmonary arterial hypertension (PAH). Common types of left heart disease that result in PAH include heart failure, left ventricular systolic dysfunction, left ventricular diastolic dysfunction and valvular disease. It is currently believed that mechanical pressure caused by high pulmonary venous pressure is the main cause of myocardial infarction (MI) in individuals with ischemic cardiomyopathy and left ventricular systolic dysfunction. In the presence of decreased cardiac function, vascular remodeling of pulmonary vessels in response to long-term stimulation by high pressure in turn leads to exacerbation of PAH. However, the underlying pathological mechanisms remain unclear. Elucidating the association between the development of MI and PAH may lead to a better understanding of potential risk factors and better disease treatment. In this article, the pathophysiological effects of multiple systems in individuals with MI and PAH were reviewed in order to provide a general perspective on various potential interactions between cardiomyocytes and pulmonary vascular cells.
Collapse
Affiliation(s)
- Wenfeng Ye
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Haixu Guo
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Jinrong Xu
- Department of Cardiovascular Internal Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Shuyun Cai
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yuan He
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Xiaorong Shui
- Laboratory of Vascular Surgery, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Shian Huang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory‑Zhanjiang, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Wei Lei
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
26
|
Novel Molecular Mechanisms of Pulmonary Hypertension: A Search for Biomarkers and Novel Drug Targets-From Bench to Bed Site. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7265487. [PMID: 32566097 PMCID: PMC7261339 DOI: 10.1155/2020/7265487] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/11/2020] [Indexed: 12/23/2022]
Abstract
Pulmonary hypertension (PH) is defined as increased mean pulmonary artery pressure (mPAP) above 25 mmHg, measured at rest by right heart catheterization. The exact global prevalence of PH is difficult to estimate, mainly due to the complex aetiology, and its spread may be underestimated. To date, numerous studies on the aetiology and pathophysiology of PH at molecular level were conducted. Simultaneously, some clinical studies have shown potential usefulness of well-known and widely recognized cardiovascular biomarkers, but their potential clinical usefulness in diagnosis and management of PH is poor due to their low specificity accompanied with numerous other cardiovascular comorbidities of PH subjects. On the other hand, a large body of basic research-based studies provides us with novel molecular pathomechanisms, biomarkers, and drug targets, according to the evidence-based medicine principles. Unfortunately, the simple implementation of these results to clinical practice is impossible due to a large heterogeneity of the PH pathophysiology, where the clinical symptoms constitute only a common denominator and a final result of numerous crosstalking metabolic pathways. Therefore, future studies, based mostly on translational medicine, are needed in order to both organize better the pathophysiological classification of various forms of PH and define precisely the optimal diagnostic markers and therapeutic targets in particular forms of PH. This review paper summarizes the current state of the art regarding the molecular background of PH with respect to its current classification. Novel therapeutic strategies and potential biomarkers are discussed with respect to their limitations in use in common clinical practice.
Collapse
|
27
|
New Insights into the Implication of Mitochondrial Dysfunction in Tissue, Peripheral Blood Mononuclear Cells, and Platelets during Lung Diseases. J Clin Med 2020; 9:jcm9051253. [PMID: 32357474 PMCID: PMC7287602 DOI: 10.3390/jcm9051253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022] Open
Abstract
Lung diseases such as chronic obstructive pulmonary disease, asthma, pulmonary arterial hypertension, or idiopathic pulmonary fibrosis are major causes of morbidity and mortality. Complex, their physiopathology is multifactorial and includes lung mitochondrial dysfunction and enhanced reactive oxygen species (ROS) release, which deserves increased attention. Further, and importantly, circulating blood cells (peripheral blood mononuclear cells-(PBMCs) and platelets) likely participate in these systemic diseases. This review presents the data published so far and shows that circulating blood cells mitochondrial oxidative capacity are likely to be reduced in chronic obstructive pulmonary disease (COPD), but enhanced in asthma and pulmonary arterial hypertension in a context of increased oxidative stress. Besides such PBMCs or platelets bioenergetics modifications, mitochondrial DNA (mtDNA) changes have also been observed in patients. These new insights open exciting challenges to determine their role as biomarkers or potential guide to a new therapeutic approach in lung diseases.
Collapse
|
28
|
Kandhi S, Alruwaili N, Wolin MS, Sun D, Huang A. Reciprocal actions of constrictor prostanoids and superoxide in chronic hypoxia-induced pulmonary hypertension: roles of EETs. Pulm Circ 2019; 9:2045894019895947. [PMID: 31908769 DOI: 10.1177/2045894019895947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 11/25/2019] [Indexed: 12/11/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are synthesized from arachidonic acid by CYP/epoxygenase and metabolized by soluble epoxide hydrolase (sEH). Roles of EETs in hypoxia-induced pulmonary hypertension (HPH) remain elusive. The present study aimed to investigate the underlying mechanisms, by which EETs potentiate HPH. Experiments were conducted on sEH knockout (sEH-KO) and wild type (WT) mice after exposure to hypoxia (10% oxygen) for three weeks. In normal/normoxic conditions, WT and sEH-KO mice exhibited comparable pulmonary artery acceleration time (PAAT), ejection time (ET), PAAT/ET ratio, and velocity time integral (VTI), along with similar right ventricular systolic pressure (RVSP). Chronic hypoxia significantly reduced PAAT, ET, and VTI, coincided with an increase in RVSP; these impairments were more severe in sEH-KO than WT mice. Hypoxia elicited downregulation of sEH and upregulation of CYP2C9 accompanied with elevation of CYP-sourced superoxide, leading to enhanced pulmonary EETs in hypoxic mice with significantly higher levels in sEH-KO mice. Isometric tension of isolated pulmonary arteries was recorded. In addition to downregulation of eNOS-induced impairment of vasorelaxation to ACh, HPH mice displayed upregulation of thromboxane A2 (TXA2) receptor, paralleled with enhanced pulmonary vasocontraction to a TXA2 analog (U46619) in an sEH-KO predominant manner. Inhibition of COX-1 or COX-2 significantly prevented the enhancement by ∼50% in both groups of vessels, and the remaining incremental components were eliminated by scavenging of superoxide with Tiron. In conclusion, hypoxia-driven increases in EETs, intensified COXs/TXA2 signaling, great superoxide sourced from activated CYP2C9, and impaired NO bioavailability work in concert, to potentiate HPH development.
Collapse
Affiliation(s)
- Sharath Kandhi
- Departments of Physiology, New York Medical College, Valhalla, NY, USA
| | - Norah Alruwaili
- Departments of Physiology, New York Medical College, Valhalla, NY, USA
| | - Michael S Wolin
- Departments of Physiology, New York Medical College, Valhalla, NY, USA
| | - Dong Sun
- Departments of Physiology, New York Medical College, Valhalla, NY, USA
| | - An Huang
- Departments of Physiology, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
29
|
Rood K, Lopez V, La Frano MR, Fiehn O, Zhang L, Blood AB, Wilson SM. Gestational Hypoxia and Programing of Lung Metabolism. Front Physiol 2019; 10:1453. [PMID: 31849704 PMCID: PMC6895135 DOI: 10.3389/fphys.2019.01453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/11/2019] [Indexed: 12/12/2022] Open
Abstract
Gestational hypoxia is a risk factor in the development of pulmonary hypertension in the newborn and other sequela, however, the mechanisms associated with the disease remain poorly understood. This review highlights disruption of metabolism by antenatal high altitude hypoxia and the impact this has on pulmonary hypertension in the newborn with discussion of model organisms and human populations. There is particular emphasis on modifications in glucose and lipid metabolism along with alterations in mitochondrial function. Additional focus is placed on increases in oxidative stress and the progression of pulmonary vascular disease in the newborn and on the need for further exploration using a combination of contemporary and classical approaches.
Collapse
Affiliation(s)
- Kristiana Rood
- Lawrence D. Longo MD Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Vanessa Lopez
- Lawrence D. Longo MD Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Michael R La Frano
- Department of Food Science and Nutrition, Center for Health Research, California Polytechnic State University, San Luis Obispo, CA, United States.,Center for Health Research, California Polytechnic State University, San Luis Obispo, CA, United States
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, University of California, Davis, Davis, CA, United States.,Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, United States
| | - Lubo Zhang
- Lawrence D. Longo MD Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Arlin B Blood
- Lawrence D. Longo MD Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Sean M Wilson
- Lawrence D. Longo MD Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| |
Collapse
|
30
|
Žaloudíková M, Vytášek R, Rašková M, Vízek M, Uhlík J, Hampl V. The effect of exposure to hypoxia on superoxide formation by alveolar macrophages is indirect. Life Sci 2019; 236:116864. [PMID: 31518607 DOI: 10.1016/j.lfs.2019.116864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/05/2019] [Accepted: 09/09/2019] [Indexed: 11/30/2022]
Abstract
AIMS To elucidate the role of alveolar macrophages (AM) in the pathogenesis of hypoxic pulmonary hypertension (HPH), we tested the effects of sustained hypoxia on AM polarization and on the formation of superoxide by AM in vivo and in vitro. MAIN METHODS Rat AM were obtained by bronchoalveolar lavage. 4-day exposure to hypoxia (10% O2) was carried out in vivo (rats in isobaric hypoxic chamber, controls kept in air) or in vitro (control AM in 21% O2 and 5% CO2). Superoxide production was measured by luminol-orthovanadate chemiluminescence, AM polarization was detected immunocytochemically. To ascertain the effect of substances contained in the alveolar environment, we cultivated cells also in the presence of non-cellular components of the bronchoalveolar lavage fluid (BALF) either from controls or from rats exposed to 4 days of hypoxia. KEY FINDINGS In vivo, but not in vitro, hypoxia increased AM superoxide production. Both types of hypoxia polarized AM into M2 (pro-proliferative) type. While the presence of control BALF attenuated superoxide production in AM cultivated in normoxia, BALF from the hypoxia-exposed rats had no effect. In AM cultivated in hypoxia, superoxide production was not altered by control BALF and elevated by BALF obtained from hypoxic rats. SIGNIFICANCE Hypoxia does not influence superoxide production by AM directly but rather by modulating their milieu and their sensitivity to external influences.
Collapse
Affiliation(s)
- Marie Žaloudíková
- Department of Physiology, Second Faculty of Medicine, Charles University, Czech Republic.
| | - Richard Vytášek
- Department of Physiology, Second Faculty of Medicine, Charles University, Czech Republic
| | - Marcela Rašková
- Department of Physiology, Second Faculty of Medicine, Charles University, Czech Republic
| | - Martin Vízek
- Department of Pathophysiology, Second Faculty of Medicine, Charles University, Czech Republic
| | - Jiří Uhlík
- Department of Histology and Embryology, Second Faculty of Medicine, Charles University, Czech Republic
| | - Václav Hampl
- Department of Physiology, Second Faculty of Medicine, Charles University, Czech Republic
| |
Collapse
|
31
|
Zhao RZ, Jiang S, Zhang L, Yu ZB. Mitochondrial electron transport chain, ROS generation and uncoupling (Review). Int J Mol Med 2019; 44:3-15. [PMID: 31115493 PMCID: PMC6559295 DOI: 10.3892/ijmm.2019.4188] [Citation(s) in RCA: 523] [Impact Index Per Article: 87.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 04/19/2019] [Indexed: 12/18/2022] Open
Abstract
The mammalian mitochondrial electron transport chain (ETC) includes complexes I-IV, as well as the electron transporters ubiquinone and cytochrome c. There are two electron transport pathways in the ETC: Complex I/III/IV, with NADH as the substrate and complex II/III/IV, with succinic acid as the substrate. The electron flow is coupled with the generation of a proton gradient across the inner membrane and the energy accumulated in the proton gradient is used by complex V (ATP synthase) to produce ATP. The first part of this review briefly introduces the structure and function of complexes I-IV and ATP synthase, including the specific electron transfer process in each complex. Some electrons are directly transferred to O2 to generate reactive oxygen species (ROS) in the ETC. The second part of this review discusses the sites of ROS generation in each ETC complex, including sites IF and IQ in complex I, site IIF in complex II and site IIIQo in complex III, and the physiological and pathological regulation of ROS. As signaling molecules, ROS play an important role in cell proliferation, hypoxia adaptation and cell fate determination, but excessive ROS can cause irreversible cell damage and even cell death. The occurrence and development of a number of diseases are closely related to ROS overproduction. Finally, proton leak and uncoupling proteins (UCPS) are discussed. Proton leak consists of basal proton leak and induced proton leak. Induced proton leak is precisely regulated and induced by UCPs. A total of five UCPs (UCP1-5) have been identified in mammalian cells. UCP1 mainly plays a role in the maintenance of body temperature in a cold environment through non-shivering thermogenesis. The core role of UCP2-5 is to reduce oxidative stress under certain conditions, therefore exerting cytoprotective effects. All diseases involving oxidative stress are associated with UCPs.
Collapse
Affiliation(s)
- Ru-Zhou Zhao
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Shuai Jiang
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lin Zhang
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zhi-Bin Yu
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
32
|
New Nitric Oxide Donor NCX 1443: Therapeutic Effects on Pulmonary Hypertension in the SAD Mouse Model of Sickle Cell Disease. J Cardiovasc Pharmacol 2019; 71:283-292. [PMID: 29438213 DOI: 10.1097/fjc.0000000000000570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Nitric oxide (NO) donors may be useful for treating pulmonary hypertension (PH) complicating sickle cell disease (SCD), as endogenous NO is inactivated by hemoglobin released by intravascular hemolysis. Here, we investigated the effects of the new NO donor NCX1443 on PH in transgenic SAD mice, which exhibit mild SCD without severe hemolytic anemia. In SAD and wild-type (WT) mice, the pulmonary pressure response to acute hypoxia was similar and was abolished by 100 mg/kg NCX1443. The level of PH was also similar in SAD and WT mice exposed to chronic hypoxia (9% O2) alone or with SU5416 and was similarly reduced by daily NCX1443 gavage. Compared with WT mice, SAD mice exhibited higher levels of HO-1, endothelial NO synthase, and PDE5 but similar levels of lung cyclic guanosine monophosphate. Cultured pulmonary artery smooth muscle cells from SAD mice grew faster than those from WT mice and had higher PDE5 protein levels. Combining NCX1443 and a PDE5 inhibitor suppressed the growth rate difference between SAD and WT cells and induced a larger reduction in hypoxic PH severity in SAD than in WT mice. By amplifying endogenous protective mechanisms, NCX1443 in combination with PDE5 inhibition may prove useful for treating PH complicating SCD.
Collapse
|
33
|
Adi SD, Eiza N, Bejar J, Shefer H, Toledano S, Kessler O, Neufeld G, Toubi E, Vadasz Z. Semaphorin 3A Is Effective in Reducing Both Inflammation and Angiogenesis in a Mouse Model of Bronchial Asthma. Front Immunol 2019; 10:550. [PMID: 30967873 PMCID: PMC6439418 DOI: 10.3389/fimmu.2019.00550] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/28/2019] [Indexed: 01/15/2023] Open
Abstract
Semaphorin 3A (sema3A) belongs to the sub-family of the immune semaphorins that function as regulators of immune-mediated inflammation. Sema3A is a membrane associated molecule on T regulatory cells and on B regulatory cells. Being transiently ligated to the cell surface of these cells it is suggested to be a useful marker for evaluating their functional status. In earlier studies, we found that reduced sema3A concentration in the serum of asthma patients as well as reduced expression by Treg cells correlates with asthma disease severity. Stimulation of Treg cells with recombinant sema3A induced a significant increase in FoxP3 and IL-10 expression. To find out if sema3A can be of benefit to asthma patients, we evaluated the effect of sema3A injection in a mouse model of asthma. BALB\c-mice were sensitized using ovalbumin (OVA) + adjuvant for 15 days followed by OVA aerosol inhalation over five consecutive days. Four hours following air ways sensitization on each of the above days- 15 of these mice were injected intraperitoneally with 50 μg per mouse of recombinant human sema3A-FR and the remaining 15 mice were injected with a similarly purified vehicle. Five days later the mice were sacrificed, broncheo-alveolar lavage (BAL) was collected and formalin-fixed lung biopsies taken and analyzed. In sema3A treated mice, only 20% of the bronchioles and arterioles were infiltrated by inflammatory cells as compared to 90% in the control group (p = 0.0079). In addition, eosinophil infiltration was also significantly increased in the control group as compared with the sema3A treated mice. In sema3A treated mice we noticed only a small number of mononuclear and neutrophil cells in the BAL while in the control mice, the BAL was enriched with mononuclear and neutrophil cells. Finally, in the control mice, angiogenesis was significantly increased in comparison with sema3A treated mice as evidenced by the reduced concentration of microvessels in the lungs of sema3A treated mice. To conclude, we find that in this asthma model, sema3A functions as a potent suppressor of asthma related inflammation that has the potential to be further developed as a new therapeutic for the treatment of asthma.
Collapse
Affiliation(s)
- Sabag D Adi
- Proteomic Unit, The Division of Clinical Immunology and Allergy, Bnai-Zion Medical Center, Haifa, Israel
| | - Nasren Eiza
- Proteomic Unit, The Division of Clinical Immunology and Allergy, Bnai-Zion Medical Center, Haifa, Israel
| | - Jacob Bejar
- The Department of Pathology, Faculty of Medicine, Bnai-Zion Medical Center, Haifa, Israel
| | - Hila Shefer
- The Department of Pathology, Faculty of Medicine, Bnai-Zion Medical Center, Haifa, Israel
| | - Shira Toledano
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Ofra Kessler
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Gera Neufeld
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Elias Toubi
- Proteomic Unit, The Division of Clinical Immunology and Allergy, Bnai-Zion Medical Center, Haifa, Israel
| | - Zahava Vadasz
- Proteomic Unit, The Division of Clinical Immunology and Allergy, Bnai-Zion Medical Center, Haifa, Israel
| |
Collapse
|
34
|
Hua C, Zhao J, Wang H, Chen F, Meng H, Chen L, Zhang Q, Yan J, Yuan L. Apple polyphenol relieves hypoxia-induced pulmonary arterial hypertension via pulmonary endothelium protection and smooth muscle relaxation: In vivo and in vitro studies. Biomed Pharmacother 2018; 107:937-944. [DOI: 10.1016/j.biopha.2018.08.080] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/15/2018] [Accepted: 08/15/2018] [Indexed: 12/25/2022] Open
|
35
|
Hao S, Jiang L, Fu C, Wu X, Liu Z, Song J, Lu H, Wu X, Li S. 2-Methoxyestradiol attenuates chronic-intermittent-hypoxia-induced pulmonary hypertension through regulating microRNA-223. J Cell Physiol 2018; 234:6324-6335. [PMID: 30246291 DOI: 10.1002/jcp.27363] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/17/2018] [Indexed: 12/12/2022]
Abstract
Pulmonary hypertension (PH) is prevalent in patients with obstructive sleep apnea (OSA) syndrome, and coexistence of PH and OSA indicates a worse prognosis and higher mortality. Chronic intermittent hypoxia (CIH) is the key pathogenesis of OSA. Also, microRNA-223 (miR-223) plays a role in the regulation of CIH-induced PH process. However, the detailed mechanism of CIH inducing PH is still unclear. This study aimed to investigate the pathological process of CIH associated PH and explore the potential therapeutic methods. In this study, adult Sprague-Dawley rats were exposed to CIH or normoxic (N) conditions with 2-methoxyestradiol (2-Me) or vehicle treatment for 6 weeks. The results showed that 2-Me treatment reduced the progression of pulmonary angiogenesis in CIH rats, and alleviated proliferation, cellular migration, and reactive oxygen species formation was induced by CIH in pulmonary artery smooth muscle cells (PASMCs). CIH decreased the expression of miR-223, whereas 2-Me reversed the downregulation of miR-223 both in vivo and in vitro. Furthermore, the antiangiogenic effect of 2-Me observed in PASMCs was abrogated by miR-223 inhibitor, while enhanced by miR-223 mimic. These findings suggested that miR-223 played an important role in the process of CIH inducing PH, and 2-Me might reverse CIH-induced PH via upregulating miR-223.
Collapse
Affiliation(s)
- Shengyu Hao
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Clinical Centre for Sleep Breathing Disorder and Snoring, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liyan Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cuiping Fu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Clinical Centre for Sleep Breathing Disorder and Snoring, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xu Wu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zilong Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jieqiong Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huan Lu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Clinical Centre for Sleep Breathing Disorder and Snoring, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaodan Wu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Clinical Centre for Sleep Breathing Disorder and Snoring, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shanqun Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Clinical Centre for Sleep Breathing Disorder and Snoring, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Heyman SN, Abassi Z, Rosenberger C, Yaseen H, Skarjinski G, Shina A, Mathia S, Krits N, Khamaisi M. Cyclosporine A induces endothelin-converting enzyme-1: Studies in vivo and in vitro. Acta Physiol (Oxf) 2018; 223:e13033. [PMID: 29330945 DOI: 10.1111/apha.13033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 01/04/2018] [Accepted: 01/08/2018] [Indexed: 11/27/2022]
Abstract
AIM Cyclosporine A (CsA) induces renal vasoconstriction and hypoxia and enhances the expression of endothelin-1 (ET-1) pro-hormone (pre-pro-ET-1), plausibly leading to a feed-forward loop of renal vasoconstriction, hypoxia and enhanced synthesis of the potent vasoconstrictor ET-1. Endothelin-converting enzyme (ECE)-1 cleaves big endothelin to generate endothelin (ET)-1 and is upregulated by hypoxia via hypoxia-inducible factor (HIF). We hypothesized that in addition to the direct induction of ET-1 synthesis, CsA might also intensify renal ECE-1 expression, thus contributing to enhanced ET-1 synthesis following CsA. METHODS CsA was administered to Sprague Dawley rats (120 mg/kg/SC) for 4 days, and renal HIF and ECE-1 expression were assessed with Western blots and immunostaining. Human umbilical vein endothelial cells (HUVEC) and proximal tubular cell line (HK-2) were subjected to CsA, and ECE-1 induction was evaluated using real-time mRNA PCR and Western blots. RESULTS Cyclosporine A intensified renal parenchymal ECE-1 expression in the rat kidney, particularly in distal nephron segments, along with renal hypoxia (detected by pimonidazole adducts) and HIF expression, in line with our recent observations showing episodic hypoxia in mice subjected to CsA. Furthermore, in cultured normoxic HUVEC and HK-2 cells, CsA dose-dependently induced both pre-pro-ET-1 and ECE-1 mRNA and protein expression, with enhanced ET-1 generation. CONCLUSION CsA induces ECE-1 via both hypoxic and non-hypoxic pathways. ECE-1 may contribute to increased renal ET-1 generation following CsA, participating in a feed-forward loop of renal parenchymal hypoxia and ET synthesis.
Collapse
Affiliation(s)
- S. N. Heyman
- Department of Medicine; Hadassah Hebrew University Hospital; Jerusalem Israel
| | - Z. Abassi
- Department of Physiology, Ruth and Bruce Rappaport Faculty of Medicine; Technion-IIT; Haifa Israel
- Department of Laboratory Medicine; Rambam Health Care Campus; Haifa Israel
| | - C. Rosenberger
- Department of Nephrology and Medical Intensive Care; Charité - Universitätsmedizin; Berlin Germany
| | - H. Yaseen
- Department of Medicine D; Rambam Health Care Campus; Haifa Israel
- Ruth and Bruce Rappaport Faculty of Medicine; Technion-IIT; Haifa Israel
| | - G. Skarjinski
- Department of Medicine; Hadassah Hebrew University Hospital; Jerusalem Israel
| | - A. Shina
- Department of Medicine; Hadassah Hebrew University Hospital; Jerusalem Israel
| | - S. Mathia
- Department of Nephrology and Medical Intensive Care; Charité - Universitätsmedizin; Berlin Germany
| | - N. Krits
- Department of Medicine D; Rambam Health Care Campus; Haifa Israel
- Ruth and Bruce Rappaport Faculty of Medicine; Technion-IIT; Haifa Israel
| | - M. Khamaisi
- Department of Medicine D; Rambam Health Care Campus; Haifa Israel
- Ruth and Bruce Rappaport Faculty of Medicine; Technion-IIT; Haifa Israel
| |
Collapse
|
37
|
Astorga CR, González-Candia A, Candia AA, Figueroa EG, Cañas D, Ebensperger G, Reyes RV, Llanos AJ, Herrera EA. Melatonin Decreases Pulmonary Vascular Remodeling and Oxygen Sensitivity in Pulmonary Hypertensive Newborn Lambs. Front Physiol 2018; 9:185. [PMID: 29559926 PMCID: PMC5845624 DOI: 10.3389/fphys.2018.00185] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/20/2018] [Indexed: 12/16/2022] Open
Abstract
Background: Chronic hypoxia and oxidative stress during gestation lead to pulmonary hypertension of the neonate (PHN), a condition characterized by abnormal pulmonary arterial reactivity and remodeling. Melatonin has strong antioxidant properties and improves pulmonary vascular function. Here, we aimed to study the effects of melatonin on the function and structure of pulmonary arteries from PHN lambs. Methods: Twelve lambs (Ovis aries) gestated and born at highlands (3,600 m) were instrumented with systemic and pulmonary catheters. Six of them were assigned to the control group (CN, oral vehicle) and 6 were treated with melatonin (MN, 1 mg.kg−1.d−1) during 10 days. At the end of treatment, we performed a graded oxygenation protocol to assess cardiopulmonary responses to inspired oxygen variations. Further, we obtained lung and pulmonary trunk samples for histology, molecular biology, and immunohistochemistry determinations. Results: Melatonin reduced the in vivo pulmonary pressor response to oxygenation changes. In addition, melatonin decreased cellular density of the media and diminished the proliferation marker KI67 in resistance vessels and pulmonary trunk (p < 0.05). This was associated with a decreased in the remodeling markers α-actin (CN 1.28 ± 0.18 vs. MN 0.77 ± 0.04, p < 0.05) and smoothelin-B (CN 2.13 ± 0.31 vs. MN 0.88 ± 0.27, p < 0.05). Further, melatonin increased vascular density by 134% and vascular luminal surface by 173% (p < 0.05). Finally, melatonin decreased nitrotyrosine, an oxidative stress marker, in small pulmonary vessels (CN 5.12 ± 0.84 vs. MN 1.14 ± 0.34, p < 0.05). Conclusion: Postnatal administration of melatonin blunts the cardiopulmonary response to hypoxia, reduces the pathological vascular remodeling, and increases angiogenesis in pulmonary hypertensive neonatal lambs.These effects improve the pulmonary vascular structure and function in the neonatal period under chronic hypoxia.
Collapse
Affiliation(s)
- Cristian R Astorga
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Alejandro González-Candia
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Alejandro A Candia
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Department for the Woman and Newborn Health Promotion, Universidad de Chile, Santiago, Chile
| | - Esteban G Figueroa
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Daniel Cañas
- Department of Mechanical Engineering, Faculty of Engineering, Universidad de Santiago de Chile, Santiago, Chile
| | - Germán Ebensperger
- Perinatal Physiology and Pathophysiology Unit, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| | - Roberto V Reyes
- Perinatal Physiology and Pathophysiology Unit, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| | - Aníbal J Llanos
- Perinatal Physiology and Pathophysiology Unit, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| | - Emilio A Herrera
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Perinatal Physiology and Pathophysiology Unit, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| |
Collapse
|