1
|
Kim TW, Ko SG. Anti-Inflammatory and Anticancer Effects of Kaurenoic Acid in Overcoming Radioresistance in Breast Cancer Radiotherapy. Nutrients 2024; 16:4320. [PMID: 39770941 PMCID: PMC11677055 DOI: 10.3390/nu16244320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Peroxisome proliferator-activated receptor γ (PPARγ) plays a key role in mediating anti-inflammatory and anticancer effects in the tumor microenvironment. Kaurenoic acid (KA), a diterpene compound isolated from Sphagneticola trilobata (L.) Pruski, has been demonstrated to exert anti-inflammatory, anticancer, and antihuman immunodeficiency virus effects. Methods: In this study, we identified KA as a novel activator of PPARγ with potent anti-inflammatory and antitumor effects both in vitro and in vivo. Given the potential of PPARγ regulators in overcoming radioresistance and chemoresistance in cancer therapies, we hypothesized that KA may enhance the efficacy of breast cancer radiotherapy. Results: In a lipopolysaccharide (LPS)-induced mouse inflammation model, KA treatment reduced the levels of pro-inflammatory cytokines, including COX-2, IL-6, IL-1β, and TNFα. In a xenograft mouse mode of breast cancer, KA treatment inhibited tumor growth. Specifically, KA treatment enhanced caspase-3 activity and cytotoxicity against MDA-MB-231 and MCF-7 breast cancer cells. When KA was co-treated with a caspase inhibitor, Z-VAD-FMK, caspase-dependent apoptosis was suppressed in these cells. KA was found to induce the generation of cytosolic calcium ions (Ca2+) and reactive oxygen species (ROS), triggering endoplasmic reticulum (ER) stress via the PERK-ATF4-CHOP axis. Hence, the ER stressor thapsigargin (TG) synergized with KA treatment to enhance apoptosis in these cells, while the loss of the PERK or CHOP function inhibited this phenomenon. KA treatment was shown to induce oxidative stress via the NADPH oxidase 4 (NOX4) and stimulate ROS production. Specifically, NOX4 knockdown (KD) and antioxidant treatment (N-acetyl cysteine or diphenyleneiodonium) suppressed such ER stress-mediated apoptosis by inhibiting KA-enhanced caspase-3 activity, cytotoxicity, and intracellular ROS production in the treated cells. In radioresistant MDA-MB-231R and MCF-7R cells, KA combined with 2 Gy radiation overcame radioresistance by upregulating PPARγ and modulating epithelial-mesenchymal transition (EMT) markers, such as E-cadherin, N-cadherin, and vimentin. In PPARγ KD MDA-MB-231R and MCF-7R cells, this phenomenon was inhibited due to reduced PPARγ and NOX4 expression. Conclusions: In conclusion, these findings demonstrated KA as a novel PPARγ regulator with promising potential to enhance the efficacy of breast cancer radiotherapy.
Collapse
Affiliation(s)
- Tae Woo Kim
- Department of Biopharmaceutical Engineering, Dongguk University-WISE, Gyeongju, Gyeongbuk 38066, Republic of Korea
| | - Seong-Gyu Ko
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| |
Collapse
|
2
|
Zhou Y, Zhang X, Baker JS, Davison GW, Yan X. Redox signaling and skeletal muscle adaptation during aerobic exercise. iScience 2024; 27:109643. [PMID: 38650987 PMCID: PMC11033207 DOI: 10.1016/j.isci.2024.109643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Redox regulation is a fundamental physiological phenomenon related to oxygen-dependent metabolism, and skeletal muscle is mainly regarded as a primary site for oxidative phosphorylation. Several studies have revealed the importance of reactive oxygen and nitrogen species (RONS) in the signaling process relating to muscle adaptation during exercise. To date, improving knowledge of redox signaling in modulating exercise adaptation has been the subject of comprehensive work and scientific inquiry. The primary aim of this review is to elucidate the molecular and biochemical pathways aligned to RONS as activators of skeletal muscle adaptation and to further identify the interconnecting mechanisms controlling redox balance. We also discuss the RONS-mediated pathways during the muscle adaptive process, including mitochondrial biogenesis, muscle remodeling, vascular angiogenesis, neuron regeneration, and the role of exogenous antioxidants.
Collapse
Affiliation(s)
- Yingsong Zhou
- Faculty of Sports Science, Ningbo University, Ningbo, China
| | - Xuan Zhang
- School of Wealth Management, Ningbo University of Finance and Economics, Ningbo, China
| | - Julien S. Baker
- Centre for Health and Exercise Science Research, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong
| | - Gareth W. Davison
- Sport and Exercise Sciences Research Institute, Ulster University, Belfast BT15 IED, UK
| | - Xiaojun Yan
- School of Marine Sciences, Ningbo University, Ningbo, China
| |
Collapse
|
3
|
Li S, Lyu Q, Shi Q, Bai Y, Ren X, Ma J. Intermittent short-duration reoxygenation relieves high-altitude pulmonary hypertension via NOX4/H2O2/PPAR-γ axis. Clin Sci (Lond) 2024; 138:103-115. [PMID: 38237016 PMCID: PMC10830432 DOI: 10.1042/cs20231508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024]
Abstract
High-altitude pulmonary hypertension (HAPH) is a severe and progressive disease that can lead to right heart failure. Intermittent short-duration reoxygenation at high altitude is effective in alleviating HAPH; however, the underlying mechanisms are unclear. In the present study, a simulated 5,000-m hypoxia rat model and hypoxic cultured pulmonary artery smooth muscle cells (PASMCs) were used to evaluate the effect and mechanisms of intermittent short-duration reoxygenation. The results showed that intermittent 3-h/per day reoxygenation (I3) effectively attenuated chronic hypoxia-induced pulmonary hypertension and reduced the content of H2O2 and the expression of NADPH oxidase 4 (NOX4) in lung tissues. In combination with I3, while the NOX inhibitor apocynin did not further alleviate HAPH, the mitochondrial antioxidant MitoQ did. Furthermore, in PASMCs, I3 attenuated hypoxia-induced PASMCs proliferation and reversed the activated HIF-1α/NOX4/PPAR-γ axis under hypoxia. Targeting this axis offset the protective effect of I3 on hypoxia-induced PASMCs proliferation. The present study is novel in revealing a new mechanism for preventing HAPH and provides insights into the optimization of intermittent short-duration reoxygenation.
Collapse
Affiliation(s)
- Shaohua Li
- Department of Aerospace Physiology, Air Force Medical University, Xi’an 710032, China
| | - Qiang Lyu
- Department of Nephrology, Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing 100853, China
| | - Qixin Shi
- Department of Aerospace Physiology, Air Force Medical University, Xi’an 710032, China
| | - Yungang Bai
- Department of Aerospace Physiology, Air Force Medical University, Xi’an 710032, China
| | - Xinling Ren
- Department of Respiratory and Critical Care Medicine, Shenzhen University General Hospital, Shenzhen 518071, China
| | - Jin Ma
- Department of Aerospace Physiology, Air Force Medical University, Xi’an 710032, China
| |
Collapse
|
4
|
Visnagri A, Oexner RR, Kmiotek-Wasylewska K, Zhang M, Zoccarato A, Shah AM. Nicotinamide Adenosine Dinucleotide Phosphate Oxidase-Mediated Signaling in Cardiac Remodeling. Antioxid Redox Signal 2023; 38:371-387. [PMID: 36656669 DOI: 10.1089/ars.2022.0176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Significance: Reactive oxygen species (ROS) play a key role in the pathogenesis of cardiac remodeling and the subsequent progression to heart failure (HF). Nicotinamide adenosine dinucleotide phosphate (NADPH) oxidases (NOXs) are one of the major sources of ROS and are expressed in different heart cell types, including cardiomyocytes, endothelial cells, fibroblasts, and inflammatory cells. Recent Advances: NOX-derived ROS are usually produced in a regulated and spatially confined fashion and typically linked to specific signaling. The two main cardiac isoforms, namely nicotinamide adenine dinucleotide phosphate oxidase isoform 2 (NOX2) and nicotinamide adenine dinucleotide phosphate oxidase isoform 4 (NOX4), possess different biochemical and (patho)physiological properties and exert distinct effects on the cardiac phenotype in many settings. Recent work has defined important cell-specific effects of NOX2 that contribute to pathological cardiac remodeling and dysfunction. NOX4, on the other hand, may exert protective effects by stimulating adaptive stress responses, with recent data showing that NOX4-mediated signaling regulates transcription and metabolism in the heart. Critical Issues: The inhibition of NOX2 appears to be a very promising therapeutic target to ameliorate pathological cardiac remodeling. If the beneficial effects of NOX4 can be enhanced, this might be a unique approach to boosting adaptive responses and thereby impact cell survival, activation, contractility, and growth. Future Directions: Increasing knowledge regarding the intricacies of NOX-mediated signaling may yield tractable therapeutic targets, in contrast to the non-specific targeting of oxidative stress. Antioxid. Redox Signal. 38, 371-387.
Collapse
Affiliation(s)
- Asjad Visnagri
- British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, United Kingdom
| | - Rafael R Oexner
- British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, United Kingdom
| | - Katarzyna Kmiotek-Wasylewska
- British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, United Kingdom
| | - Min Zhang
- British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, United Kingdom
| | - Anna Zoccarato
- British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, United Kingdom
| | - Ajay M Shah
- British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, United Kingdom
| |
Collapse
|
5
|
Yu Z, Wang L, Wu S, Zhao W. Dissecting the potential mechanism of antihypertensive effects of RVPSL on spontaneously hypertensive rats via widely targeted kidney metabolomics. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:428-436. [PMID: 36373790 DOI: 10.1002/jsfa.12157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 07/26/2022] [Accepted: 07/30/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Our previous study has demonstrated that the egg-white-derived peptide RVPSL can lower blood pressure in spontaneously hypertensive rats (SHRs), but its potential action mechanism remains unclear. In this work, the underlying mechanism of the antihypertensive effects of RVPSL in SHRs was elucidated using the widely targeted kidney metabolomics approach. RESULTS Ten SHRs were divided into two groups: SHR-Untreated group (0.9% saline) and SHR-RVPSL group (50 mg kg-1 body weight RVPSL) for 4 weeks. After 4 weeks, kidney samples were collected and widely targeted (liquid chromatography-electrospray ionization-tandem mass spectrometry) metabolomics was used to detect metabolites. Fifty-six biomarkers were identified that may be associated with hypertension. Among them, 17 biomarkers were upregulated and 39 biomarkers were downregulated. The results suggested that eight potential biomarkers were identified in kidney samples: O-phospho-l-serine, tyramine, citric acid, 3-hydroxybutyrate, O-acetyl-l-serine, 15-oxo-5Z,8Z,11Z,13E-eicosatetraenoic acid (15-oxoETE), dopaquinone and 3,3',5-triiodo-l-thyronine. These potential biomarkers mainly involved carbon metabolism, thyroid hormone signaling pathway, tyrosine metabolism and arachidonic acid metabolism. CONCLUSION The study suggested that RVPSL may exert antihypertensive effects through upregulation of O-phospho-l-serine, 3-hydroxybutyrate and 15-oxoETE, and downregulation of tyramine, citric acid, O-acetyl-l-serine, 3,3',5-triiodo-l-thyronine and dopaquinone. The antihypertensive effects of RVPSL may be related to carbon metabolism, thyroid hormone signaling pathway, tyrosine metabolism and arachidonic acid metabolism. RVPSL exhibited a potent antihypertensive effect, and the antihypertensive effects were associated with inhibition of vascular smooth muscle cell proliferation, vascular remodeling, vascular endothelium dysfunction, restoring reactive oxygen species, oxidative stress, inflammation and immune reaction. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhipeng Yu
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Li Wang
- College of Food Science and Engineering, Bohai University, Jinzhou, PR China
| | - Sijia Wu
- Laboratory of Nutrition and Functional Food, Jilin University, Changchun, PR China
| | - Wenzhu Zhao
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| |
Collapse
|
6
|
Chen X, Zheng L, Zhang B, Deng Z, Li H. Synergistic protection of quercetin and lycopene against oxidative stress via SIRT1-Nox4-ROS axis in HUVEC cells. Curr Res Food Sci 2022; 5:1985-1993. [PMID: 36304485 PMCID: PMC9593281 DOI: 10.1016/j.crfs.2022.10.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/26/2022] [Accepted: 10/14/2022] [Indexed: 11/19/2022] Open
Abstract
Oxidative stress is a potential factor in the promotion of endothelial dysfunction. In this research, flavonoids (quercetin, luteolin) combined with carotenoids (lycopene, lutein), especially quercetin-lycopene combination (molar ratio 5:1), prevented the oxidative stress in HUVEC cells by reducing the reactive oxygen species (ROS) and suppressing the expression of NADPH oxidase 4 (Nox4), a major source of ROS production. RNA-seq analysis indicated quercetin-lycopene combination downregulated inflammatory genes induced by H2O2, such as IL-17 and NF-κB. The expression of NF-κB p65 was activated by H2O2 but inhibited by the quercetin-lycopene combination. Moreover, the quercetin and lycopene combination promoted the thermostability of Sirtuin 1 (SIRT1) and activated SIRT1 deacetyl activity. SIRT1 inhibitor EX-527 attenuated the inhibitory effects of quercetin, lycopene, and their combination on the expression of p65, Nox4 enzyme, and ROS. Quercetin-lycopene combination could interact with SIRT1 to inhibit Nox4 and prevent endothelial oxidative stress, potentially contributing to the prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Xuan Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Liufeng Zheng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Bing Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, Jiangxi, China
- Institute for Advanced Study, Nanchang University, Nanchang, 330031, Jiangxi, China
- Corresponding author. State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, Jiangxi, China.
| | - Hongyan Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, Jiangxi, China
- Corresponding author.
| |
Collapse
|
7
|
Ning M, Liu Y, Wang D, Wei J, Hu G, Xing P. Knockdown of TRIM27 alleviated sepsis-induced inflammation, apoptosis, and oxidative stress via suppressing ubiquitination of PPARγ and reducing NOX4 expression. Inflamm Res 2022; 71:1315-1325. [PMID: 35962797 PMCID: PMC9375190 DOI: 10.1007/s00011-022-01625-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Sepsis is a global fatal disease and leads to severe lung injury due to dysfunction of inflammation response. TRIM27 is closely related to the diseased with dysfunction of inflammation response. The aim of this study was to clarify the role and mechanism of TRIM27 in sepsis-induced lung injury. METHODS The lipopolysaccharide (LPS)-induced septic mouse model was successfully established. The lung injury was evaluated by lung wet/dry (W/D) ratio and hematoxylin-eosin (H&E) staining. The cell apoptosis was evaluated by TUNEL assay. The inflammatory cytokines were measured by quantitative real time-PCR (qRT-PCR) assay and commercial enzyme-linked immunosorbent assay (ELISA). The oxidative stress was assessed by the contents of superoxide dismutase (SOD) and malondialdehyde (MDA), and the expression of dihydroethidium (DHE). RESULTS In this study, we demonstrated that TRIM27 was up-regulated in LPS-induced septic mice. In loss-of-function experiments, knockdown of TRIM27 alleviated sepsis-induced lung injury, inflammation, apoptosis, and oxidative stress. More importantly, knockdown of TRIM27 was observed to reduce p-p65/NOX4 expression via suppressing ubiquitination of PPARγ. In rescue experiments, overexpression of NOX4 abolished the effect of sh-TRIM27 on alleviating sepsis-induced inflammation, apoptosis, and oxidative stress. CONCLUSION These findings highlighted that knockdown of TRIM27 alleviated sepsis-induced inflammation, oxidative stress and apoptosis via suppressing ubiquitination of PPARγ and reducing NOX4 expression, which supports the potential utility of TRIM27 as a therapeutic target in septic lung injury.
Collapse
Affiliation(s)
- Meng Ning
- Department of Heart Center, Tianjin Third Central Hospital, Tianjin, 300170, China.,Department of Heart Center, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, 300170, China.,Department of Heart Center, Artificial Cell Engineering Technology Research Center, Tianjin, 300170, China
| | - Yingwu Liu
- Department of Heart Center, Tianjin Third Central Hospital, Tianjin, 300170, China.,Department of Heart Center, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, 300170, China.,Department of Heart Center, Artificial Cell Engineering Technology Research Center, Tianjin, 300170, China
| | - Donglian Wang
- Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 222, West Three Road Around Lake, Nanhui New Town, Pudong New Area, Shanghai, China
| | - Jin Wei
- Department of Heart Center, Tianjin Third Central Hospital, Tianjin, 300170, China.,Department of Heart Center, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, 300170, China.,Department of Heart Center, Artificial Cell Engineering Technology Research Center, Tianjin, 300170, China
| | - Guoyong Hu
- Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 222, West Three Road Around Lake, Nanhui New Town, Pudong New Area, Shanghai, China
| | - Pengcheng Xing
- Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 222, West Three Road Around Lake, Nanhui New Town, Pudong New Area, Shanghai, China.
| |
Collapse
|
8
|
Yang C, Rong R, Li Y, Cheng M, Luo Y. Decrease in LINC00963 attenuates the progression of pulmonary arterial hypertension via microRNA-328-3p/profilin 1 axis. J Clin Lab Anal 2022; 36:e24383. [PMID: 35349725 PMCID: PMC9102517 DOI: 10.1002/jcla.24383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/08/2022] [Accepted: 03/19/2022] [Indexed: 12/23/2022] Open
Abstract
Background Pulmonary arterial hypertension (PAH) is a severe cardiopulmonary disease characterized by vascular hyperplasia and remodeling. Long noncoding RNA LINC00963 can regulate cell proliferation and metastasis in nonsmall cell lung cancer. However, the function of LINC00963 on PAH progression is rarely reported. Methods Quantitative real‐time PCR was used to determine the expression levels of LINC00963, microRNA (miRNA)‐328‐3p, and profilin 1 (PFN1), as well as vascular endothelial growth factor (VEGF), fibroblast growth factor 2 (FGF‐2), and hypoxia‐inducible factor (HIF)‐α. The protein level of PFN1 was measured by western blotting. The viability and migration of hypoxia‐induced pulmonary arterial smooth muscle cells (PASMCs) were assessed by 3‐(4, 5‐dimethyl‐2‐thiazolyl)‐2, 5‐diphenyl‐2‐h‐tetrazolium bromide, and transwell assays, respectively. The target relationships between miR‐328‐3p and LINC00963/PFN1 were confirmed by dual‐luciferase reporter assay. A PAH mouse model was conducted to explore the effects of hypoxia on cardiopulmonary functions. Results In hypoxia‐induced PASMCs and PAH mouse model, high expression levels of LINC00963 and PFN1, and low expression of miR‐328‐3p, were determined. The viability, migration of hypoxia‐induced PASMCs, the expression of VEGF, FGF‐2, and HIF‐α were significantly repressed by transfection of si‐LINC00963 or miR‐328‐3p mimics. The inhibitory effects of LINC00963 silencing on cell viability, migration, and the levels of VEGF, FGF‐2, and HIF‐α were partly eliminated by miR‐328‐3p inhibitor or increasing the expression of PFN1. Hypoxia treatment increased the levels of RVSP, mPAP, and RV/(LV+S), as well as the thickness of pulmonary artery wall. Conclusions Silencing of LINC00963 ameliorates PAH via modulating miR‐328‐3p/PFN1.
Collapse
Affiliation(s)
- Chengpeng Yang
- Cardiothoracic Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi City, China
| | - Rong Rong
- Department of Physics Diagnosis, First Affiliated Hospital of Jiamusi University, Jiamusi City, China
| | - Yuze Li
- Department of Nephrology, First Affiliated Hospital of Jiamusi University, Jiamusi City, China
| | - Mingxun Cheng
- Vascular Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi City, China
| | - Yanzhuo Luo
- Ministry of Continuing Education, First Affiliated Hospital of Jiamusi University, Jiamusi City, China
| |
Collapse
|
9
|
MicroRNA 148a Suppresses Tuberculous Fibrosis by Targeting NOX4 and POLDIP2. Int J Mol Sci 2022; 23:ijms23062999. [PMID: 35328424 PMCID: PMC8954251 DOI: 10.3390/ijms23062999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/22/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023] Open
Abstract
Extracellular matrix production by pleural mesothelial cells in response to Mycobacterium tuberculosis contributes to tuberculous fibrosis. NOX4 is involved in the pathogenesis of tuberculous fibrosis. In this study, we evaluated whether NOX4 gene-targeting microRNAs showed protective effects in tuberculosis fibrosis. TargetScan prediction software was used to identify candidate microRNAs that bind the 3′ UTRs of NOX4, and microRNA-148a (miR-148a) was selected as the best miRNA candidate. A repressed and forced expression assay in Met5A cells was performed to investigate the causal relationship between miR-148a and NOX4. The role of miR-148a in tuberculous pleural fibrosis was studied using a murine model of Mycobacterium bovis bacillus Calmette–Guérin (BCG) pleural infection. Heat-killed M. tuberculosis (HKMT) induces NOX4 and POLDIP2 expression. We demonstrated the inhibitory effect of miR-148a on NOX4 and POLDIP2 expression. The increased expression of miR-148a suppressed HKMT-induced collagen-1A synthesis in PMC cells. In the BCG pleurisy model, miR-148a significantly reduced fibrogenesis and epithelial mesenchymal transition. High levels of miR-148a in tuberculous pleural effusion can be interpreted as a self-limiting homeostatic response. Our data indicate that miR-148a may protect against tuberculous pleural fibrosis by regulating NOX4 and POLDIP2.
Collapse
|
10
|
Zhang F, Guo X, Xia Y, Mao L. An update on the phenotypic switching of vascular smooth muscle cells in the pathogenesis of atherosclerosis. Cell Mol Life Sci 2021; 79:6. [PMID: 34936041 PMCID: PMC11072026 DOI: 10.1007/s00018-021-04079-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/20/2021] [Accepted: 12/03/2021] [Indexed: 12/11/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are involved in phenotypic switching in atherosclerosis. This switching is characterized by VSMC dedifferentiation, migration, and transdifferentiation into other cell types. VSMC phenotypic transitions have historically been considered bidirectional processes. Cells can adopt a physiological contraction phenotype or an alternative "synthetic" phenotype in response to injury. However, recent studies, including lineage tracing and single-cell sequencing studies, have shown that VSMCs downregulate contraction markers during atherosclerosis while adopting other phenotypes, including macrophage-like, foam cell, mesenchymal stem-like, myofibroblast-like, and osteochondral-like phenotypes. However, the molecular mechanism and processes regulating the switching of VSMCs at the onset of atherosclerosis are still unclear. This systematic review aims to review the critical outstanding challenges and issues that need further investigation and summarize the current knowledge in this field.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoqing Guo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuanpeng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
11
|
Taylor JP, Tse HM. The role of NADPH oxidases in infectious and inflammatory diseases. Redox Biol 2021; 48:102159. [PMID: 34627721 PMCID: PMC8487856 DOI: 10.1016/j.redox.2021.102159] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX) are enzymes that generate superoxide or hydrogen peroxide from molecular oxygen utilizing NADPH as an electron donor. There are seven enzymes in the NOX family: NOX1-5 and dual oxidase (DUOX) 1-2. NOX enzymes in humans play important roles in diverse biological functions and vary in expression from tissue to tissue. Importantly, NOX2 is involved in regulating many aspects of innate and adaptive immunity, including regulation of type I interferons, the inflammasome, phagocytosis, antigen processing and presentation, and cell signaling. DUOX1 and DUOX2 play important roles in innate immune defenses at epithelial barriers. This review discusses the role of NOX enzymes in normal physiological processes as well as in disease. NOX enzymes are important in autoimmune diseases like type 1 diabetes and have also been implicated in acute lung injury caused by infection with SARS-CoV-2. Targeting NOX enzymes directly or through scavenging free radicals may be useful therapies for autoimmunity and acute lung injury where oxidative stress contributes to pathology.
Collapse
Affiliation(s)
- Jared P Taylor
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hubert M Tse
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
12
|
Korbecki J, Simińska D, Gąssowska-Dobrowolska M, Listos J, Gutowska I, Chlubek D, Baranowska-Bosiacka I. Chronic and Cycling Hypoxia: Drivers of Cancer Chronic Inflammation through HIF-1 and NF-κB Activation: A Review of the Molecular Mechanisms. Int J Mol Sci 2021; 22:ijms221910701. [PMID: 34639040 PMCID: PMC8509318 DOI: 10.3390/ijms221910701] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic (continuous, non-interrupted) hypoxia and cycling (intermittent, transient) hypoxia are two types of hypoxia occurring in malignant tumors. They are both associated with the activation of hypoxia-inducible factor-1 (HIF-1) and nuclear factor κB (NF-κB), which induce changes in gene expression. This paper discusses in detail the mechanisms of activation of these two transcription factors in chronic and cycling hypoxia and the crosstalk between both signaling pathways. In particular, it focuses on the importance of reactive oxygen species (ROS), reactive nitrogen species (RNS) together with nitric oxide synthase, acetylation of HIF-1, and the action of MAPK cascades. The paper also discusses the importance of hypoxia in the formation of chronic low-grade inflammation in cancerous tumors. Finally, we discuss the effects of cycling hypoxia on the tumor microenvironment, in particular on the expression of VEGF-A, CCL2/MCP-1, CXCL1/GRO-α, CXCL8/IL-8, and COX-2 together with PGE2. These factors induce angiogenesis and recruit various cells into the tumor niche, including neutrophils and monocytes which, in the tumor, are transformed into tumor-associated neutrophils (TAN) and tumor-associated macrophages (TAM) that participate in tumorigenesis.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (D.S.); (I.G.); (D.C.)
| | - Donata Simińska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (D.S.); (I.G.); (D.C.)
| | - Magdalena Gąssowska-Dobrowolska
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland;
| | - Joanna Listos
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a St., 20-093 Lublin, Poland;
| | - Izabela Gutowska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (D.S.); (I.G.); (D.C.)
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (D.S.); (I.G.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (D.S.); (I.G.); (D.C.)
- Correspondence: ; Tel.: +48-(91)-466-1515
| |
Collapse
|
13
|
Cannabinoids-A New Perspective in Adjuvant Therapy for Pulmonary Hypertension. Int J Mol Sci 2021; 22:ijms221810048. [PMID: 34576212 PMCID: PMC8472313 DOI: 10.3390/ijms221810048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
Currently, no treatment can completely cure pulmonary hypertension (PH), which can lead to right ventricular failure and, consequently, death. Therefore, searching for new therapies remains important. Increased resistance in pulmonary circulation is mainly caused by the excessive contraction and proliferation of small pulmonary arteries. Cannabinoids, a group of lipophilic compounds that all interact with cannabinoid receptors, exert a pulmonary vasodilatory effect through several different mechanisms, including mechanisms that depend on vascular endothelium and/or receptor-based mechanisms, and may also have anti-proliferative and anti-inflammatory properties. The vasodilatory effect is important in regulating pulmonary resistance, which can improve patients’ quality of life. Moreover, experimental studies on the effects of cannabidiol (plant-derived, non-psychoactive cannabinoid) in animal PH models have shown that cannabidiol reduces right ventricular systolic pressure and excessive remodelling and decreases pulmonary vascular hypertrophy and pulmonary vascular resistance. Due to the potentially beneficial effects of cannabinoids on pulmonary circulation and PH, in this work, we review whether cannabinoids can be used as an adjunctive therapy for PH. However, clinical trials are still needed to recommend the use of cannabinoids in the treatment of PH.
Collapse
|
14
|
Dou X, Ma Y, Qin Y, Dong Q, Zhang S, Tian R, Pan M. NEAT1 silencing alleviates pulmonary arterial smooth muscle cell migration and proliferation under hypoxia through regulation of miR‑34a‑5p/KLF4 in vitro. Mol Med Rep 2021; 24:749. [PMID: 34468014 DOI: 10.3892/mmr.2021.12389] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/29/2021] [Indexed: 11/06/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe vascular disease that adversely affects patient health and can be life threatening. The present study aimed to investigate the detailed role of nuclear paraspeckle assembly transcript 1 (NEAT1) in PAH. Using RT‑qPCR, the expression levels of NEAT1, microRNA (miR)‑34a‑5p, and Krüppel‑like factor 4 (KLF4) were detected in both hypoxia‑treated pulmonary arterial smooth muscle cells (PASMCs) and serum from PAH patients. Then, the interactions among miR‑34a‑5p, NEAT1, and KLF4 were evaluated by dual‑luciferase reporter assay. The detailed role of the NEAT1/miR‑34a‑5p/KLF4 axis in PAH pathogenesis was further explored using MTT, Transwell, and western blot assays. The results revealed that NEAT1 targeted miR‑34a‑5p and miR‑34a‑5p targeted KLF4. In hypoxia‑treated PASMCs and serum from PAH patients, high NEAT1 and KLF4 expression levels and low miR‑34a‑5p expression were observed. The proliferation and migration of hypoxia‑treated PASMCs were reduced by transfection with sh‑NEAT1 or miR‑34a‑5p mimics. The suppressive effects of NEAT1 knockdown on the proliferation and migration of hypoxia‑treated PASMCs were reversed by knock down of miR‑34a‑5p expression and increased KLF4 expression. NEAT1 was not only highly expressed in the serum of PAH patients but its silencing also alleviated PAH by regulating miR‑34a‑5p/KLF4 in vitro. The present study highlighted a potential new therapeutic target and diagnostic biomarker for PAH.
Collapse
Affiliation(s)
- Xiuli Dou
- Department of Emergency, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Yuxiao Ma
- Department of Emergency, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Yijie Qin
- Department of Emergency, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Qinglin Dong
- Department of Emergency, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Shouwei Zhang
- Department of Medical, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Rui Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Mingyu Pan
- Department of Cardiology, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| |
Collapse
|
15
|
Zolty R. Novel Experimental Therapies for Treatment of Pulmonary Arterial Hypertension. J Exp Pharmacol 2021; 13:817-857. [PMID: 34429666 PMCID: PMC8380049 DOI: 10.2147/jep.s236743] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 07/07/2021] [Indexed: 12/18/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and devastating disease characterized by pulmonary artery vasoconstriction and vascular remodeling leading to vascular rarefaction with elevation of pulmonary arterial pressures and pulmonary vascular resistance. Often PAH will cause death from right heart failure. Current PAH-targeted therapies improve functional capacity, pulmonary hemodynamics and reduce hospitalization. Nevertheless, today PAH still remains incurable and is often refractory to medical therapy, underscoring the need for further research. Over the last three decades, PAH has evolved from a disease of unknown pathogenesis devoid of effective therapy to a condition whose cellular, genetic and molecular underpinnings are unfolding. This article provides an update on current knowledge and summarizes the progression in recent advances in pharmacological therapy in PAH.
Collapse
Affiliation(s)
- Ronald Zolty
- Pulmonary Hypertension Program, University of Nebraska Medical Center, Lied Transplant Center, Omaha, NE, USA
| |
Collapse
|
16
|
Shimoda LA. Cellular Pathways Promoting Pulmonary Vascular Remodeling by Hypoxia. Physiology (Bethesda) 2021; 35:222-233. [PMID: 32490752 DOI: 10.1152/physiol.00039.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Exposure to hypoxia increases pulmonary vascular resistance, leading to elevated pulmonary arterial pressure and, potentially, right heart failure. Vascular remodeling is an important contributor to the increased pulmonary vascular resistance. Hyperproliferation of smooth muscle, endothelial cells, and fibroblasts, and deposition of extracellular matrix lead to increased wall thickness, extension of muscle into normally non-muscular arterioles, and vascular stiffening. This review highlights intrinsic and extrinsic modulators contributing to the remodeling process.
Collapse
Affiliation(s)
- Larissa A Shimoda
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
17
|
Gorelova A, Berman M, Al Ghouleh I. Endothelial-to-Mesenchymal Transition in Pulmonary Arterial Hypertension. Antioxid Redox Signal 2021; 34:891-914. [PMID: 32746619 PMCID: PMC8035923 DOI: 10.1089/ars.2020.8169] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/14/2022]
Abstract
Endothelial-to-mesenchymal transition (EndMT) is a process that encompasses extensive transcriptional reprogramming of activated endothelial cells leading to a shift toward mesenchymal cellular phenotypes and functional responses. Initially observed in the context of embryonic development, in the last few decades EndMT is increasingly recognized as a process that contributes to a variety of pathologies in the adult organism. Within the settings of cardiovascular biology, EndMT plays a role in various diseases, including atherosclerosis, heart valvular disease, cardiac fibrosis, and myocardial infarction. EndMT is also being progressively implicated in development and progression of pulmonary hypertension (PH) and pulmonary arterial hypertension (PAH). This review covers the current knowledge about EndMT in PH and PAH, and provides comprehensive overview of seminal discoveries. Topics covered include evidence linking EndMT to factors associated with PAH development, including hypoxia responses, inflammation, dysregulation of bone-morphogenetic protein receptor 2 (BMPR2), and redox signaling. This review amalgamates these discoveries into potential insights for the identification of underlying mechanisms driving EndMT in PH and PAH, and discusses future directions for EndMT-based therapeutic strategies in disease management.
Collapse
Affiliation(s)
- Anastasia Gorelova
- Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mariah Berman
- Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Imad Al Ghouleh
- Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
18
|
Alatshan A, Benkő S. Nuclear Receptors as Multiple Regulators of NLRP3 Inflammasome Function. Front Immunol 2021; 12:630569. [PMID: 33717162 PMCID: PMC7952630 DOI: 10.3389/fimmu.2021.630569] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Nuclear receptors are important bridges between lipid signaling molecules and transcription responses. Beside their role in several developmental and physiological processes, many of these receptors have been shown to regulate and determine the fate of immune cells, and the outcome of immune responses under physiological and pathological conditions. While NLRP3 inflammasome is assumed as key regulator for innate and adaptive immune responses, and has been associated with various pathological events, the precise impact of the nuclear receptors on the function of inflammasome is hardly investigated. A wide variety of factors and conditions have been identified as modulators of NLRP3 inflammasome activation, and at the same time, many of the nuclear receptors are known to regulate, and interact with these factors, including cellular metabolism and various signaling pathways. Nuclear receptors are in the focus of many researches, as these receptors are easy to manipulate by lipid soluble molecules. Importantly, nuclear receptors mediate regulatory mechanisms at multiple levels: not only at transcription level, but also in the cytosol via non-genomic effects. Their importance is also reflected by the numerous approved drugs that have been developed in the past decade to specifically target nuclear receptors subtypes. Researches aiming to delineate mechanisms that regulate NLRP3 inflammasome activation draw a wide range of attention due to their unquestionable importance in infectious and sterile inflammatory conditions. In this review, we provide an overview of current reports and knowledge about NLRP3 inflammasome regulation from the perspective of nuclear receptors, in order to bring new insight to the potentially therapeutic aspect in targeting NLRP3 inflammasome and NLRP3 inflammasome-associated diseases.
Collapse
Affiliation(s)
- Ahmad Alatshan
- Departments of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cellular and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilvia Benkő
- Departments of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cellular and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
19
|
Luo X, Wu J, Wu G. PPARγ activation suppresses the expression of MMP9 by downregulating NF-κB post intracerebral hemorrhage. Neurosci Lett 2021; 752:135770. [PMID: 33636289 DOI: 10.1016/j.neulet.2021.135770] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/19/2021] [Indexed: 01/23/2023]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPARγ) is critical in protecting against inflammatory and oxidative stresses post brain injury. We have previously reported that rosiglitazone, an agonist of PPARγ, was effective to prevent microglia from apoptosis and ameliorate neuronal injuries post intracerebral hemorrhage (ICH) with suppression of matrix metalloproteinase-9 (MMP9) expression. However, molecular mechanisms linking how PPARγ decreases MMP9 remain unknown. Here, we hypothesize that PPARγ downregulates MMP9 expression post hemorrhage by inhibiting nuclear factor kappa B (NF-κB), an upstream regulator of MMPs gene and also key transcription factor involved in the control of immune and neuroinflammatory responses. We found both in vivo and in vitro that PPARγ was significantly downregulated post ICH with prominent increases of NF-κB and MMP9. Activation of PPARγ using rosiglitazone decreased the expression of both NF-κB and MMP9, while reversed effects were observed when administrating the PPARγ antagonist GW9662. Besides, inhibiting NF-κB by JSH-23 also suppressed the expression of MMP9, with only limited effect on PPARγ. Further studies revealed prominent colocalizations of NF-κB with PPARγ and MMP9, respectively. Finally, direct interactions of NF-κB with PPARγ and MMP9 gene were also confirmed, respectively, by protein and chromatin immunoprecipitations. These results suggested a role of NF-κB in mediating the reduction of MMP9 by PPARγ, potentially providing a new therapeutic target for brain hemorrhage.
Collapse
Affiliation(s)
- Xingmei Luo
- The Second Affiliated Hospital of Suzhou University, Suzhou, China; Department of Comprehensive Ward, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jing Wu
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Guofeng Wu
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| |
Collapse
|
20
|
Maietta V, Reyes-García J, Yadav VR, Zheng YM, Peng X, Wang YX. Cellular and Molecular Processes in Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:21-38. [PMID: 34019261 DOI: 10.1007/978-3-030-68748-9_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pulmonary hypertension (PH) is a progressive lung disease characterized by persistent pulmonary vasoconstriction. Another well-recognized characteristic of PH is the muscularization of peripheral pulmonary arteries. This pulmonary vasoremodeling manifests in medial hypertrophy/hyperplasia of smooth muscle cells (SMCs) with possible neointimal formation. The underlying molecular processes for these two major vascular responses remain not fully understood. On the other hand, a series of very recent studies have shown that the increased reactive oxygen species (ROS) seems to be an important player in mediating pulmonary vasoconstriction and vasoremodeling, thereby leading to PH. Mitochondria are a primary site for ROS production in pulmonary artery (PA) SMCs, which subsequently activate NADPH oxidase to induce further ROS generation, i.e., ROS-induced ROS generation. ROS control the activity of multiple ion channels to induce intracellular Ca2+ release and extracellular Ca2+ influx (ROS-induced Ca2+ release and influx) to cause PH. ROS and Ca2+ signaling may synergistically trigger an inflammatory cascade to implicate in PH. Accordingly, this paper explores the important roles of ROS, Ca2+, and inflammatory signaling in the development of PH, including their reciprocal interactions, key molecules, and possible therapeutic targets.
Collapse
Affiliation(s)
- Vic Maietta
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Jorge Reyes-García
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA.,Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Vishal R Yadav
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Yun-Min Zheng
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA.
| | - Xu Peng
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, TX, USA.
| | - Yong-Xiao Wang
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
21
|
Segatto M, Szokoll R, Fittipaldi R, Bottino C, Nevi L, Mamchaoui K, Filippakopoulos P, Caretti G. BETs inhibition attenuates oxidative stress and preserves muscle integrity in Duchenne muscular dystrophy. Nat Commun 2020; 11:6108. [PMID: 33257646 PMCID: PMC7705749 DOI: 10.1038/s41467-020-19839-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/31/2020] [Indexed: 12/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) affects 1 in 3500 live male births. To date, there is no effective cure for DMD, and the identification of novel molecular targets involved in disease progression is important to design more effective treatments and therapies to alleviate DMD symptoms. Here, we show that protein levels of the Bromodomain and extra-terminal domain (BET) protein BRD4 are significantly increased in the muscle of the mouse model of DMD, the mdx mouse, and that pharmacological inhibition of the BET proteins has a beneficial outcome, tempering oxidative stress and muscle damage. Alterations in reactive oxygen species (ROS) metabolism are an early event in DMD onset and they are tightly linked to inflammation, fibrosis, and necrosis in skeletal muscle. By restoring ROS metabolism, BET inhibition ameliorates these hallmarks of the dystrophic muscle, translating to a beneficial effect on muscle function. BRD4 direct association to chromatin regulatory regions of the NADPH oxidase subunits increases in the mdx muscle and JQ1 administration reduces BRD4 and BRD2 recruitment at these regions. JQ1 treatment reduces NADPH subunit transcript levels in mdx muscles, isolated myofibers and DMD immortalized myoblasts. Our data highlight novel functions of the BET proteins in dystrophic skeletal muscle and suggest that BET inhibitors may ameliorate the pathophysiology of DMD.
Collapse
Affiliation(s)
- Marco Segatto
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy.,Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche (Is), Italy
| | - Roberta Szokoll
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Raffaella Fittipaldi
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Cinzia Bottino
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Lorenzo Nevi
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Kamel Mamchaoui
- Sorbonne Université, Inserm, Institut de Myologie, U974, Center for Research in Myology, 47 Boulevard de l'hôpital, 75013, Paris, France
| | - Panagis Filippakopoulos
- Structural Genomics Consortium, Old Road Campus Research Building, Nuffield Department of Medicine, Oxford, OX3 7DQ, UK
| | - Giuseppina Caretti
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
22
|
Xu T, Shao L, Wang A, Liang R, Lin Y, Wang G, Zhao Y, Hu J, Liu S. CD248 as a novel therapeutic target in pulmonary arterial hypertension. Clin Transl Med 2020; 10:e175. [PMID: 32997414 PMCID: PMC7507048 DOI: 10.1002/ctm2.175] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/05/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022] Open
Abstract
Pulmonary vascular remodeling is the most important pathological characteristic of pulmonary arterial hypertension (PAH). No effective treatment for PAH is currently available because the mechanism underlying vascular remodeling is not completely clear. CD248, also known as endosialin, is a transmembrane protein that is highly expressed in pericytes and fibroblasts. Here, we evaluated the role of CD248 in pulmonary vascular remodeling and the processes of PAH pathogenesis. Activation of CD248 in pulmonary artery smooth muscle cells (PASMCs) was found to be proportional to the severity of PAH. CD248 contributed to platelet-derived growth factor-BB (PDGF-BB)-induced PASMC proliferation and migration along with the shift to more synthetic phenotypes. In contrast, treatment with Cd248 siRNA or the anti-CD248 therapeutic antibody (ontuxizumab) significantly inhibited the PDGF signaling pathway, obstructed NF-κB p65-mediated transcription of Nox4, and decreased reactive oxygen species production induced by PDGF-BB in PAMSCs. In addition, knockdown of CD248 alleviated pulmonary vascular remodeling in rat PAH models. This study provides novel insights into the dysfunction of PASMCs leading to pulmonary vascular remodeling, and provides evidence for anti-remodeling treatment for PAH via the immediate targeting of CD248.
Collapse
Affiliation(s)
- Tao Xu
- Life Science InstituteJinzhou Medical UniversityJinzhouP. R. China
| | - Lei Shao
- Department of CardiologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinP. R. China
| | - Aimei Wang
- Department of PhysiologyJinzhou Medical UniversityJinzhouP. R. China
| | - Rui Liang
- Department of PhysiologyJinzhou Medical UniversityJinzhouP. R. China
| | - Yuhan Lin
- Department of PhysiologyJinzhou Medical UniversityJinzhouP. R. China
| | - Guan Wang
- Life Science InstituteJinzhou Medical UniversityJinzhouP. R. China
| | - Yan Zhao
- Life Science InstituteJinzhou Medical UniversityJinzhouP. R. China
| | - Jing Hu
- Life Science InstituteJinzhou Medical UniversityJinzhouP. R. China
| | - Shuangyue Liu
- Department of PhysiologyJinzhou Medical UniversityJinzhouP. R. China
| |
Collapse
|
23
|
Li ZM, Xu SY, Feng YZ, Cheng YR, Xiong JB, Zhou Y, Guan CX. The role of NOX4 in pulmonary diseases. J Cell Physiol 2020; 236:1628-1637. [PMID: 32780450 DOI: 10.1002/jcp.30005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/26/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022]
Abstract
Nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) is a subtype of the NOX family, which is mainly expressed in the pulmonary vasculature and pulmonary endothelial cells in the respiratory system. NOX4 has unique characteristics, and is a constitutively active enzyme that primarily produces hydrogen peroxide. The signaling pathways associated with NOX4 are complicated. Negative and positive feedback play significant roles in regulating NOX4 expression. The role of NOX4 is controversial because NOX4 plays a protective or damaging role in different respiratory diseases. This review summarizes the structure, enzymatic properties, regulation, and signaling pathways of NOX4. This review then introduces the roles of NOX4 in different diseases in the respiratory system, such as acute respiratory distress syndrome, chronic obstructive pulmonary disease, and pulmonary fibrosis.
Collapse
Affiliation(s)
- Zi-Ming Li
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Sheng-Ya Xu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yi-Zhuo Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yu-Rui Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jian-Bing Xiong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
24
|
Poznyak AV, Grechko AV, Orekhova VA, Khotina V, Ivanova EA, Orekhov AN. NADPH Oxidases and Their Role in Atherosclerosis. Biomedicines 2020; 8:biomedicines8070206. [PMID: 32664404 PMCID: PMC7399834 DOI: 10.3390/biomedicines8070206] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
The current view on atherosclerosis positions it as a multifactorial disorder that results from the interplay between lipid metabolism disturbances and inflammatory processes. Oxidative stress is proven to be one of the initiating factors in atherosclerosis development, being implicated both in the inflammatory response and in atherogenic modifications of lipoproteins that facilitate lipid accumulation in the arterial wall. The hallmark of oxidative stress is the elevated level of reactive oxygen species (ROS). Correspondingly, the activity of major ROS-generating enzymes, including nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, xanthine oxidases, and cyclooxygenases, is an important element in atherosclerosis development. In particular, the role of NADPH oxidases in atherosclerosis development has become a subject of intensive research. Aberrant activity of NADPH oxidases was shown to be associated with cardiovascular disease in humans. With regard to atherosclerosis, several important pathological components of the disease development, including endothelial dysfunction, inflammation, and vascular remodeling, involve aberrations in NADPH oxidases functioning. In humans, NADPH oxidases are represented by four isoforms expressed in vascular tissues, where they serve as the main source of ROS during atherogenesis. Moreover, recent studies have demonstrated their impact on vascular remodeling processes. Interestingly, one of the NADPH oxidase isoforms, NOX4, was shown to have an atheroprotective effect. Despite the growing evidence of the crucial involvement of NADPH oxidases in atherosclerosis pathogenesis, the available data still remains controversial. In this narrative review, we summarize the current knowledge of the role of NADPH oxidases in atherosclerosis and outline the future directions of research.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Department of Basic Research, Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia; (A.V.P.); (E.A.I.)
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 14-3 Solyanka Street, 109240 Moscow, Russia;
| | - Varvara A. Orekhova
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Street, 121552 Moscow, Russia;
| | - Victoria Khotina
- Laboratory of Infectious Pathology and Molecular Microecology, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia;
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8, Baltiyskaya St., 125315 Moscow, Russia
| | - Ekaterina A. Ivanova
- Department of Basic Research, Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia; (A.V.P.); (E.A.I.)
| | - Alexander N. Orekhov
- Laboratory of Infectious Pathology and Molecular Microecology, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia;
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8, Baltiyskaya St., 125315 Moscow, Russia
- Correspondence: ; Tel./Fax: +7-(495)-4159594
| |
Collapse
|
25
|
Pereira BP, do Valle GT, Salles BCC, Costa KCM, Ângelo ML, Torres LHL, Novaes RD, Ruginsk SG, Tirapelli CR, de Araújo Paula FB, Ceron CS. Pyrrolidine dithiocarbamate reduces alloxan-induced kidney damage by decreasing nox4, inducible nitric oxide synthase, and metalloproteinase-2. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:1899-1910. [PMID: 32440769 DOI: 10.1007/s00210-020-01906-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/10/2020] [Indexed: 12/18/2022]
Abstract
We examined the effect of the NFκB inhibitor pyrrolidine-1-carbodithioic acid (PDTC) on inducible nitric oxide synthase (iNOS), matrix metalloproteinase-2 (MMP-2) activity, and oxidative and inflammatory kidney damage in alloxan-induced diabetes. Two weeks after diabetes induction (alloxan-130 mg/kg), control and diabetic rats received PDTC (100 mg/kg) or vehicle for 8 weeks. Body weight, glycemia, urea, and creatinine were measured. Kidney changes were measured in hematoxylin/eosin sections and ED1 by immunohistochemistry. Kidney thiobarbituric acid reactive substances (TBARS), superoxide anion (O2-), and nitrate/nitrite (NOx) levels, and catalase and superoxide dismutase (SOD) activities were analyzed. Also, kidney nox4 and iNOS expression, and NFkB nuclear translocation were measured by western blot, and MMP-2 by zymography. Glycemia and urea increased in alloxan rats, which were not modified by PDTC treatment. However, PDTC attenuated kidney structural alterations and macrophage infiltration in diabetic rats. While diabetes increased both TBARS and O2- levels, PDTC treatment reduced TBARS in diabetic and O2- in control kidneys. A decrease in NOx levels was found in diabetic kidneys, which was prevented by PDTC. Diabetes reduced catalase activity, and PDTC increased catalase and SOD activities in both control and diabetic kidneys. PDTC treatment reduced MMP-2 activity and iNOS and p65 NFκB nuclear expression found increased in diabetic kidneys. Our results show that the NFκB inhibitor PDTC reduces renal damage through reduction of Nox4, iNOS, macrophages, and MMP-2 in the alloxan-induced diabetic model. These findings suggest that PDTC inhibits alloxan kidney damage via antioxidative and anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Bruna Pinheiro Pereira
- Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais,, Brazil
| | - Gabriel Tavares do Valle
- Escola de Enfermagem de Ribeirão Preto (EERP), Universidade de São Paulo - USP, Sao Paulo, Brazil
| | - Bruno César Côrrea Salles
- Departamento de Análises Clínicas, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais, Brazil
| | - Karla Cristinne Mancini Costa
- Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais,, Brazil
| | - Marilene Lopes Ângelo
- Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais,, Brazil
| | - Larissa Helena Lobo Torres
- Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais,, Brazil
| | - Rômulo Dias Novaes
- Departamento de Biologia Estrutural, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais, Brazil
| | - Sílvia Graciela Ruginsk
- Departamento de Ciências Fisiológicas, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais, Brazil
| | - Carlos Renato Tirapelli
- Escola de Enfermagem de Ribeirão Preto (EERP), Universidade de São Paulo - USP, Sao Paulo, Brazil
| | | | - Carla Speroni Ceron
- Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais,, Brazil.
| |
Collapse
|
26
|
Han Y, Liu Y, Yang C, Gao C, Guo X, Cheng J. LncRNA CASC2 inhibits hypoxia-induced pulmonary artery smooth muscle cell proliferation and migration by regulating the miR-222/ING5 axis. Cell Mol Biol Lett 2020; 25:21. [PMID: 32206065 PMCID: PMC7079380 DOI: 10.1186/s11658-020-00215-y] [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: 09/10/2019] [Accepted: 03/04/2020] [Indexed: 02/07/2023] Open
Abstract
Background Pulmonary arterial hypertension (PAH) is often characterized by cell proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs). LncRNA cancer susceptibility candidate 2 (CASC2) has been revealed to be involved in PASMC injury in hypoxia-induced pulmonary hypertension. However, the exact molecular mechanisms whereby CASC2 regulates PASMC proliferation and migration are still incompletely understood. Methods The expression levels of CASC2, miR-222 and inhibitor of growth 5 (ING5) were measured using quantitative real-time polymerase chain reaction (qRT-PCR) or western blot, respectively. Cell proliferation was analyzed by Cell Counting Kit-8 (CCK-8) assay. Wound healing assay was used to analyze cell migration ability. The relationship between miR-222 and CASC2 or ING5 was confirmed using bioinformatics analysis, luciferase reporter assay and RNA immunoprecipitation assay. Results CASC2 was down-regulated in hypoxia-induced PASMCs in a dose- and time-dependent manner. Functional experiments showed that CASC2 overexpression could reverse hypoxia-induced proliferation and migration of PASMCs. Bioinformatics analysis indicated that CASC2 acted as a competing endogenous RNA of miR-222, thereby regulating the expression of ING5, the downstream target of miR-222, in PASMCs. In addition, rescue assay suggested that the inhibition mediated by CASC2 of hypoxia-induced PASMC proliferation and migration could be attenuated by miR-222 inhibition or ING5 overexpression. Conclusion CASC2 attenuated hypoxia-induced PASMC proliferation and migration by regulating the miR-222/ING5 axis to prevent vascular remodeling and the development of PAH, providing a novel insight and therapeutic strategy for hypoxia-induced PAH.
Collapse
Affiliation(s)
- Yan Han
- Department of Cardiology, Henan Province People's Hospital, Huazhongfuwai Hospital, No. 7, Weiwu Road, Jinshui area, Zhengzhou City, Henan P.R. China
| | - Yuhao Liu
- Department of Cardiology, Henan Province People's Hospital, Huazhongfuwai Hospital, No. 7, Weiwu Road, Jinshui area, Zhengzhou City, Henan P.R. China
| | - Chaokuan Yang
- Department of Cardiology, Henan Province People's Hospital, Huazhongfuwai Hospital, No. 7, Weiwu Road, Jinshui area, Zhengzhou City, Henan P.R. China
| | - Chuanyu Gao
- Department of Cardiology, Henan Province People's Hospital, Huazhongfuwai Hospital, No. 7, Weiwu Road, Jinshui area, Zhengzhou City, Henan P.R. China
| | - Xiaoyan Guo
- Department of Cardiology, Henan Province People's Hospital, Huazhongfuwai Hospital, No. 7, Weiwu Road, Jinshui area, Zhengzhou City, Henan P.R. China
| | - Jiangtao Cheng
- Department of Cardiology, Henan Province People's Hospital, Huazhongfuwai Hospital, No. 7, Weiwu Road, Jinshui area, Zhengzhou City, Henan P.R. China
| |
Collapse
|
27
|
Tseng V, Sutliff RL, Hart CM. Redox Biology of Peroxisome Proliferator-Activated Receptor-γ in Pulmonary Hypertension. Antioxid Redox Signal 2019; 31:874-897. [PMID: 30582337 PMCID: PMC6751396 DOI: 10.1089/ars.2018.7695] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Peroxisome proliferator-activated receptor-gamma (PPARγ) maintains pulmonary vascular health through coordination of antioxidant defense systems, inflammation, and cellular metabolism. Insufficient PPARγ contributes to pulmonary hypertension (PH) pathogenesis, whereas therapeutic restoration of PPARγ activity attenuates PH in preclinical models. Recent Advances: Numerous studies in the past decade have elucidated the complex mechanisms by which PPARγ in the pulmonary vasculature and right ventricle (RV) protects against PH. The scope of PPARγ-interconnected pathways continues to expand and includes induction of antioxidant genes, transrepression of inflammatory signaling, regulation of mitochondrial biogenesis and bioenergetic integrity, control of cell cycle and proliferation, and regulation of vascular tone through interactions with nitric oxide and endogenous vasoactive molecules. Furthermore, PPARγ interacts with an extensive regulatory network of transcription factors and microRNAs leading to broad impact on cell signaling. Critical Issues: Abundant evidence suggests that targeting PPARγ exerts diverse salutary effects in PH and represents a novel and potentially translatable therapeutic strategy. However, progress has been slowed by an incomplete understanding of how specific PPARγ pathways are critically disrupted across PH disease subtypes and lack of optimal pharmacological ligands. Future Directions: Recent studies indicate that ligand-induced post-translational modifications of the PPARγ receptor differentially induce therapeutic benefits versus adverse side effects of PPARγ receptor activation. Strategies to selectively target PPARγ activity in diseased cells of pulmonary circulation and RV, coupled with development of ligands designed to specifically regulate post-translational PPARγ modifications, may unlock the full therapeutic potential of this versatile master transcriptional and metabolic regulator in PH.
Collapse
Affiliation(s)
- Victor Tseng
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia.,Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Roy L Sutliff
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia.,Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - C Michael Hart
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia.,Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| |
Collapse
|
28
|
Alruwaili N, Kandhi S, Sun D, Wolin MS. Metabolism and Redox in Pulmonary Vascular Physiology and Pathophysiology. Antioxid Redox Signal 2019; 31:752-769. [PMID: 30403147 PMCID: PMC6708269 DOI: 10.1089/ars.2018.7657] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: This review considers how some systems controlling pulmonary vascular function are potentially regulated by redox processes to examine how and why conditions such as prolonged hypoxia, pathological mediators, and other factors promoting vascular remodeling contribute to the development of pulmonary hypertension (PH). Recent Advances and Critical Issues: Aspects of vascular remodeling induction mechanisms described are associated with shifts in glucose metabolism through the pentose phosphate pathway and increased cytosolic NADPH generation by glucose-6-phosphate dehydrogenase, increased glycolysis generation of cytosolic NADH and lactate, mitochondrial dysfunction associated with superoxide dismutase-2 depletion, changes in reactive oxygen species and iron metabolism, and redox signaling. Future Directions: The regulation and impact of hypoxia-inducible factor and the function of cGMP-dependent and redox regulation of protein kinase G are considered for their potential roles as key sensors and coordinators of redox and metabolic processes controlling the progression of vascular pathophysiology in PH, and how modulating aspects of metabolic and redox regulatory systems potentially function in beneficial therapeutic approaches.
Collapse
Affiliation(s)
- Norah Alruwaili
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Sharath Kandhi
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Dong Sun
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Michael S Wolin
- Department of Physiology, New York Medical College, Valhalla, New York
| |
Collapse
|
29
|
Yeligar SM, Kang BY, Bijli KM, Kleinhenz JM, Murphy TC, Torres G, San Martin A, Sutliff RL, Hart CM. PPARγ Regulates Mitochondrial Structure and Function and Human Pulmonary Artery Smooth Muscle Cell Proliferation. Am J Respir Cell Mol Biol 2019; 58:648-657. [PMID: 29182484 DOI: 10.1165/rcmb.2016-0293oc] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive disorder that causes significant morbidity and mortality despite existing therapies. PH pathogenesis is characterized by metabolic derangements that increase pulmonary artery smooth muscle cell (PASMC) proliferation and vascular remodeling. PH-associated decreases in peroxisome proliferator-activated receptor γ (PPARγ) stimulate PASMC proliferation, and PPARγ in coordination with PPARγ coactivator 1α (PGC1α) regulates mitochondrial gene expression and biogenesis. To further examine the impact of decreases in PPARγ expression on human PASMC (HPASMC) mitochondrial function, we hypothesized that depletion of either PPARγ or PGC1α perturbs mitochondrial structure and function to stimulate PASMC proliferation. To test this hypothesis, HPASMCs were exposed to hypoxia and treated pharmacologically with the PPARγ antagonist GW9662 or with siRNA against PPARγ or PGC1α for 72 hours. HPASMC proliferation (cell counting), target mRNA levels (qRT-PCR), target protein levels (Western blotting), mitochondria-derived H2O2 (confocal immunofluorescence), mitochondrial mass and fragmentation, and mitochondrial bioenergetic profiling were determined. Hypoxia or knockdown of either PPARγ or PGC1α increased HPASMC proliferation, enhanced mitochondria-derived H2O2, decreased mitochondrial mass, stimulated mitochondrial fragmentation, and impaired mitochondrial bioenergetics. Taken together, these findings provide novel evidence that loss of PPARγ diminishes PGC1α and stimulates derangements in mitochondrial structure and function that cause PASMC proliferation. Overexpression of PGC1α reversed hypoxia-induced HPASMC derangements. This study identifies additional mechanistic underpinnings of PH, and provides support for the notion of activating PPARγ as a novel therapeutic strategy in PH.
Collapse
Affiliation(s)
- Samantha M Yeligar
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| | - Bum-Yong Kang
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| | - Kaiser M Bijli
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| | - Jennifer M Kleinhenz
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| | - Tamara C Murphy
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| | - Gloria Torres
- 3 Division of Cardiology, Department of Medicine, Emory University, Atlanta, Georgia
| | - Alejandra San Martin
- 3 Division of Cardiology, Department of Medicine, Emory University, Atlanta, Georgia
| | - Roy L Sutliff
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| | - C Michael Hart
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| |
Collapse
|
30
|
Bromodomain and Extraterminal (BET) Protein Inhibition Restores Redox Balance and Inhibits Myofibroblast Activation. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1484736. [PMID: 31119153 PMCID: PMC6500679 DOI: 10.1155/2019/1484736] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/04/2019] [Accepted: 04/01/2019] [Indexed: 12/28/2022]
Abstract
Background and Objective Progressive pulmonary fibrosis is the main cause of death in patients with systemic sclerosis (SSc) with interstitial lung disease (ILD) and in those with idiopathic pulmonary fibrosis (IPF). Transforming growth factor-β (TGF-β) and NADPH oxidase- (NOX-) derived reactive oxygen species (ROS) are drivers of lung fibrosis. We aimed to determine the role of the epigenetic readers, bromodomain and extraterminal (BET) proteins in the regulation of redox balance in activated myofibroblasts. Methods In TGF-β-stimulated fibroblasts, we investigated the effect of the BET inhibitor JQ1 on the mRNA expression of the prooxidant gene NOX4 and the antioxidant gene superoxide dismutase (SOD2) by quantitative RT-PCR, the antioxidant transcription factor NF-E2-related factor 2 (Nrf2) activity by a reporter assay, and intracellular ROS levels by dichlorofluorescein staining. Myofibroblast activation was determined by α-smooth muscle actin immunocytochemistry. The role of specific BET protein isoforms in NOX4 gene regulation was studied by siRNA silencing and chromatin-immunoprecipitation. Results and Conclusions Affymetrix gene array analysis revealed increased NOX4 and reduced SOD2 expression in SSc and IPF fibroblasts. SOD2 silencing in non-ILD control fibroblasts induced a profibrotic phenotype. TGF-β increased NOX4 and inhibited SOD2 expression, while increasing ROS production and myofibroblast differentiation. JQ1 reversed the TGF-β-mediated NOX4/SOD2 imbalance and Nrf2 inactivation and attenuated ROS production and myofibroblast differentiation. The BET proteins Brd3 and Brd4 were shown to bind to the NOX4 promoter and drive TGF-β-induced NOX4 expression. Our data indicate a critical role of BET proteins in promoting redox imbalance and pulmonary myofibroblast activation and support BET bromodomain inhibitors as a potential therapy for fibrotic lung disease.
Collapse
|
31
|
Huetsch JC, Suresh K, Shimoda LA. Regulation of Smooth Muscle Cell Proliferation by NADPH Oxidases in Pulmonary Hypertension. Antioxidants (Basel) 2019; 8:antiox8030056. [PMID: 30841544 PMCID: PMC6466559 DOI: 10.3390/antiox8030056] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/25/2019] [Accepted: 03/01/2019] [Indexed: 02/07/2023] Open
Abstract
Hyperproliferation of pulmonary arterial smooth muscle cells is a key component of vascular remodeling in the setting of pulmonary hypertension (PH). Numerous studies have explored factors governing the changes in smooth muscle cell phenotype that lead to the increased wall thickness, and have identified various potential candidates. A role for reactive oxygen species (ROS) has been well documented in PH. ROS can be generated from a variety of sources, including mitochondria, uncoupled nitric oxide synthase, xanthine oxidase, and reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. In this article, we will review recent data supporting a role for ROS generated from NADPH oxidases in promoting pulmonary arterial smooth muscle cell proliferation during PH.
Collapse
Affiliation(s)
- John C Huetsch
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21224, USA.
| | - Karthik Suresh
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21224, USA.
| | - Larissa A Shimoda
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21224, USA.
| |
Collapse
|
32
|
Chen F, Wang H, Zhao J, Yan J, Meng H, Zhan H, Chen L, Yuan L. Grape seed proanthocyanidin inhibits monocrotaline-induced pulmonary arterial hypertension via attenuating inflammation: in vivo and in vitro studies. J Nutr Biochem 2019; 67:72-77. [PMID: 30856466 DOI: 10.1016/j.jnutbio.2019.01.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 01/16/2019] [Accepted: 01/29/2019] [Indexed: 12/13/2022]
Abstract
Inflammation in pulmonary arterioles initiates and maintains pathological processes in pulmonary arterial hypertension (PAH), and inhibition of it attenuates PAH development. Grape seed proanthocyanidin (GSP) is believed to be effective in protecting vascular system via inhibiting inflammation, while its effect on pulmonary circulation remains inconclusive. In this study, we made observations in monocrotaline (MCT)-induced PAH rats and found decreases in mean pulmonary arterial pressure, pulmonary vessel resistance, right ventricular hypertrophy index, percentage of medial wall thickness, percentage of medial wall area, and lung weight of wet and dry tissue ratio after GSP administration in vivo. At the cellular and molecular levels, we also found several effects of GSP on MCT-induced PAH: (a) endothelial nitric oxide synthase expression in lung tissue and plasma NO level were increased; (b) Ca2+ level in pulmonary arterial smooth muscle cell (PASMC) was decreased; (c) transcription of inflammatory factors such as myeloperoxidase, interleukin (IL)-1β, IL-6 and tumor necrosis factor alpha (TNF-α) was down-regulated in lung tissue; (d) nuclear factor-κB pathway was inhibited as IκBα was less phosphorylated; (e) TNFα-induced PASMC overproliferation could be inhibited. These results indicated a possible mechanism of GSP reversing pulmonary vascular remodeling and vascular contraction by inhibiting inflammation, and it may be useful for preventing PAH development.
Collapse
Affiliation(s)
- Fangzheng Chen
- The First Clinical College, Wenzhou Medical University, Wenzhou, P.R. China.
| | - Heng Wang
- Optometry & Ophthalmology College, Wenzhou Medical University, Wenzhou, P.R. China.
| | - Jie Zhao
- The First Clinical College, Wenzhou Medical University, Wenzhou, P.R. China.
| | - Junjie Yan
- Pharmacy College, Wenzhou Medical University, Wenzhou, P.R. China.
| | - Hanyan Meng
- Department of Pediatric, The Maternity Hospital of Zhejiang University, Hangzhou, P.R. China.
| | - Huilu Zhan
- The Second Clinical College, Wenzhou Medical University, Wenzhou, P.R. China.
| | - Luowei Chen
- The First Clinic Medicine College, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P.R. China.
| | - Linbo Yuan
- Department of Physiology, Basic Medicine School, Wenzhou Medical University, Wenzhou, P.R. China.
| |
Collapse
|
33
|
Rashid J, Nozik-Grayck E, McMurtry IF, Stenmark KR, Ahsan F. Inhaled combination of sildenafil and rosiglitazone improves pulmonary hemodynamics, cardiac function, and arterial remodeling. Am J Physiol Lung Cell Mol Physiol 2019; 316:L119-L130. [PMID: 30307312 PMCID: PMC6383494 DOI: 10.1152/ajplung.00381.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/01/2018] [Accepted: 10/01/2018] [Indexed: 01/15/2023] Open
Abstract
Currently, dual- or triple-drug combinations comprising different vasodilators are the mainstay for the treatment of pulmonary arterial hypertension (PAH). However, the patient outcome continues to be disappointing because the existing combination therapy cannot restrain progression of the disease. Previously, we have shown that when given as a monotherapy, long-acting inhaled formulations of sildenafil (a phosphodiesterase-5 inhibitor) and rosiglitazone (a peroxisome proliferator receptor-γ agonist) ameliorate PAH in rats. Thus, with a goal to develop a new combination therapy, we prepared and characterized poly(lactic-co-glycolic acid) (PLGA)-based long-acting inhalable particles of sildenafil and rosiglitazone. We then assessed the efficacy of the combinations of sildenafil and rosiglitazone, given in plain forms or as PLGA particles, in reducing mean pulmonary arterial pressure (mPAP) and improving pulmonary arterial remodeling and right ventricular hypertrophy (RVH) in Sugen 5416 plus hypoxia-induced PAH rats. After intratracheal administration of the formulations, we catheterized the rats and measured mPAP, cardiac output, total pulmonary resistance, and RVH. We also conducted morphometric studies using lung tissue samples and assessed the degree of muscularization, the arterial medial wall thickening, and the extent of collagen deposition. Compared with the plain drugs, given via the pulmonary or oral route as a single or dual combination, PLGA particles of the drugs, although given at a longer dosing interval compared with the plain drugs, caused more pronounced reduction in mPAP without affecting mean systemic pressure, improved cardiac function, slowed down right heart remodeling, and reduced arterial muscularization. Overall, PLGA particles of sildenafil and rosiglitazone, given as an inhaled combination, could be a viable alternative to currently available vasodilator-based combination therapy for PAH.
Collapse
Affiliation(s)
- Jahidur Rashid
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center , Amarillo, Texas
| | - Eva Nozik-Grayck
- Department of Pediatrics, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Ivan F McMurtry
- Department of Pharmacology, Center for Lung Biology, University of South Alabama , Mobile, Alabama
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Fakhrul Ahsan
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center , Amarillo, Texas
| |
Collapse
|
34
|
Murtaza G, Paddenberg R, Pfeil U, Goldenberg A, Mermer P, Kummer W. Hypoxia-induced pulmonary vasoconstriction of intra-acinar arteries is impaired in NADPH oxidase 4 gene-deficient mice. Pulm Circ 2018; 8:2045894018808240. [PMID: 30284509 PMCID: PMC6236867 DOI: 10.1177/2045894018808240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We show that genetic deficiency of the reactive oxygen species generating enzyme
NADPH oxidase 4 (NOX4) impairs hypoxic pulmonary vasoconstriction in small
(25–40 µm) intra-acinar, but not pre-acinar, arteries in murine precision cut
lung slices. These data suggest an involvement of NOX4 in ventilation-perfusion
matching at the acinar level.
Collapse
Affiliation(s)
- Ghulam Murtaza
- 1 Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany.,2 Department of Zoology, University of Gujrat, Gujrat, Pakistan
| | - Renate Paddenberg
- 1 Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Uwe Pfeil
- 1 Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Anna Goldenberg
- 1 Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Petra Mermer
- 1 Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Wolfgang Kummer
- 1 Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany.,3 German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
35
|
Rashid J, Alobaida A, Al-Hilal TA, Hammouda S, McMurtry IF, Nozik-Grayck E, Stenmark KR, Ahsan F. Repurposing rosiglitazone, a PPAR-γ agonist and oral antidiabetic, as an inhaled formulation, for the treatment of PAH. J Control Release 2018; 280:113-123. [PMID: 29723610 DOI: 10.1016/j.jconrel.2018.04.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/28/2018] [Indexed: 12/20/2022]
Abstract
Peroxisome-proliferator-activated-receptor-gamma (PPAR-γ) is implicated, in some capacity, in the pathogenesis of pulmonary arterial hypertension (PAH). Rosiglitazone, an oral antidiabetic and PPAR-γ agonist, has the potential to dilate pulmonary arteries and to attenuate arterial remodeling in PAH. Here, we sought to test the hypothesis that rosiglitazone can be repurposed as inhaled formulation for the treatment of PAH. We have tested this conjecture by preparing and optimizing poly(lactic-co-glycolic) acid (PLGA) based particles of rosiglitazone, assessing the drug particles for pulmonary absorption, investigating the efficacy of the plain versus particulate drug formulation in improving the respiratory hemodynamics in PAH animals, and finally studying the effect of the drug in regulating the molecular markers associated with PAH pathogenesis. The optimized particles were slightly porous and spherical, and released 87.9% ± 6.7% of the drug in 24 h. The elimination half-life of the drug formulated in PLGA particles was 2.5-fold greater than that of the plain drug administered via the same route at the same dose. The optimized formulation, given via the pulmonary route, produced pulmonary selective vasodilation in PAH animals, but oral rosiglitazone had no effect in pulmonary hemodynamics. Rosiglitazone ameliorates the pathogenesis of PAH by balancing the molecular regulators involved in the vasoconstriction and vasodilation of human pulmonary arterial smooth muscle cells. All in all, data generated using intact animal and cellular models point to the conclusion that PLGA particles of an antidiabetic drug can be used for the treatment of a different disease, PAH.
Collapse
Affiliation(s)
- Jahidur Rashid
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX 79106, USA
| | - Ahmad Alobaida
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX 79106, USA
| | - Taslim A Al-Hilal
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX 79106, USA
| | - Samia Hammouda
- The School of Sciences and Engineering, The American University in Cairo, Cairo, Egypt
| | - Ivan F McMurtry
- Department of Pharmacology, The Center for Lung Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Eva Nozik-Grayck
- Department of Pediatrics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Fakhrul Ahsan
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX 79106, USA.
| |
Collapse
|
36
|
The Role of Nicotinamide Adenine Dinucleotide Phosphate Oxidases in Lung Architecture Remodeling. Antioxidants (Basel) 2017; 6:antiox6040104. [PMID: 29257052 PMCID: PMC5745514 DOI: 10.3390/antiox6040104] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 02/07/2023] Open
Abstract
Chronic lung disorders, such as pulmonary artery hypertension (PAH), chronic obstructive pulmonary disease (COPD), asthma and neonatal bronchopulmonary dysplasia (BPD), are characterized by airway and/or vascular remodeling. Despite differences in the pathology, reactive oxygen species (ROS) have been highlighted as a critical contributor to the initiation and development of airway and vascular remodeling. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Nox) appear to play a pivotal role in lung signaling, leading to marked changes in pulmonary airway and vascular cell phenotypes, including proliferation, hypertrophy and apoptosis. In this review, we summarized the current literature regarding the role of Nox in the airway and vascular remodeling.
Collapse
|
37
|
You B, Liu Y, Chen J, Huang X, Peng H, Liu Z, Tang Y, Zhang K, Xu Q, Li X, Cheng G, Shi R, Zhang G. Vascular peroxidase 1 mediates hypoxia-induced pulmonary artery smooth muscle cell proliferation, apoptosis resistance and migration. Cardiovasc Res 2017; 114:188-199. [PMID: 29186367 DOI: 10.1093/cvr/cvx234] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 11/25/2017] [Indexed: 11/13/2022] Open
Abstract
Abstract
Aims
Reactive oxygen species (ROS) play essential roles in the pulmonary vascular remodelling associated with hypoxia-induced pulmonary hypertension (PH). Vascular peroxidase 1 (VPO1) is a newly identified haeme-containing peroxidase that accelerates oxidative stress development in the vasculature. This study aimed to determine the potential role of VPO1 in hypoxia-induced PH-related vascular remodelling.
Methods and results
The vascular morphology and VPO1 expression were assessed in the pulmonary arteries of Sprague–Dawley (SD) rats. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4) and VPO1 expression and HOCl production were significantly increased in hypoxic rats, which also exhibited obvious vascular remodelling. Furthermore, a hypoxia-induced PH model was generated by exposing primary rat pulmonary artery smooth muscle cells (PASMCs) to hypoxic conditions (3% O2, 48 h), which significantly increased the expression of NOX4 and VPO1 and the production of HOCl. These hypoxic changes were accompanied by enhanced proliferation, apoptosis resistance, and migration. In PASMCs, hypoxia-induced changes, including effects on the expression of cell cycle regulators (cyclin B1 and cyclin D1), apoptosis-related proteins (bax, bcl-2, and cleaved caspase-3), migration promoters (matrix metalloproteinases 2 and 9), and NF-κB expression, as well as the production of HOCl, were all inhibited by silencing VPO1 with small interfering RNAs. Moreover, treatment with HOCl under hypoxic conditions upregulated NF-κB expression and enhanced proliferation, apoptosis resistance, and migration in PASMCs, whereas BAY 11-7082 (an inhibitor of NF-κB) significantly inhibited these effects.
Conclusion
Collectively, these results demonstrate that VPO1 promotes hypoxia-induced proliferation, apoptosis resistance, and migration in PASMCs via the NOX4/VPO1/HOCl/NF-κB signalling pathway.
Collapse
Affiliation(s)
- Baiyang You
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yanbo Liu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Jia Chen
- Department of Humanistic Nursing, Xiangya Nursing School, Central South University, Changsha, China
| | - Xiao Huang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Huihui Peng
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zhaoya Liu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yixin Tang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Kai Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Xu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaohui Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Guangjie Cheng
- Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ruizheng Shi
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Guogang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
38
|
LeBlanc AJ, Kelm NQ. Thrombospondin-1, Free Radicals, and the Coronary Microcirculation: The Aging Conundrum. Antioxid Redox Signal 2017; 27:785-801. [PMID: 28762749 PMCID: PMC5647494 DOI: 10.1089/ars.2017.7292] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
SIGNIFICANCE Successful matching of cardiac metabolism to perfusion is accomplished primarily through vasodilation of the coronary resistance arterioles, but the mechanism that achieves this effect changes significantly as aging progresses and involves the contribution of reactive oxygen species (ROS). Recent Advances: A matricellular protein, thrombospondin-1 (Thbs-1), has been shown to be a prolific contributor to the production and modulation of ROS in large conductance vessels and in the peripheral circulation. Recently, the presence of physiologically relevant circulating Thbs-1 levels was proven to also disrupt vasodilation to nitric oxide (NO) in coronary arterioles from aged animals, negatively impacting coronary blood flow reserve. CRITICAL ISSUES This review seeks to reconcile how ROS can be successfully utilized as a substrate to mediate vasoreactivity in the coronary microcirculation as "normal" aging progresses, but will also examine how Thbs-1-induced ROS production leads to dysfunctional perfusion and eventual ischemia and why this is more of a concern in advancing age. FUTURE DIRECTIONS Current therapies that may effectively disrupt Thbs-1 and its receptor CD47 in the vascular wall and areas for future exploration will be discussed. Antioxid. Redox Signal. 27, 785-801.
Collapse
Affiliation(s)
- Amanda J LeBlanc
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville , Louisville, Kentucky
| | - Natia Q Kelm
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville , Louisville, Kentucky
| |
Collapse
|
39
|
Meitzler JL, Makhlouf HR, Antony S, Wu Y, Butcher D, Jiang G, Juhasz A, Lu J, Dahan I, Jansen-Dürr P, Pircher H, Shah AM, Roy K, Doroshow JH. Decoding NADPH oxidase 4 expression in human tumors. Redox Biol 2017; 13:182-195. [PMID: 28578276 PMCID: PMC5458090 DOI: 10.1016/j.redox.2017.05.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 05/23/2017] [Indexed: 12/27/2022] Open
Abstract
NADPH oxidase 4 (NOX4) is a redox active, membrane-associated protein that contributes to genomic instability, redox signaling, and radiation sensitivity in human cancers based on its capacity to generate H2O2 constitutively. Most studies of NOX4 in malignancy have focused on the evaluation of a small number of tumor cell lines and not on human tumor specimens themselves; furthermore, these studies have often employed immunological tools that have not been well characterized. To determine the prevalence of NOX4 expression across a broad range of solid tumors, we developed a novel monoclonal antibody that recognizes a specific extracellular region of the human NOX4 protein, and that does not cross-react with any of the other six members of the NOX gene family. Evaluation of 20 sets of epithelial tumors revealed, for the first time, high levels of NOX4 expression in carcinomas of the head and neck (15/19 patients), esophagus (12/18 patients), bladder (10/19 patients), ovary (6/17 patients), and prostate (7/19 patients), as well as malignant melanoma (7/15 patients) when these tumors were compared to histologically-uninvolved specimens from the same organs. Detection of NOX4 protein upregulation by low levels of TGF-β1 demonstrated the sensitivity of this new probe; and immunofluorescence experiments found that high levels of endogenous NOX4 expression in ovarian cancer cells were only demonstrable associated with perinuclear membranes. These studies suggest that NOX4 expression is upregulated, compared to normal tissues, in a well-defined, and specific group of human carcinomas, and that its expression is localized on intracellular membranes in a fashion that could modulate oxidative DNA damage.
Collapse
Affiliation(s)
- Jennifer L Meitzler
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Hala R Makhlouf
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Smitha Antony
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yongzhong Wu
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Donna Butcher
- Pathology/Histotechnology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21702, USA
| | - Guojian Jiang
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Agnes Juhasz
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jiamo Lu
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Iris Dahan
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research and Center for Molecular Biosciences Innsbruck (CMBI), Universität Innsbruck, 6020 Innsbruck, Austria
| | - Haymo Pircher
- Institute for Biomedical Aging Research and Center for Molecular Biosciences Innsbruck (CMBI), Universität Innsbruck, 6020 Innsbruck, Austria
| | - Ajay M Shah
- King's College London British Heart Foundation Centre, Cardiovascular Division, James Black Centre, London SE5 9NU, United Kingdom
| | - Krishnendu Roy
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - James H Doroshow
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA; Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
40
|
Yu B, Liu Z, Fu Y, Wang Y, Zhang L, Cai Z, Yu F, Wang X, Zhou J, Kong W. CYLD Deubiquitinates Nicotinamide Adenine Dinucleotide Phosphate Oxidase 4 Contributing to Adventitial Remodeling. Arterioscler Thromb Vasc Biol 2017; 37:1698-1709. [PMID: 28751569 DOI: 10.1161/atvbaha.117.309859] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 07/13/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Transdifferentiation of adventitial fibroblasts (AFs) into myofibroblasts plays a critical role during the vascular remodeling that occurs during atherosclerosis, restenosis, and aortic aneurysm. The ubiquitination/deubiquitination regulatory system is essential for the quality control of proteins. The involvement of ubiquitination/deubiquitination during AF transdifferentiation remains largely unknown. In this study, we determined the role of cylindromatosis (CYLD), a deubiquitinase, in the process of AF differentiation and activation in vitro and in vivo. APPROACH AND RESULTS Transforming growth factor-β1 and homocysteine, 2 known inducers of AF transdifferentiation, greatly upregulated CYLD expression in a time- and dose-dependent manner. The silencing of CYLD significantly inhibited AF transdifferentiation and activation as evidenced by the expression of contractile proteins, the production of the proinflammatory cytokines MCP-1 (monocyte chemotactic protein 1) and IL-6 (interleukin-6), the deposition of extracellular matrix, and cell migration. We further asked whether CYLD mediates AF activation via the regulation of nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) as it is an essential factor during AF transdifferentiation. Indeed, the silencing of CYLD repressed transforming growth factor-β1-induced and homocysteine-induced Nox4 upregulation and reactive oxygen species production, whereas Nox4 overexpression greatly rescued the inhibitory effect on AF activation by CYLD silencing. Most interestingly, transforming growth factor-β1 and homocysteine repressed Nox4 ubiquitination and prolonged the half-life of Nox4. Moreover, Nox4 was deubiquitinated via a direct interaction with the ubiquitin-specific protease domain of CYLD. In accordance, hyperhomocysteinemia significantly increased adventitial CYLD and Nox4 expression, promoted AF transdifferentiation, and aggravated CaPO4-induced abdominal aortic aneurysm in mice. These effects were abolished in CYLD-/- mice. CONCLUSIONS CYLD contributes to the transdifferentiation of AFs via deubiquitinating Nox4 and may play a role in vascular remodeling.
Collapse
Affiliation(s)
- Bing Yu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Ziyi Liu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Yi Fu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Yingbao Wang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Lu Zhang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Zeyu Cai
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Fang Yu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Xian Wang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Jun Zhou
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.).
| | - Wei Kong
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.).
| |
Collapse
|
41
|
Thrombospondins: A Role in Cardiovascular Disease. Int J Mol Sci 2017; 18:ijms18071540. [PMID: 28714932 PMCID: PMC5536028 DOI: 10.3390/ijms18071540] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/05/2017] [Accepted: 07/13/2017] [Indexed: 12/16/2022] Open
Abstract
Thrombospondins (TSPs) represent extracellular matrix (ECM) proteins belonging to the TSP family that comprises five members. All TSPs have a complex multidomain structure that permits the interaction with various partners including other ECM proteins, cytokines, receptors, growth factors, etc. Among TSPs, TSP1, TSP2, and TSP4 are the most studied and functionally tested. TSP1 possesses anti-angiogenic activity and is able to activate transforming growth factor (TGF)-β, a potent profibrotic and anti-inflammatory factor. Both TSP2 and TSP4 are implicated in the control of ECM composition in hypertrophic hearts. TSP1, TSP2, and TSP4 also influence cardiac remodeling by affecting collagen production, activity of matrix metalloproteinases and TGF-β signaling, myofibroblast differentiation, cardiomyocyte apoptosis, and stretch-mediated enhancement of myocardial contraction. The development and evaluation of TSP-deficient animal models provided an option to assess the contribution of TSPs to cardiovascular pathology such as (myocardial infarction) MI, cardiac hypertrophy, heart failure, atherosclerosis, and aortic valve stenosis. Targeting of TSPs has a significant therapeutic value for treatment of cardiovascular disease. The activation of cardiac TSP signaling in stress and pressure overload may be therefore beneficial.
Collapse
|
42
|
Kang BY, Park K, Kleinhenz JM, Murphy TC, Sutliff RL, Archer D, Hart CM. Peroxisome Proliferator-Activated Receptor γ Regulates the V-Ets Avian Erythroblastosis Virus E26 Oncogene Homolog 1/microRNA-27a Axis to Reduce Endothelin-1 and Endothelial Dysfunction in the Sickle Cell Mouse Lung. Am J Respir Cell Mol Biol 2017; 56:131-144. [PMID: 27612006 DOI: 10.1165/rcmb.2016-0166oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pulmonary hypertension (PH), a serious complication of sickle cell disease (SCD), causes significant morbidity and mortality. Although a recent study determined that hemin release during hemolysis triggers endothelial dysfunction in SCD, the pathogenesis of SCD-PH remains incompletely defined. This study examines peroxisome proliferator-activated receptor γ (PPARγ) regulation in SCD-PH and endothelial dysfunction. PH and right ventricular hypertrophy were studied in Townes humanized sickle cell (SS) and littermate control (AA) mice. In parallel studies, SS or AA mice were gavaged with the PPARγ agonist, rosiglitazone (RSG), 10 mg/kg/day, or vehicle for 10 days. In vitro, human pulmonary artery endothelial cells (HPAECs) were treated with vehicle or hemin for 72 hours, and selected HPAECs were treated with RSG. SS mice developed PH and right ventricular hypertrophy associated with reduced lung levels of PPARγ and increased levels of microRNA-27a (miR-27a), v-ets avian erythroblastosis virus E26 oncogene homolog 1 (ETS1), endothelin-1 (ET-1), and markers of endothelial dysfunction (platelet/endothelial cell adhesion molecule 1 and E selectin). HPAECs treated with hemin had increased ETS1, miR-27a, ET-1, and endothelial dysfunction and decreased PPARγ levels. These derangements were attenuated by ETS1 knockdown, inhibition of miR-27a, or PPARγ overexpression. In SS mouse lung or in hemin-treated HPAECs, activation of PPARγ with RSG attenuated reductions in PPARγ and increases in miR-27a, ET-1, and markers of endothelial dysfunction. In SCD-PH pathogenesis, ETS1 stimulates increases in miR-27a levels that reduce PPARγ and increase ET-1 and endothelial dysfunction. PPARγ activation attenuated SCD-associated signaling derangements, suggesting a novel therapeutic approach to attenuate SCD-PH pathogenesis.
Collapse
Affiliation(s)
- Bum-Yong Kang
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - Kathy Park
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - Jennifer M Kleinhenz
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - Tamara C Murphy
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - Roy L Sutliff
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - David Archer
- 2 Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - C Michael Hart
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| |
Collapse
|
43
|
The Role of NOX4 and TRX2 in Angiogenesis and Their Potential Cross-Talk. Antioxidants (Basel) 2017; 6:antiox6020042. [PMID: 28594389 PMCID: PMC5488022 DOI: 10.3390/antiox6020042] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022] Open
Abstract
The nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) family is the major source of reactive oxygen species (ROS) in the vascular system. In this family, NOX4, a constitutive active form of NOXs, plays an important role in angiogenesis. Thioredoxin 2 (TRX2) is a key mitochondrial redox protein that maintains normal protein function and also provides electrons to peroxiredoxin 3 (PRX3) to scavenge H₂O₂ in mitochondria. Angiogenesis, a process of new blood vessel formation, is involved in a variety of physiological processes and pathological conditions. It seems to be paradoxical for ROS-producing NOX4 and ROS-scavenging TRX2 to have a similar role in promoting angiogenesis. In this review, we will focus on data supporting the role of NOX4 and TRX2 in angiogenesis and their cross-talks and discuss how ROS can positively or negatively regulate angiogenesis, depending on their species, levels and locations. NOX4 and TRX2-mediated ROS signaling could be promising targets for the treatment of angiogenesis-related diseases.
Collapse
|
44
|
Teixeira G, Szyndralewiez C, Molango S, Carnesecchi S, Heitz F, Wiesel P, Wood JM. Therapeutic potential of NADPH oxidase 1/4 inhibitors. Br J Pharmacol 2017; 174:1647-1669. [PMID: 27273790 PMCID: PMC5446584 DOI: 10.1111/bph.13532] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 12/16/2022] Open
Abstract
The NADPH oxidase (NOX) family of enzymes produces ROS as their sole function and is becoming recognized as key modulators of signal transduction pathways with a physiological role under acute stress and a pathological role after excessive activation under chronic stress. The seven isoforms differ in their regulation, tissue and subcellular localization and ROS products. The most studied are NOX1, 2 and 4. Genetic deletion of NOX1 and 4, in contrast to NOX2, has revealed no significant spontaneous pathologies and a pathogenic relevance of both NOX1 and 4 across multiple organs in a wide range of diseases and in particular inflammatory and fibrotic diseases. This has stimulated interest in NOX inhibitors for therapeutic application. GKT136901 and GKT137831 are two structurally related compounds demonstrating a preferential inhibition of NOX1 and 4 that have suitable properties for in vivo studies and have consequently been evaluated across a range of disease models and compared with gene deletion. In contrast to gene deletion, these inhibitors do not completely suppress ROS production, maintaining some basal level of ROS. Despite this and consistent with most gene deletion studies, these inhibitors are well tolerated and slow or prevent disease progression in a range of models of chronic inflammatory and fibrotic diseases by modulating common signal transduction pathways. Clinical trials in patients with GKT137831 have demonstrated excellent tolerability and reduction of various markers of chronic inflammation. NOX1/4 inhibition may provide a safe and effective therapeutic strategy for a range of inflammatory and fibrotic diseases. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- G Teixeira
- Evotec International GmbHGoettingenGermany
| | | | - S Molango
- Genkyotex SAPlan les OuatesSwitzerland
| | | | - F Heitz
- Genkyotex SAPlan les OuatesSwitzerland
| | - P Wiesel
- Genkyotex SAPlan les OuatesSwitzerland
| | | |
Collapse
|
45
|
Forte M, Palmerio S, Yee D, Frati G, Sciarretta S. Functional Role of Nox4 in Autophagy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:307-326. [DOI: 10.1007/978-3-319-55330-6_16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
46
|
Green DE, Murphy TC, Kang BY, Bedi B, Yuan Z, Sadikot RT, Hart CM. Peroxisome proliferator-activated receptor-γ enhances human pulmonary artery smooth muscle cell apoptosis through microRNA-21 and programmed cell death 4. Am J Physiol Lung Cell Mol Physiol 2017; 313:L371-L383. [PMID: 28522568 DOI: 10.1152/ajplung.00532.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 05/01/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive disorder whose cellular pathogenesis involves enhanced smooth muscle cell (SMC) proliferation and resistance to apoptosis signals. Existing evidence demonstrates that the tumor suppressor programmed cell death 4 (PDCD4) affects patterns of cell growth and repair responses in the systemic vasculature following experimental injury. In the current study, the regulation PDCD4 and its functional effects on growth and apoptosis susceptibility in pulmonary artery smooth muscle cells were explored. We previously demonstrated that pharmacological activation of the nuclear transcription factor peroxisome proliferator-activated receptor-γ (PPARγ) attenuated hypoxia-induced proliferation of human pulmonary artery smooth muscle cells (HPASMCs) by inhibiting the expression and mitogenic functions of microRNA-21 (miR-21). In the current study, we hypothesize that PPARγ stimulates PDCD4 expression and HPASMC apoptosis by inhibiting miR-21. Our findings demonstrate that PDCD4 is reduced in the mouse lung upon exposure to chronic hypoxia (10% O2 for 3 wk) and in hypoxia-exposed HPASMCs (1% O2). HPASMC apoptosis was reduced by hypoxia, by miR-21 overexpression, or by siRNA-mediated PPARγ and PDCD4 depletion. Activation of PPARγ inhibited miR-21 expression and resultant proliferation, while restoring PDCD4 levels and apoptosis to baseline. Additionally, pharmacological activation of PPARγ with rosiglitazone enhanced PDCD4 protein expression and apoptosis in a dose-dependent manner as demonstrated by increased annexin V detection by flow cytometry. Collectively, these findings demonstrate that PPARγ confers growth-inhibitory signals in hypoxia-exposed HPASMCs through suppression of miR-21 and the accompanying derepression of PDCD4 that augments HPASMC susceptibility to undergo apoptosis.
Collapse
Affiliation(s)
- David E Green
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| | - Tamara C Murphy
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| | - Bum-Yong Kang
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| | - Brahmchetna Bedi
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| | - Zhihong Yuan
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| | - Ruxana T Sadikot
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| | - C Michael Hart
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| |
Collapse
|
47
|
Xie X, Li S, Zhu Y, Liu L, Pan Y, Wang J, Shi W, Song Y, Yang L, Gao L, Zang W, Li M. MicroRNA-27a/b mediates endothelin-1-induced PPARγ reduction and proliferation of pulmonary artery smooth muscle cells. Cell Tissue Res 2017; 369:527-539. [PMID: 28484848 DOI: 10.1007/s00441-017-2625-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/05/2017] [Indexed: 11/27/2022]
Abstract
The down-regulation of peroxisome proliferator-activated receptor γ (PPARγ) expression has been found to correlate with the proliferation of pulmonary artery smooth muscle cells (PASMC), pulmonary vascular remodeling and pulmonary hypertension, while the molecular mechanisms underlying PPARγ reduction in PASMC remain largely unclear. The aim of the current study is to address this issue. Endothelin-1 (ET-1) dose- and time-dependently resulted in PPARγ reduction and proliferation of primary cultured rat PASMC, which was accompanied by the activation of nuclear factor-kappaB (NF-κB) and subsequent induction of microRNA-27a/b (miR-27a/b) expression. Chromatin immunoprecipitation assay revealed that NF-κB directly bound to the promoter regions of miR-27a/b. Luciferase reporter assay identified that miR-27a/b directly regulates the expression of PPARγ in PASMC. Further study indicated that the presence of either NF-κB inhibitor pyrrolidinedithiocarbamate or prior silencing miR-27a/b with anti-miRNA oligonucleotides suppressed ET-1-induced PPARγ reduction and proliferation of PASMC, while overexpression of miR-27a/b reduced PPARγ expression and enhanced PASMC proliferation. Taken together, our study demonstrates that ET-1 stimulates miR-27a/b expression by activation of the NF-κB pathway, which in turn results in PPARγ reduction and contributes to ET-1-induced PASMC proliferation.
Collapse
Affiliation(s)
- Xinming Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Yanting Zhu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Lu Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Yilin Pan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Yang Song
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Lan Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Li Gao
- Division of Allergy and Clinical Immunology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Weijin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xian Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061.
| |
Collapse
|
48
|
Bedi B, Maurice NM, Ciavatta VT, Lynn KS, Yuan Z, Molina SA, Joo M, Tyor WR, Goldberg JB, Koval M, Hart CM, Sadikot RT. Peroxisome proliferator-activated receptor-γ agonists attenuate biofilm formation by Pseudomonas aeruginosa. FASEB J 2017; 31:3608-3621. [PMID: 28442545 DOI: 10.1096/fj.201700075r] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/11/2017] [Indexed: 12/20/2022]
Abstract
Pseudomonas aeruginosa is a significant contributor to recalcitrant multidrug-resistant infections, especially in immunocompromised and hospitalized patients. The pathogenic profile of P. aeruginosa is related to its ability to secrete a variety of virulence factors and to promote biofilm formation. Quorum sensing (QS) is a mechanism wherein P. aeruginosa secretes small diffusible molecules, specifically acyl homo serine lactones, such as N-(3-oxo-dodecanoyl)-l-homoserine lactone (3O-C12-HSL), that promote biofilm formation and virulence via interbacterial communication. Strategies that strengthen the host's ability to inhibit bacterial virulence would enhance host defenses and improve the treatment of resistant infections. We have recently shown that peroxisome proliferator-activated receptor γ (PPARγ) agonists are potent immunostimulators that play a pivotal role in host response to virulent P. aeruginosa Here, we show that QS genes in P. aeruginosa (strain PAO1) and 3O-C12-HSL attenuate PPARγ expression in bronchial epithelial cells. PAO1 and 3O-C12-HSL induce barrier derangements in bronchial epithelial cells by lowering the expression of junctional proteins, such as zonula occludens-1, occludin, and claudin-4. Expression of these proteins was restored in cells that were treated with pioglitazone, a PPARγ agonist, before infection with PAO1 and 3O-C12-HSL. Barrier function and bacterial permeation studies that have been performed in primary human epithelial cells showed that PPARγ agonists are able to restore barrier integrity and function that are disrupted by PAO1 and 3O-C12-HSL. Mechanistically, we show that these effects are dependent on the induction of paraoxonase-2, a QS hydrolyzing enzyme, that mitigates the effects of QS molecules. Importantly, our data show that pioglitazone, a PPARγ agonist, significantly inhibits biofilm formation on epithelial cells by a mechanism that is mediated via paraoxonase-2. These findings elucidate a novel role for PPARγ in host defense against P. aeruginosa Strategies that activate PPARγ can provide a therapeutic complement for treatment of resistant P. aeruginosa infections.-Bedi, B., Maurice, N. M., Ciavatta, V. T., Lynn, K. S., Yuan, Z., Molina, S. A., Joo, M., Tyor, W. R., Goldberg, J. B., Koval, M., Hart, C. M., Sadikot, R. T. Peroxisome proliferator-activated receptor-γ agonists attenuate biofilm formation by Pseudomonas aeruginosa.
Collapse
Affiliation(s)
- Brahmchetna Bedi
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA.,Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, Georgia, USA
| | - Nicholas M Maurice
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA.,Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, Georgia, USA
| | - Vincent T Ciavatta
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA.,Department of Ophthalmology, Emory University, Atlanta, Georgia, USA
| | - K Sabrina Lynn
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, Georgia, USA
| | - Zhihong Yuan
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA.,Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, Georgia, USA
| | - Samuel A Molina
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, Georgia, USA.,Emory + Children's Center for Cystic Fibrosis and Airway Disease Research, Emory University, Atlanta, Georgia, USA
| | - Myungsoo Joo
- Department of Immunology, Pusan University, Yangsan, South Korea
| | - William R Tyor
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA.,Department of Neurology, Emory University, Atlanta, Georgia, USA
| | | | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, Georgia, USA.,Emory + Children's Center for Cystic Fibrosis and Airway Disease Research, Emory University, Atlanta, Georgia, USA.,Department of Cell Biology, Emory University, Atlanta, Georgia, USA
| | - C Michael Hart
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA.,Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, Georgia, USA
| | - Ruxana T Sadikot
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA; .,Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
49
|
Zhang L, Li Z, Feng D, Shen H, Tian X, Li H, Wang Z, Chen G. Involvement of Nox2 and Nox4 NADPH oxidases in early brain injury after subarachnoid hemorrhage. Free Radic Res 2017; 51:316-328. [PMID: 28330417 DOI: 10.1080/10715762.2017.1311015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Li Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhen Li
- Department of Neurosurgery, Jingjiang People’s Hospital, Jiangsu, Jiangsu Province, China
| | | | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiaodi Tian
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
50
|
Chaudhry A, Carthan KA, Kang BY, Calvert J, Sutliff RL, Michael Hart C. PPARγ attenuates hypoxia-induced hypertrophic transcriptional pathways in the heart. Pulm Circ 2017; 7:98-107. [PMID: 28680569 PMCID: PMC5448534 DOI: 10.1086/689749] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/15/2016] [Indexed: 02/01/2023] Open
Abstract
Chronic hypoxia-induced pulmonary hypertension (PH) is characterized by increased pressure and resistance in the pulmonary vasculature and hypertrophy of the right ventricle (RV). The transcription factors, nuclear factor activated T-cells (NFAT), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB/p65) contribute to RV hypertrophy (RVH). Because peroxisome proliferator-activated receptor gamma (PPARγ) activation attenuates hypoxia-induced PH and RVH, we hypothesized that PPARγ inhibits activation of RV hypertrophic transcriptional signaling mechanisms. C57BL/6J mice were exposed to normoxia (21% O2) or hypoxia (10% O2) for 21 days. During the final 10 days of exposure, selected mice were treated with the PPARγ ligand, pioglitazone. RV systolic pressure (RVSP) and RVH were measured, and NFATc2 and NF-kB/p65 protein levels were measured in RV and LV nuclear and cytosolic fractions. Cardiomyocyte hypertrophy was assessed with wheatgerm agglutinin staining. NFAT activation was also examined with luciferase reporter mice and analysis of protein levels of selected transcriptional targets. Chronic-hypoxia increased: (1) RVH, RVSP, and RV cardiomyocyte hypertrophy; (2) NFATc2 and NF-κB activation in RV nuclear homogenates; (3) RV and LV NFAT luciferase activity; and (4) RV protein levels of brain natriuretic peptide (BNP) and β-myosin heavy chain (β-MyHC). Treatment with pioglitazone attenuated hypoxia-induced increases in both RV and LV NFAT luciferase activity. Chronic hypoxia caused sustained RV NFATc2 and NF-κB activation. Pioglitazone attenuated PH, RVH, cardiomyocyte hypertrophy, and activation of RV hypertrophic signaling and also attenuated LV NFAT activation. PPARγ favorably modulates signaling derangements in the heart as well as in the pulmonary vascular wall.
Collapse
Affiliation(s)
- Abubakr Chaudhry
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| | - Kristal A Carthan
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| | - Bum-Yong Kang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| | - John Calvert
- Department of Surgery, Emory University, Atlanta, GA, USA
| | - Roy L Sutliff
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| | - C Michael Hart
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| |
Collapse
|