1
|
Guers JJ, Heffernan KS, Campbell SC. Getting to the Heart of the Matter: Exploring the Intersection of Cardiovascular Disease, Sex and Race and How Exercise, and Gut Microbiota Influence these Relationships. Rev Cardiovasc Med 2025; 26:26430. [PMID: 40026503 PMCID: PMC11868917 DOI: 10.31083/rcm26430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/16/2024] [Accepted: 11/28/2024] [Indexed: 03/05/2025] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, with physical inactivity being a known contributor to the global rates of CVD incidence. CVD incidence, however, is not uniform with recognized sex differences as well and racial and ethnic differences. Furthermore, gut microbiota have been associated with CVD, sex, and race/ethnicity. Researchers have begun to examine the interplay of these complicated yet interrelated topics. This review will present evidence that CVD (risk and development), and gut microbiota are distinct between the sexes and racial/ethnic groups, which appear to be influenced by acculturation, discrimination, stress, and lifestyle factors like exercise. Furthermore, this review will address the beneficial impacts of exercise on the cardiovascular system and will provide recommendations for future research in the field.
Collapse
Affiliation(s)
- John J. Guers
- Department of Health Sciences and Nursing, Rider University, Lawrenceville, NJ 08648, USA
| | - Kevin S. Heffernan
- Department of Biobehavioral Sciences, Teachers College, Columbia University, New York, NY 10027, USA
| | - Sara C. Campbell
- Department of Kinesiology and Health, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Centers for Human Nutrition, Exercise, and Metabolism, Nutrition, Microbiome, and Health, and Lipid Research, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
2
|
Poyatos P, Gratacós M, Samuel K, Orriols R, Tura-Ceide O. Oxidative Stress and Antioxidant Therapy in Pulmonary Hypertension. Antioxidants (Basel) 2023; 12:1006. [PMID: 37237872 PMCID: PMC10215203 DOI: 10.3390/antiox12051006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive disease characterized by elevated artery pressures and pulmonary vascular resistance. Underlying mechanisms comprise endothelial dysfunction, pulmonary artery remodeling and vasoconstriction. Several studies have shown evidence of the critical role of oxidative stress in PH pathophysiology. Alteration of redox homeostasis produces excessive generation of reactive oxygen species, inducing oxidative stress and the subsequent alteration of biological molecules. Exacerbations in oxidative stress production can lead to alterations in nitric oxide signaling pathways, contributing to the proliferation of pulmonary arterial endothelial cells and smooth muscle cells, inducing PH development. Recently, antioxidant therapy has been suggested as a novel therapeutic strategy for PH pathology. However, the favorable outcomes observed in preclinical studies have not been consistently reproduced in clinical practice. Therefore, targeting oxidative stress as a therapeutic intervention for PH is an area that is still being explored. This review summarizes the contribution of oxidative stress to the pathogenesis of the different types of PH and suggests antioxidant therapy as a promising strategy for PH treatment.
Collapse
Affiliation(s)
- Paula Poyatos
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain; (P.P.); (M.G.)
- Department of Medical Sciences, Faculty of Medicine, University of Girona, 17003 Girona, Spain
| | - Miquel Gratacós
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain; (P.P.); (M.G.)
| | - Kay Samuel
- Scottish National Blood Transfusion Service, NHS National Services Scotland, Edinburgh EH14 4BE, UK
| | - Ramon Orriols
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain; (P.P.); (M.G.)
- Department of Medical Sciences, Faculty of Medicine, University of Girona, 17003 Girona, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain; (P.P.); (M.G.)
- Department of Medical Sciences, Faculty of Medicine, University of Girona, 17003 Girona, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
3
|
Cai B, Yang L, Do Jung Y, Zhang Y, Liu X, Zhao P, Li J. PTEN: An Emerging Potential Target for Therapeutic Intervention in Respiratory Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4512503. [PMID: 35814272 PMCID: PMC9262564 DOI: 10.1155/2022/4512503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 04/22/2022] [Accepted: 05/19/2022] [Indexed: 12/13/2022]
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a potent tumor suppressor that regulates several key cellular processes, including proliferation, survival, genomic integrity, migration, and invasion, via PI3K-dependent and independent mechanisms. A subtle decrease in PTEN levels or catalytic activity is implicated not only in cancer but also in a wide spectrum of other diseases, including various respiratory diseases. A systemic overview of the advances in the molecular and cellular mechanisms of PTEN involved in the initiation and progression of respiratory diseases may offer novel targets for the development of effective therapeutics for the treatment of respiratory diseases. In the present review, we highlight the novel findings emerging from current research on the role of PTEN expression and regulation in airway pathological conditions such as asthma/allergic airway inflammation, pulmonary hypertension (PAH), chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and other acute lung injuries (ALI). Moreover, we discuss the clinical implications of PTEN alteration and recently suggested therapeutic possibilities for restoration of PTEN expression and function in respiratory diseases.
Collapse
Affiliation(s)
- Bangrong Cai
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P.R. Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, China
- Henan Research Center for Special Processing Technology of Chinese Medicine, School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Liu Yang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P.R. Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, China
| | - Young Do Jung
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Republic of Korea
| | - Ying Zhang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xinguang Liu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P.R. Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, China
| | - Peng Zhao
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P.R. Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, China
| | - Jiansheng Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P.R. Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, China
- Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| |
Collapse
|
4
|
Abstract
The development of pulmonary hypertension (PH) is common and has adverse prognostic implications in patients with heart failure due to left heart disease (LHD), and thus far, there are no known treatments specifically for PH-LHD, also known as group 2 PH. Diagnostic thresholds for PH-LHD, and clinical classification of PH-LHD phenotypes, continue to evolve and, therefore, present a challenge for basic and translational scientists actively investigating PH-LHD in the preclinical setting. Furthermore, the pathobiology of PH-LHD is not well understood, although pulmonary vascular remodeling is thought to result from (1) increased wall stress due to increased left atrial pressures; (2) hemodynamic congestion-induced decreased shear stress in the pulmonary vascular bed; (3) comorbidity-induced endothelial dysfunction with direct injury to the pulmonary microvasculature; and (4) superimposed pulmonary arterial hypertension risk factors. To ultimately be able to modify disease, either by prevention or treatment, a better understanding of the various drivers of PH-LHD, including endothelial dysfunction, abnormalities in vascular tone, platelet aggregation, inflammation, adipocytokines, and systemic complications (including splanchnic congestion and lymphatic dysfunction) must be further investigated. Here, we review the diagnostic criteria and various hemodynamic phenotypes of PH-LHD, the potential biological mechanisms underlying this disorder, and pressing questions yet to be answered about the pathobiology of PH-LHD.
Collapse
Affiliation(s)
- Jessica H Huston
- Division of Cardiology, Department of Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA (J.H.H.)
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.)
| |
Collapse
|
5
|
Liu S, Yan Y. Animal models of pulmonary hypertension due to left heart disease. Animal Model Exp Med 2022; 5:197-206. [PMID: 35234367 PMCID: PMC9240728 DOI: 10.1002/ame2.12214] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/13/2022] [Accepted: 01/23/2022] [Indexed: 01/02/2023] Open
Abstract
Pulmonary hypertension due to left heart disease (PH‐LHD) is regarded as the most prevalent form of pulmonary hypertension (PH). Indeed, PH is an independent risk factor and predicts adverse prognosis for patients with left heart disease (LHD). Clinically, there are no drugs or treatments that directly address PH‐LHD, and treatment of LHD alone will not also ameliorate PH. To target the underlying physiopathological alterations of PH‐LHD and to develop novel therapeutic approaches for this population, animal models that simulate the pathophysiology of PH‐LHD are required. There are several available models for PH‐LHD that have been successfully employed in rodents or large animals by artificially provoking an elevated pressure load on the left heart, which by transduction elicits an escalated pressure in pulmonary artery. In addition, metabolic derangement combined with aortic banding or vascular endothelial growth factor receptor antagonist is also currently applied to reproduce the phenotype of PH‐LHD. As of today, none of the animal models exactly recapitulates the condition of patients with PH‐LHD. Nevertheless, the selection of an appropriate animal model is essential in basic and translational studies of PH‐LHD. Therefore, this review will summarize the characteristics of each PH‐LHD animal model and discuss the advantages and limitations of the different models.
Collapse
Affiliation(s)
- Shao‐Fei Liu
- Charité—Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin Berlin Germany
| | - Yi Yan
- Institute for Cardiovascular Prevention (IPEK) Ludwig‐Maximilians‐University Munich Munich Germany
- DZHK (German Centre for Cardiovascular Research) Partner Site Munich Heart Alliance Munich Germany
| |
Collapse
|
6
|
Bartolo MA, Qureshi MU, Colebank MJ, Chesler NC, Olufsen MS. Numerical predictions of shear stress and cyclic stretch in pulmonary hypertension due to left heart failure. Biomech Model Mechanobiol 2022; 21:363-381. [PMID: 35037114 PMCID: PMC10174274 DOI: 10.1007/s10237-021-01538-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022]
Abstract
Isolated post-capillary pulmonary hypertension (Ipc-PH) occurs due to left heart failure, which contributes to 1 out of every 9 deaths in the United States. In some patients, through unknown mechanisms, Ipc-PH transitions to combined pre-/post-capillary PH (Cpc-PH) and is associated with a dramatic increase in mortality. Altered mechanical forces and subsequent biological signaling in the pulmonary vascular bed likely contribute to the transition from Ipc-PH to Cpc-PH. However, even in a healthy pulmonary circulation, the mechanical forces in the smallest vessels (the arterioles, capillary bed, and venules) have not been quantitatively defined. This study is the first to examine this question via a computational fluid dynamics model of the human pulmonary arteries, arterioles, venules, and veins. Using this model, we predict temporal and spatial dynamics of cyclic stretch and wall shear stress with healthy and diseased hemodynamics. In the normotensive case for large vessels, numerical simulations show that large arteries have higher pressure and flow than large veins, as well as more pronounced changes in area throughout the cardiac cycle. In the microvasculature, shear stress increases and cyclic stretch decreases as vessel radius decreases. When we impose an increase in left atrial pressure to simulate Ipc-PH, shear stress decreases and cyclic stretch increases as compared to the healthy case. Overall, this model predicts pressure, flow, shear stress, and cyclic stretch that providing a way to analyze and investigate hypotheses related to disease progression in the pulmonary circulation.
Collapse
Affiliation(s)
- Michelle A Bartolo
- Department of Mathematics, North Carolina State University, Raleigh, NC, 27695, USA
| | - M Umar Qureshi
- Department of Mathematics, North Carolina State University, Raleigh, NC, 27695, USA
| | - Mitchel J Colebank
- Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, CA, 92697, USA
| | - Naomi C Chesler
- Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, CA, 92697, USA
| | - Mette S Olufsen
- Department of Mathematics, North Carolina State University, Raleigh, NC, 27607, USA.
| |
Collapse
|
7
|
Peng H, Zhou L, Li H, Zhang Y, Cheng S, Chen Z, Yu S, Hu S, Chen W, Ouyang M, Xue J, Zeng W. The therapeutic effect and mechanism of Rapamycin combined with HO-3867 on monocrotaline-induced pulmonary hypertension in rats. Eur J Pharm Sci 2021; 170:106102. [PMID: 34958883 DOI: 10.1016/j.ejps.2021.106102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 01/11/2023]
Abstract
This study test was designed to investigate the possible modulatory effect of rapamycin combined with HO-3867 in monocrotaline(MCT)-induced pulmonary arterial hypertension in rats. We hypothesized that combined treatment with rapamycin and HO-3867 is superior to either alone in attenuating MCT-induced rat pulmonary arterial hypertension (PAH). Pulmonary arterial hypertension was induced by a single intraperitoneal injection of monocrotaline (60 mg/kg). 2 weeks later, rapamycin (2 mg/kg i.p.) and HO3867 (10 mg/kg i.h.) were administered daily, alone and in combination, for 2 weeks. Right ventricular systolic pressure, echocardiography were recorded and then rats were sacrificed. Histological analysis of pulmonary arteries medial wall thickness, right ventricular hypertrophy index (RVHI), the ratio of right ventricular to body weight, and collagen volume fraction (CVF) of right ventricular were performed. Moreover, the expression of t-STAT3, p-STAT3, t-Akt, p-Akt in lung and t-STAT3, p-STAT3, t-S6, p-S6 in right ventricular were examined. The result showed that combined treatment provided a considerable improvement toward maintaining hemodynamic changes, lung vascular remodeling as well as amending RV remodeling and function. Furthermore, Combined treatment can normalize the protein levels of two signal pathways in lung and heart tissue, where p-S6 or p-Akt significantly decreased compared to HO-3867 alone, or p-STAT3 significantly reduced compared to rapamycin alone. In conclusion, combined treatment with rapamycin and HO-3867 is superior to either alone in attenuating MCT-induced PAH in rats.
Collapse
Affiliation(s)
- Huajing Peng
- Cardiovascular Department of the Sixth Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong Province, China
| | - Ling Zhou
- Ultrasonic Department, Hospital of South China University of Technology, 510000, Guangzhou, China
| | - Huayang Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 510000, Guangzhou, Guangdong Province, China
| | - Yitao Zhang
- Cardiovascular Department of the Sixth Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong Province, China
| | - Shiyao Cheng
- Cardiovascular Department of the Sixth Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong Province, China
| | - Zhichong Chen
- Cardiovascular Department of the Sixth Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong Province, China
| | - Shuqi Yu
- Cardiovascular Department of the Sixth Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong Province, China
| | - Sutian Hu
- Cardiovascular Department of the Sixth Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong Province, China
| | - Wenzeng Chen
- Department of Cardiac Surgery, Sun Yet-sen Memorial Hospital, 510000, Guangzhou, China
| | - Mao Ouyang
- Cardiovascular Department of the Sixth Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong Province, China
| | - Jiaojie Xue
- Cardiovascular Department of the Sixth Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong Province, China.
| | - Weijie Zeng
- Cardiovascular Department of the Sixth Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong Province, China.
| |
Collapse
|
8
|
Ravi Y, Sai-Sudhakar CB, Kuppusamy P. PTEN as a Therapeutic Target in Pulmonary Hypertension Secondary to Left-heart Failure: Effect of HO-3867 and Supplemental Oxygenation. Cell Biochem Biophys 2021; 79:593-607. [PMID: 34133009 DOI: 10.1007/s12013-021-01010-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2021] [Indexed: 01/27/2023]
Abstract
Pulmonary hypertension (PH) is a condition when the pressure in the lung blood vessels is elevated. This leads to increase in thickness of the blood vessels and increases the workload of the heart and lungs. The incidence and prevalence of PH has been on the increase in the last decade. It is estimated that PH affects about 1% of the global population and about 10% of individuals >65 years of age. Of the various types, Group 2 PH is the most common type seen in the elderly population. Fixed PH or PH refractive to therapies is considered a contraindication for heart transplantation; the 30-day mortality in heart transplant recipients is significantly increased in the subset of this population. In general, the pathobiology of PH involves multiple factors including hypoxia, oxidative stress, growth factor receptors, vascular stress, etc. Hence, it is challenging and important to identify specific mechanisms, diagnosis and develop effective therapeutic strategies. The focus of this manuscript is to review some of the important pathobiological processes and mechanisms in the development of PH. Results from our previously reported studies, including targeted treatments along with some new data on PH secondary to left-heart failure, are presented.
Collapse
Affiliation(s)
- Yazhini Ravi
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Periannan Kuppusamy
- Departments of Radiology and Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
9
|
Daneva Z, Marziano C, Ottolini M, Chen YL, Baker TM, Kuppusamy M, Zhang A, Ta HQ, Reagan CE, Mihalek AD, Kasetti RB, Shen Y, Isakson BE, Minshall RD, Zode GS, Goncharova EA, Laubach VE, Sonkusare SK. Caveolar peroxynitrite formation impairs endothelial TRPV4 channels and elevates pulmonary arterial pressure in pulmonary hypertension. Proc Natl Acad Sci U S A 2021; 118:e2023130118. [PMID: 33879616 PMCID: PMC8092599 DOI: 10.1073/pnas.2023130118] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent studies have focused on the contribution of capillary endothelial TRPV4 channels to pulmonary pathologies, including lung edema and lung injury. However, in pulmonary hypertension (PH), small pulmonary arteries are the focus of the pathology, and endothelial TRPV4 channels in this crucial anatomy remain unexplored in PH. Here, we provide evidence that TRPV4 channels in endothelial cell caveolae maintain a low pulmonary arterial pressure under normal conditions. Moreover, the activity of caveolar TRPV4 channels is impaired in pulmonary arteries from mouse models of PH and PH patients. In PH, up-regulation of iNOS and NOX1 enzymes at endothelial cell caveolae results in the formation of the oxidant molecule peroxynitrite. Peroxynitrite, in turn, targets the structural protein caveolin-1 to reduce the activity of TRPV4 channels. These results suggest that endothelial caveolin-1-TRPV4 channel signaling lowers pulmonary arterial pressure, and impairment of endothelial caveolin-1-TRPV4 channel signaling contributes to elevated pulmonary arterial pressure in PH. Thus, inhibiting NOX1 or iNOS activity, or lowering endothelial peroxynitrite levels, may represent strategies for restoring vasodilation and pulmonary arterial pressure in PH.
Collapse
Affiliation(s)
- Zdravka Daneva
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Corina Marziano
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Matteo Ottolini
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| | - Yen-Lin Chen
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Thomas M Baker
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Maniselvan Kuppusamy
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Aimee Zhang
- Department of Surgery, University of Virginia, Charlottesville, VA 22908
| | - Huy Q Ta
- Department of Surgery, University of Virginia, Charlottesville, VA 22908
| | - Claire E Reagan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | - Andrew D Mihalek
- Department of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA 22908
| | - Ramesh B Kasetti
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Yuanjun Shen
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908
| | - Richard D Minshall
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Gulab S Zode
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Elena A Goncharova
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213
| | - Victor E Laubach
- Department of Surgery, University of Virginia, Charlottesville, VA 22908
| | - Swapnil K Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908;
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
10
|
Protective Effects of Curcumin on Pulmonary Arterial Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1328:213-221. [DOI: 10.1007/978-3-030-73234-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
11
|
Yan S, Resta TC, Jernigan NL. Vasoconstrictor Mechanisms in Chronic Hypoxia-Induced Pulmonary Hypertension: Role of Oxidant Signaling. Antioxidants (Basel) 2020; 9:E999. [PMID: 33076504 PMCID: PMC7602539 DOI: 10.3390/antiox9100999] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
Elevated resistance of pulmonary circulation after chronic hypoxia exposure leads to pulmonary hypertension. Contributing to this pathological process is enhanced pulmonary vasoconstriction through both calcium-dependent and calcium sensitization mechanisms. Reactive oxygen species (ROS), as a result of increased enzymatic production and/or decreased scavenging, participate in augmentation of pulmonary arterial constriction by potentiating calcium influx as well as activation of myofilament sensitization, therefore mediating the development of pulmonary hypertension. Here, we review the effects of chronic hypoxia on sources of ROS within the pulmonary vasculature including NADPH oxidases, mitochondria, uncoupled endothelial nitric oxide synthase, xanthine oxidase, monoamine oxidases and dysfunctional superoxide dismutases. We also summarize the ROS-induced functional alterations of various Ca2+ and K+ channels involved in regulating Ca2+ influx, and of Rho kinase that is responsible for myofilament Ca2+ sensitivity. A variety of antioxidants have been shown to have beneficial therapeutic effects in animal models of pulmonary hypertension, supporting the role of ROS in the development of pulmonary hypertension. A better understanding of the mechanisms by which ROS enhance vasoconstriction will be useful in evaluating the efficacy of antioxidants for the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
| | | | - Nikki L. Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (S.Y.); (T.C.R.)
| |
Collapse
|
12
|
Zsidó BZ, Balog M, Erős N, Poór M, Mohos V, Fliszár-Nyúl E, Hetényi C, Nagane M, Hideg K, Kálai T, Bognár B. Synthesis of Spin-Labelled Bergamottin: A Potent CYP3A4 Inhibitor with Antiproliferative Activity. Int J Mol Sci 2020; 21:ijms21020508. [PMID: 31941150 PMCID: PMC7013880 DOI: 10.3390/ijms21020508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/13/2022] Open
Abstract
Bergamottin (BM, 1), a component of grapefruit juice, acts as an inhibitor of some isoforms of the cytochrome P450 (CYP) enzyme, particularly CYP3A4. Herein, a new bergamottin containing a nitroxide moiety (SL-bergamottin, SL-BM, 10) was synthesized; chemically characterized, evaluated as a potential inhibitor of the CYP2C19, CYP3A4, and CYP2C9 enzymes; and compared to BM and known inhibitors such as ketoconazole (KET) (3A4), warfarin (WAR) (2C9), and ticlopidine (TIC) (2C19). The antitumor activity of the new SL-bergamottin was also investigated. Among the compounds studied, BM showed the strongest inhibition of the CYP2C9 and 2C19 enzymes. SL-BM is a more potent inhibitor of CYP3A4 than the parent compound; this finding was also supported by docking studies, suggesting that the binding positions of BM and SL-BM to the active site of CYP3A4 are very similar, but that SL-BM had a better ∆Gbind value than that of BM. The nitroxide moiety markedly increased the antitumor activity of BM toward HeLa cells and marginally increased its toxicity toward a normal cell line. In conclusion, modification of the geranyl sidechain of BM can result in new CYP3A4 enzyme inhibitors with strong antitumor effects.
Collapse
Affiliation(s)
- Balázs Zoltán Zsidó
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Medical School, Szigeti út 12, H-7624 Pécs, Hungary (C.H.)
| | - Mária Balog
- Institute of Organic and Medicinal Chemistry, University of Pécs, Medical School, Honvéd utca 1, H-7624 Pécs, Hungary; (M.B.); (N.E.); (K.H.); (T.K.)
| | - Nikolett Erős
- Institute of Organic and Medicinal Chemistry, University of Pécs, Medical School, Honvéd utca 1, H-7624 Pécs, Hungary; (M.B.); (N.E.); (K.H.); (T.K.)
| | - Miklós Poór
- Department of Pharmacology, University of Pécs, Faculty of Pharmacy, Szigeti út 12, H-7624 Pécs, Hungary; (M.P.); (V.M.); (E.F.-N.)
- János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - Violetta Mohos
- Department of Pharmacology, University of Pécs, Faculty of Pharmacy, Szigeti út 12, H-7624 Pécs, Hungary; (M.P.); (V.M.); (E.F.-N.)
- János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - Eszter Fliszár-Nyúl
- Department of Pharmacology, University of Pécs, Faculty of Pharmacy, Szigeti út 12, H-7624 Pécs, Hungary; (M.P.); (V.M.); (E.F.-N.)
- János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - Csaba Hetényi
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Medical School, Szigeti út 12, H-7624 Pécs, Hungary (C.H.)
| | - Masaki Nagane
- Department of Biochemistry, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan;
| | - Kálmán Hideg
- Institute of Organic and Medicinal Chemistry, University of Pécs, Medical School, Honvéd utca 1, H-7624 Pécs, Hungary; (M.B.); (N.E.); (K.H.); (T.K.)
| | - Tamás Kálai
- Institute of Organic and Medicinal Chemistry, University of Pécs, Medical School, Honvéd utca 1, H-7624 Pécs, Hungary; (M.B.); (N.E.); (K.H.); (T.K.)
- János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - Balázs Bognár
- Institute of Organic and Medicinal Chemistry, University of Pécs, Medical School, Honvéd utca 1, H-7624 Pécs, Hungary; (M.B.); (N.E.); (K.H.); (T.K.)
- Correspondence: or ; Tel.: +36-536-220
| |
Collapse
|
13
|
Zhao M, Chen N, Li X, Lin L, Chen X. MiR-19a modulates hypoxia-mediated cell proliferation and migration via repressing PTEN in human pulmonary arterial smooth muscle. Life Sci 2019; 239:116928. [PMID: 31682848 DOI: 10.1016/j.lfs.2019.116928] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/23/2019] [Accepted: 10/01/2019] [Indexed: 12/16/2022]
Abstract
AIM The dysfunction of human pulmonary arterial smooth muscle cells (HPASMCs) has been suggested to participate in the pathophysiology of pulmonary arterial hypertension (PAH). This study determined miR-19a expression in hypoxia-induced HPASMCs and explored the mechanistic actions of miR-19a in hypoxia-induced HPASMC proliferation and migration. METHODS QRT-PCR and western blot assays respectively determined the mRNA and protein expression of miR-19a, phosphatase and tensin homolog (PTEN) and hypoxia-inducible factor-1 alpha (HIF-1α). In vitro functional assays determined HPASMC proliferation and migration, respectively. Luciferase reporter assay determined interaction between miR-19a and PTEN. The knockdown effects of miR-19a on PAH were confirmed in in vivo mice model. RESULTS Hypoxia treatment time-dependently up-regulated miR-19a expression and enhanced cell proliferation in HPASMCs. MiR-19a overexpression increased cell proliferation and migration of HPASMCs, while repression of miR-19a reduced cell proliferative and migratory potentials of hypoxia-treated HPASMCs. Bioinformatics analysis and luciferase reporter assay showed that PTEN 3' untranslated region was targeted by miR-19a, and miR-19a repressed the mRNA and protein expression of PTEN in HPASMCs. Further rescue studies revealed that miR-19a regulated proliferative and migratory potentials of hypoxia-treated HPASMCs via suppressing PTEN expression. In addition, HIF-1α was identified as one of the mediators for the hypoxia-induced aberrant expression levels of miR-19a and PTEN. MiR-19a overexpression enhanced PI3K/AKT signaling, which was attenuated by enforced expression of PTEN in HPASMCs. More importantly, knockdown of miR-19 attenuated the chronic hypoxia-induced PAH in in vivo mice model. CONCLUSION This study presented a novel mechanistic action of miR-19a-mediated cell proliferation and migration of HPASMCs.
Collapse
Affiliation(s)
- Mei Zhao
- Department of Pharmacy, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya City, Hainan Province, China.
| | - Ni Chen
- Department of Pharmacy, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China.
| | - Xuelian Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin City, Heilongjiang Province, China.
| | - Ling Lin
- Department of Cardiovascular Medicine, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya City, Hainan Province, China.
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, China.
| |
Collapse
|
14
|
Nagane M, Yamashita T, Vörös P, Kálai T, Hideg K, Bognár B. Synthesis and evaluation of paramagnetic caffeic acid phenethyl ester (CAPE) analogs. MONATSHEFTE FUR CHEMIE 2019. [DOI: 10.1007/s00706-019-02458-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
15
|
The pseudogene PTENP1 regulates smooth muscle cells as a competing endogenous RNA. Clin Sci (Lond) 2019; 133:1439-1455. [PMID: 31235554 DOI: 10.1042/cs20190156] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/30/2019] [Accepted: 06/24/2019] [Indexed: 02/06/2023]
Abstract
The long non-coding RNA (lncRNA) PTENP1 is a pseudogene of phosphatase and tensin homologue deleted on chromosome ten (PTEN), has been implicated in smooth muscle cell (SMC) proliferation and apoptosis. PTENP1 is the pseudogene of PTEN. However, it is unclear whether and how PTENP1 functions in the proliferation and apoptosis of human aortic SMCs (HASMCs). Here, we hypothesised that PTENP1 inhibits HASMC proliferation and enhances apoptosis by promoting PTEN expression. PCR analysis and Western blot assays respectively showed that both PTENP1 and PTEN were up-regulated in human aortic dissection (AD) samples. PTENP1 overexpression significantly increased the protein expression of PTEN, promoted apoptosis and inhibited the proliferation of HASMCs. PTENP1 silencing exhibited the opposite effects and mitigated H2O2-induced apoptosis of HASMCs. In an angiotensin II (Ang II)-induced mouse aortic aneurysm (AA) model, PTENP1 overexpression potentiated aortic SMC apoptosis, exacerbated aneurysm formation. Mechanistically, RNA pull-down assay and a series of luciferase reporter assays using miR-21 mimics or inhibitors identified PTENP1 as a molecular sponge for miR-21 to endogenously compete for the binding between miR-21 and the PTEN transcript, releasing PTEN expression. This finding was further supported by in vitro immunofluorescent evidence showing decreased cell apoptosis upon miR-21 mimic administration under baseline PTENP1 overexpression. Ex vivo rescue of PTEN significantly mitigated the SMC apoptosis induced by PTENP1 overexpression. Finally, Western blot assays showed substantially reduced Akt phosphorylation and cyclin D1 and cyclin E levels with up-regulated PTENP1 in HASMCs. Our study identified PTENP1 as a mediator of HASMC homeostasis and suggests that PTENP1 is a potential target in AD or AA intervention.
Collapse
|
16
|
Zhu G, Zhang W, Liu Y, Wang S. miR‑371b‑5p inhibits endothelial cell apoptosis in monocrotaline‑induced pulmonary arterial hypertension via PTEN/PI3K/Akt signaling pathways. Mol Med Rep 2018; 18:5489-5501. [PMID: 30387816 PMCID: PMC6236307 DOI: 10.3892/mmr.2018.9614] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 03/16/2018] [Indexed: 01/25/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a clinical hemodynamic syndrome. It is characterized by elevated PA pressure and pulmonary vascular resistance. In the present study, the role of microRNA (miRNA/miR)-371b-5p in monocrotaline-induced PAH and the underlying mechanisms were investigated. In a monocrotaline-induced PAH rat model, gene chip and reverse transcription-quantitative polymerase chain reaction were employed to measure miRNA expression levels. The results revealed that miR-371b-5p was downregulated in PAH rats compared with the control group. In addition, in vitro results demonstrated that an miR-371b-5p inhibitor reduced miR-371b-5p expression levels, increased apoptosis and reduced proliferation of pulmonary arterial endothelial cells (PAECs) in rats with monocrotaline-induced PAH. Furthermore, inhibition of miR-371b-5p induced phosphatase and tensin homolog (PTEN) protein expression and suppressed that of phosphoinositide 3-kinase (PI3K) and phosphorylated (p)-Akt in the PAECs. In addition, VO-OHpic, a PTEN inhibitor, reduced the protein expression levels of PTEN in the PAECs and inhibited the effects of anti-miR-371b-5p on cell apoptosis. In addition, LY294002, a PI3K inhibitor, reduced the PI3K and p-Akt protein expression in the PAECs and reversed the effects of miR-371b-5p overexpression on the apoptosis of PAECs in rats with monocrotaline-induced PAH. Collectively, the results of the present study indicate that, in this animal model of PAH, miR-371b-5p inhibits apoptosis of PAECs via PTEN/PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Guangfa Zhu
- Department of Pulmonary and Critical Care Medicine, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Wenmei Zhang
- Department of Pulmonary and Critical Care Medicine, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Yan Liu
- Department of Infectious Diseases, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Shenghao Wang
- Department of Infectious Diseases, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| |
Collapse
|
17
|
Lu R, Zhang YG, Sun J. STAT3 activation in infection and infection-associated cancer. Mol Cell Endocrinol 2017; 451:80-87. [PMID: 28223148 PMCID: PMC5469714 DOI: 10.1016/j.mce.2017.02.023] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 12/23/2022]
Abstract
The Janus kinase/signal transducers and activators for transcription (JAK/STAT) pathway plays crucial roles in regulating apoptosis, proliferation, differentiation, and the inflammatory response. The JAK/STAT families are composed of four JAK family members and seven STAT family members. STAT3 plays a key role in inducing and maintaining a pro-carcinogenic inflammatory microenvironment. Recent evidence suggests that STAT3 regulates diverse biological functions in pathogenesis of diseases, such as infection and cancer. In the current review, we will summarize the research progress of STAT3 activation in infection and cancers. We highlight our recent study on the novel role of STAT3 in Salmonella infection-associated colon cancer. Infection with bacterial AvrA-expressing Salmonella activates the STAT3 pathway, which induces the β-catenin signals and enhances colonic tumorigenesis. STAT3 may be a promising target in developing prevention and treatment for infectious diseases and infection-associated cancers.
Collapse
Affiliation(s)
- Rong Lu
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Yong-Guo Zhang
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
18
|
ElNaggar AC, Saini U, Naidu S, Wanner R, Sudhakar M, Fowler J, Nagane M, Kuppusamy P, Cohn DE, Selvendiran K. Anticancer potential of diarylidenyl piperidone derivatives, HO-4200 and H-4318, in cisplatin resistant primary ovarian cancer. Cancer Biol Ther 2017; 17:1107-1115. [PMID: 27415751 DOI: 10.1080/15384047.2016.1210733] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We have previously developed a novel class of bi-functional compounds based on a diarylidenyl-piperidone (DAP) backbone conjugated to an N-hydroxypyrroline (-NOH; a nitroxide precursor) group capable of selectively inhibiting STAT3 activation, translocation, and DNA binding activity. HO-4200 and H-4318 are 2 such derivatives capable of inducing apoptosis in ovarian cancer cells through this mechanism and demonstrated efficacy in platinum resistant primary ovarian cancer cell populations and tumor tissues. The improved absorption and cellular uptake of HO-4200 by cancer cells was determined using optical and electron paramagnetic resonance spectrometry. Treatment of ovarian cancer cells with HO-4200 and H-4318 resulted in cleavage of caspase proteins 3, 7, and 9, as well as PARP and inhibition of the pro-survival protein, Bcl-xL, resulting in significantly decreased cell survival and increased apoptosis. HO-4200 and H-4318 significantly inhibit fatty acid synthase (FAS) and pSTAT3 and decreased the expression of STAT3 target proteins: Survivin, c-myc, Bcl-xl, Bcl-2, cyclin D1/D2, and VEGF were suppressed as analyzed using quantitative real time PCR. In addition, HO-4200 and H-4318 significantly inhibited migration/invasion, in primary ovarian cancer cell populations isolated from primary and recurrent ovarian cancer patients. Treatment of freshly collected human ovarian tumor sections with HO-4200 demonstrated significant suppression of pSTAT3 Tyr 705, angiogenesis (VEFG), and markers of proliferation (Ki-67) in ex vivo models. We have shown, for the first time, that the DAP compounds, HO-4200 and H-4318, inhibit cell migration/invasion and induce apoptosis by targeting FAS/STAT3 in human ovarian cancer cells, including primary ovarian cancer cell populations and tumor tissues. Therefore, our results highlight the clinical anti-cancer potential of HO-4200 and H-4318.
Collapse
Affiliation(s)
- Adam C ElNaggar
- a Division of Gynecologic Oncology , The Ohio State University Comprehensive Cancer Center- Arthur G. James Cancer Hospital and Richard J. Solve Research Institute , Columbus , OH , USA
| | - Uksha Saini
- b Division of Gynecologic Oncology , Comprehensive Cancer Center and Solid Tumor Biology Program, The Ohio State University Wexner Medical Center , Columbus , OH , USA
| | - Shan Naidu
- b Division of Gynecologic Oncology , Comprehensive Cancer Center and Solid Tumor Biology Program, The Ohio State University Wexner Medical Center , Columbus , OH , USA
| | - Ross Wanner
- b Division of Gynecologic Oncology , Comprehensive Cancer Center and Solid Tumor Biology Program, The Ohio State University Wexner Medical Center , Columbus , OH , USA
| | - Millie Sudhakar
- b Division of Gynecologic Oncology , Comprehensive Cancer Center and Solid Tumor Biology Program, The Ohio State University Wexner Medical Center , Columbus , OH , USA
| | | | - Masaki Nagane
- c Department of Radiology , Dartmouth College Geisel School of Medicine , Hanover , NH , USA
| | - Periannan Kuppusamy
- c Department of Radiology , Dartmouth College Geisel School of Medicine , Hanover , NH , USA
| | - David E Cohn
- a Division of Gynecologic Oncology , The Ohio State University Comprehensive Cancer Center- Arthur G. James Cancer Hospital and Richard J. Solve Research Institute , Columbus , OH , USA
| | - Karuppaiyah Selvendiran
- b Division of Gynecologic Oncology , Comprehensive Cancer Center and Solid Tumor Biology Program, The Ohio State University Wexner Medical Center , Columbus , OH , USA
| |
Collapse
|
19
|
HOTAIR functions as a competing endogenous RNA to regulate PTEN expression by inhibiting miR-19 in cardiac hypertrophy. Mol Cell Biochem 2017; 432:179-187. [PMID: 28316060 DOI: 10.1007/s11010-017-3008-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 03/07/2017] [Indexed: 01/18/2023]
Abstract
Sustained cardiac hypertrophy (CH) is related to a variety of physiological as well as pathological stimuli and eventually increases the risk of heart failure. HOTAIR has been identified as a competing endogenous RNA in multiple human biological processes. Whether lncRNA-HOTAIR is involved in the progress of CH and how it works still remain unknown. Herein, we found that HOTAIR was down-regulated, while miR-19 was up-regulated in both heart tissues from TAC-operated mice in vivo and cultural cardiomyocytes treated with Ang-II in vitro by real-time PCR. Meanwhile, HOTAIR expression was negatively correlated with miR-19 in TAC-operated mice. HOTAIR overexpression reduced cell surface area and the expression of hypertrophic markers ANP, BNP, and β-MHC in response to Ang-II stimulation as well as knockdown of miR-19. The further molecular mechanisms of HOTAIR action in CH demonstrated that HOTAIR may act as a competing endogenous RNA (ceRNA) for miR-19, thereby modulating the dis-inhibition of its endogenous target PTEN and playing an important role in inhibiting CH progress. These findings reveal a novel function of LncRNAs, which conduce to an extensive understanding of CH and provide novel research directions and therapeutic options for treating this disease.
Collapse
|
20
|
Transforming growth factor-beta1 upregulation triggers pulmonary artery smooth muscle cell proliferation and apoptosis imbalance in rats with hypoxic pulmonary hypertension via the PTEN/AKT pathways. Int J Biochem Cell Biol 2016; 77:141-154. [DOI: 10.1016/j.biocel.2016.06.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 12/15/2022]
|
21
|
Guazzi M, Phillips SA, Arena R, Lavie CJ. Endothelial dysfunction and lung capillary injury in cardiovascular diseases. Prog Cardiovasc Dis 2014; 57:454-62. [PMID: 25446556 DOI: 10.1016/j.pcad.2014.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cardiac dysfunction of both systolic and diastolic origins leads to increased left atrial pressure, lung capillary injury and increased resistance to gas transfer. Acutely, pressure-induced trauma disrupts the endothelial and alveolar anatomical configuration and definitively causes an impairment of cellular pathways involved in fluid-flux regulation and gas exchange efficiency, a process well identified as stress failure of the alveolar-capillary membrane. In chronic heart failure (HF), additional stimuli other than pressure may trigger the true remodeling process of capillaries and small arteries characterized by endothelial dysfunction, proliferation of myofibroblasts, fibrosis and extracellular matrix deposition. In parallel there is a loss of alveolar gas diffusion properties due to the increased path from air to blood (thickening of extracellular matrix) and loss of fine molecular mechanism involved in fluid reabsorption and clearance. Deleterious changes in gas transfer not only reflect the underlying lung tissue damage but also portend independent prognostic information and may play a role in the pathogenesis of exercise limitation and ventilatory abnormalities observed in these patients. Few currently approved treatments for chronic HF have the potential to positively affect structural remodeling of the lung capillary network; angiotensin-converting enzyme inhibitors are one of the few currently established options. Recently, more attention has been paid to novel therapies specifically targeting the nitric oxide pathway as a suitable target to improve endothelial function and permeability as well as alveolar gas exchange properties.
Collapse
Affiliation(s)
- Marco Guazzi
- Heart Failure Unit, I.R.C.C.S. Policlinico San Donato, University of Milano, San Donato Milanese, Italy.
| | - Shane A Phillips
- Department of Physical Therapy and Integrative Physiology Laboratory, College of Applied Health Sciences, University of Illinois, Chicago, IL, USA
| | - Ross Arena
- Department of Physical Therapy and Integrative Physiology Laboratory, College of Applied Health Sciences, University of Illinois, Chicago, IL, USA
| | - Carl J Lavie
- Department of Cardiovascular Diseases, John Ochsner Heart and Vascular Institute, Ochsner Clinical School-The University of Queensland School of Medicine, New Orleans, LA, USA
| |
Collapse
|
22
|
McCann GA, Naidu S, Rath KS, Bid HK, Tierney BJ, Suarez A, Varadharaj S, Zhang J, Hideg K, Houghton P, Kuppusamy P, Cohn DE, Selvendiran K. Targeting constitutively-activated STAT3 in hypoxic ovarian cancer, using a novel STAT3 inhibitor. Oncoscience 2014; 1:216-28. [PMID: 25594014 PMCID: PMC4278289 DOI: 10.18632/oncoscience.26] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 03/31/2014] [Indexed: 01/04/2023] Open
Abstract
Tumor hypoxia, a feature of many solid tumors including ovarian cancer, is associated with resistance to therapies. We previously demonstrated that hypoxic exposure results in increased expression of phosphorylated signal transducer and activator of transcription 3 (pSTAT3). We hypothesized the activation of STAT3 could lead to chemotherapeutic resistance in ovarian cancer cells in hypoxic conditions. In this study, we demonstrate the level of pSTAT3 Tyr705 is increased in the hypoxic regions of human epithelial ovarian cancer (EOC) specimens, as determined by HIF-1α and CD-31 staining. In vitro mutagenesis studies proved that pSTAT3 Tyr705 is necessary for cell survival and proliferation under hypoxic conditions. In addition, we show that S1PR1, a regulator of STAT3 transcription via the JAK/STAT pathway, is highly expressed in hypoxic ovarian cancer cells (HOCCs). Knock down of S1PR1 in HOCCs reduced pSTAT3 Tyr705 levels and was associated with decreased cell survival. Treatment of HOCCs with the STAT3 inhibitor HO-3867 resulted in a rapid and dramatic decrease in pSTAT3 Tyr705 levels as a result of ubiquitin proteasome degradation. STAT3-target proteins Bcl-xL, cyclin D2 and VEGF showed similar decreases in HO-3867 treated cells. Taken together, these findings suggest that activation of STAT3 Tyr705 promotes cell survival and proliferation in HOCCs, and that S1PR1 is involved in the initiation of STAT3 activation. Targeting hypoxia-mediated STAT3 activation represents a therapeutic option for ovarian cancer and other solid tumors.
Collapse
Affiliation(s)
- Georgia A McCann
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Shan Naidu
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Kellie S Rath
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Brent J Tierney
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Adrian Suarez
- Department of Pathology, Divisions of Gynecological Pathology and Cytopathology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Saradhadevi Varadharaj
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Jianying Zhang
- Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Kálmán Hideg
- Institute of Organic and Medicinal Chemistry, University of Pécs, Pécs, Hungary
| | | | | | - David E Cohn
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Karuppaiyah Selvendiran
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| |
Collapse
|
23
|
Bonini MG, Consolaro MEL, Hart PC, Mao M, de Abreu ALP, Master AM. Redox control of enzymatic functions: The electronics of life's circuitry. IUBMB Life 2014; 66:167-181. [PMID: 24668617 DOI: 10.1002/iub.1258] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 03/06/2014] [Indexed: 12/22/2022]
Abstract
The field of redox biology has changed tremendously over the past 20 years. Formerly regarded as bi-products of the aerobic metabolism exclusively involved in tissue damage, reactive oxygen species (ROS) are now recognized as active participants of cell signaling events in health and in disease. In this sense, ROS and the more recently defined reactive nitrogen species (RNS) are, just like hormones and second messengers, acting as fundamental orchestrators of cell signaling pathways. The chemical modification of enzymes by ROS and RNS (that result in functional enzymatic alterations) accounts for a considerable fraction of the transient and persistent perturbations imposed by variations in oxidant levels. Upregulation of ROS and RNS in response to stress is a common cellular response that foments adaptation to a variety of physiologic alterations (hypoxia, hyperoxia, starvation, and cytokine production). Frequently, these are beneficial and increase the organisms' resistance against subsequent acute stress (preconditioning). Differently, the sustained ROS/RNS-dependent rerouting of signaling produces irreversible alterations in cellular functioning, often leading to pathogenic events. Thus, the duration and reversibility of protein oxidations define whether complex organisms remain "electronically" healthy. Among the 20 essential amino acids, four are particularly susceptible to oxidation: cysteine, methionine, tyrosine, and tryptophan. Here, we will critically review the mechanisms, implications, and repair systems involved in the redox modifications of these residues in proteins while analyzing well-characterized prototypic examples. Occasionally, we will discuss potential consequences of amino acid oxidation and speculate on the biologic necessity for such events in the context of adaptative redox signaling. © 2014 IUBMB Life, 66(3):167-181, 2014.
Collapse
Affiliation(s)
- Marcelo G Bonini
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Programa de Biociencias Aplicadas a Farmacia (PBF), Universidade Estadual de Maringa, Maringa, Parana, Brazil
| | - Marcia E L Consolaro
- Programa de Biociencias Aplicadas a Farmacia (PBF), Universidade Estadual de Maringa, Maringa, Parana, Brazil
| | - Peter C Hart
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Mao Mao
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Andre Luelsdorf Pimenta de Abreu
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Programa de Biociencias Aplicadas a Farmacia (PBF), Universidade Estadual de Maringa, Maringa, Parana, Brazil
| | - Alyssa M Master
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
24
|
Horita H, Furgeson SB, Ostriker A, Olszewski KA, Sullivan T, Villegas LR, Levine M, Parr JE, Cool CD, Nemenoff RA, Weiser-Evans MCM. Selective inactivation of PTEN in smooth muscle cells synergizes with hypoxia to induce severe pulmonary hypertension. J Am Heart Assoc 2013; 2:e000188. [PMID: 23727701 PMCID: PMC3698782 DOI: 10.1161/jaha.113.000188] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Pulmonary vascular remodeling in pulmonary hypertension (PH) is characterized by increased vascular smooth muscle cell (SMC) and adventitial fibroblast proliferation, small vessel occlusion, and inflammatory cell accumulation. The underlying molecular mechanisms driving progression remain poorly defined. We have focused on loss of the phosphatase PTEN in SMCs as a major driver of pathological vascular remodeling. Our goal was to define the role of PTEN in human PH and in hypoxia‐induced PH using a mouse model with inducible deletion of PTEN in SMCs. Methods and Results Staining of human biopsies demonstrated enhanced inactive PTEN selectively in the media from hypertensive patients compared to controls. Mice with induced deletion of PTEN in SMCs were exposed to normoxia or hypoxia for up to 4 weeks. Under normoxia, SMC PTEN depletion was sufficient to induce features of PH similar to those observed in wild‐type mice exposed to chronic hypoxia. Under hypoxia, PTEN depletion promoted an irreversible progression of PH characterized by increased pressure, extensive pulmonary vascular remodeling, formation of complex vascular lesions, and increased macrophage accumulation associated with synergistic increases in proinflammatory cytokines and proliferation of both SMCs and nonSMCs. Conclusions Chronic inactivation of PTEN selectively in SMC represents a critical mediator of PH progression, leading to cell autonomous events and increased production of factors correlated to proliferation and recruitment of adventitial and inflammatory cells, resulting in irreversible progression of the disease.
Collapse
Affiliation(s)
- Henrick Horita
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|