1
|
Ichimura K, Gross A, Mathew RO, Salman L, Reddy S, Spiekerkoetter E, Sidhu MS. Cardiorenal Syndrome in Right Heart Failure Due to Pulmonary Arterial Hypertension-The Right Ventricle as a Therapeutic Target to Improve Renal Function. Cardiovasc Drugs Ther 2025; 39:373-384. [PMID: 38847906 DOI: 10.1007/s10557-024-07588-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 03/30/2025]
Abstract
Cardiorenal syndrome (CRS) due to right ventricular (RV) failure is a disease entity emerging as a key indicator of morbidity and mortality. The multifactorial aspects of CRS and the left-right ventricular interdependence complicate the link between RV failure and renal function. RV failure has a direct pathophysiological link to renal dysfunction by leading to systemic venous congestion in certain circumstances and low cardiac output in other situations, both leading to impaired renal perfusion. Indeed, renal dysfunction is known to be an independent predictor of mortality in patients with pulmonary arterial hypertension (PAH) and RV failure. Thus, it is important to further understand the interaction between the RV and renal function. RV adaptation is critical to long-term survival in patients with PAH. The RV is also known for its remarkable capacity to recover once the aggravating factor is addressed or mitigated. However, less is known about the renal potential for recovery following the resolution of chronic RV failure. In this review, we provide an overview of the intricate relationship between RV dysfunction and the subsequent development of CRS, with a particular emphasis on PAH. Additionally, we summarize potential RV-targeted therapies and their potential beneficial impact on renal function.
Collapse
Affiliation(s)
- Kenzo Ichimura
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Stanford University, 1701 Page Mill Road, Palo Alto, CA, 94304, USA.
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, 94305, USA.
- Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA.
| | - Adam Gross
- Albany Medical College, Albany, NY, 12208, USA
| | - Roy O Mathew
- Department of Medicine, Loma Linda VA Health Care System, Loma Linda, CA, 92357, USA
| | - Loay Salman
- Division of Nephrology, Department of Medicine, Albany Medical College, Albany, NY, 12208, USA
| | - Sushma Reddy
- Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, CA, 94305, USA
| | - Edda Spiekerkoetter
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Stanford University, 1701 Page Mill Road, Palo Alto, CA, 94304, USA
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, 94305, USA
- Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Mandeep S Sidhu
- Division of Cardiology, Department of Medicine, Department of Medical Education, Albany Medical College, Albany, NY, 12208, USA
| |
Collapse
|
2
|
Lai YJ, Yeh YH, Huang YL, De Almeida C, Chang GJ, Chen WJ, Hsu HH. Empagliflozin Attenuates Pulmonary Arterial Remodeling Through Peroxisome Proliferator-Activated Receptor Gamma Activation. ACS Pharmacol Transl Sci 2024; 7:2725-2738. [PMID: 39296270 PMCID: PMC11406702 DOI: 10.1021/acsptsci.4c00127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/21/2024]
Abstract
The loss of peroxisome proliferator-activated receptor gamma (PPARγ) exacerbates pulmonary arterial hypertension (PAH), while its upregulation reduces cell proliferation and vascular remodeling, thereby decreasing PAH severity. SGLT2 inhibitors, developed for type 2 diabetes, might also affect signal transduction in addition to modulating sodium-glucose cotransporters. Pulmonary arterial smooth muscle cells (PASMCs) isolated from patients with idiopathic pulmonary arterial hypertension (IPAH) were treated with three SGLT2 inhibitors, canagliflozin (Cana), dapagliflozin (Dapa), and empagliflozin (Empa), to investigate their antiproliferative effects. To assess the impact of Empa on PPARγ, luciferase reporter assays and siRNA-mediated PPARγ knockdown were employed to examine regulation of the γ-secretase complex and its downstream target Notch3. Therapy involving daily administration of Empa was initiated 21 days after inducing hypoxia-induced PAH in mice. Empa exhibited significant antiproliferative effects on fast-growing IPAH PASMCs. Empa activated PPARγ to prevent formation of the γ-secretase complex, with specific impacts on presenilin enhancer 2 (PEN2), which plays a crucial role in maintaining γ-secretase complex stability, thereby inhibiting Notch3. Similar results were obtained in lung tissue of chronically hypoxic mice. Empa attenuated pulmonary arterial remodeling and right ventricle hypertrophy in a hypoxic PAH mouse model. Moreover, PPARγ expression was significantly decreased and PEN2, and Notch3 levels were increased in lung tissue from PAH patients compared with non-PAH lung tissue. Empa reverses vascular remodeling by activating PPARγ to suppress the γ-secretase-Notch3 axis. We propose Empa as a PPARγ activator and potential therapeutic for PAH.
Collapse
Affiliation(s)
- Ying-Ju Lai
- Cardiovascular Division, Chang Gung Memorial Hospital, Tao-Yuan 333, Taiwan
- Department of Respiratory Therapy, Chang Gung University College of Medicine, Chang-Gung University, Tao-Yuan 33353, Taiwan
- Department of Respiratory Care, Chang-Gung University of Science and Technology, Chia-Yi 61363, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Division, Chang Gung Memorial Hospital, Tao-Yuan 333, Taiwan
| | - Yen-Lin Huang
- Department of Pathology, National Taiwan University Cancer Center and National Taiwan University College of Medicine, Taipei 106328, Taiwan
| | - Celina De Almeida
- Department of Respiratory Therapy, Chang Gung University College of Medicine, Chang-Gung University, Tao-Yuan 33353, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Tao-Yuan 33353, Taiwan
| | - Gwo-Jyh Chang
- Cardiovascular Division, Chang Gung Memorial Hospital, Tao-Yuan 333, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Tao-Yuan 33353, Taiwan
| | - Wei-Jan Chen
- Cardiovascular Division, Chang Gung Memorial Hospital, Tao-Yuan 333, Taiwan
| | - Hsao-Hsun Hsu
- Division of Thoracic Surgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 10002, Taiwan
| |
Collapse
|
3
|
Li Y, Wei X, Xiao R, Chen Y, Xiong T, Fang ZM, Huo B, Guo X, Luo H, Wu X, Liu L, Zhu XH, Hu Q, Jiang DS, Yi X. SMYD2-Methylated PPARγ Facilitates Hypoxia-Induced Pulmonary Hypertension by Activating Mitophagy. Circ Res 2024; 135:93-109. [PMID: 38770649 DOI: 10.1161/circresaha.124.323698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Hyperproliferation of pulmonary arterial smooth muscle cells (PASMCs) and consequent pulmonary vascular remodeling are the crucial pathological features of pulmonary hypertension (PH). Protein methylation has been shown to be critically involved in PASMC proliferation and PH, but the underlying mechanism remains largely unknown. METHODS PH animal models were generated by treating mice/rats with chronic hypoxia for 4 weeks. SMYD2-vTg mice (vascular smooth muscle cell-specific suppressor of variegation, enhancer of zeste, trithorax and myeloid Nervy DEAF-1 (deformed epidural auto-regulatory factor-1) domain-containing protein 2 transgenic) or wild-type rats and mice treated with LLY-507 (3-cyano-5-{2-[4-[2-(3-methylindol-1-yl)ethyl]piperazin-1-yl]-phenyl}-N-[(3-pyrrolidin-1-yl)propyl]benzamide) were used to investigate the function of SMYD2 (suppressor of variegation, enhancer of zeste, trithorax and myeloid Nervy DEAF-1 domain-containing protein 2) on PH development in vivo. Primary cultured rat PASMCs with SMYD2 knockdown or overexpression were used to explore the effects of SMYD2 on proliferation and to decipher the underlying mechanism. RESULTS We demonstrated that the expression of the lysine methyltransferase SMYD2 was upregulated in the smooth muscle cells of pulmonary arteries from patients with PH and hypoxia-exposed rats/mice and in the cytoplasm of hypoxia-induced rat PASMCs. More importantly, targeted inhibition of SMYD2 by LLY-507 significantly attenuated hypoxia-induced pulmonary vascular remodeling and PH development in both male and female rats in vivo and reduced rat PASMC hyperproliferation in vitro. In contrast, SMYD2-vTg mice exhibited more severe PH phenotypes and related pathological changes than nontransgenic mice after 4 weeks of chronic hypoxia treatment. Furthermore, SMYD2 overexpression promoted, while SMYD2 knockdown suppressed, the proliferation of rat PASMCs by affecting the cell cycle checkpoint between S and G2 phases. Mechanistically, we revealed that SMYD2 directly interacted with and monomethylated PPARγ (peroxisome proliferator-activated receptor gamma) to inhibit the nuclear translocation and transcriptional activity of PPARγ, which further promoted mitophagy to facilitate PASMC proliferation and PH development. Furthermore, rosiglitazone, a PPARγ agonist, largely abolished the detrimental effects of SMYD2 overexpression on PASMC proliferation and PH. CONCLUSIONS Our results demonstrated that SMYD2 monomethylates nonhistone PPARγ and inhibits its nuclear translocation and activation to accelerate PASMC proliferation and PH by triggering mitophagy, indicating that targeting SMYD2 or activating PPARγ are potential strategies for the prevention of PH.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Rats
- Cell Proliferation
- Cells, Cultured
- Histone-Lysine N-Methyltransferase/metabolism
- Histone-Lysine N-Methyltransferase/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/genetics
- Hypoxia/complications
- Hypoxia/metabolism
- Methylation
- Mice, Inbred C57BL
- Mice, Transgenic
- Mitophagy
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- PPAR gamma/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/metabolism
- Rats, Sprague-Dawley
- Vascular Remodeling
Collapse
Affiliation(s)
- Yi Li
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Xiang Wei
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
- Key Laboratory of Organ Transplantation, Ministry of Education (X. Wei, D.-S.J.), Chinese Academy of Medical Sciences, Wuhan, China
- NHC Key Laboratory of Organ Transplantation (X. Wei, D.-S.J.), Chinese Academy of Medical Sciences, Wuhan, China
- Key Laboratory of Organ Transplantation (X. Wei, D.-S.J.), Chinese Academy of Medical Sciences, Wuhan, China
| | - Rui Xiao
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Wuhan (R.X., Q.H.)
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China (R.X., Q.H.)
| | - Yongjie Chen
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, China (Y.C.)
| | - Tianxin Xiong
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Ze-Min Fang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Bo Huo
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Xian Guo
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Hanshen Luo
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Xingliang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, China (X. Wu, L.L., X.Y.)
| | - Liyuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, China (X. Wu, L.L., X.Y.)
| | - Xue-Hai Zhu
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Qinghua Hu
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Wuhan (R.X., Q.H.)
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China (R.X., Q.H.)
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
- Key Laboratory of Organ Transplantation, Ministry of Education (X. Wei, D.-S.J.), Chinese Academy of Medical Sciences, Wuhan, China
- NHC Key Laboratory of Organ Transplantation (X. Wei, D.-S.J.), Chinese Academy of Medical Sciences, Wuhan, China
- Key Laboratory of Organ Transplantation (X. Wei, D.-S.J.), Chinese Academy of Medical Sciences, Wuhan, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, China (X. Wu, L.L., X.Y.)
| |
Collapse
|
4
|
Zanotto TM, Gonçalves AEDSS, Saad MJA. Pulmonary hypertension and insulin resistance: a mechanistic overview. Front Endocrinol (Lausanne) 2024; 14:1283233. [PMID: 38239990 PMCID: PMC10794542 DOI: 10.3389/fendo.2023.1283233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/08/2023] [Indexed: 01/22/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a vascular remodeling disease, characterized by increased blood pressure levels in pulmonary circulation, leading to a restriction in the circulation flow and heart failure. Although the emergence of new PAH therapies has increased survival rates, this disease still has a high mortality and patients that receive diagnosis die within a few years. The pathogenesis of PAH involves multiple pathways, with a complex interaction of local and distant cytokines, hormones, growth factors, and transcription factors, leading to an inflammation that changes the vascular anatomy in PAH patients. These abnormalities involve more than just the lungs, but also other organs, and between these affected organs there are different metabolic dysfunctions implied. Recently, several publications demonstrated in PAH patients a disturbance in glucose metabolism, demonstrated by higher levels of glucose, insulin, and lipids in those patients. It is possible that a common molecular mechanism can have a significant role in this connection. In this regard, this narrative review intends to focus on the recent papers that mainly discuss the molecular determinants between insulin resistance (IR) associated PAH, which included obesity subclinical inflammation induced IR, PPAR gamma and Adiponectin, BMPR2, mitochondrial dysfunction and endoplasmic reticulum stress. Therefore, the following review will summarize some of the existing data for IR associated PAH, focusing on the better understanding of PAH molecular mechanisms, for the development of new translational therapies.
Collapse
Affiliation(s)
- Tamires M. Zanotto
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
- Departament of Medical Clinics, Obesity and Comorbidities Research Centre (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | - Mario J. A. Saad
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
- Departament of Medical Clinics, Obesity and Comorbidities Research Centre (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
5
|
Körbelin J, Klein J, Matuszcak C, Runge J, Harbaum L, Klose H, Hennigs JK. Transcription factors in the pathogenesis of pulmonary arterial hypertension-Current knowledge and therapeutic potential. Front Cardiovasc Med 2023; 9:1036096. [PMID: 36684555 PMCID: PMC9853303 DOI: 10.3389/fcvm.2022.1036096] [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: 09/03/2022] [Accepted: 11/21/2022] [Indexed: 01/09/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a disease characterized by elevated pulmonary vascular resistance and pulmonary artery pressure. Mortality remains high in severe cases despite significant advances in management and pharmacotherapy. Since currently approved PAH therapies are unable to significantly reverse pathological vessel remodeling, novel disease-modifying, targeted therapeutics are needed. Pathogenetically, PAH is characterized by vessel wall cell dysfunction with consecutive remodeling of the pulmonary vasculature and the right heart. Transcription factors (TFs) regulate the process of transcribing DNA into RNA and, in the pulmonary circulation, control the response of pulmonary vascular cells to macro- and microenvironmental stimuli. Often, TFs form complex protein interaction networks with other TFs or co-factors to allow for fine-tuning of gene expression. Therefore, identification of the underlying molecular mechanisms of TF (dys-)function is essential to develop tailored modulation strategies in PAH. This current review provides a compendium-style overview of TFs and TF complexes associated with PAH pathogenesis and highlights their potential as targets for vasculoregenerative or reverse remodeling therapies.
Collapse
Affiliation(s)
- Jakob Körbelin
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,*Correspondence: Jakob Körbelin,
| | - Julius Klein
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christiane Matuszcak
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Runge
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Harbaum
- Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans Klose
- Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan K. Hennigs
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Jan K. Hennigs,
| |
Collapse
|
6
|
Zare E, Kafshbani P, Chenaghlou M, Noori M, Ghaemmaghami Z, Amin A, Taghavi S, Naderi N. Prognostic significance of insulin resistance in pulmonary hypertension. ESC Heart Fail 2022; 9:318-326. [PMID: 34904389 PMCID: PMC8788000 DOI: 10.1002/ehf2.13752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 07/31/2021] [Accepted: 11/24/2021] [Indexed: 11/08/2022] Open
Abstract
AIMS The relationship between insulin resistance (IR) and glucose intolerance with pulmonary hypertension (PH) has been suggested in recent investigations. In the present study, we aimed to show the prevalence of IR and its correlation with haemodynamic variables as well as its prognostic significance in this group of patients. METHODS AND RESULTS Among 100 new and returning patients with PH, scheduled for right heart catheterization (RHC), 59 non-diabetic patients were enrolled. The homeostasis model assessment of insulin resistance (HOMA-IR) was used to assess IR. The study population were followed up for a median (interquartile range) of 48 (23-48) months for all-cause mortalities. Most of the study population [mean (standard deviation) age of 45.9 (17.3)] were classified as class I of PH classification (47.5%). Overall, 27% of our study population had IR considering the Iranian cut points of HOMA-IR. The prevalence of IR in non-diabetic, non-metabolic syndrome patients with precapillary PH (PAH) was 34.2%, which was higher than the prevalence of IR in non-diabetic, non-metabolic syndrome Iranian population (24.1%). There was no difference between IR and insulin sensitive (IS) groups regarding demographic and clinical findings, 6 min walk test, and laboratory and haemodynamic data in univariable and multivariable analyses. The mortality rate in the follow-up period was 44.1%. The survival of patient with IR was slightly lower than IS patients; however, IR was not an independent predictor of death. CONCLUSIONS The glucose metabolism is dysregulated in patients with PH, and IR may increase the risk of adverse events among these patients.
Collapse
Affiliation(s)
- Elahe Zare
- Yazd Cardiovascular Research CenterShahid Sadoughi University of Medical SciencesYazdIran
| | - Parvaneh Kafshbani
- Rajaie Cardiovascular Research CenterIran University of Medical SciencesTehranIran
| | - Maryam Chenaghlou
- Cardiovascular Research CenterTabriz University of Medical SciencesTehranIran
| | - Mehdi Noori
- Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| | - Zahra Ghaemmaghami
- Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| | - Ahmad Amin
- Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| | - Sepideh Taghavi
- Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| | - Nasim Naderi
- Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| |
Collapse
|
7
|
Zhao K, Wu T, Yang C, Pan H, Xu T, Zhang J, Guo X, Tu J, Zhang D, Kong X, Zhou B, Sun W. Low-intensity pulsed ultrasound prevents angiotensin II-induced aortic smooth muscle cell phenotypic switch via hampering miR-17-5p and enhancing PPAR-γ. Eur J Pharmacol 2021; 911:174509. [PMID: 34547245 DOI: 10.1016/j.ejphar.2021.174509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 10/20/2022]
Abstract
Vascular events can trigger a pathological phenotypic switch in vascular smooth muscle cells (VSMCs), decreasing and disrupting the plasticity and diversity of vascular networks. The development of novel therapeutic approaches is necessary to prevent these changes. We aimed to investigate the effects and associated mechanisms of low-intensity pulsed ultrasound (LIPUS) irradiation on the angiotensin II (AngII)-induced phenotypic switch in VSMCs. In vivo, AngII was infused subcutaneously for 4 weeks to stimulate vascular remodeling in mice, and LIPUS irradiation was applied for 20 min every 2 days for 4 weeks. In vitro, cultured rat aortic VSMCs (RAVSMCs) were pretreated once with LIPUS irradiation for 20 min before 48-h AngII stimulation. Our results showed that LIPUS irradiation prevents AngII-induced vascular remodeling of the whole wall artery without discriminating between adventitia and media in vivo and RAVSMC phenotypic switching in vitro. LIPUS irradiation downregulated miR-17-5p expression and upregulated peroxisome proliferator-activated receptor gamma (PPAR-γ) expression. The PPAR-γ activator rosiglitazone could mimic the favorable effects of LIPUS irradiation on AngII-treated RAVSMCs. In contrast, GW9662 could impede the LIPUS-mediated downregulation of RAVSMC proliferation and inflammation under AngII stimulation conditions in vivo and in vitro. Also, the miR-17-5p agomir has the same effects as GW9662 in vitro. Besides, the inhibitory effects of GW9662 against the anti-remodeling effects of LIPUS irradiation in AngII-induced RAVSMCs could be blocked by pretreatment with the miR-17-5p antagomir. Overall, LIPUS irradiation prevents AngII-induced RAVSMCs phenotypic switching through hampering miR-17-5p and enhancing PPAR-γ, suggesting a new approach for the treatment of vascular disorders.
Collapse
MESH Headings
- Animals
- MicroRNAs/genetics
- MicroRNAs/metabolism
- PPAR gamma/metabolism
- PPAR gamma/genetics
- Angiotensin II/pharmacology
- Male
- Rats
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/radiation effects
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/radiation effects
- Phenotype
- Ultrasonic Waves
- Aorta/drug effects
- Aorta/metabolism
- Aorta/cytology
- Vascular Remodeling/drug effects
- Vascular Remodeling/radiation effects
- Cells, Cultured
- Mice
- Rats, Sprague-Dawley
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Kun Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Tingting Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Chuanxi Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China; Department of Cardiology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200082, China
| | - Haotian Pan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Tianhua Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jing Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Xiasheng Guo
- Key Laboratory of Modern Acoustics, Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Juan Tu
- Key Laboratory of Modern Acoustics, Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Dong Zhang
- Key Laboratory of Modern Acoustics, Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Bin Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China; Department of Genetics, Pediatrics and Medicine Cardiology, Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
8
|
PPARγ and TGFβ-Major Regulators of Metabolism, Inflammation, and Fibrosis in the Lungs and Kidneys. Int J Mol Sci 2021; 22:ijms221910431. [PMID: 34638771 PMCID: PMC8508998 DOI: 10.3390/ijms221910431] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 01/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a type II nuclear receptor, initially recognized in adipose tissue for its role in fatty acid storage and glucose metabolism. It promotes lipid uptake and adipogenesis by increasing insulin sensitivity and adiponectin release. Later, PPARγ was implicated in cardiac development and in critical conditions such as pulmonary arterial hypertension (PAH) and kidney failure. Recently, a cluster of different papers linked PPARγ signaling with another superfamily, the transforming growth factor beta (TGFβ), and its receptors, all of which play a major role in PAH and kidney failure. TGFβ is a multifunctional cytokine that drives inflammation, fibrosis, and cell differentiation while PPARγ activation reverses these adverse events in many models. Such opposite biological effects emphasize the delicate balance and complex crosstalk between PPARγ and TGFβ. Based on solid experimental and clinical evidence, the present review summarizes connections and their implications for PAH and kidney failure, highlighting the similarities and differences between lung and kidney mechanisms as well as discussing the therapeutic potential of PPARγ agonist pioglitazone.
Collapse
|
9
|
Derrett-Smith E, Clark KEN, Shiwen X, Abraham DJ, Hoyles RK, Lacombe O, Broqua P, Junien JL, Konstantinova I, Ong VH, Denton CP. The pan-PPAR agonist lanifibranor reduces development of lung fibrosis and attenuates cardiorespiratory manifestations in a transgenic mouse model of systemic sclerosis. Arthritis Res Ther 2021; 23:234. [PMID: 34488870 PMCID: PMC8419933 DOI: 10.1186/s13075-021-02592-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/26/2021] [Indexed: 12/02/2022] Open
Abstract
Background The TβRII∆k-fib transgenic (TG) mouse model of scleroderma replicates key fibrotic and vasculopathic complications of systemic sclerosis through fibroblast-directed upregulation of TGFβ signalling. We have examined peroxisome proliferator-activated receptor (PPAR) pathway perturbation in this model and explored the impact of the pan-PPAR agonist lanifibranor on the cardiorespiratory phenotype. Methods PPAR pathway gene and protein expression differences from TG and WT sex-matched littermate mice were determined at baseline and following administration of one of two doses of lanifibranor (30 mg/kg or 100 mg/kg) or vehicle administered by daily oral gavage up to 4 weeks. The prevention of bleomycin-induced lung fibrosis and SU5416-induced pulmonary hypertension by lanifibranor was explored. Results Gene expression data were consistent with the downregulation of the PPAR pathway in the TβRII∆k-fib mouse model. TG mice treated with high-dose lanifibranor demonstrated significant protection from lung fibrosis after bleomycin and from right ventricular hypertrophy following induction of pulmonary hypertension by SU5416, despite no significant change in right ventricular systolic pressure. Conclusions In the TβRII∆k-fib mouse strain, treatment with 100 mg/kg lanifibranor reduces the development of lung fibrosis and right ventricular hypertrophy induced by bleomycin or SU5416, respectively. Reduced PPAR activity may contribute to the exaggerated fibroproliferative response to tissue injury in this transgenic model of scleroderma and its pulmonary complications.
Collapse
Affiliation(s)
- Emma Derrett-Smith
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, Rowland Hill St., London, NW3 2PF, UK
| | - Kristina E N Clark
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, Rowland Hill St., London, NW3 2PF, UK
| | - Xu Shiwen
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, Rowland Hill St., London, NW3 2PF, UK
| | - David J Abraham
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, Rowland Hill St., London, NW3 2PF, UK
| | | | | | | | | | | | - Voon H Ong
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, Rowland Hill St., London, NW3 2PF, UK
| | - Christopher P Denton
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, Rowland Hill St., London, NW3 2PF, UK.
| |
Collapse
|
10
|
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
|
11
|
Ali MK, Ichimura K, Spiekerkoetter E. Promising therapeutic approaches in pulmonary arterial hypertension. Curr Opin Pharmacol 2021; 59:127-139. [PMID: 34217109 DOI: 10.1016/j.coph.2021.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/12/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a debilitating multifactorial disease characterized by progressive pulmonary vascular remodeling, elevated pulmonary arterial pressure, and pulmonary vascular resistance, resulting in right ventricular failure and subsequent death. Current available therapies do not reverse the disease, resulting in a persistent high morbidity and mortality. Thus, there is an urgent unmet medical need for novel effective therapies to better treat patients with PAH. Over the past few years, enthusiastic attempts have been made to identify novel effective therapies that address the essential roots of PAH with targeting key signaling pathways in both preclinical models and patients with PAH. This review aims to discuss the most emerging and promising therapeutic interventions in PAH pathogenesis.
Collapse
Affiliation(s)
- Md Khadem Ali
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford Medical School, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, USA
| | - Kenzo Ichimura
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford Medical School, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, USA
| | - Edda Spiekerkoetter
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford Medical School, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, USA.
| |
Collapse
|
12
|
Yao D, He Q, Sun J, Cai L, Wei J, Cai G, Liu J, Lin Y, Wang L, Huang X. FGF21 attenuates hypoxia‑induced dysfunction and inflammation in HPAECs via the microRNA‑27b‑mediated PPARγ pathway. Int J Mol Med 2021; 47:116. [PMID: 33907846 PMCID: PMC8083827 DOI: 10.3892/ijmm.2021.4949] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/02/2021] [Indexed: 12/20/2022] Open
Abstract
Pulmonary arterial hypertension (PAH), is a chronic and progressive disorder characterized by pulmonary vascular remodeling, including endothelial cell dysfunction and inflammation. MicroRNAs (miRNAs or miRs) play an important role in the development of PAH. In addition, fibroblast growth factor 21 (FGF21) has been found to have marked anti-dysfunction and anti-inflammatory properties. Therefore, the present study aimed to investigate the latent effects of FGF21 against PAH through the miR-27b/peroxisome proliferator-activated receptor γ (PPARγ) axis. Human pulmonary arterial endothelial cells (HPAECs) subjected to hypoxia were used as PAH models. The results revealed that PPARγ expression was downregulated and miR-27b expression was upregulated in the HPAECs exposed to hypoxia. Luciferase assay suggested that PPARγ was a target gene of miR-27b. Furthermore, miR-27b inhibited the expression of the PPARγ gene, thereby aggravating hypoxia-induced HPAEC dysfunction. Moreover, miR-27b activated the nuclear factor-κB signaling pathway and the expression of inflammatory factors [interleukin (IL)-1β, IL-6 and tumor necrosis factor-α] by targeting PPARγ. In addition, the expression of miR-27b decreased following treatment of the hypoxia-exposed HPAECs with FGF21. Furthermore, FGF21 alleviated hypoxia-induced HPAEC dysfunction and inflammation by inhibiting miR-27b expression and thereby promoting PPARγ expression. On the whole, the findings of the present study suggest that FGF21 may serve as a therapeutic target for managing PAH through the miR-27b-mediated PPARγ pathway.
Collapse
Affiliation(s)
- Dan Yao
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Qinlian He
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Junwei Sun
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Luqiong Cai
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Jinqiu Wei
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Gexiang Cai
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Jingjing Liu
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Yinuo Lin
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Liangxing Wang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiaoying Huang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
13
|
Papathanasiou AE, Spyropoulos F, Michael Z, Joung KE, Briana DD, Malamitsi-Puchner A, Mantzoros CS, Christou H. Adipokines and Metabolic Regulators in Human and Experimental Pulmonary Arterial Hypertension. Int J Mol Sci 2021; 22:ijms22031435. [PMID: 33535425 PMCID: PMC7867052 DOI: 10.3390/ijms22031435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Pulmonary hypertension (PH) is associated with meta-inflammation related to obesity but the role of adipose tissue in PH pathogenesis is unknown. We hypothesized that adipose tissue-derived metabolic regulators are altered in human and experimental PH. We measured circulating levels of fatty acid binding protein 4 (FABP-4), fibroblast growth factor -21 (FGF-21), adiponectin, and the mRNA levels of FABP-4, FGF-21, and peroxisome proliferator-activated receptor γ (PPARγ) in lung tissue of patients with idiopathic PH and healthy controls. We also evaluated lung and adipose tissue expression of these mediators in the three most commonly used experimental rodent models of pulmonary hypertension. Circulating levels of FABP-4, FGF-21, and adiponectin were significantly elevated in PH patients compared to controls and the mRNA levels of these regulators and PPARγ were also significantly increased in human PH lungs and in the lungs of rats with experimental PH compared to controls. These findings were coupled with increased levels of adipose tissue mRNA of genes related to glucose uptake, glycolysis, tricarboxylic acid cycle, and fatty acid oxidation in experimental PH. Our results support that metabolic alterations in human PH are recapitulated in rodent models of the disease and suggest that adipose tissue may contribute to PH pathogenesis.
Collapse
Affiliation(s)
- Aimilia Eirini Papathanasiou
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (A.E.P.); (F.S.); (K.E.J.)
- Harvard Medical School, Boston, MA 02215, USA;
- Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 10679 Athens, Greece; (D.D.B.); (A.M.-P.)
| | - Fotios Spyropoulos
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (A.E.P.); (F.S.); (K.E.J.)
- Harvard Medical School, Boston, MA 02215, USA;
| | - Zoe Michael
- Harvard Medical School, Boston, MA 02215, USA;
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02215, USA
| | - Kyoung E. Joung
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (A.E.P.); (F.S.); (K.E.J.)
- Harvard Medical School, Boston, MA 02215, USA;
| | - Despina D. Briana
- Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 10679 Athens, Greece; (D.D.B.); (A.M.-P.)
| | - Ariadne Malamitsi-Puchner
- Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 10679 Athens, Greece; (D.D.B.); (A.M.-P.)
| | - Christos S. Mantzoros
- Harvard Medical School, Boston, MA 02215, USA;
- Division of Endocrinology Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02215, USA
- Correspondence: (C.S.M.); (H.C.)
| | - Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (A.E.P.); (F.S.); (K.E.J.)
- Harvard Medical School, Boston, MA 02215, USA;
- Correspondence: (C.S.M.); (H.C.)
| |
Collapse
|
14
|
Gonçalves AEDSS, Rocha GZ, Marin R, Camargo RL, dos Santos A, do Carmo H, Guadagnini D, Petrucci O, Moysés ZP, Salemi VMC, Oliveira AG, Saad MJA. Pulmonary Hypertension in Obese Mice Is Accompanied by a Reduction in PPAR-γ Expression in Pulmonary Artery. Front Endocrinol (Lausanne) 2021; 12:701994. [PMID: 34552556 PMCID: PMC8450870 DOI: 10.3389/fendo.2021.701994] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/16/2021] [Indexed: 11/14/2022] Open
Abstract
Obesity and insulin resistance (IR) are well-studied risk factors for systemic cardiovascular disease, but their impact on pulmonary hypertension (PH) is not well clarified. This study aims to investigate if diet-induced obesity induces PH and if peroxisome-proliferator-activated receptor (PPAR-γ) and/or endoplasmic reticulum (ER) stress are involved in this process. Mice were maintained on a high-fat diet (HFD) for 4 months, and IR and PH were confirmed. In a separate group, after 4 months of HFD, mice were treated with pioglitazone (PIO) or 4-phenylbutyric acid for the last month. The results demonstrated that HFD for at least 4 months is able to increase pulmonary artery pressure, which is maintained, and this animal model can be used to investigate the link between IR and PH, without changes in ER stress in the pulmonary artery. There was also a reduction in circulating adiponectin and in perivascular adiponectin expression in the pulmonary artery, associated with a reduction in PPAR-γ expression. Treatment with PIO improved IR and PH and reversed the lower expression of adiponectin and PPAR-γ in the pulmonary artery, highlighting this drug as potential benefit for this poorly recognized complication of obesity.
Collapse
Affiliation(s)
| | - Guilherme Zweig Rocha
- Department of Internal Medicine, Faculty of Medicine, State University of Campinas, Campinas, Brazil
| | - Rodrigo Marin
- Department of Internal Medicine, Faculty of Medicine, State University of Campinas, Campinas, Brazil
| | - Rafael Ludemann Camargo
- Department of Internal Medicine, Faculty of Medicine, State University of Campinas, Campinas, Brazil
| | - Andrey dos Santos
- Department of Internal Medicine, Faculty of Medicine, State University of Campinas, Campinas, Brazil
| | - Helison do Carmo
- Department of Internal Medicine, Faculty of Medicine, State University of Campinas, Campinas, Brazil
| | - Dioze Guadagnini
- Department of Internal Medicine, Faculty of Medicine, State University of Campinas, Campinas, Brazil
| | - Orlando Petrucci
- Department of Internal Medicine, Faculty of Medicine, State University of Campinas, Campinas, Brazil
| | - Zenaide Providello Moysés
- Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Vera Maria Cury Salemi
- Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - Mario José Abdalla Saad
- Department of Internal Medicine, Faculty of Medicine, State University of Campinas, Campinas, Brazil
- *Correspondence: Mario José Abdalla Saad,
| |
Collapse
|
15
|
Shi XF, Su YC. Vascular Metabolic Mechanisms of Pulmonary Hypertension. Curr Med Sci 2020; 40:444-454. [PMID: 32681249 DOI: 10.1007/s11596-020-2198-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/10/2020] [Indexed: 02/07/2023]
Abstract
Pulmonary hypertension (PH) is a severe and progressive disease characterized by increased pulmonary vascular resistance leading to right heart failure and death. In PH, the cellular metabolisms including those of the three major nutrients (carbohydrate, lipid and protein) are aberrant in pulmonary vascular cells. Glucose uptake, glycolysis, insulin resistance, sphingolipid S1P, PGE2, TXA2, leukotrienes and glutaminolysis are upregulated, and phospholipid-prostacyclin and L-arginine-nitric oxide pathway are compromised in lung vascular cells. Fatty acid metabolism is disordered in lung endothelial cells and smooth muscle cells. These molecular mechanisms are integrated to promote PH-specific abnormal vascular cell proliferation and vascular remodeling. This review summarizes the recent advances in the metabolic reprogramming of glucose, fatty acid, and amino acid metabolism in pulmonary vascular remodeling in PH and the mechanisms for how these alterations affect vascular cell fate and impact the course of PH.
Collapse
Affiliation(s)
- Xiao-Fan Shi
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA, 30912, USA
| | - Yun-Chao Su
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA, 30912, USA. .,Department of Medicine, Augusta University, Augusta, GA, 30912, USA. .,Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
16
|
PPARγ is a gatekeeper for extracellular matrix and vascular cell homeostasis: beneficial role in pulmonary hypertension and renal/cardiac/pulmonary fibrosis. Curr Opin Nephrol Hypertens 2020; 29:171-179. [PMID: 31815758 DOI: 10.1097/mnh.0000000000000580] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE OF REVIEW Pulmonary arterial hypertension (PAH) is characterized by pulmonary arterial endothelial cell (PAEC) dysfunction and apoptosis, pulmonary arterial smooth muscle cell (PASMC) proliferation, inflammation, vasoconstriction, and metabolic disturbances that include disrupted bone morphogenetic protein receptor (BMPR2)-peroxisome proliferator-activated receptor gamma (PPARγ) axis and DNA damage. Activation of PPARγ improves many of these mechanisms, although erroneous reports on potential adverse effects of thiazolidinedione (TZD)-class PPARγ agonists reduced their clinical use in the past decade. Here, we review recent findings in heart, lung, and kidney research related to the pathobiology of vascular remodeling and tissue fibrosis, and also potential therapeutic effects of the PPARγ agonist pioglitazone. RECENT FINDINGS Independent of its metabolic effects (improved insulin sensitivity and fatty acid handling), PPARγ activation rescues BMPR2 dysfunction, inhibits TGFβ/Smad3/CTGF and TGFβ/pSTAT3/pFoxO1 pathways, and induces the PPARγ/apoE axis, inhibiting vascular remodeling. PPARγ activation dampens mtDNA damage via PPARγ/UBR5/ATM pathway, improves function of endothelial progenitor cells (EPCs), and decrease renal fibrosis by repressing TGFβ/pSTAT3 and TGFβ/EGR1. SUMMARY Pharmacological PPARγ activation improves many hallmarks of PAH, including dysfunction of BMPR2-PPARγ axis, PAEC, PASMC, EPC, mitochondria/metabolism, and inflammation. Recent randomized controlled trials, including IRIS (Insulin Resistance Intervention After Stroke Trial), emphasize the beneficial effects of PPARγ agonists in PAH patients, leading to recent revival for clinical use.
Collapse
|
17
|
Duan D, Gu C, Jun JC. Altered metabolism in pulmonary hypertension: fuelling the fire or just smoke? Eur Respir J 2020; 55:2000447. [PMID: 32273332 PMCID: PMC7573924 DOI: 10.1183/13993003.00447-2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 02/29/2020] [Indexed: 11/05/2022]
Affiliation(s)
- Daisy Duan
- Division of Endocrinology, Diabetes and Metabolism, Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chenjuan Gu
- Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan C Jun
- Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Liu J, Hu S, Zhu B, Shao S, Yuan L. Grape seed procyanidin suppresses inflammation in cigarette smoke-exposed pulmonary arterial hypertension rats by the PPAR-γ/COX-2 pathway. Nutr Metab Cardiovasc Dis 2020; 30:347-354. [PMID: 31791634 DOI: 10.1016/j.numecd.2019.09.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 08/27/2019] [Accepted: 09/19/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND AIM Pulmonary arterial hypertension (PAH) is characterized by pulmonary vascular remodeling, which is mainly caused by inflammation. Inhibiting inflammation can relieve PAH. Grape seed procyanidin (GSP) possesses remarkable anti-inflammatory property and vascular protective function. In this experiment, we verified the anti-inflammatory property of GSP in cigarette smoke-exposed PAH rats and revealed its molecular mechanism. METHODS AND RESULTS In vivo, 45 Sprague Dawley (SD) rats were divided into 5 groups randomly, treated with normoxia/cigarette smoke (CS)/GSP + CS/CS + solvent/GSP. After GSP + CS administration, a decrease in mPAP, PVR, RVHI, WT%, and WA% was detected in the rats as compared to those treated with CS. In vitro, the proliferation of pulmonary arterial smooth muscle cells (PASMCs) caused by cigarette smoke extract (CSE) was effectively attenuated with GSP + CSE administration. Furthermore, GSP significantly increased the expression of peroxisome proliferator-activated receptor γ (PPAR-γ) together with the lowered expression level of cyclooxygenase 2 (COX-2) in PASMCs co-incubated with CSE. CONCLUSION These findings indicate that GSP ameliorates inflammation by the PPAR-γ/COX-2 pathway and finally inhibits the proliferation of PASMCs, which leads to pulmonary vascular remodeling.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Cell Proliferation/drug effects
- Cells, Cultured
- Cigarette Smoking
- Cyclooxygenase 2/metabolism
- Disease Models, Animal
- Grape Seed Extract/pharmacology
- Inflammation/enzymology
- Inflammation/etiology
- Inflammation/physiopathology
- Inflammation/prevention & control
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- PPAR gamma/metabolism
- Proanthocyanidins/pharmacology
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/enzymology
- Pulmonary Arterial Hypertension/etiology
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/enzymology
- Pulmonary Artery/pathology
- Rats, Sprague-Dawley
- Signal Transduction
- Vascular Remodeling/drug effects
- Ventricular Function, Right/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Jiantao Liu
- The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, PR China
| | - Songli Hu
- The Renji College, Wenzhou Medical University, Wenzhou, PR China
| | - Bingqing Zhu
- The Renji College, Wenzhou Medical University, Wenzhou, PR China
| | - Siming Shao
- The Renji College, Wenzhou Medical University, Wenzhou, PR China
| | - Linbo Yuan
- Department of Physiology, Basic Medical Science School, Wenzhou Medical University, Wenzhou, PR China.
| |
Collapse
|
19
|
Hansmann G, Calvier L, Risbano MG, Chan SY. Activation of the Metabolic Master Regulator PPARγ: A Potential PIOneering Therapy for Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2020; 62:143-156. [PMID: 31577451 PMCID: PMC6993553 DOI: 10.1165/rcmb.2019-0226ps] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/02/2019] [Indexed: 12/24/2022] Open
Abstract
Translational research is essential to the development of reverse-remodeling strategies for the treatment of pulmonary vascular disease, pulmonary hypertension, and heart failure via mechanistic in vivo studies using animal models resembling human pulmonary arterial hypertension (PAH), cardiovascular remodeling, and progressive right heart failure. Since 2007, peroxisome proliferator-activated receptor γ (PPARγ) agonists have emerged as promising novel, antiproliferative, antiinflammatory, insulin-sensitizing, efficient medications for the treatment of PAH. However, early diabetes study results, their subsequent misinterpretations, errors in published review articles, and rumors regarding potential adverse effects in the literature have dampened enthusiasm for considering pharmacological PPARγ activation for the treatment of cardiovascular diseases, including PAH. Most recently, the thiazolidinedione class PPARγ agonist pioglitazone underwent a clinical revival, especially based on the IRIS (Insulin Resistance Intervention After Stroke) study, a randomized controlled trial in 3,876 patients without diabetes status post-transient ischemic attack/ischemic stroke who were clinically followed for 4.8 years. We discuss preclinical basic translational findings and randomized controlled trials related to the beneficial and adverse effects of PPARγ agonists of the thiazolidinedione class, with a particular focus on the last 5 years. The objective is a data-driven approach to set the preclinical and clinical study record straight. The convincing recent clinical trial data on the lack of significant toxicity in high-risk populations justify the timely conduct of clinical studies to achieve "repurposing" or "repositioning" of pioglitazone for the treatment of clinical PAH.
Collapse
Affiliation(s)
- Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany; and
| | - Laurent Calvier
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany; and
| | - Michael G. Risbano
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, and
| | - Stephen Y. Chan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, and
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
20
|
Khaing P, Pandit P, Awsare B, Summer R. Pulmonary Circulation in Obesity, Diabetes, and Metabolic Syndrome. Compr Physiol 2019; 10:297-316. [DOI: 10.1002/cphy.c190018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
21
|
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
|
22
|
Legchenko E, Chouvarine P, Borchert P, Fernandez-Gonzalez A, Snay E, Meier M, Maegel L, Mitsialis SA, Rog-Zielinska EA, Kourembanas S, Jonigk D, Hansmann G. PPARγ agonist pioglitazone reverses pulmonary hypertension and prevents right heart failure via fatty acid oxidation. Sci Transl Med 2019; 10:10/438/eaao0303. [PMID: 29695452 DOI: 10.1126/scitranslmed.aao0303] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/18/2017] [Accepted: 03/12/2018] [Indexed: 12/13/2022]
Abstract
Right ventricular (RV) heart failure is the leading cause of death in pulmonary arterial hypertension (PAH). Peroxisome proliferator-activated receptor γ (PPARγ) acts as a vasoprotective metabolic regulator in smooth muscle and endothelial cells; however, its role in the heart is unclear. We report that deletion of PPARγ in cardiomyocytes leads to biventricular systolic dysfunction and intramyocellular lipid accumulation in mice. In the SU5416/hypoxia (SuHx) rat model, oral treatment with the PPARγ agonist pioglitazone completely reverses severe PAH and vascular remodeling and prevents RV failure. Failing RV cardiomyocytes exhibited mitochondrial disarray and increased intramyocellular lipids (lipotoxicity) in the SuHx heart, which was prevented by pioglitazone. Unbiased ventricular microRNA (miRNA) arrays, mRNA sequencing, and lipid metabolism studies revealed dysregulation of cardiac hypertrophy, fibrosis, myocardial contractility, fatty acid transport/oxidation (FAO), and transforming growth factor-β signaling in the failing RV. These epigenetic, transcriptional, and metabolic alterations were modulated by pioglitazone through miRNA/mRNA networks previously not associated with PAH/RV dysfunction. Consistently, pre-miR-197 and pre-miR-146b repressed genes that drive FAO (Cpt1b and Fabp4) in primary cardiomyocytes. We recapitulated our major pathogenic findings in human end-stage PAH: (i) in the pressure-overloaded failing RV (miR-197 and miR-146b up-regulated), (ii) in peripheral pulmonary arteries (miR-146b up-regulated, miR-133b down-regulated), and (iii) in plexiform vasculopathy (miR-133b up-regulated, miR-146b down-regulated). Together, PPARγ activation can normalize epigenetic and transcriptional regulation primarily related to disturbed lipid metabolism and mitochondrial morphology/function in the failing RV and the hypertensive pulmonary vasculature, representing a therapeutic approach for PAH and other cardiovascular/pulmonary diseases.
Collapse
Affiliation(s)
- Ekaterina Legchenko
- Department of Pediatric Cardiology and Critical Care, Pulmonary Vascular Research Center, Hannover Medical School, Hannover, Germany
| | - Philippe Chouvarine
- Department of Pediatric Cardiology and Critical Care, Pulmonary Vascular Research Center, Hannover Medical School, Hannover, Germany
| | - Paul Borchert
- Department of Pediatric Cardiology and Critical Care, Pulmonary Vascular Research Center, Hannover Medical School, Hannover, Germany
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Erin Snay
- Division of Nuclear Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Martin Meier
- Small Animal Imaging Center, Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Lavinia Maegel
- Institute of Pathology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Hannover, Germany.,The German Center for Lung Research (Deutsches Zentrum für Lungenforschung DZL), Giessen, Germany
| | - S Alex Mitsialis
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Eva A Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Hannover, Germany.,The German Center for Lung Research (Deutsches Zentrum für Lungenforschung DZL), Giessen, Germany
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Pulmonary Vascular Research Center, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
23
|
Abstract
Pulmonary hypertension (PH) and its severe subtype pulmonary arterial hypertension (PAH) encompass a set of multifactorial diseases defined by sustained elevation of pulmonary arterial pressure and pulmonary vascular resistance leading to right ventricular failure and subsequent death. Pulmonary hypertension is characterized by vascular remodeling in association with smooth muscle cell proliferation of the arterioles, medial thickening, and plexiform lesion formation. Despite our recent advances in understanding its pathogenesis and related therapeutic discoveries, PH still remains a progressive disease without a cure. Nevertheless, development of drugs that specifically target molecular pathways involved in disease pathogenesis has led to improvement in life quality and clinical outcomes in patients with PAH. There are presently more than 12 Food and Drug Administration-approved vasodilator drugs in the United States for the treatment of PAH; however, mortality with contemporary therapies remains high. More recently, there have been exuberant efforts to develop new pharmacologic therapies that target the fundamental origins of PH and thus could represent disease-modifying opportunities. This review aims to summarize recent developments on key signaling pathways and molecular targets that drive PH disease progression, with emphasis on new therapeutic options under development.
Collapse
Affiliation(s)
- Chen-Shan Chen Woodcock
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen Y. Chan
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
24
|
Li Q, Wu J, Xu Y, Liu L, Xie J. Role of RASEF hypermethylation in cigarette smoke-induced pulmonary arterial smooth muscle remodeling. Respir Res 2019; 20:52. [PMID: 30845941 PMCID: PMC6407244 DOI: 10.1186/s12931-019-1014-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/21/2019] [Indexed: 01/06/2023] Open
Abstract
Background Pulmonary hypertension (PH) is a progressive and fatal disease. While cigarette smoke can change DNA methylation status, the role of such molecular alterations in smoke-associated PH is unclear. Methods A PH rat model was developed by exposing animals to cigarette smoke for 3 months. Right ventricular systolic pressure was measured with a right heart catheter. Histological changes (right ventricular hypertrophy index, medial wall thickness in pulmonary arteries (PAs)) and DNMT1 protein levels in rat PAs or primary human PA smooth muscle cells (HPASMCs) exposed to cigarette smoke extract were assessed. Methylation sequencing and MassArray® were used to detect genomic and RASEF promoter methylation status, respectively. After DNMT1 knockdown and cigarette smoke extract exposure, HPASMCs behavior (proliferation, migration) and RASEF methylation status were examined; RASEF mRNA expression was evaluated by real-time-polymerase chain reaction. RASEF overexpression viral vectors were used to assess the impact of RASEF on rat PH and HPASMCs remodeling. Results Higher right ventricular systolic pressure, medial wall thickness, and right ventricular hypertrophy index values were observed in the smoking group rats. Smoke exposure increased DNMT1 expression and RASEF methylation levels in rat PAs and HPASMCs. Cigarette smoke extract induced HPASMCs behavioral changes and RASEF hypermethylation followed by silencing, while DNMT1 knockdown markedly inhibited these changes. RASEF overexpression distinctly inhibited PH and HPASMCs remodeling, possibly through phospho-AKT (Ser473), PCNA, and MMP9 downregulation. Conclusions Cigarette smoke caused PA remodeling in PH rats related to RASEF hypermethylation. These results expand our understanding of key epigenetic mechanisms in cigarette smoke-associated PH and potentially provide a novel therapeutic target for PH. Electronic supplementary material The online version of this article (10.1186/s12931-019-1014-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qinghai Li
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Pulmonary Medicine, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266011, China
| | - Jixing Wu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yongjian Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lu Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
25
|
Caglayan E, Trappiel M, Behringer A, Berghausen EM, Odenthal M, Wellnhofer E, Kappert K. Pulmonary arterial remodelling by deficiency of peroxisome proliferator-activated receptor-γ in murine vascular smooth muscle cells occurs independently of obesity-related pulmonary hypertension. Respir Res 2019; 20:42. [PMID: 30813929 PMCID: PMC6391752 DOI: 10.1186/s12931-019-1003-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/11/2019] [Indexed: 11/30/2022] Open
Abstract
Background Obesity is associated with cardiovascular complications, including pulmonary hypertension (PH). Reports suggest that peroxisome proliferator-activated receptor-γ (PPARγ) has direct action in preventing vascular remodelling in PH. Here we dissected the specific role of high-fat-diet (HFD)-induced obesity and vascular smooth muscle cell (VSMC)-PPARγ for remodelling of small pulmonary arteries. Methods Wild-type (WT) and VSMC-specific PPARγ-knockout (SmPparγ−/−) mice were fed a low-fat-diet (LFD, 10% kcal from fat) or HFD (60% kcal from fat) for 24 weeks. Mice were metabolically phenotyped (e.g. weight development, insulin/glucose tolerance) at the beginning, and after 12 and 24 weeks, respectively. At 24 weeks additionally pulmonary pressure, heart structure, pulmonary vascular muscularization together with gene and protein expression in heart and lung tissues were determined. Results HFD increased right ventricular systolic pressure (RVSP) to a similar extent in WT and SmPparγ−/− mice. HFD decreased glucose tolerance and insulin sensitivity in both WT and SmPparγ−/− mice. Importantly, the increase in RVSP correlated with the degree of insulin resistance. However, VSMC-PPARγ deficiency increased pulmonary vascular muscularization independently of the diet-induced rise in RVSP. This increase was associated with elevated expression of early growth response protein 1 in heart and osteopontin in lung tissue. Conclusions Here we demonstrate a correlation of insulin resistance and pulmonary pressure. Further, deficiency of PPARγ in VSMCs diet-independently leads to increased pulmonary vascular muscularization.
Collapse
Affiliation(s)
- Evren Caglayan
- Klinik III für Innere Medizin, University of Cologne Heart Center, Cologne, Germany.,Center for Molecular Medine Cologne (CMMC), Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany.,Department of Cardiology, University Medicine Rostock, Rostock, Germany
| | - Manuela Trappiel
- Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Center for Cardiovascular Research (CCR), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Arnica Behringer
- Klinik III für Innere Medizin, University of Cologne Heart Center, Cologne, Germany.,Center for Molecular Medine Cologne (CMMC), Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Eva Maria Berghausen
- Klinik III für Innere Medizin, University of Cologne Heart Center, Cologne, Germany
| | | | - Ernst Wellnhofer
- Department of Cardiology, German Heart Center Berlin, Berlin, Germany
| | - Kai Kappert
- Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Center for Cardiovascular Research (CCR), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
26
|
Pulmonary Hypertension and Obesity: Focus on Adiponectin. Int J Mol Sci 2019; 20:ijms20040912. [PMID: 30791536 PMCID: PMC6412189 DOI: 10.3390/ijms20040912] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 02/15/2019] [Accepted: 02/16/2019] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension is an umbrella term including many different disorders causing an increase of the mean pulmonary arterial pressure (mPAP) ≥ 25 mmHg. Recent data revealed a strong association between obesity and pulmonary hypertension. Adiponectin is a protein synthetized by the adipose tissue with pleiotropic effects on inflammation and cell proliferation, with a potential protective role on the pulmonary vasculature. Both in vivo and in vitro studies documented that adiponectin is an endogenous modulator of NO production and interferes with AMP-activated protein kinase (AMPK) activation, mammalian target of rapamycin (mTOR), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κβ) signaling preventing endothelial dysfunction and proliferation. Furthermore, adiponectin ameliorates insulin resistance by mediating the biological effects of peroxisome proliferator-activated receptor-gamma (PPARγ). Therefore, adiponectin modulation emerged as a theoretical target for the treatment of pulmonary hypertension, currently under investigation. Recently, consistent data showed that hypoglycemic agents targeting PPARγ as well as renin–angiotensin system inhibitors and mineralocorticoid receptor blockers may influence pulmonary hemodynamics in different models of pulmonary hypertension.
Collapse
|
27
|
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: 14] [Impact Index Per Article: 2.3] [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
|
28
|
Gosemann JH, Friedmacher F, Hofmann A, Zimmer J, Kuebler JF, Rittinghausen S, Suttkus A, Lacher M, Alvarez L, Corcionivoschi N, Puri P. Prenatal treatment with rosiglitazone attenuates vascular remodeling and pulmonary monocyte influx in experimental congenital diaphragmatic hernia. PLoS One 2018; 13:e0206975. [PMID: 30418988 PMCID: PMC6231640 DOI: 10.1371/journal.pone.0206975] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/23/2018] [Indexed: 01/29/2023] Open
Abstract
Introduction Extensive vascular remodeling causing pulmonary hypertension (PH) represents a major cause of mortality in patients with congenital diaphragmatic hernia (CDH). The chemokine monocyte chemoattractant protein-1 (MCP-1) is a biomarker for the severity of PH and its activation is accompanied by pulmonary influx of monocytes and extensive vascular remodeling. MCP-1 activation can be reversed by application of rosiglitazone (thiazolidinedione). We performed this study to evaluate the role of MCP-1 for the pathogenesis of PH in experimental CDH. We hypothesized that vascular remodeling and MCP-1 activation is accompanied by pulmonary influx of fetal monocytes and can be attenuated by prenatal treatment with rosiglitazone. Methods In a first set of experiments pregnant rats were treated with either nitrofen or vehicle on gestational day 9 (D9). Fetal lungs were harvested on D21 and divided into CDH and control. Quantitative real-time polymerase chain reaction, Western blot (WB), and immunohistochemistry (IHC) were used to evaluate MCP-1 expression, activation, and localization. Quantification and localization of pulmonary monocytes/macrophages were carried out by IHC. In a second set of experiments nitrofen-exposed dams were randomly assigned to prenatal treatment with rosiglitazone or placebo on D18+D19. Fetal lungs were harvested on D21, divided into control, CDH+rosiglitazone, and CDH+placebo and evaluated by WB as well as IHC. Results Increased thickness of pulmonary arteries of CDH fetuses was accompanied by increased systemic and perivascular MCP-1 protein expression and significantly higher amounts of pulmonary monocytes/macrophages compared to controls (p<0.01). These effects were reversed by prenatal treatment with rosiglitazone (p<0.01 vs. CDH+P; control). Conclusion Prenatal treatment with rosiglitazone has the potential to attenuate activation of pulmonary MCP-1, pulmonary monocyte influx, and vascular remodeling in experimental CDH. These results provide a basis for future research on prenatal immunomodulation as a novel treatment strategy to decrease secondary effects of PH in CDH.
Collapse
MESH Headings
- Animals
- Chemokine CCL2/blood
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Disease Models, Animal
- Female
- Gene Expression
- Hernias, Diaphragmatic, Congenital/drug therapy
- Hernias, Diaphragmatic, Congenital/etiology
- Hernias, Diaphragmatic, Congenital/metabolism
- Hernias, Diaphragmatic, Congenital/pathology
- Immunohistochemistry
- Lung/metabolism
- Lung/pathology
- Macrophages/immunology
- Macrophages/metabolism
- Monocytes/drug effects
- Monocytes/metabolism
- Phenyl Ethers/adverse effects
- Pregnancy
- Prenatal Care
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rosiglitazone/pharmacology
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Jan-Hendrik Gosemann
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
- * E-mail:
| | - Florian Friedmacher
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland
- The Royal London Hospital, London, United Kingdom
| | - Alejandro Hofmann
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Julia Zimmer
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Joachim F. Kuebler
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Susanne Rittinghausen
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | - Anne Suttkus
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Martin Lacher
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Luis Alvarez
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland
- Wellcome Centre Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Nicolae Corcionivoschi
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland
- Agri-Food and Biosciences Institute, Belfast, Northern Ireland, United Kingdom
| | - Prem Puri
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland
- School of Medicine and Medical Science and Conway Institute of Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
29
|
Liu J, Cai G, Li M, Fan S, Yao B, Ping W, Huang Z, Cai H, Dai Y, Wang L, Huang X. Fibroblast growth factor 21 attenuates hypoxia-induced pulmonary hypertension by upregulating PPARγ expression and suppressing inflammatory cytokine levels. Biochem Biophys Res Commun 2018; 504:478-484. [PMID: 30197006 DOI: 10.1016/j.bbrc.2018.09.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/01/2018] [Indexed: 11/26/2022]
Abstract
Hypoxia-induced pulmonary hypertension (HPH) is a progressive disease characterized by a sustained, elevated pulmonary arterial pressure and vascular remodeling. The latter pathogenesis mainly involves overproliferation of pulmonary artery smooth muscle cells (PASMCs). Fibroblast growth factor 21 (FGF21) has recently emerged as a novel regulator that prevents cardiac hypertrophic remodeling. However, its possible role in pulmonary remodeling remains unclear. The activation of peroxisome proliferator activated receptor γ (PPARγ) is reported to attenuate HPH by suppressing proliferative signals. Loss of PPARγ in the lung contributes to abnormal proliferation of PASMCs. FGF21 is a key regulator of PPARγ activity in adipocytes, but its role has not been elucidated in PASMCs. Therefore, we hypothesized that FGF21 may confer therapeutic effects in HPH by upregulating the expression of PPARγ. Sprague-Dawley rats were exposed to hypoxia and treated with FGF21 for 4 weeks. In parallel, hypoxic conditions and FGF21 were administered to rat PASMCs for 48 h. FGF21 attenuated the hypoxia-induced elevation in mean pulmonary arterial pressure (mPAP), right ventricular hypertrophy (RVH), medial thickening and overproliferation of PASMCs. Furthermore, FGF21 abrogated the reductions in PPARγ expression and increases in TNF-α, IL-1 and IL-6 levels in PASMC culture media. Collectively, these results demonstrate that FGF21 could potentially attenuate the pathogenic derangements of HPH by targeting PPARγ and inflammatory cytokines.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, 325000, PR China
| | - Gexiang Cai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, 325000, PR China
| | - Manxiang Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Shanxi, 710061, PR China
| | - Shiqian Fan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, 325000, PR China
| | - Boyang Yao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, 325000, PR China
| | - Weidong Ping
- Chinese People's Liberation Army 117 Hospital, Zhejiang, 310013, PR China
| | - Zhifeng Huang
- Key Laboratory of Biotechnology and Pharmaceutical Engineering of Zhejiang Province, Wenzhou Medical University, Zhejiang, 325000, PR China
| | - Hui Cai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, 325000, PR China
| | - Yongyue Dai
- Department of Pathophysiology, Wenzhou Medical University, Zhejiang, 325000, PR China
| | - Liangxing Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, 325000, PR China.
| | - Xiaoying Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, 325000, PR China.
| |
Collapse
|
30
|
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
|
31
|
Chan SY, Rubin LJ. Metabolic dysfunction in pulmonary hypertension: from basic science to clinical practice. Eur Respir Rev 2017; 26:26/146/170094. [PMID: 29263174 PMCID: PMC5842433 DOI: 10.1183/16000617.0094-2017] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/28/2017] [Indexed: 01/29/2023] Open
Abstract
Pulmonary hypertension (PH) is an often-fatal vascular disease of unclear molecular origins. The pulmonary vascular remodelling which occurs in PH is characterised by elevated vasomotor tone and a pro-proliferative state, ultimately leading to right ventricular dysfunction and heart failure. Guided in many respects by prior evidence from cancer biology, recent investigations have identified metabolic aberrations as crucial components of the disease process in both the pulmonary vessels and the right ventricle. Given the need for improved diagnostic and therapeutic options for PH, the development or repurposing of metabolic tracers and medications could provide an effective avenue for preventing or even reversing disease progression. In this review, we describe the metabolic mechanisms that are known to be dysregulated in PH; we explore the advancing diagnostic testing and imaging modalities that are being developed to improve diagnostic capability for this disease; and we discuss emerging drugs for PH which target these metabolic pathways. Understanding metabolic pathways in PH provides opportunities for improved diagnostic and therapeutic optionshttp://ow.ly/pFQb30guez6
Collapse
Affiliation(s)
- Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Dept of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Lewis J Rubin
- University of California, San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
32
|
Xu Y, Gu Q, Liu N, Yan Y, Yang X, Hao Y, Qu C. PPARγ Alleviates Right Ventricular Failure Secondary to Pulmonary Arterial Hypertension in Rats. Int Heart J 2017; 58:948-956. [PMID: 29151490 DOI: 10.1536/ihj.16-591] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by pulmonary vascular remodeling leading to right ventricular hypertrophy (RVH) and failure. Peroxisome proliferator-activated receptor γ (PPARγ), a member of nuclear receptors, has been proved to ameliorate PAH. However, its effect on PAH-induced right ventricular failure (RVF) remains unknown. Therefore, we investigated the therapeutic potential of PPARγ in preventing monocrotaline (MCT)-induced RV dysfunction. The PAH model was induced by MCT administration. Male rats were administered with MCT to develop PAH and RVF formed by approximately day 30. Significant increase in RV area, RVAW resulted in an ascending RV index. However, the LV function including EF, FS, and LVID did not change significantly. PPARγ agonist prevented PAH-induced RVF by preserving RV index and preventing RVH. PPARγ's beneficial effects seem to result from various factors, including anti-apoptosis, preservation RV index, reversal of inflammation, improvement of glucolipid metabolism, reduction of ROS. In a word, PPARγ agonist prevents the development of RVF.
Collapse
Affiliation(s)
- Ying Xu
- Intensive Care Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University
| | - Qin Gu
- Intensive Care Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University
| | - Ning Liu
- Intensive Care Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University
| | - Yan Yan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University
| | - Xilan Yang
- Department of Geriatric Medicine, The Second Affiliated Hospital of Nanjing Medical University
| | - Yingying Hao
- Intensive Care Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University
| | - Chen Qu
- Department of Geriatric Medicine, The Second Affiliated Hospital of Nanjing Medical University
| |
Collapse
|
33
|
Lecarpentier Y, Schussler O, Claes V, Vallée A. The Myofibroblast: TGFβ-1, A Conductor which Plays a Key Role in Fibrosis by Regulating the Balance between PPARγ and the Canonical WNT Pathway. NUCLEAR RECEPTOR RESEARCH 2017. [DOI: 10.11131/2017/101299] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l’Est Francilien (GHEP), Meaux, France
| | - Olivier Schussler
- Department of Cardiovascular Surgery, Cardiovascular Research Laboratory, HUG/CMU, Geneva, Switzerland
| | - Victor Claes
- Department of Pharmaceutical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, Poitiers, France
| |
Collapse
|
34
|
Calvier L, Chouvarine P, Legchenko E, Hoffmann N, Geldner J, Borchert P, Jonigk D, Mozes MM, Hansmann G. PPARγ Links BMP2 and TGFβ1 Pathways in Vascular Smooth Muscle Cells, Regulating Cell Proliferation and Glucose Metabolism. Cell Metab 2017; 25:1118-1134.e7. [PMID: 28467929 DOI: 10.1016/j.cmet.2017.03.011] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 12/21/2016] [Accepted: 03/20/2017] [Indexed: 01/24/2023]
Abstract
BMP2 and TGFβ1 are functional antagonists of pathological remodeling in the arteries, heart, and lung; however, the mechanisms in VSMCs, and their disturbance in pulmonary arterial hypertension (PAH), are unclear. We found a pro-proliferative TGFβ1-Stat3-FoxO1 axis in VSMCs, and PPARγ as inhibitory regulator of TGFβ1-Stat3-FoxO1 and TGFβ1-Smad3/4, by physically interacting with Stat3 and Smad3. TGFβ1 induces fibrosis-related genes and miR-130a/301b, suppressing PPARγ. Conversely, PPARγ inhibits TGFβ1-induced mitochondrial activation and VSMC proliferation, and regulates two glucose metabolism-related enzymes, platelet isoform of phosphofructokinase (PFKP, a PPARγ target, via miR-331-5p) and protein phosphatase 1 regulatory subunit 3G (PPP1R3G, a Smad3 target). PPARγ knockdown/deletion in VSMCs activates TGFβ1 signaling. The PPARγ agonist pioglitazone reverses PAH and inhibits the TGFβ1-Stat3-FoxO1 axis in TGFβ1-overexpressing mice. We identified PPARγ as a missing link between BMP2 and TGFβ1 pathways in VSMCs. PPARγ activation can be beneficial in TGFβ1-associated diseases, such as PAH, parenchymal lung diseases, and Marfan's syndrome.
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Philippe Chouvarine
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Ekaterina Legchenko
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Nadine Hoffmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Jonas Geldner
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Paul Borchert
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover 30625, Germany
| | - Miklos M Mozes
- Department of Pathophysiology, Semmelweis University, Budapest 1089, Hungary
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany.
| |
Collapse
|
35
|
Di Mise A, Wang YX, Zheng YM. Role of Transcription Factors in Pulmonary Artery Smooth Muscle Cells: An Important Link to Hypoxic Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:13-32. [PMID: 29047078 DOI: 10.1007/978-3-319-63245-2_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypoxia, namely a lack of oxygen in the blood, induces pulmonary vasoconstriction and vasoremodeling, which serve as essential pathologic factors leading to pulmonary hypertension (PH). The underlying molecular mechanisms are uncertain; however, pulmonary artery smooth muscle cells (PASMCs) play an essential role in hypoxia-induced pulmonary vasoconstriction, vasoremodeling, and PH. Hypoxia causes oxidative damage to DNAs, proteins, and lipids. This damage (oxidative stress) modulates the activity of ion channels and elevates the intracellular calcium concentration ([Ca2+]i, Ca2+ signaling) of PASMCs. The oxidative stress and increased Ca2+ signaling mutually interact with each other, and synergistically results in a variety of cellular responses. These responses include functional and structural abnormalities of mitochondria, sarcoplasmic reticulum, and nucleus; cell contraction, proliferation, migration, and apoptosis, as well as generation of vasoactive substances, inflammatory molecules, and growth factors that mediate the development of PH. A number of studies reveal that various transcription factors (TFs) play important roles in hypoxia-induced oxidative stress, disrupted PAMSC Ca2+ signaling and the development and progress of PH. It is believed that in the pathogenesis of PH, hypoxia facilitates these roles by mediating the expression of multiple genes. Therefore, the identification of specific genes and their transcription factors implicated in PH is necessary for the complete understanding of the underlying molecular mechanisms. Moreover, this identification may aid in the development of novel and effective therapeutic strategies for PH.
Collapse
Affiliation(s)
- Annarita Di Mise
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Yong-Xiao Wang
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| | - Yun-Min Zheng
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
36
|
Rashid J, Ahsan F. A highly sensitive LC-MS/MS method for concurrent determination of sildenafil and rosiglitazone in rat plasma. J Pharm Biomed Anal 2016; 129:21-27. [PMID: 27392173 DOI: 10.1016/j.jpba.2016.06.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/10/2016] [Accepted: 06/13/2016] [Indexed: 12/28/2022]
Abstract
Patients with pulmonary arterial hypertension (PAH) are currently treated with more than one drug. Sildenafil, a phosphodiesterase type 5 (PDE-5) inhibitor, and rosiglitazone, a peroxisome proliferator-activated receptor γ (PPAR-γ) activator, is one of those combinations that could be used in PAH. To monitor the pharmacokinetics of sildenafil in the presence of rosiglitazone, we have developed and validated a sensitive, specific and rapid liquid chromatography-tandem mass spectrometric (LC-MS/MS) method. We have used this validated method to study the pharmacokinetics of sildenafil and rosiglitazone after intravenous administration of sildenafil alone or a combination of sildenafil plus rosiglitazone to adult male Sprague-Dawley rats. Sildenafil and rosiglitazone were extracted from plasma by protein precipitation with methanol. With an octadeuterated sildenafil as the internal standard, the drugs were separated via gradient elution using a C18 column and formic acid in methanol or in water as the mobile phase with a flow rate of 0.25mL/min. Both sildenafil and rosiglitazone samples in rat plasma produced linear response, when the concentration ranged between 5 and 1000ng/mL (r(2)>0.99). The pharmacokinetics study suggests that intravenous co-administration rosiglitazone plus sildenafil increases the plasma concentration of sildenafil and extends the drug's elimination half-life.
Collapse
Affiliation(s)
- Jahidur Rashid
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 Coulter St., Amarillo, TX 79106, United States
| | - Fakhrul Ahsan
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 Coulter St., Amarillo, TX 79106, United States.
| |
Collapse
|
37
|
Hussain N, Charalampopoulos A, Ramjug S, Condliffe R, Elliot CA, O'Toole L, Swift A, Kiely DG. Pulmonary hypertension in patients with heart failure and preserved ejection fraction: differential diagnosis and management. Pulm Circ 2016; 6:3-14. [PMID: 27162611 PMCID: PMC4860544 DOI: 10.1086/685021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The most common cause of pulmonary hypertension (PH) due to left heart disease (LHD) was previously rheumatic mitral valve disease. However, with the disappearance of rheumatic fever and an aging population, nonvalvular LHD is now the most common cause of group 2 PH in the developed world. In this review, we examine the challenge of investigating patients who have PH and heart failure with preserved ejection fraction (HF-pEF), where differentiating between pulmonary arterial hypertension (PAH) and PH-LHD can be difficult, and also discuss the entity of combined precapillary and postcapillary PH. Given the proven efficacy of targeted therapy for the treatment of PAH, there is increasing interest in whether these treatments may benefit selected patients with PH associated with HF-pEF, and we review current trial data.
Collapse
Affiliation(s)
- Nehal Hussain
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield National Health Service (NHS) Teaching Hospitals Foundation Trust, Sheffield, United Kingdom
| | - Athanasios Charalampopoulos
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield National Health Service (NHS) Teaching Hospitals Foundation Trust, Sheffield, United Kingdom
| | - Sheila Ramjug
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield National Health Service (NHS) Teaching Hospitals Foundation Trust, Sheffield, United Kingdom
| | - Robin Condliffe
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield National Health Service (NHS) Teaching Hospitals Foundation Trust, Sheffield, United Kingdom
| | - Charlie A Elliot
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield National Health Service (NHS) Teaching Hospitals Foundation Trust, Sheffield, United Kingdom
| | - Laurence O'Toole
- Department of Cardiology, Northern General Hospital, Sheffield NHS Teaching Hospitals Foundation Trust, Sheffield, United Kingdom
| | - Andrew Swift
- Academic Unit of Radiology, University of Sheffield, Royal Hallamshire Hospital, Sheffield NHS Teaching Hospitals Foundation Trust, Sheffield, United Kingdom
| | - David G Kiely
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield National Health Service (NHS) Teaching Hospitals Foundation Trust, Sheffield, United Kingdom
| |
Collapse
|
38
|
Abstract
Previously considered a disease isolated to the pulmonary circulation, pulmonary arterial hypertension is now being recognized as a systemic disorder that is associated with significant metabolic dysfunction. Numerous animal models have demonstrated the development of pulmonary arterial hypertension following the onset of insulin resistance, indicating that insulin resistance may be causal. Recent publications highlighting alterations in aerobic glycolysis, fatty acid oxidation, and the tricarboxylic acid cycle in the pulmonary circulation and right ventricle have expanded our understanding of the complex pathobiology of this disease. By targeting these derangements in metabolism, numerous researchers are investigating noninvasive techniques to monitor disease activity and therapeutics that address the underlying metabolic condition. In the following review, we will explore pre-clinical and clinical studies investigating the metabolic dysfunction seen in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Tufik R Assad
- Division of Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, T1218 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232, USA
| | | |
Collapse
|
39
|
Ye L, Jiang Y, Zuo X. Farnesoid-X-receptor expression in monocrotaline-induced pulmonary arterial hypertension and right heart failure. Biochem Biophys Res Commun 2015; 467:164-70. [PMID: 26392308 DOI: 10.1016/j.bbrc.2015.09.067] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 09/11/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The farnesoid-X-receptor (FXR) is a metabolic nuclear receptor superfamily member that is highly expressed in enterohepatic tissue and is also expressed in the cardiovascular system. Multiple nuclear receptors, including FXR, play a pivotal role in cardiovascular disease (CVD). Pulmonary arterial hypertension (PAH) is an untreatable cardiovascular system disease that leads to right heart failure (RHF). However, the potential physiological/pathological roles of FXR in PAH and RHF are unknown. We therefore compared FXR expression in the cardiovascular system in PAH, RHF and a control. METHODS AND RESULTS Hemodynamic parameters and morphology were assessed in blank solution-exposed control, monocrotaline (MCT)-exposed PAH (4 weeks) and RHF (7 weeks) Sprague-Dawley rats. Real-time reverse transcription polymerase chain reaction (real-time RT-PCR), Western blot (WB), immunohistochemistry (IHC) analysis and immunofluorescence (IF) analysis were performed to assess FXR levels in the lung and heart tissues of MCT-induced PAH and RHF rats. In normal rats, low FXR levels were detected in the heart, and nearly no FXR was expressed in rat lungs. However, FXR expression was significantly elevated in PAH and RHF rat lungs but reduced in PAH and RHF rat right ventricular (RV) tissues. FXR expression was reduced only in RHF rat left ventricular (LV) tissues. CONCLUSIONS The differential expression of FXR in MCT-induced PAH lungs and heart tissues in parallel with PAH pathophysiological processes suggests that FXR contributes to PAH.
Collapse
Affiliation(s)
- Lusi Ye
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Department of Rheumatology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325015, China
| | - Ying Jiang
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Xiaoxia Zuo
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China.
| |
Collapse
|
40
|
Abernethy AD, Stackhouse K, Hart S, Devendra G, Bashore TM, Dweik R, Krasuski RA. Impact of diabetes in patients with pulmonary hypertension. Pulm Circ 2015; 5:117-23. [PMID: 25992276 DOI: 10.1086/679705] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 08/26/2014] [Indexed: 12/20/2022] Open
Abstract
Diabetes complicates management in a number of disease states and adversely impacts survival; how diabetes affects patients with pulmonary hypertension (PH) has not been well characterized. With insulin resistance having recently been demonstrated in PH, we sought to examine the impact of diabetes in these patients. Demographic characteristics, echo data, and invasive hemodynamic data were prospectively collected for 261 patients with PH referred for initial hemodynamic assessment. Diabetes was defined as documented insulin resistance or treatment with antidiabetic medications. Fifty-five patients (21%) had diabetes, and compared with nondiabetic patients, they were older (mean years ± SD, 61 ± 13 vs. 56 ± 16; [Formula: see text]), more likely to be black (29% vs. 14%; [Formula: see text]) and hypertensive (71% vs. 30%; [Formula: see text]), and had higher mean (±SD) serum creatinine levels (1.1 ± 0.5 vs. 1.0 ± 0.4; [Formula: see text]). Diabetic patients had similar World Health Organization functional class at presentation but were more likely to have pulmonary venous etiology of PH (24% vs. 10%; [Formula: see text]). Echo findings, including biventricular function, tricuspid regurgitation, and pressure estimates were similar. Invasive pulmonary pressures and cardiac output were similar, but right atrial pressure was appreciably higher (14 ± 8 mmHg vs. 10 ± 5 mmHg; [Formula: see text]). Despite similar management, survival was markedly worse and remained so after statistical adjustment. In summary, diabetic patients referred for assessment of PH were more likely to have pulmonary venous disease than nondiabetic patients with PH, with hemodynamics suggesting greater right-sided diastolic dysfunction. The markedly worse survival in these patients merits further study.
Collapse
Affiliation(s)
- Abraham D Abernethy
- Department of Internal Medicine/Pediatrics, University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Kathryn Stackhouse
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Stephen Hart
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Ganesh Devendra
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Thomas M Bashore
- Department of Cardiovascular Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Raed Dweik
- Department of Pulmonary Medicine, Cleveland Clinic Respiratory Institute, Cleveland, Ohio, USA
| | - Richard A Krasuski
- Department of Cardiovascular Medicine, Cleveland Clinic Heart and Vascular Institute, Cleveland, Ohio, USA
| |
Collapse
|
41
|
Gopal DM, Santhanakrishnan R, Wang YC, Ayalon N, Donohue C, Rahban Y, Perez AJ, Downing J, Liang CS, Gokce N, Colucci WS, Ho JE. Impaired right ventricular hemodynamics indicate preclinical pulmonary hypertension in patients with metabolic syndrome. J Am Heart Assoc 2015; 4:e001597. [PMID: 25758604 PMCID: PMC4392440 DOI: 10.1161/jaha.114.001597] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Metabolic disease can lead to intrinsic pulmonary hypertension in experimental models. The contributions of metabolic syndrome (MetS) and obesity to pulmonary hypertension and right ventricular dysfunction in humans remain unclear. We investigated the association of MetS and obesity with right ventricular structure and function in patients without cardiovascular disease. Methods and Results A total of 156 patients with MetS (mean age 44 years, 71% women, mean body mass index 40 kg/m2), 45 similarly obese persons without MetS, and 45 nonobese controls underwent echocardiography, including pulsed wave Doppler measurement of pulmonary artery acceleration time (PAAT) and ejection time. Pulmonary artery systolic pressure was estimated from PAAT using validated equations. MetS was associated with lower tricuspid valve e′ (right ventricular diastolic function parameter), shorter PAAT, shorter ejection time, and larger pulmonary artery diameter compared with controls (P<0.05 for all). Estimated pulmonary artery systolic pressure based on PAAT was 42±12 mm Hg in participants with MetS compared with 32±9 and 32±10 mm Hg in obese and nonobese controls (P for ANOVA <0.0001). After adjustment for age, sex, hypertension, diabetes, body mass index, and triglycerides, MetS remained associated with a 20‐ms–shorter PAAT (β=−20.4, SE=6.5, P=0.002 versus obese). This association persisted after accounting for left ventricular structure and function and after exclusion of participants with obstructive sleep apnea. Conclusions MetS is associated with abnormal right ventricular and pulmonary artery hemodynamics, as shown by shorter PAAT and subclinical right ventricular diastolic dysfunction. Estimated pulmonary artery systolic pressures are higher in MetS and preclinical metabolic heart disease and raise the possibility that pulmonary hypertension contributes to the pathophysiology of metabolic heart disease.
Collapse
Affiliation(s)
- Deepa M Gopal
- Cardiology Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (D.M.G.)
| | - Rajalakshmi Santhanakrishnan
- Cardiovascular Medicine Section, Boston University School of Medicine, Boston, MA (R.S., N.A., C.D., A.J.P., J.D., C.L., N.G., W.S.C., J.E.H.)
| | - Yi-Chih Wang
- Cardiovascular Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan (Y.C.W.)
| | - Nir Ayalon
- Cardiovascular Medicine Section, Boston University School of Medicine, Boston, MA (R.S., N.A., C.D., A.J.P., J.D., C.L., N.G., W.S.C., J.E.H.)
| | - Courtney Donohue
- Cardiovascular Medicine Section, Boston University School of Medicine, Boston, MA (R.S., N.A., C.D., A.J.P., J.D., C.L., N.G., W.S.C., J.E.H.)
| | - Youssef Rahban
- Department of Medicine, Boston University School of Medicine, Boston, MA (Y.R.)
| | - Alejandro J Perez
- Cardiovascular Medicine Section, Boston University School of Medicine, Boston, MA (R.S., N.A., C.D., A.J.P., J.D., C.L., N.G., W.S.C., J.E.H.)
| | - Jill Downing
- Cardiovascular Medicine Section, Boston University School of Medicine, Boston, MA (R.S., N.A., C.D., A.J.P., J.D., C.L., N.G., W.S.C., J.E.H.)
| | - Chang-seng Liang
- Cardiovascular Medicine Section, Boston University School of Medicine, Boston, MA (R.S., N.A., C.D., A.J.P., J.D., C.L., N.G., W.S.C., J.E.H.)
| | - Noyan Gokce
- Cardiovascular Medicine Section, Boston University School of Medicine, Boston, MA (R.S., N.A., C.D., A.J.P., J.D., C.L., N.G., W.S.C., J.E.H.)
| | - Wilson S Colucci
- Cardiovascular Medicine Section, Boston University School of Medicine, Boston, MA (R.S., N.A., C.D., A.J.P., J.D., C.L., N.G., W.S.C., J.E.H.)
| | - Jennifer E Ho
- Cardiovascular Medicine Section, Boston University School of Medicine, Boston, MA (R.S., N.A., C.D., A.J.P., J.D., C.L., N.G., W.S.C., J.E.H.)
| |
Collapse
|
42
|
Bertero T, Cottrill K, Krauszman A, Lu Y, Annis S, Hale A, Bhat B, Waxman AB, Chau BN, Kuebler WM, Chan SY. The microRNA-130/301 family controls vasoconstriction in pulmonary hypertension. J Biol Chem 2015; 290:2069-85. [PMID: 25505270 PMCID: PMC4303661 DOI: 10.1074/jbc.m114.617845] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/26/2014] [Indexed: 12/19/2022] Open
Abstract
Pulmonary hypertension (PH) is a complex disorder, spanning several known vascular cell types. Recently, we identified the microRNA-130/301 (miR-130/301) family as a regulator of multiple pro-proliferative pathways in PH, but the true breadth of influence of the miR-130/301 family across cell types in PH may be even more extensive. Here, we employed targeted network theory to identify additional pathogenic pathways regulated by miR-130/301, including those involving vasomotor tone. Guided by these predictions, we demonstrated, via gain- and loss-of-function experimentation in vitro and in vivo, that miR-130/301-specific control of the peroxisome proliferator-activated receptor γ regulates a panel of vasoactive factors communicating between diseased pulmonary vascular endothelial and smooth muscle cells. Of these, the vasoconstrictive factor endothelin-1 serves as an integral point of communication between the miR-130/301-peroxisome proliferator-activated receptor γ axis in endothelial cells and contractile function in smooth muscle cells. Thus, resulting from an in silico analysis of the architecture of the PH disease gene network coupled with molecular experimentation in vivo, these findings clarify the expanded role of the miR-130/301 family in the global regulation of PH. They further emphasize the importance of molecular cross-talk among the diverse cellular populations involved in PH.
Collapse
Affiliation(s)
- Thomas Bertero
- From the Divisions of Cardiovascular and Network Medicine and
| | | | - Adrienn Krauszman
- the Keenan Research Centre for Biomedical Science of St. Michael's, University of Toronto, Toronto, Ontario M5R 0A3, Canada
| | - Yu Lu
- From the Divisions of Cardiovascular and Network Medicine and
| | - Sofia Annis
- From the Divisions of Cardiovascular and Network Medicine and
| | - Andrew Hale
- From the Divisions of Cardiovascular and Network Medicine and
| | | | - Aaron B Waxman
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - B Nelson Chau
- Regulus Therapeutics, San Diego, California 92121, and
| | - Wolfgang M Kuebler
- the Keenan Research Centre for Biomedical Science of St. Michael's, University of Toronto, Toronto, Ontario M5R 0A3, Canada, the Department of Physiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Stephen Y Chan
- From the Divisions of Cardiovascular and Network Medicine and
| |
Collapse
|
43
|
Bertero T, Lu Y, Annis S, Hale A, Bhat B, Saggar R, Saggar R, Wallace WD, Ross DJ, Vargas SO, Graham BB, Kumar R, Black SM, Fratz S, Fineman JR, West JD, Haley KJ, Waxman AB, Chau BN, Cottrill KA, Chan SY. Systems-level regulation of microRNA networks by miR-130/301 promotes pulmonary hypertension. J Clin Invest 2014; 124:3514-28. [PMID: 24960162 PMCID: PMC4109523 DOI: 10.1172/jci74773] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 05/08/2014] [Indexed: 01/16/2023] Open
Abstract
Development of the vascular disease pulmonary hypertension (PH) involves disparate molecular pathways that span multiple cell types. MicroRNAs (miRNAs) may coordinately regulate PH progression, but the integrative functions of miRNAs in this process have been challenging to define with conventional approaches. Here, analysis of the molecular network architecture specific to PH predicted that the miR-130/301 family is a master regulator of cellular proliferation in PH via regulation of subordinate miRNA pathways with unexpected connections to one another. In validation of this model, diseased pulmonary vessels and plasma from mammalian models and human PH subjects exhibited upregulation of miR-130/301 expression. Evaluation of pulmonary arterial endothelial cells and smooth muscle cells revealed that miR-130/301 targeted PPARγ with distinct consequences. In endothelial cells, miR-130/301 modulated apelin-miR-424/503-FGF2 signaling, while in smooth muscle cells, miR-130/301 modulated STAT3-miR-204 signaling to promote PH-associated phenotypes. In murine models, induction of miR-130/301 promoted pathogenic PH-associated effects, while miR-130/301 inhibition prevented PH pathogenesis. Together, these results provide insight into the systems-level regulation of miRNA-disease gene networks in PH with broad implications for miRNA-based therapeutics in this disease. Furthermore, these findings provide critical validation for the evolving application of network theory to the discovery of the miRNA-based origins of PH and other diseases.
Collapse
Affiliation(s)
- Thomas Bertero
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yu Lu
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sofia Annis
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew Hale
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Balkrishen Bhat
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rajan Saggar
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rajeev Saggar
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - W. Dean Wallace
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David J. Ross
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sara O. Vargas
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian B. Graham
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rahul Kumar
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen M. Black
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sohrab Fratz
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey R. Fineman
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James D. West
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kathleen J. Haley
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Aaron B. Waxman
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - B. Nelson Chau
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Katherine A. Cottrill
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen Y. Chan
- Divisions of Cardiovascular Medicine and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. Regulus Therapeutics, San Diego, California, USA. Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA. Department of Medicine, University of Arizona Medical Center, Tuscon, Arizona, USA. Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA. Program in Translational Lung Research, University of Colorado, Denver, Aurora, Colorado, USA. Vascular Biology Center, Pulmonary Disease Program, Georgia Regents University, August, Georgia, USA. Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Munich, Germany. Department of Pediatrics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA. Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Hansmann G. Interdisciplinary networks for the treatment of childhood pulmonary vascular disease: what pulmonary hypertension doctors can learn from pediatric oncologists. Pulm Circ 2014; 3:792-801. [PMID: 25006395 DOI: 10.1086/674766] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 08/22/2013] [Indexed: 01/10/2023] Open
Abstract
The pathobiology of pulmonary arterial hypertension (PAH) is complex and multifactorial. None of the current therapies has been shown to be universally effective or able to reverse advanced pulmonary vascular disease, characterized by plexiform vascular lesions, or to prevent right ventricular failure in advanced PAH. It is thus unlikely that only one factor, pathway, or gene mutation will explain all forms and cases. Pediatric oncologists recognized a need for intensified, collaborative research within their field more than 40 years ago and implemented major clinical and translational networks worldwide to achieve evidence-based "tailored therapies." The similarities in the pathobiology (e.g., increased proliferation and resistance to apoptosis in vascular cells and perivascular inflammation) and the uncertainties in the proper treatment of both cancer and pulmonary hypertension (PH) have led to the idea of building interdisciplinary networks among PH centers to achieve rapid translation of basic research findings, optimal diagnostic algorithms, and significant, sustained treatment results. Such networks leading to patient registries, clinical trials, drug development, and innovative, effective therapies are urgently needed for the care of children with PH. This article reviews the current status, limitations, and recent developments in the field of pediatric PH. It is suggested that the oncologists' exemplary networks, concepts, and results in the treatment of acute lymphoblastic leukemia are applicable to future networks and innovative therapies for pediatric pulmonary hypertensive vascular disease and right ventricular dysfunction.
Collapse
Affiliation(s)
- Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
| |
Collapse
|
45
|
Affiliation(s)
- Roxane Paulin
- From the Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
46
|
Li H, Lu W, Cai WW, Wang PJ, Zhang N, Yu CP, Wang DL, Liu BC, Sun W. Telmisartan attenuates monocrotaline-induced pulmonary artery endothelial dysfunction through a PPAR gamma-dependent PI3K/Akt/eNOS pathway. Pulm Pharmacol Ther 2014; 28:17-24. [DOI: 10.1016/j.pupt.2013.11.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 11/04/2013] [Accepted: 11/11/2013] [Indexed: 01/11/2023]
|
47
|
Abstract
Pulmonary arterial hypertension (PAH) is a vascular remodeling disease of the lungs resulting in heart failure and premature death. Although, until recently, it was thought that PAH pathology is restricted to pulmonary arteries, several extrapulmonary organs are also affected. The realization that these tissues share a common metabolic abnormality (i.e., suppression of mitochondrial glucose oxidation and increased glycolysis) is important for our understanding of PAH, if not a paradigm shift. Here, we discuss an emerging metabolic theory, which proposes that PAH should be viewed as a syndrome involving many organs sharing a mitochondrial abnormality and explains many PAH features and provides novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Gopinath Sutendra
- Department of Medicine, University of Alberta, 2C2 Walter Mackenzie Centre, 8440 112 Street Northwest, Edmonton, AB T6G 2P4, Canada
| | - Evangelos D Michelakis
- Department of Medicine, University of Alberta, 2C2 Walter Mackenzie Centre, 8440 112 Street Northwest, Edmonton, AB T6G 2P4, Canada.
| |
Collapse
|
48
|
Rosiglitazone Attenuated Endothelin-1-Induced Vasoconstriction of Pulmonary Arteries in the Rat Model of Pulmonary Arterial Hypertension via Differential Regulation of ET-1 Receptors. PPAR Res 2014; 2014:374075. [PMID: 24701204 PMCID: PMC3950948 DOI: 10.1155/2014/374075] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 01/03/2014] [Indexed: 01/10/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease characterized by a progressive increase in pulmonary arterial pressure leading to right ventricular failure and death. Activation of the endothelin (ET)-1 system has been demonstrated in plasma and lung tissue of PAH patients as well as in animal models of PAH. Recently, peroxisome proliferator-activated receptor γ (PPARγ) agonists have been shown to ameliorate PAH. The present study aimed to investigate the mechanism for the antivasoconstrictive effects of rosiglitazone in response to ET-1 in PAH. Sprague-Dawley rats were exposed to chronic hypoxia (10% oxygen) for 3 weeks. Pulmonary arteries from PAH rats showed an enhanced vasoconstriction in response to ET-1. Treatment with PPARγ agonist rosiglitazone (20 mg/kg per day) with oral gavage for 3 days attenuated the vasocontractive effect of ET-1. The effect of rosiglitazone was lost in the presence of L-NAME, indicating a nitric oxide-dependent mechanism. Western blotting revealed that rosiglitazone increased ETBR but decreased ETAR level in pulmonary arteries from PAH rats. ETBR antagonist A192621 diminished the effect of rosiglitazone on ET-1-induced contraction. These results demonstrated that rosiglitazone attenuated ET-1-induced pulmonary vasoconstriction in PAH through differential regulation of the subtypes of ET-1 receptors and, thus, provided a new mechanism for the therapeutic use of PPARγ agonists in PAH.
Collapse
|
49
|
Abstract
The clinical recognition of pulmonary arterial hypertension (PAH) is increasing, and with recent therapeutic advances, short-term survival has improved. In spite of these advances, however, PAH remains a disease with substantial morbidity and long-term mortality. The pathogenesis of PAH involves a complex interaction of local and distant cytokines, growth factors, co-factors, and transcription factors occurring in the right genetic and environmental setting. These factors ultimately lead to the detrimental changes in vascular anatomy and function seen in PAH patients. An important association between obesity/insulin resistance and PAH has recently been identified. Both conditions occur in the presence of a chronic low-grade inflammatory state, and although it is unlikely that a single pathway is solely responsible for the observed association, deficiencies in adiponectin, apolipoprotein E (ApoE) and peroxisome proliferator-activator receptor gamma (PPAR-γ) activity likely play a prominent role. Although incompletely understood, it is clear that further investigation is warranted and the role of weight loss and improved glycemic control in the treatment of at-risk patients with PAH and obesity should be determined.
Collapse
Affiliation(s)
- Elisa A Bradley
- Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center and Nationwide Children's Hospital, Columbus, OH, USA
| | - David Bradley
- Division of Endocrinology, Diabetes and Metabolism, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
50
|
Fessel JP, Flynn CR, Robinson LJ, Penner NL, Gladson S, Kang CJ, Wasserman DH, Hemnes AR, West JD. Hyperoxia synergizes with mutant bone morphogenic protein receptor 2 to cause metabolic stress, oxidant injury, and pulmonary hypertension. Am J Respir Cell Mol Biol 2013; 49:778-87. [PMID: 23742019 DOI: 10.1165/rcmb.2012-0463oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) has been associated with a number of different but interrelated pathogenic mechanisms. Metabolic and oxidative stresses have been shown to play important pathogenic roles in a variety of model systems. However, many of these relationships remain at the level of association. We sought to establish a direct role for metabolic stress and oxidant injury in the pathogenesis of PAH. Mice that universally express a disease-causing mutation in bone morphogenic protein receptor 2 (Bmpr2) were exposed to room air or to brief daily hyperoxia (95% oxygen for 3 h) for 6 weeks, and were compared with wild-type animals undergoing identical exposures. In both murine tissues and cultured endothelial cells, the expression of mutant Bmpr2 was sufficient to cause oxidant injury that was particularly pronounced in mitochondrial membranes. With the enhancement of mitochondrial generation of reactive oxygen species by hyperoxia, oxidant injury was substantially enhanced in mitochondrial membranes, even in tissues distant from the lung. Hyperoxia, despite its vasodilatory actions in the pulmonary circulation, significantly worsened the PAH phenotype (elevated right ventricular systolic pressure, decreased cardiac output, and increased pulmonary vascular occlusion) in Bmpr2 mutant animals. These experiments demonstrate that oxidant injury and metabolic stress contribute directly to disease development, and provide further evidence for PAH as a systemic disease with life-limiting cardiopulmonary manifestations.
Collapse
Affiliation(s)
- Joshua P Fessel
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|