1
|
Zhao P, Li S, He Z, Ma X. Physiological and Genetic Basis of High-Altitude Indigenous Animals' Adaptation to Hypoxic Environments. Animals (Basel) 2024; 14:3031. [PMID: 39457960 PMCID: PMC11505238 DOI: 10.3390/ani14203031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Adaptation is one of the fundamental characteristics of life activities; humans and animals inhabiting high altitudes are well adapted to hypobaric hypoxic environments, and studies on the mechanisms of this adaptation emerged a hundred years ago. Based on these studies, this paper reviews the adaptive changes in hypoxia-sensitive tissues and organs, as well as at the molecular genetic level, such as pulmonary, cardiovascular, O2-consuming tissues, and the hemoglobin and HIF pathway, that occur in animals in response to the challenge of hypobaric hypoxia. High-altitude hypoxia adaptation may be due to the coordinated action of genetic variants in multiple genes and, as a result, adaptive changes in multiple tissues and organs at the physiological and biochemical levels. Unraveling their mechanisms of action can provide a reference for the prevention and treatment of multiple diseases caused by chronic hypoxia.
Collapse
Affiliation(s)
- Pengfei Zhao
- Faculty of Chemistry and Life Sciences, Gansu Minzu Normal University, Hezuo 747000, China;
| | - Shaobin Li
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China;
| | - Zhaohua He
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China;
| | - Xiong Ma
- Faculty of Chemistry and Life Sciences, Gansu Minzu Normal University, Hezuo 747000, China;
| |
Collapse
|
2
|
Imano H, Hayashi T, Nomura A, Tanaka S, Kohda Y, Yamaguchi T, Izumi Y, Yoshiyama M, Hirose Y, Ohta-Ogo K, Ishibashi-Ueda H, Kato R. Suppressing the expression of steroidogenic acute regulatory protein (StAR) in the myocardium by spironolactone contributes to the improvement of right ventricular remodeling in pulmonary arterial hypertension. Hypertens Res 2024:10.1038/s41440-024-01908-z. [PMID: 39367269 DOI: 10.1038/s41440-024-01908-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/06/2024] [Accepted: 08/24/2024] [Indexed: 10/06/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive condition that frequently leads to right ventricular (RV) remodeling. Aldosterone promotes vascular and RV remodeling. The upregulation of steroidogenic acute regulatory protein (StAR) stimulates aldosterone synthesis. However, the expression of StAR in the myocardium under PAH conditions remains unknown. To investigate the expression of StAR in the myocardium and its association with RV remodeling in PAH, utilizing spironolactone as a treatment. A PAH model was created using male Sprague-Dawley rats, which received a subcutaneous injection of Sugen5416 (20 mg/kg) and were exposed to hypoxia (10% O2) for 2 weeks, followed by 2 weeks of normoxia. The animals were then divided into two groups, with one group receiving spironolactone (25 mg/kg/day) for an additional 4 weeks, while the other group did not. H9c2 cells were cultured under hypoxic conditions (37 °C, 1% O2, 5% CO2) with or without spironolactone treatment. In the model rats, RV systolic pressure and the Fulton index, both of which increased upon exposure to Sugen5416 and hypoxia, significantly decreased with spironolactone treatment. In H9c2 cells, hypoxic exposure elevated aldosterone levels, while spironolactone treatment significantly suppressed aldosterone production. Suppression of StAR expression in the myocardium via spironolactone contributes to the improvement of RV remodeling in PAH. Spironolactone may offer a valuable therapeutic strategy for RV remodeling in patients with PAH.
Collapse
Affiliation(s)
- Hideki Imano
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Tetsuya Hayashi
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Atsuo Nomura
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyoto, Japan
| | - Saori Tanaka
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Yuka Kohda
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Takehiro Yamaguchi
- Department of Pharmacology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yasukatsu Izumi
- Department of Pharmacology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Minoru Yoshiyama
- Department of Cardiovascular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yoshinobu Hirose
- Department of Pathology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Keiko Ohta-Ogo
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Hatsue Ishibashi-Ueda
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
- Department of Diagnostic Pathology, Hokusetsu General Hospital, Osaka, Japan
| | - Ryuji Kato
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka, Japan.
| |
Collapse
|
3
|
Durrer I, Ackermann D, Klossner R, Grössl M, Vögel C, Du Toit T, Vogt B, Jamin H, Mohaupt MG, Gennari-Moser C. No extra-adrenal aldosterone production in various human cell lines. J Mol Endocrinol 2024; 72:e230100. [PMID: 38175924 PMCID: PMC10895282 DOI: 10.1530/jme-23-0100] [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: 08/04/2023] [Accepted: 01/04/2024] [Indexed: 01/06/2024]
Abstract
Extra-adrenal de novo aldosterone (Aldo) production has been described inconsistently. Systematic data based upon state-of-the-art technology including validated controls are sparse. We hypothesized that aldosterone synthase (CYP11B2) expression and de novo Aldo production are absent in nonadrenal human cell lines, either immortalized cell lines or commercially available primary cell lines, including peripheral blood mononuclear cells (PBMCs) of individuals without and with primary hyperaldosteronism (PA). CYP11B2-transfected COS-7 and endogenous CYP11B2 expressing adrenal H295R cells served as positive controls. Various well-characterized, purchased, immortalized (BeWo, HEK293, HTR-8/SVneo, JEG-3) and primary (HAEC, HLEC, HRGEC, HRMC, HUAEC, HUVEC, PBMC) cell lines as well as self-isolated PBMCs from PA patients (n = 5) were incubated with the steroid hormone substrates progesterone, deoxycorticosterone, corticosterone or 18-OH-corticosterone with and without Ang II for 24 h to assess CYP11B2 enzymatic activity. CYP11B2 expression was analyzed by real-time PCR and liquid chromatography-mass spectrometry was used to quantify Aldo production. Pronounced CYP11B2 mRNA expression and Aldo production were observed in both positive controls, which followed an incremental time course. Neither substrates alone nor coincubation with Ang II significantly stimulated CYP11B2 expression or Aldo production in various immortalized and primary cell lines and PBMCs of PA patients. These results strongly support the absence of relevant de novo extra-adrenal Aldo production in nonadrenal cells, including blood mononuclear cells, irrespective of the absence or presence of autonomous adrenal Aldo production.
Collapse
Affiliation(s)
- Isabelle Durrer
- Department of Nephrology and Hypertension University of Bern, Berne, Switzerland
| | - Daniel Ackermann
- Department of Nephrology and Hypertension University of Bern, Berne, Switzerland
| | - Rahel Klossner
- Department of Nephrology and Hypertension University of Bern, Berne, Switzerland
- Department of Internal Medicine, Sonnenhof, Lindenhofgruppe, Berne, Switzerland
| | - Michael Grössl
- Department of Nephrology and Hypertension University of Bern, Berne, Switzerland
| | - Clarissa Vögel
- Department of Nephrology and Hypertension University of Bern, Berne, Switzerland
| | - Therina Du Toit
- Department for BioMedical Research University of Bern, Berne, Switzerland
| | - Bruno Vogt
- Department of Nephrology and Hypertension University of Bern, Berne, Switzerland
| | - Heidi Jamin
- Department of Nephrology and Hypertension University of Bern, Berne, Switzerland
- Department for BioMedical Research University of Bern, Berne, Switzerland
| | - Markus G Mohaupt
- Department of Internal Medicine, Sonnenhof, Lindenhofgruppe, Berne, Switzerland
- Department for BioMedical Research University of Bern, Berne, Switzerland
| | - Carine Gennari-Moser
- Department of Nephrology and Hypertension University of Bern, Berne, Switzerland
- Department for BioMedical Research University of Bern, Berne, Switzerland
| |
Collapse
|
4
|
Hua R, Gao H, He C, Xin S, Wang B, Zhang S, Gao L, Tao Q, Wu W, Sun F, Xu J. An emerging view on vascular fibrosis molecular mediators and relevant disorders: from bench to bed. Front Cardiovasc Med 2023; 10:1273502. [PMID: 38179503 PMCID: PMC10764515 DOI: 10.3389/fcvm.2023.1273502] [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: 08/06/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Vascular fibrosis is a widespread pathologic condition that arises during vascular remodeling in cardiovascular dysfunctions. According to previous studies, vascular fibrosis is characterized by endothelial matrix deposition and vascular wall thickening. The RAAS and TGF-β/Smad signaling pathways have been frequently highlighted. It is, however, far from explicit in terms of understanding the cause and progression of vascular fibrosis. In this review, we collected and categorized a large number of molecules which influence the fibrosing process, in order to acquire a better understanding of vascular fibrosis, particularly of pathologic dysfunction. Furthermore, several mediators that prevent vascular fibrosis are discussed in depth in this review, with the aim that this will contribute to the future prevention and treatment of related conditions.
Collapse
Affiliation(s)
- Rongxuan Hua
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Han Gao
- Department of Clinical Laboratory, Aerospace Center Hospital, Peking University, Beijing, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Boya Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Sitian Zhang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lei Gao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Qiang Tao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Wenqi Wu
- Experimental Center for Morphological Research Platform, Capital Medical University, Beijing, China
| | - Fangling Sun
- Department of Experimental Animal Laboratory, Xuan-Wu Hospital of Capital Medical University, Beijing, China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Elhage Hassan M, Vinales J, Perkins S, Sandesara P, Aggarwal V, Jaber WA. Pathogenesis, Diagnosis, and Management of Chronic Thromboembolic Pulmonary Hypertension. Interv Cardiol Clin 2023; 12:e37-e49. [PMID: 38964822 DOI: 10.1016/j.iccl.2024.04.003] [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] [Indexed: 07/06/2024]
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is thought to occur as a sequelae of thromboembolic processes in the pulmonary vasculature. The pathophysiology of CTEPH is multifactorial, including impaired fibrinolysis, endothelial dysregulation, and hypoxic adaptations. The diagnosis of CTEPH is typically delayed considering the nonspecific nature of the symptoms, lack of screening, and relatively low incidence. Diagnostic tools include ventilation-perfusion testing, echocardiography, cardiac catheterization, and pulmonary angiography. The only potentially curative treatment for CTEPH is pulmonary endarterectomy However, approximately 40% of patients are inoperable. Currently, only Riociguat is Food and Drug Administration approved specifically for CTEPH, with additional drug trials underway.
Collapse
Affiliation(s)
- Malika Elhage Hassan
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road Northeast Suite F606, Atlanta, GA 30322, USA
| | - Jorge Vinales
- Department of Medicine, University of Michigan Medical School, 1301 Catherine Street, Ann Arbor, MI 48109, USA
| | - Sidney Perkins
- Department of Internal Medicine, University of Michigan Medical School, 1500 E Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Pratik Sandesara
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road Northeast Suite F606, Atlanta, GA 30322, USA
| | - Vikas Aggarwal
- Department of Cardiology, Henry Ford Medical Center, 2799 W Grand Blvd, K-2 Cath Admin Suite, Detroit, MI 48206, USA
| | - Wissam A Jaber
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road Northeast Suite F606, Atlanta, GA 30322, USA.
| |
Collapse
|
6
|
Crompton M, Skinner LJ, Satchell SC, Butler MJ. Aldosterone: Essential for Life but Damaging to the Vascular Endothelium. Biomolecules 2023; 13:1004. [PMID: 37371584 PMCID: PMC10296074 DOI: 10.3390/biom13061004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
The renin angiotensin aldosterone system is a key regulator of blood pressure. Aldosterone is the final effector of this pathway, acting predominantly via mineralocorticoid receptors. Aldosterone facilitates the conservation of sodium and, with it, water and acts as a powerful stimulus for potassium excretion. However, evidence for the pathological impact of excess mineralocorticoid receptor stimulation is increasing. Here, we discussed how in the heart, hyperaldosteronism is associated with fibrosis, cardiac dysfunction, and maladaptive hypertrophy. In the kidney, aldosterone was shown to cause proteinuria and fibrosis and may contribute to the progression of kidney disease. More recently, studies suggested that aldosterone excess damaged endothelial cells. Here, we reviewed how damage to the endothelial glycocalyx may contribute to this process. The endothelial glycocalyx is a heterogenous, negatively charged layer on the luminal surface of cells. Aldosterone exposure alters this layer. The resulting structural changes reduced endothelial reactivity in response to protective shear stress, altered permeability, and increased immune cell trafficking. Finally, we reviewed current therapeutic strategies for limiting endothelial damage and suggested that preventing glycocalyx remodelling in response to aldosterone exposure may provide a novel strategy, free from the serious adverse effect of hyperkalaemia seen in response to mineralocorticoid blockade.
Collapse
Affiliation(s)
| | | | | | - Matthew J. Butler
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| |
Collapse
|
7
|
Wertheim BM, Wang RS, Guillermier C, Hütter CV, Oldham WM, Menche J, Steinhauser ML, Maron BA. Proline and glucose metabolic reprogramming supports vascular endothelial and medial biomass in pulmonary arterial hypertension. JCI Insight 2023; 8:163932. [PMID: 36626231 PMCID: PMC9977503 DOI: 10.1172/jci.insight.163932] [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: 08/01/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
In pulmonary arterial hypertension (PAH), inflammation promotes a fibroproliferative pulmonary vasculopathy. Reductionist studies emphasizing single biochemical reactions suggest a shift toward glycolytic metabolism in PAH; however, key questions remain regarding the metabolic profile of specific cell types within PAH vascular lesions in vivo. We used RNA-Seq to profile the transcriptome of pulmonary artery endothelial cells (PAECs) freshly isolated from an inflammatory vascular injury model of PAH ex vivo, and these data were integrated with information from human gene ontology pathways. Network medicine was then used to map all aa and glucose pathways to the consolidated human interactome, which includes data on 233,957 physical protein-protein interactions. Glucose and proline pathways were significantly close to the human PAH disease module, suggesting that these pathways are functionally relevant to PAH pathobiology. To test this observation in vivo, we used multi-isotope imaging mass spectrometry to map and quantify utilization of glucose and proline in the PAH pulmonary vasculature at subcellular resolution. Our findings suggest that elevated glucose and proline avidity underlie increased biomass in PAECs and the media of fibrosed PAH pulmonary arterioles. Overall, these data show that anabolic utilization of glucose and proline are fundamental to the vascular pathology of PAH.
Collapse
Affiliation(s)
| | - Rui-Sheng Wang
- Division of Cardiovascular Medicine, Department of Medicine.,Channing Division of Network Medicine, Department of Medicine; and
| | - Christelle Guillermier
- Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Center for NanoImaging, Cambridge, Massachusetts, USA
| | - Christiane Vr Hütter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Vienna, Austria.,Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and the Medical University of Vienna, Vienna, Austria
| | - William M Oldham
- Division of Pulmonary and Critical Medicine, Department of Medicine
| | - Jörg Menche
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Vienna, Austria.,Faculty of Mathematics, University of Vienna, Vienna, Austria
| | - Matthew L Steinhauser
- Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Center for NanoImaging, Cambridge, Massachusetts, USA.,Division of Cardiovascular Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
8
|
Mamazhakypov A, Lother A. Therapeutic targeting of mineralocorticoid receptors in pulmonary hypertension: Insights from basic research. Front Cardiovasc Med 2023; 10:1118516. [PMID: 36793473 PMCID: PMC9922727 DOI: 10.3389/fcvm.2023.1118516] [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: 12/07/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
Pulmonary hypertension (PH) is characterized by pulmonary vascular remodeling and associated with adverse outcomes. In patients with PH, plasma aldosterone levels are elevated, suggesting that aldosterone and its receptor, the mineralocorticoid receptor (MR), play an important role in the pathophysiology of PH. The MR plays a crucial role in adverse cardiac remodeling in left heart failure. A series of experimental studies from the past few years indicate that MR activation promotes adverse cellular processes that lead to pulmonary vascular remodeling, including endothelial cell apoptosis, smooth muscle cell (SMC) proliferation, pulmonary vascular fibrosis, and inflammation. Accordingly, in vivo studies have demonstrated that pharmacological inhibition or cell-specific deletion of the MR can prevent disease progression and partially reverse established PH phenotypes. In this review, we summarize recent advances in MR signaling in pulmonary vascular remodeling based on preclinical research and discuss the potential, but also the challenges, in bringing MR antagonists (MRAs) into clinical application.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany,Faculty of Medicine, Interdisciplinary Medical Intensive Care, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany,*Correspondence: Achim Lother,
| |
Collapse
|
9
|
Wolter NL, Jaffe IZ. Emerging vascular cell-specific roles for mineralocorticoid receptor: implications for understanding sex differences in cardiovascular disease. Am J Physiol Cell Physiol 2023; 324:C193-C204. [PMID: 36440858 PMCID: PMC9902217 DOI: 10.1152/ajpcell.00372.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/04/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022]
Abstract
As growing evidence implicates extrarenal mineralocorticoid receptor (MR) in cardiovascular disease (CVD), recent studies have defined both cell- and sex-specific roles. MR is expressed in vascular smooth muscle (SMC) and endothelial cells (ECs). This review integrates published data from the past 5 years to identify novel roles for vascular MR in CVD, with a focus on understanding sex differences. Four areas are reviewed in which there is recently expanded understanding of the cell type- or sex-specific role of MR in 1) obesity-induced microvascular endothelial dysfunction, 2) vascular inflammation in atherosclerosis, 3) pulmonary hypertension, and 4) chronic kidney disease (CKD)-related CVD. The review focuses on preclinical data on each topic describing new mechanistic paradigms, cell type-specific mechanisms, sexual dimorphism if addressed, and clinical implications are then considered. New data support that MR drives vascular dysfunction induced by cardiovascular risk factors via sexually dimorphic mechanisms. In females, EC-MR contributes to obesity-induced endothelial dysfunction by regulating epithelial sodium channel expression and by inhibiting estrogen-induced nitric oxide production. In males with hyperlipidemia, EC-MR promotes large vessel inflammation by genomic regulation of leukocyte adhesion molecules, which is inhibited by the estrogen receptor. In pulmonary hypertension models, MRs in EC and SMC contribute to distinct components of disease pathologies including pulmonary vessel remodeling and RV dysfunction. Despite a female predominance in pulmonary hypertension, sex-specific roles for MR have not been explored. Vascular MR has also been directly implicated in CKD-related vascular dysfunction, independent of blood pressure. Despite these advances, sex differences in MR function remain understudied.
Collapse
Affiliation(s)
- Nicole L Wolter
- Molecular Cardiology Research Institute, https://ror.org/002hsbm82Tufts Medical Center, Boston, Massachusetts
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, https://ror.org/002hsbm82Tufts Medical Center, Boston, Massachusetts
| |
Collapse
|
10
|
Physiology and Proteomic Basis of Lung Adaptation to High-Altitude Hypoxia in Tibetan Sheep. Animals (Basel) 2022; 12:ani12162134. [PMID: 36009723 PMCID: PMC9405401 DOI: 10.3390/ani12162134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary As an indigenous animal living in the Tibetan plateau, the Tibetan sheep is well adapted to high-altitude hypoxia, and the lungs play an important role in overcoming the hypoxic environment. To reveal the physiological and proteomic basis of Tibetan sheep lungs during their adaptation to hypoxia, we studied the lungs of Tibetan sheep at different altitudes using light and electron microscopy and proteome sequencing. The results showed that in the lungs of Tibetan sheep occurred a series of physiological changes with increasing altitude, and some important proteins and pathways identified by proteome sequencing further support these physiology findings. These changes at the physiological and molecular levels may facilitate the adaptation of Tibetan sheep to high-altitude hypoxia. In conclusion, these findings may provide a reference for the prevention of altitude sickness in humans. Abstract The Tibetan sheep is an indigenous animal of the Tibetan plateau, and after a long period of adaptation have adapted to high-altitude hypoxia. Many physiological changes occur in Tibetan sheep as they adapt to high-altitude hypoxia, especially in the lungs. To reveal the physiological changes and their molecular mechanisms in the lungs of Tibetan sheep during adaptation to high altitudes, we selected Tibetan sheep from three altitudes (2500 m, 3500 m, and 4500 m) and measured blood-gas indicators, observed lung structures, and compared lung proteome changes. The results showed that the Tibetan sheep increased their O2-carrying capacity by increasing the hemoglobin (Hb) concentration and Hematocrit (Hct) at an altitude of 3500 m. While at altitude of 4500 m, Tibetan sheep decreased their Hb concentration and Hct to avoid pulmonary hypertension and increased the efficiency of air-blood exchange and O2 transfer by increasing the surface area of gas exchange and half-saturation oxygen partial pressure. Besides these, some important proteins and pathways related to gas transport, oxidative stress, and angiogenesis identified by proteome sequencing further support these physiology findings, including HBB, PRDX2, GPX1, GSTA1, COL14A1, and LTBP4, etc. In conclusion, the lungs of Tibetan sheep are adapted to different altitudes by different strategies; these findings are valuable for understanding the basis of hypoxic adaptation in Tibetan sheep.
Collapse
|
11
|
Manna PR, Ahmed AU, Molehin D, Narasimhan M, Pruitt K, Reddy PH. Hormonal and Genetic Regulatory Events in Breast Cancer and Its Therapeutics: Importance of the Steroidogenic Acute Regulatory Protein. Biomedicines 2022; 10:biomedicines10061313. [PMID: 35740335 PMCID: PMC9220045 DOI: 10.3390/biomedicines10061313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023] Open
Abstract
Estrogen promotes the development and survival of the majority of breast cancers (BCs). Aromatase is the rate-limiting enzyme in estrogen biosynthesis, and it is immensely expressed in both cancerous and non-cancerous breast tissues. Endocrine therapy based on estrogen blockade, by aromatase inhibitors, has been the mainstay of BC treatment in post-menopausal women; however, resistance to hormone therapy is the leading cause of cancer death. An improved understanding of the molecular underpinnings is the key to develop therapeutic strategies for countering the most prevalent hormone receptor positive BCs. Of note, cholesterol is the precursor of all steroid hormones that are synthesized in a variety of tissues and play crucial roles in diverse processes, ranging from organogenesis to homeostasis to carcinogenesis. The rate-limiting step in steroid biosynthesis is the transport of cholesterol from the outer to the inner mitochondrial membrane, a process that is primarily mediated by the steroidogenic acute regulatory (StAR) protein. Advances in genomic and proteomic technologies have revealed a dynamic link between histone deacetylases (HDACs) and StAR, aromatase, and estrogen regulation. We were the first to report that StAR is abundantly expressed, along with large amounts of 17β-estradiol (E2), in hormone-dependent, but not hormone-independent, BCs, in which StAR was also identified as a novel acetylated protein. Our in-silico analyses of The Cancer Genome Atlas (TCGA) datasets, for StAR and steroidogenic enzyme genes, revealed an inverse correlation between the amplification of the StAR gene and the poor survival of BC patients. Additionally, we reported that a number of HDAC inhibitors, by altering StAR acetylation patterns, repress E2 synthesis in hormone-sensitive BC cells. This review highlights the current understanding of molecular pathogenesis of BCs, especially for luminal subtypes, and their therapeutics, underlining that StAR could serve not only as a prognostic marker, but also as a therapeutic candidate, in the prevention and treatment of this life-threatening disease.
Collapse
Affiliation(s)
- Pulak R. Manna
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
- Correspondence: ; Tel.: +1-806-743-3573; Fax: +1-806-743-3143
| | - Ahsen U. Ahmed
- Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA;
| | - Deborah Molehin
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (D.M.); (K.P.)
| | - Madhusudhanan Narasimhan
- Neuroscience and Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Kevin Pruitt
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (D.M.); (K.P.)
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
- Neuroscience and Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
- Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
12
|
Behrendt T, Bielitzki R, Behrens M, Herold F, Schega L. Effects of Intermittent Hypoxia-Hyperoxia on Performance- and Health-Related Outcomes in Humans: A Systematic Review. SPORTS MEDICINE - OPEN 2022; 8:70. [PMID: 35639211 PMCID: PMC9156652 DOI: 10.1186/s40798-022-00450-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/17/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Intermittent hypoxia applied at rest or in combination with exercise promotes multiple beneficial adaptations with regard to performance and health in humans. It was hypothesized that replacing normoxia by moderate hyperoxia can increase the adaptive response to the intermittent hypoxic stimulus. OBJECTIVE Our objective was to systematically review the current state of the literature on the effects of chronic intermittent hypoxia-hyperoxia (IHH) on performance- and health-related outcomes in humans. METHODS PubMed, Web of Science™, Scopus, and Cochrane Library databases were searched in accordance with PRISMA guidelines (January 2000 to September 2021) using the following inclusion criteria: (1) original research articles involving humans, (2) investigation of the chronic effect of IHH, (3) inclusion of a control group being not exposed to IHH, and (4) articles published in peer-reviewed journals written in English. RESULTS Of 1085 articles initially found, eight studies were included. IHH was solely performed at rest in different populations including geriatric patients (n = 1), older patients with cardiovascular (n = 3) and metabolic disease (n = 2) or cognitive impairment (n = 1), and young athletes with overtraining syndrome (n = 1). The included studies confirmed the beneficial effects of chronic exposure to IHH, showing improvements in exercise tolerance, peak oxygen uptake, and global cognitive functions, as well as lowered blood glucose levels. A trend was discernible that chronic exposure to IHH can trigger a reduction in systolic and diastolic blood pressure. The evidence of whether IHH exerts beneficial effects on blood lipid levels and haematological parameters is currently inconclusive. A meta-analysis was not possible because the reviewed studies had a considerable heterogeneity concerning the investigated populations and outcome parameters. CONCLUSION Based on the published literature, it can be suggested that chronic exposure to IHH might be a promising non-pharmacological intervention strategy for improving peak oxygen consumption, exercise tolerance, and cognitive performance as well as reducing blood glucose levels, and systolic and diastolic blood pressure in older patients with cardiovascular and metabolic diseases or cognitive impairment. However, further randomized controlled trials with adequate sample sizes are needed to confirm and extend the evidence. This systematic review was registered on the international prospective register of systematic reviews (PROSPERO-ID: CRD42021281248) ( https://www.crd.york.ac.uk/prospero/ ).
Collapse
Affiliation(s)
- Tom Behrendt
- Department of Sport Science, Chair for Health and Physical Activity, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39104 Magdeburg, Germany
| | - Robert Bielitzki
- Department of Sport Science, Chair for Health and Physical Activity, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39104 Magdeburg, Germany
| | - Martin Behrens
- Department of Sport Science, Chair for Health and Physical Activity, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39104 Magdeburg, Germany
- Department of Orthopaedics, Rostock University Medical Center, Doberaner Str. 142, 18057 Rostock, Germany
| | - Fabian Herold
- Research Group Degenerative and Chronic Disease, Movement, Faculty of Health Sciences, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany
| | - Lutz Schega
- Department of Sport Science, Chair for Health and Physical Activity, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39104 Magdeburg, Germany
| |
Collapse
|
13
|
Zhao P, Zhao F, Hu J, Wang J, Liu X, Zhao Z, Xi Q, Sun H, Li S, Luo Y. Physiology and Transcriptomics Analysis Reveal the Contribution of Lungs on High-Altitude Hypoxia Adaptation in Tibetan Sheep. Front Physiol 2022; 13:885444. [PMID: 35634140 PMCID: PMC9133604 DOI: 10.3389/fphys.2022.885444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/12/2022] [Indexed: 01/10/2023] Open
Abstract
The Tibetan sheep is an indigenous species on the Tibetan plateau with excellent adaptability to high-altitude hypoxia and is distributed at altitudes of 2500–5000 m. The high-altitude hypoxia adaptation of Tibetan sheep requires adaptive reshaping of multiple tissues and organs, especially the lungs. To reveal the mechanisms of adaptation at the tissue and molecular levels in the lungs of Tibetan sheep under hypoxic conditions at different altitudes, we performed light and electron microscopic observations, transcriptomic sequencing, and enzyme-linked immunosorbent assay studies on the lungs of Tibetan sheep from three altitudes (2500, 3500, and 4500 m). The results showed that in addition to continuous increase in pulmonary artery volume, thickness, and elastic fiber content with altitude, Tibetan sheep increase the hemoglobin concentration at an altitude of 3500 m, while they decrease the Hb concentration and increase the surface area of gas exchange and capacity of the blood at an altitude of 4500 m. Other than that, some important differentially expressed genes related to angiogenesis (FNDC1, HPSE, and E2F8), vasomotion and fibrogenesis (GJA4, FAP, COL1A1, COL1A2, COL3A1, and COL14A1), and gas transport (HBB, HBA1, APOLD1, and CHL1) were also identified; these discoveries at the molecular level explain to some extent the physiological findings. In conclusion, the lungs of Tibetan sheep adopt different strategies when adapting to different altitudes, and these findings are valuable for understanding the basis of survival of indigenous species on the Tibetan plateau.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Shaobin Li
- *Correspondence: Shaobin Li, ; Yuzhu Luo,
| | - Yuzhu Luo
- *Correspondence: Shaobin Li, ; Yuzhu Luo,
| |
Collapse
|
14
|
Bauersachs J, Lother A. Mineralocorticoid receptor activation and antagonism in cardiovascular disease: cellular and molecular mechanisms. Kidney Int Suppl (2011) 2022; 12:19-26. [DOI: 10.1016/j.kisu.2021.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 02/08/2023] Open
|
15
|
Lu M, Chen LY, Gairhe S, Mazer AJ, Anderson SA, Nelson JN, Noguchi A, Siddique MAH, Dougherty EJ, Zou Y, Johnston KA, Yu ZX, Wang H, Wang S, Sun J, Solomon SB, Vanderpool RR, Solomon MA, Danner RL, Elinoff JM. Mineralocorticoid receptor antagonist treatment of established pulmonary arterial hypertension improves interventricular dependence in the SU5416-hypoxia rat model. Am J Physiol Lung Cell Mol Physiol 2022; 322:L315-L332. [PMID: 35043674 PMCID: PMC8858673 DOI: 10.1152/ajplung.00238.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Treatment with mineralocorticoid receptor (MR) antagonists beginning at the outset of disease, or early thereafter, prevents pulmonary vascular remodeling in preclinical models of pulmonary arterial hypertension (PAH). However, the efficacy of MR blockade in established disease, a more clinically relevant condition, remains unknown. Therefore, we investigated the effectiveness of two MR antagonists, eplerenone (EPL) and spironolactone (SPL), after the development of severe right ventricular (RV) dysfunction in the rat SU5416-hypoxia (SuHx) PAH model. Cardiac magnetic resonance imaging (MRI) in SuHx rats at the end of week 5, before study treatment, confirmed features of established disease including reduced RV ejection fraction and RV hypertrophy, pronounced septal flattening with impaired left ventricular filling and reduced cardiac index. Five weeks of treatment with either EPL or SPL improved left ventricular filling and prevented the further decline in cardiac index compared with placebo. Interventricular septal displacement was reduced by EPL whereas SPL effects were similar, but not significant. Although MR antagonists did not significantly reduce pulmonary artery pressure or vessel remodeling in SuHx rats with established disease, animals with higher drug levels had lower pulmonary pressures. Consistent with effects on cardiac function, EPL treatment tended to suppress MR and proinflammatory gene induction in the RV. In conclusion, MR antagonist treatment led to modest, but consistent beneficial effects on interventricular dependence after the onset of significant RV dysfunction in the SuHx PAH model. These results suggest that measures of RV structure and/or function may be useful endpoints in clinical trials of MR antagonists in patients with PAH.
Collapse
Affiliation(s)
- Mengyun Lu
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Li-Yuan Chen
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Salina Gairhe
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Adrien J. Mazer
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Stasia A. Anderson
- 2Animal MRI Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Jasmine N.H. Nelson
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Audrey Noguchi
- 3Murine Phenotyping Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Edward J. Dougherty
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Yvette Zou
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Kathryn A. Johnston
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Zu-Xi Yu
- 4Pathology Core Facility, National Heart, Lung, and Blood
Institute, National Institutes of Health, Bethesda, Maryland
| | - Honghui Wang
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Shuibang Wang
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Junfeng Sun
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Steven B. Solomon
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Rebecca R. Vanderpool
- 6Department of Medicine and Biomedical Engineering, University of Arizona College of Medicine, Tucson, Arizona
| | - Michael A. Solomon
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland,5Cardiology Branch, National Heart, Lung, and Blood
Institute, National Institutes of Health, Bethesda, Maryland
| | - Robert L. Danner
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Jason M. Elinoff
- 1Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
16
|
Xue HM, Sun WT, Chen HX, He GW, Yang Q. Targeting IRE1α-JNK-c-Jun/AP-1-sEH Signaling Pathway Improves Myocardial and Coronary Endothelial Function Following Global Myocardial Ischemia/Reperfusion. Int J Med Sci 2022; 19:1460-1472. [PMID: 36035373 PMCID: PMC9413556 DOI: 10.7150/ijms.74533] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/28/2022] [Indexed: 11/26/2022] Open
Abstract
Objectives: Endoplasmic reticulum (ER) stress and soluble epoxide hydrolase (sEH) upregulation/activation have been implicated in myocardial ischemia/reperfusion (I/R) injury. We previously reported that ER stress mediates angiotensin II-induced sEH upregulation in coronary endothelium, whether and how ER stress regulates sEH expression to affect postischemic cardiac function remain unexplored. This study aimed to unravel the signaling linkage between ER stress and sEH in an ex vivo model of myocardial I/R injury. Methods: Hearts from male Wistar-Kyoto rats were mounted on a Langendorff apparatus and randomly allocated to 7 groups, including control, I/R (30-min ischemia and 60-min reperfusion), and I/R groups pretreated with one of the following inhibitors: 4-PBA (targeting: ER stress), GSK2850163 (IRE1α), SP600125 (JNK), SR11302 (AP-1), and DCU (sEH). The inhibitor was administered for 15 min before ischemia with a peristaltic pump. Hemodynamic parameters including left ventricular systolic pressure (LVSP), left ventricular end-diastolic pressure (LVEDP), and maximal velocity of contraction (+dp/dtmax) and relaxation (-dp/dtmax) of the left ventricle were continuously recorded using an intraventricular balloon. Endothelial dilator function of the left anterior descending artery was studied in a wire myograph upon completion of reperfusion. The expression of ER stress molecules, JNK, c-Jun, and sEH was determined by western-blot. Results: I/R decreased LVSP (105.5±6.4 vs. 146.9±13.4 mmHg), and increased LVEDP (71.4±3.0 vs. 6.0±2.7 mmHg), with a resultant decreased LVDP (34.1±9.2 vs. 140.9±13.1 mmHg). I/R attenuated +dp/dtmax (651.7±142.1 vs. 2806.6±480.6 mmHg/s) and -dp/dtmax (-580.0±109.6 vs. -2118.0±244.9 mmHg/s) (all ps<0.001). The I/R-induced cardiac dysfunction could be alleviated by 4-PBA (LVSP 119.5±15.6 mmHg, p<0.01; LVEDP 21.2±4.2 mmHg, LVDP 98.3±12.0 mmHg, +dp/dtmax 2166.7±208.4 mmHg/s, and -dp/dtmax -1350.9±99.8 mmHg/s, all ps<0.001), GSK2850163 (LVSP 113.4±10.9 mmHg, p<0.01; LVEDP 37.1±3.1 mmHg, LVDP 76.3±13.9 mmHg, +dp/dtmax 1586.5±263.3 mmHg/s, -dp/dtmax -1127.7±159.9 mmHg/s, all ps<0.001), SP600125 (LVSP 113.9±5.6 mmHg, LVDP 40.5±3.3 mmHg, +dp/dtmax 970.1±89.8 mmHg/s, all ps<0.01), SR11302 (LVSP 97.9±7.5 mmHg, p<0.01; LVEDP 52.7±8.6mmHg, p<0.001; LVDP 45.2±9.8mmHg, p<0.05; +dp/dtmax 1231.5±196.6 mmHg/s, p<0.01; -dp/dtmax -658.3±68.9 mmHg/s, p<0.05), or DCU (LVSP 109.9±4.1 mmHg, p<0.01; LVEDP 11.7±1.8 mmHg, LVDP 98.2±4.9 mmHg, +dp/dtmax 1869.8±121.9 mmHg/s, and -dp/dtmax -1492.3±30.8 mmHg/s, all ps<0.001). The relaxant response of the coronary artery to acetylcholine was decreased after I/R in terms of both magnitude and sensitivity (p<0.001). All inhibitors improved acetylcholine-induced relaxation. Global I/R increased sEH expression and induced ER stress in both myocardium and coronary artery. Inhibition of ER stress or IRE1α downregulated I/R-induced sEH expression and inhibited JNK and c-Jun phosphorylation. Both JNK and AP-1 inhibitors lowered sEH level in myocardium and coronary artery in I/R-injured hearts. Conclusions: This study deciphered the molecular linkage between ER stress and sEH regulation in global I/R insult by uncovering a novel signaling axis of IRE1α-JNK-c-Jun/AP-1-sEH, which provided basis for future research on the therapeutic potential of targeting the IRE1α-JNK-c-Jun/AP-1-sEH axis for ischemic myocardial injury.
Collapse
Affiliation(s)
- Hong-Mei Xue
- The Institute of Cardiovascular Diseases & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, China.,Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wen-Tao Sun
- The Institute of Cardiovascular Diseases & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, China.,University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Huan-Xin Chen
- The Institute of Cardiovascular Diseases & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, China
| | - Guo-Wei He
- The Institute of Cardiovascular Diseases & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, China.,Drug Research and Development Center, Wannan Medical College, Wuhu, Anhui, China.,Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA
| | - Qin Yang
- The Institute of Cardiovascular Diseases & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, China
| |
Collapse
|
17
|
Wang F, Sun C, Lv X, Sun M, Si C, Zhen Y, Guo J, Sun W, Ye Z, Wen J, Liu P. Identification of a Novel Gene Correlated With Vascular Smooth Muscle Cells Proliferation and Migration in Chronic Thromboembolic Pulmonary Hypertension. Front Physiol 2021; 12:744219. [PMID: 34858201 PMCID: PMC8632225 DOI: 10.3389/fphys.2021.744219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/05/2021] [Indexed: 01/29/2023] Open
Abstract
Objective: Chronic thromboembolic pulmonary hypertension (CTEPH) is characterized by thrombofibrotic obstruction of the proximal pulmonary arteries, which result in vascular remodeling of the distal pulmonary artery. While the cellular and molecular mechanisms underlying CTEPH pathogenesis remain incompletely understood, recent evidence implicates vascular remodeling. Here, we identify the molecular mechanisms that contribute to vascular remodeling in CTEPH. Methods: Microarray data (GSE130391) for patients with CTEPH and healthy controls were downloaded from the Gene Expression Omnibus (GEO) and screened for differentially expressed genes (DEGs). DEGs were functionally annotated using Gene Ontology (GO) functional analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. A protein-protein interaction (PPI) network was constructed to identify hub genes. Finally, pulmonary artery samples were harvested from patients with CTEPH (n = 10) and from controls (n = 10) and primary vascular smooth muscle cells (VSMCs) were cultured. Effects of the proto-oncogene FOS on VSMC proliferation and migration were assessed using expression and knockdown studies. Results: We detected a total of 292 DEGs, including 151 upregulated and 141 downregulated genes. GO analysis revealed enrichment of DEGs in biological processes of signal transduction, response to lipopolysaccharide, signal transduction, and myeloid dendritic cell differentiation. Molecular function analysis revealed enrichment in tumor necrosis factor (TNF)-activated receptor activity, transcriptional activator activity, and protein homodimerization activity. The expression of TNF-α and its receptor (sTNFR1 and sTNFR2) were significantly higher in CTEPH group, compared with control group. KEGG pathway analysis revealed enrichment in salmonella infection, pathways in cancer, osteoclast differentiation, and cytokine-cytokine receptor interaction. Hub genes in the PPI included FOS, suggesting an important role for this gene in vascular remodeling in CTEPH. Primary VSMCs derived from patients with CTEPH showed increased FOS expression and high proliferation and migration, which was attenuated by FOS inhibition. In control VSMCs, TNF-α treatment increased proliferation and migration, which FOS inhibition likewise attenuated. Conclusion: TNF-α drives CTEPH pathogenesis by promoting VSMC proliferation and migration via increased FOS expression. These results advance our understanding of the molecular mechanisms of vascular remodeling in CTEPH, and may inform the development of new therapeutic targets.
Collapse
Affiliation(s)
- Feng Wang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Congrui Sun
- Department of Cardiovascular Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xiaoshuo Lv
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Mingsheng Sun
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Chaozeng Si
- Department of Operations and Information Management, China-Japan Friendship Hospital, Beijing, China
| | - Yanan Zhen
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jing Guo
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Weiliang Sun
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Zhidong Ye
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jianyan Wen
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Peng Liu
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Gupta D, Kumar A, Mandloi A, Shenoy V. Renin angiotensin aldosterone system in pulmonary fibrosis: Pathogenesis to therapeutic possibilities. Pharmacol Res 2021; 174:105924. [PMID: 34607005 DOI: 10.1016/j.phrs.2021.105924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/21/2021] [Accepted: 09/29/2021] [Indexed: 01/12/2023]
Abstract
Pulmonary fibrosis is a devastating lung disease with multifactorial etiology characterized by alveolar injury, fibroblast proliferation and excessive deposition of extracellular matrix proteins, which progressively results in respiratory failure and death. Accumulating evidence from experimental and clinical studies supports a central role of the renin angiotensin aldosterone system (RAAS) in the pathogenesis and progression of idiopathic pulmonary fibrosis. Angiotensin II (Ang II), a key vasoactive peptide of the RAAS mediates pro-inflammatory and pro-fibrotic effects on the lungs, adversely affecting organ function. Recent years have witnessed seminal discoveries in the field of RAAS. Identification of new enzymes, peptides and receptors has led to the development of several novel concepts. Of particular interest is the establishment of a protective axis of the RAAS comprising of Angiotensin converting enzyme 2 (ACE2), Angiotensin-(1-7) [Ang-(1-7)], and the Mas receptor (the ACE2/Ang-(1-7)/Mas axis), and the discovery of a functional role for the Angiotensin type 2 (AT2) receptor. Herein, we will review our current understanding of the role of RAAS in lung fibrogenesis, provide evidence on the anti-fibrotic actions of the newly recognized RAAS components (the ACE2/Ang-(1-7)/Mas axis and AT2 receptor), discuss potential strategies and translational efforts to convert this new knowledge into effective therapeutics for PF.
Collapse
Affiliation(s)
- Dipankar Gupta
- Congenital Heart Center, Department of Pediatrics, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Ashok Kumar
- Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS, USA
| | - Avinash Mandloi
- College of Pharmacy, VNS Group of Institutions, Bhopal, India
| | - Vinayak Shenoy
- College of Pharmacy, California Health Sciences University, Clovis, CA, USA.
| |
Collapse
|
19
|
Mamazhakypov A, Hein L, Lother A. Mineralocorticoid receptors in pulmonary hypertension and right heart failure: From molecular biology to therapeutic targeting. Pharmacol Ther 2021; 231:107987. [PMID: 34480966 DOI: 10.1016/j.pharmthera.2021.107987] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/21/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022]
Abstract
Pulmonary hypertension (PH) is a devastating condition characterized by pulmonary vascular remodelling, leading to progressive increase in pulmonary artery pressure and subsequent right ventricular failure. Aldosterone and the mineralocorticoid receptor (MR), a nuclear transcription factor, are key drivers of cardiovascular disease and MR antagonists are well-established in heart failure. Now, a growing body of evidence points at a detrimental role of MR in PH. Pharmacological MR blockade attenuated PH and prevented RV failure in experimental models. Mouse models with cell selective MR deletion suggest that this effect is mediated by MR in endothelial cells. While the evidence from experimental studies appears convincing, the available clinical data on MR antagonist use in patients with PH is more controversial. Integrated analysis of clinical data together with MR-dependent molecular alterations may provide insights why some patients respond to MRA treatment while others do not. Potential ways to identify MRA 'responders' include the analysis of underlying PH causes, stage of disease, or sex, as well as new biomarkers.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany; Heart Center Freiburg University, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Germany.
| |
Collapse
|
20
|
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: 5.0] [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
|
21
|
Alba GA, Samokhin AO, Wang RS, Zhang YY, Wertheim BM, Arons E, Greenfield EA, Lundberg Slingsby MH, Ceglowski JR, Haley KJ, Bowman FP, Yu YR, Haney JC, Eng G, Mitchell RN, Sheets A, Vargas SO, Seo S, Channick RN, Leary PJ, Rajagopal S, Loscalzo J, Battinelli EM, Maron BA. NEDD9 Is a Novel and Modifiable Mediator of Platelet-Endothelial Adhesion in the Pulmonary Circulation. Am J Respir Crit Care Med 2021; 203:1533-1545. [PMID: 33523764 PMCID: PMC8483217 DOI: 10.1164/rccm.202003-0719oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 01/29/2021] [Indexed: 12/16/2022] Open
Abstract
Rationale: Data on the molecular mechanisms that regulate platelet-pulmonary endothelial adhesion under conditions of hypoxia are lacking, but may have important therapeutic implications. Objectives: To identify a hypoxia-sensitive, modifiable mediator of platelet-pulmonary artery endothelial cell adhesion and thrombotic remodeling. Methods: Network medicine was used to profile protein-protein interactions in hypoxia-treated human pulmonary artery endothelial cells. Data from liquid chromatography-mass spectrometry and microscale thermophoresis informed the development of a novel antibody (Ab) to inhibit platelet-endothelial adhesion, which was tested in cells from patients with chronic thromboembolic pulmonary hypertension (CTEPH) and three animal models in vivo. Measurements and Main Results: The protein NEDD9 was identified in the hypoxia thrombosome network in silico. Compared with normoxia, hypoxia (0.2% O2) for 24 hours increased HIF-1α (hypoxia-inducible factor-1α)-dependent NEDD9 upregulation in vitro. Increased NEDD9 was localized to the plasma-membrane surface of cells from control donors and patients with CTEPH. In endarterectomy specimens, NEDD9 colocalized with the platelet surface adhesion molecule P-selectin. Our custom-made anti-NEDD9 Ab targeted the NEDD9-P-selectin interaction and inhibited the adhesion of activated platelets to pulmonary artery endothelial cells from control donors in vitro and from patients with CTEPH ex vivo. Compared with control mice, platelet-pulmonary endothelial aggregates and pulmonary hypertension induced by ADP were decreased in NEDD9-/- mice or wild-type mice treated with the anti-NEDD9 Ab, which also decreased chronic pulmonary thromboembolic remodeling in vivo. Conclusions: The NEDD9-P-selectin protein-protein interaction is a modifiable target with which to inhibit platelet-pulmonary endothelial adhesion and thromboembolic vascular remodeling, with potential therapeutic implications for patients with disorders of increased hypoxia signaling pathways, including CTEPH.
Collapse
Affiliation(s)
- George A. Alba
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Andriy O. Samokhin
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Rui-Sheng Wang
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Ying-Yi Zhang
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Elena Arons
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | | | | | | | | | - Frederick P. Bowman
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Yen-Rei Yu
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - John. C. Haney
- Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - George Eng
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Anthony Sheets
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, and
| | - Sara O. Vargas
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Duke University, Durham, North Carolina
| | - Sachiko Seo
- Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts
| | - Richard N. Channick
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi, Japan
| | - Peter J. Leary
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Ronald Reagan UCLA Medical Center, University of California, Los Angeles, Los Angeles, California; and
| | - Sudarshan Rajagopal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Joseph Loscalzo
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Bradley A. Maron
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
22
|
Kowalski J, Deng L, Suennen C, Koca D, Meral D, Bode C, Hein L, Lother A. Eplerenone Improves Pulmonary Vascular Remodeling and Hypertension by Inhibition of the Mineralocorticoid Receptor in Endothelial Cells. Hypertension 2021; 78:456-465. [PMID: 33966455 DOI: 10.1161/hypertensionaha.120.16196] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jessica Kowalski
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany
| | - Lisa Deng
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany
| | - Chiara Suennen
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany
| | - Duygu Koca
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany
| | - David Meral
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), Cardiovascular Research Track (D.M.), University of Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, Faculty of Medicine, Heart Center Freiburg University (C.B., A.L.), University of Freiburg, Germany
| | - Lutz Hein
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies (L.H.), University of Freiburg, Germany
| | - Achim Lother
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany.,Department of Cardiology and Angiology I, Faculty of Medicine, Heart Center Freiburg University (C.B., A.L.), University of Freiburg, Germany
| |
Collapse
|
23
|
Peters EL, Bogaard HJ, Vonk Noordegraaf A, de Man FS. Neurohormonal modulation in pulmonary arterial hypertension. Eur Respir J 2021; 58:13993003.04633-2020. [PMID: 33766951 PMCID: PMC8551560 DOI: 10.1183/13993003.04633-2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/13/2021] [Indexed: 12/15/2022]
Abstract
Pulmonary hypertension is a fatal condition of elevated pulmonary pressures, complicated by right heart failure. Pulmonary hypertension appears in various forms; one of those is pulmonary arterial hypertension (PAH) and is particularly characterised by progressive remodelling and obstruction of the smaller pulmonary vessels. Neurohormonal imbalance in PAH patients is associated with worse prognosis and survival. In this back-to-basics article on neurohormonal modulation in PAH, we provide an overview of the pharmacological and nonpharmacological strategies that have been tested pre-clinically and clinically. The benefit of neurohormonal modulation strategies in PAH patients has been limited by lack of insight into how the neurohormonal system is changed throughout the disease and difficulties in translation from animal models to human trials. We propose that longitudinal and individual assessments of neurohormonal status are required to improve the timing and specificity of neurohormonal modulation strategies. Ongoing developments in imaging techniques such as positron emission tomography may become helpful to determine neurohormonal status in PAH patients in different disease stages and optimise individual treatment responses.
Collapse
Affiliation(s)
- Eva L Peters
- Dept of Pulmonology, Amsterdam UMC, Amsterdam, The Netherlands.,Dept of Physiology, Amsterdam UMC, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
24
|
Lahm T, Hess E, Barón AE, Maddox TM, Plomondon ME, Choudhary G, Maron BA, Zamanian RT, Leary PJ. Renin-Angiotensin-Aldosterone System Inhibitor Use and Mortality in Pulmonary Hypertension: Insights From the Veterans Affairs Clinical Assessment Reporting and Tracking Database. Chest 2020; 159:1586-1597. [PMID: 33031831 DOI: 10.1016/j.chest.2020.09.258] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/25/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The renin-angiotensin-aldosterone system (RAAS) contributes to pulmonary hypertension (PH) pathogenesis. Although animal data suggest that RAAS inhibition attenuates PH, it is unknown if RAAS inhibition is beneficial in PH patients. RESEARCH QUESTION Is RAAS inhibitor use associated with lower mortality in a large cohort of patients with hemodynamically confirmed PH? STUDY DESIGN AND METHODS We used the Department of Veterans Affairs Clinical Assessment Reporting and Tracking Database to study retrospectively relationships between RAAS inhibitors (angiotensin converting enzyme inhibitors [ACEIs], angiotensin receptor blockers [ARBs], and aldosterone antagonists [AAs]) and mortality in 24,221 patients with hemodynamically confirmed PH. We evaluated relationships in the full and in propensity-matched cohorts. Analyses were adjusted for demographics, socioeconomic status, comorbidities, disease severity, and comedication use in staged models. RESULTS ACEI and ARB use was associated with improved survival in unadjusted Kaplan-Meier survival analyses in the full cohort and the propensity-matched cohort. This relationship was insensitive to adjustment, independent of pulmonary artery wedge pressure, and also was observed in a cohort restricted to individuals with precapillary PH. AA use was associated with worse survival in unadjusted Kaplan-Meier survival analyses in the full cohort; however, AA use was associated less robustly with mortality in the propensity-matched cohort and was not associated with worse survival after adjustment for disease severity, indicating that AAs in real-world practice are used preferentially in sicker patients and that the unadjusted association with increased mortality may be an artifice of confounding by indication of severity. INTERPRETATION ACEI and ARB use is associated with lower mortality in veterans with PH. AA use is a marker of disease severity in PH. ACEIs and ARBs may represent a novel treatment strategy for diverse PH phenotypes.
Collapse
Affiliation(s)
- Tim Lahm
- Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN; Indiana University School of Medicine, Indianapolis, IN.
| | - Edward Hess
- Veterans Affairs Eastern Colorado Health Care System, Denver, CO
| | - Anna E Barón
- Veterans Affairs Eastern Colorado Health Care System, Denver, CO; Colorado School of Public Health, Denver, CO
| | - Thomas M Maddox
- Washington University School of Medicine Division of Cardiology and Healthcare Innovation Lab, St. Louis, MO
| | - Mary E Plomondon
- Veterans Affairs Eastern Colorado Health Care System, Denver, CO
| | - Gaurav Choudhary
- Providence Veterans Affairs Medical Center, Providence, RI; Alpert Medical School of Brown University, Providence, RI
| | - Bradley A Maron
- Veterans Affairs Boston Healthcare System, Boston, MA; Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Roham T Zamanian
- Stanford University Division of Pulmonary, Allergy, and Critical Care Medicine and Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford, CA
| | | |
Collapse
|
25
|
Klinke A, Schubert T, Müller M, Legchenko E, Zelt JGE, Shimauchi T, Napp LC, Rothman AMK, Bonnet S, Stewart DJ, Hansmann G, Rudolph V. Emerging therapies for right ventricular dysfunction and failure. Cardiovasc Diagn Ther 2020; 10:1735-1767. [PMID: 33224787 PMCID: PMC7666928 DOI: 10.21037/cdt-20-592] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022]
Abstract
Therapeutic options for right ventricular (RV) dysfunction and failure are strongly limited. Right heart failure (RHF) has been mostly addressed in the context of pulmonary arterial hypertension (PAH), where it is not possible to discern pulmonary vascular- and RV-directed effects of therapeutic approaches. In part, opposing pathomechanisms in RV and pulmonary vasculature, i.e., regarding apoptosis, angiogenesis and proliferation, complicate addressing RHF in PAH. Therapy effective for left heart failure is not applicable to RHF, e.g., inhibition of adrenoceptor signaling and of the renin-angiotensin system had no or only limited success. A number of experimental studies employing animal models for PAH or RV dysfunction or failure have identified beneficial effects of novel pharmacological agents, with most promising results obtained with modulators of metabolism and reactive oxygen species or inflammation, respectively. In addition, established PAH agents, in particular phosphodiesterase-5 inhibitors and soluble guanylate cyclase stimulators, may directly address RV integrity. Promising results are furthermore derived with microRNA (miRNA) and long non-coding RNA (lncRNA) blocking or mimetic strategies, which can target microvascular rarefaction, inflammation, metabolism or fibrotic and hypertrophic remodeling in the dysfunctional RV. Likewise, pre-clinical data demonstrate that cell-based therapies using stem or progenitor cells have beneficial effects on the RV, mainly by improving the microvascular system, however clinical success will largely depend on delivery routes. A particular option for PAH is targeted denervation of the pulmonary vasculature, given the sympathetic overdrive in PAH patients. Finally, acute and durable mechanical circulatory support are available for the right heart, which however has been tested mostly in RHF with concomitant left heart disease. Here, we aim to review current pharmacological, RNA- and cell-based therapeutic options and their potential to directly target the RV and to review available data for pulmonary artery denervation and mechanical circulatory support.
Collapse
Affiliation(s)
- Anna Klinke
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Torben Schubert
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Marion Müller
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Ekaterina Legchenko
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
| | - Jason G. E. Zelt
- Division of Cardiology, University of Ottawa Heart Institute and the Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Canada
| | - Tsukasa Shimauchi
- Pulmonary Hypertension Research Group, Centre de recherche de IUCPQ/Laval University, Quebec, Canada
| | - L. Christian Napp
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | | | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Centre de recherche de IUCPQ/Laval University, Quebec, Canada
| | - Duncan J. Stewart
- Division of Cardiology, University of Ottawa Heart Institute and the Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Canada
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
| | - Volker Rudolph
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| |
Collapse
|
26
|
Omidkhoda N, Vakilian F, Mohammadpour AH, Sathyapalan T, Sahebkar A. Aldosterone and Mineralocorticoid Receptor Antagonists on Pulmonary Hypertension and Right Ventricular Failure: A Review. Curr Pharm Des 2020; 26:3862-3870. [DOI: 10.2174/1381612826666200523171137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 04/06/2020] [Indexed: 12/23/2022]
Abstract
There is an increasing number of therapeutic agents being developed for the treatment of pulmonary
artery hypertension (PAH) which is a condition characterized by raised pulmonary artery pressure and right heart
failure. Despite our better understanding of the pathophysiology of PAH, the treatment outcomes are still suboptimal.
There is growing evidence suggesting the role of increases in the levels of aldosterone, which is a mineralocorticoid
hormone, in the pathophysiology of PAH; however, the extent to which hyperaldosteronism is associated
with PAH in patients is unclear. There are also a few studies assessing the effects of mineralocorticoid receptor
antagonists (MRA) in PAH. MRAs are a recognized treatment for heart failure and hypertension. In this review,
we focus on the relationship between aldosterone level in patients with PAH and right ventricular failure
and the effect of MRAs on the PAH severity.
Collapse
Affiliation(s)
- Navid Omidkhoda
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farveh Vakilian
- Atherosclerotic Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir H. Mohammadpour
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull HU3 2JZ, United Kingdom
| | | |
Collapse
|
27
|
Affiliation(s)
- Achim Lother
- From the Department of Cardiology and Angiology I, Faculty of Medicine, Heart Center Freiburg University, University of Freiburg, Germany (A.L.)
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany (A.L.)
| |
Collapse
|
28
|
Extra-adrenal glucocorticoid biosynthesis: implications for autoimmune and inflammatory disorders. Genes Immun 2020; 21:150-168. [PMID: 32203088 PMCID: PMC7276297 DOI: 10.1038/s41435-020-0096-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022]
Abstract
Glucocorticoid synthesis is a complex, multistep process that starts with cholesterol being delivered to the inner membrane of mitochondria by StAR and StAR-related proteins. Here its side chain is cleaved by CYP11A1 producing pregnenolone. Pregnenolone is converted to cortisol by the enzymes 3-βHSD, CYP17A1, CYP21A2 and CYP11B1. Glucocorticoids play a critical role in the regulation of the immune system and exert their action through the glucocorticoid receptor (GR). Although corticosteroids are primarily produced in the adrenal gland, they can also be produced in a number of extra-adrenal tissue including the immune system, skin, brain, and intestine. Glucocorticoid production is regulated by ACTH, CRH, and cytokines such as IL-1, IL-6 and TNFα. The bioavailability of cortisol is also dependent on its interconversion to cortisone which is inactive, by 11βHSD1/2. Local and systemic glucocorticoid biosynthesis can be stimulated by ultraviolet B, explaining its immunosuppressive activity. In this review, we want to emphasize that dysregulation of extra-adrenal glucocorticoid production can play a key role in a variety of autoimmune diseases including multiple sclerosis (MS), lupus erythematosus (LE), rheumatoid arthritis (RA), and skin inflammatory disorders such as psoriasis and atopic dermatitis (AD). Further research on local glucocorticoid production and its bioavailability may open doors into new therapies for autoimmune diseases.
Collapse
|
29
|
Maron BA, Leopold JA, Hemnes AR. Metabolic syndrome, neurohumoral modulation, and pulmonary arterial hypertension. Br J Pharmacol 2020; 177:1457-1471. [PMID: 31881099 DOI: 10.1111/bph.14968] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/03/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022] Open
Abstract
Pulmonary vascular disease, including pulmonary arterial hypertension (PAH), is increasingly recognized to be affected by systemic alterations including up-regulation of the renin-angiotensin-aldosterone system and perturbations to metabolic pathways, particularly glucose and fat metabolism. There is increasing preclinical and clinical data that each of these pathways can promote pulmonary vascular disease and right heart failure and are not simply disease markers. More recently, trials of therapeutics aimed at neurohormonal activation or metabolic dysfunction are beginning to shed light on how interventions in these pathways may affect patients with PAH. This review will focus on underlying mechanistic data that supports neurohormonal activation and metabolic dysfunction in the pathogenesis of PAH and right heart failure as well as discussing early translational data in patients with PAH.
Collapse
Affiliation(s)
- Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jane A Leopold
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Anna R Hemnes
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
30
|
Napoli C, Benincasa G, Loscalzo J. Epigenetic Inheritance Underlying Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2020; 39:653-664. [PMID: 30727752 DOI: 10.1161/atvbaha.118.312262] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In pulmonary arterial hypertension (PAH), the Warburg effect (glycolytic shift) and mitochondrial fission are determinants of phenotype alterations characteristic of the disease, such as proliferation, apoptosis resistance, migration, endothelial-mesenchymal transition, and extracellular matrix stiffness. Current therapies, focusing largely on vasodilation and antithrombotic protection, do not restore these aberrant phenotypes suggesting that additional pathways need be targeted. The multifactorial nature of PAH suggests epigenetic changes as potential determinants of vascular remodeling. Transgenerational epigenetic changes induced by hypoxia can result in permanent changes early in fetal development increasing PAH risk in adulthood. Unlike genetic mutations, epigenetic changes are pharmacologically reversible, making them an attractive target as therapeutic strategies for PAH. This review offers a landscape of the most current clinical, epigenetic-sensitive changes contributing to PAH vascular remodeling both in early and later life, with a focus on a network medicine strategy. Furthermore, we discuss the importance of the application (from morphogenesis to disease onset) of molecular network-based algorithms to dissect PAH molecular pathobiology. Additionally, we suggest an integrated network-based program for clinical disease gene discovery that may reveal novel biomarkers and novel disease targets, thus offering a truly innovative path toward redefining and treating PAH, as well as facilitating the trajectory of a comprehensive precision medicine approach to PAH.
Collapse
Affiliation(s)
- Claudio Napoli
- From the Department of Medical, Surgical, Neurological, Metabolic, and Geriatric Sciences (C.N., G.B.), University of Campania Luigi Vanvitelli, Naples, Italy
- Clinical Department of Internal Medicine and Specialistic Units AOU (C.N., G.B.), University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giuditta Benincasa
- From the Department of Medical, Surgical, Neurological, Metabolic, and Geriatric Sciences (C.N., G.B.), University of Campania Luigi Vanvitelli, Naples, Italy
- Clinical Department of Internal Medicine and Specialistic Units AOU (C.N., G.B.), University of Campania Luigi Vanvitelli, Naples, Italy
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.)
| |
Collapse
|
31
|
Prins KW, Thenappan T, Weir EK, Kalra R, Pritzker M, Archer SL. Repurposing Medications for Treatment of Pulmonary Arterial Hypertension: What's Old Is New Again. J Am Heart Assoc 2020; 8:e011343. [PMID: 30590974 PMCID: PMC6405714 DOI: 10.1161/jaha.118.011343] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kurt W Prins
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - Thenappan Thenappan
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - E Kenneth Weir
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - Rajat Kalra
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - Marc Pritzker
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | | |
Collapse
|
32
|
Elinoff JM, Mazer AJ, Cai R, Lu M, Graninger G, Harper B, Ferreyra GA, Sun J, Solomon MA, Danner RL. Meta-analysis of blood genome-wide expression profiling studies in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2019; 318:L98-L111. [PMID: 31617731 DOI: 10.1152/ajplung.00252.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Inflammatory cell infiltrates are a prominent feature of aberrant vascular remodeling in pulmonary arterial hypertension (PAH), suggesting that immune effector cells contribute to disease progression. Genome-wide blood expression profiling studies have attempted to better define this inflammatory component of PAH pathobiology but have been hampered by small sample sizes, methodological differences, and very little gene-level reproducibility. The current meta-analysis (seven studies; 156 PAH patients/110 healthy controls) was performed to assess the comparability of data across studies and to possibly derive a generalizable transcriptomic signature. Idiopathic (IPAH) compared with disease-associated PAH (APAH) displayed highly similar expression profiles with no differentially expressed genes, even after substantially relaxing selection stringency. In contrast, using a false discovery rate of ≤1% and I2 < 40% (low-to-moderate heterogeneity across studies) both IPAH and APAH differed markedly from healthy controls with the combined PAH cohort yielding 1,269 differentially expressed, unique gene transcripts. Bioinformatic analyses, including gene-set enrichment, which uses all available data independent of gene selection thresholds, identified interferon, mammalian target of rapamycin/p70S6K, stress kinase, and Toll-like receptor signaling as enriched mechanisms within the PAH gene signature. Enriched biological functions and diseases included tumorigenesis, autoimmunity, antiviral response, and cell death consistent with prevailing theories of PAH pathogenesis. Although otherwise indistinguishable, APAH (predominantly PAH due to systemic sclerosis) had a somewhat stronger interferon profile than IPAH. Meta-analysis defined a robust and generalizable transcriptomic signature in the blood of PAH patients that can help inform the identification of biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jason M Elinoff
- Critical Care Medicine Department, Clinical Center, and National Institutes of Health, Bethesda, Maryland
| | - Adrien J Mazer
- Critical Care Medicine Department, Clinical Center, and National Institutes of Health, Bethesda, Maryland
| | - Rongman Cai
- Critical Care Medicine Department, Clinical Center, and National Institutes of Health, Bethesda, Maryland
| | - Mengyun Lu
- Critical Care Medicine Department, Clinical Center, and National Institutes of Health, Bethesda, Maryland
| | - Grace Graninger
- Critical Care Medicine Department, Clinical Center, and National Institutes of Health, Bethesda, Maryland
| | - Bonnie Harper
- Critical Care Medicine Department, Clinical Center, and National Institutes of Health, Bethesda, Maryland
| | - Gabriela A Ferreyra
- Critical Care Medicine Department, Clinical Center, and National Institutes of Health, Bethesda, Maryland
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, and National Institutes of Health, Bethesda, Maryland
| | - Michael A Solomon
- Critical Care Medicine Department, Clinical Center, and National Institutes of Health, Bethesda, Maryland.,Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Robert L Danner
- Critical Care Medicine Department, Clinical Center, and National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
33
|
Udjus C, Cero FT, Halvorsen B, Behmen D, Carlson CR, Bendiksen BA, Espe EKS, Sjaastad I, Løberg EM, Yndestad A, Aukrust P, Christensen G, Skjønsberg OH, Larsen KO. Caspase-1 induces smooth muscle cell growth in hypoxia-induced pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2019; 316:L999-L1012. [PMID: 30908936 DOI: 10.1152/ajplung.00322.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Lung diseases with hypoxia are complicated by pulmonary hypertension, leading to heart failure and death. No pharmacological treatment exists. Increased proinflammatory cytokines are found in hypoxic patients, suggesting an inflammatory pathogenesis. Caspase-1, the effector of the inflammasome, mediates inflammation through activation of the proinflammatory cytokines interleukin (IL)-18 and IL-1β. Here, we investigate inflammasome-related mechanisms that can trigger hypoxia-induced pulmonary hypertension. Our aim was to examine whether caspase-1 induces development of hypoxia-related pulmonary hypertension and is a suitable target for therapy. Wild-type (WT) and caspase-1-/- mice were exposed to 10% oxygen for 14 days. Hypoxic caspase-1-/- mice showed lower pressure and reduced muscularization in pulmonary arteries, as well as reduced right ventricular remodeling compared with WT. Smooth muscle cell (SMC) proliferation was reduced in caspase-1-deficient pulmonary arteries and in WT arteries treated with a caspase-1 inhibitor. Impaired inflammation was shown in hypoxic caspase-1-/- mice by abolished pulmonary influx of immune cells and lower levels of IL-18, IL-1β, and IL-6, which were also reduced in the medium surrounding caspase-1 abrogated pulmonary arteries. By adding IL-18 or IL-1β to caspase-1-deficient pulmonary arteries, SMC proliferation was retained. Furthermore, inhibition of both IL-6 and phosphorylated STAT3 reduced proliferation of SMC in vitro, indicating IL-18, IL-6, and STAT3 as downstream mediators of caspase-1-induced SMC proliferation in pulmonary arteries. Caspase-1 induces SMC proliferation in pulmonary arteries through the caspase-1/IL-18/IL-6/STAT3 pathway, leading to pulmonary hypertension in mice exposed to hypoxia. We propose that caspase-1 inhibition is a potential target for treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Camilla Udjus
- Department of Pulmonary Medicine, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway.,Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway.,K. G. Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo , Oslo , Norway
| | - Fadila T Cero
- Department of Pulmonary Medicine, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway.,Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway.,K. G. Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo , Oslo , Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet and University of Oslo , Oslo , Norway
| | - Dina Behmen
- Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway.,K. G. Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo , Oslo , Norway
| | - Cathrine R Carlson
- Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway.,K. G. Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo , Oslo , Norway
| | - Bård A Bendiksen
- Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway.,K. G. Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo , Oslo , Norway
| | - Emil K S Espe
- Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway.,K. G. Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo , Oslo , Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway.,K. G. Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo , Oslo , Norway
| | - Else M Løberg
- Department of Pathology, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway
| | - Arne Yndestad
- K. G. Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo , Oslo , Norway.,Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet and University of Oslo , Oslo , Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet and University of Oslo , Oslo , Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet and University of Oslo , Oslo , Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway.,K. G. Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo , Oslo , Norway
| | - Ole H Skjønsberg
- Department of Pulmonary Medicine, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway
| | - Karl-Otto Larsen
- Department of Pulmonary Medicine, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway.,K. G. Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo , Oslo , Norway
| |
Collapse
|
34
|
Abstract
Pulmonary arterial hypertension (PAH) is a pulmonary vasculopathy that causes right ventricular dysfunction and exercise limitation and progresses to death. New findings from translational studies have suggested alternative pathways for treatment. These avenues include sex hormones, genetic abnormalities and DNA damage, elastase inhibition, metabolic dysfunction, cellular therapies, and anti-inflammatory approaches. Both novel and repurposed compounds with rationale from preclinical experimental models and human cells are now in clinical trials in patients with PAH. Findings from these studies will elucidate the pathobiology of PAH and may result in clinically important improvements in outcome.
Collapse
Affiliation(s)
- Edda Spiekerkoetter
- Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA; ,
| | - Steven M Kawut
- Department of Medicine and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6021, USA;
| | - Vinicio A de Jesus Perez
- Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA; ,
| |
Collapse
|
35
|
Buonafine M, Bonnard B, Jaisser F. Mineralocorticoid Receptor and Cardiovascular Disease. Am J Hypertens 2018; 31:1165-1174. [PMID: 30192914 DOI: 10.1093/ajh/hpy120] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 08/09/2018] [Indexed: 12/14/2022] Open
Abstract
Activation of the mineralocorticoid receptor (MR) in the distal nephron by its ligand, aldosterone, plays an important role in sodium reabsorption and blood pressure regulation. However, expression of the MR goes beyond the kidney. It is expressed in a variety of other tissues in which its activation could lead to tissue injury. Indeed, MR activation in the cardiovascular (CV) system has been shown to promote hypertension, fibrosis, and inflammation. Pharmacological blockade of the MR has protective effects in several animal models of CV disease. Furthermore, the use of MR antagonists is beneficial for heart failure patients, preventing mortality and morbidity. A better understanding of the implications of the MR in the setting of CV diseases is critical for refining treatments and improving patient care. The mechanisms involved in the deleterious effects of MR activation are complex and include oxidative stress, inflammation, and fibrosis. This review will discuss the pathological role of the MR in the CV system and the major mechanisms underlying it.
Collapse
Affiliation(s)
- Mathieu Buonafine
- INSERM, UMRS, Centre de Recherche des Cordeliers, Sorbonne University, Paris, France
- Paris Descartes University, Paris, France
| | - Benjamin Bonnard
- INSERM, UMRS, Centre de Recherche des Cordeliers, Sorbonne University, Paris, France
- Paris Descartes University, Paris, France
| | - Frédéric Jaisser
- INSERM, UMRS, Centre de Recherche des Cordeliers, Sorbonne University, Paris, France
- Paris Descartes University, Paris, France
- INSERM, Clinical Investigation Centre, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, RHU Fight-HF, Nancy, France
| |
Collapse
|
36
|
Badlam JB, Austin ED. Beyond oestrogens: towards a broader evaluation of the hormone profile in pulmonary arterial hypertension. Eur Respir J 2018; 51:51/6/1801058. [PMID: 29954927 DOI: 10.1183/13993003.01058-2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 06/06/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Jessica B Badlam
- University of Colorado at Denver - Anschutz Medical Campus, Aurora, CO, USA
| | - Eric D Austin
- Dept of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
37
|
Samokhin AO, Stephens T, Wertheim BM, Wang RS, Vargas SO, Yung LM, Cao M, Brown M, Arons E, Dieffenbach PB, Fewell JG, Matar M, Bowman FP, Haley KJ, Alba GA, Marino SM, Kumar R, Rosas IO, Waxman AB, Oldham WM, Khanna D, Graham BB, Seo S, Gladyshev VN, Yu PB, Fredenburgh LE, Loscalzo J, Leopold JA, Maron BA. NEDD9 targets COL3A1 to promote endothelial fibrosis and pulmonary arterial hypertension. Sci Transl Med 2018; 10:eaap7294. [PMID: 29899023 PMCID: PMC6223025 DOI: 10.1126/scitranslmed.aap7294] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 05/23/2018] [Indexed: 12/12/2022]
Abstract
Germline mutations involving small mothers against decapentaplegic-transforming growth factor-β (SMAD-TGF-β) signaling are an important but rare cause of pulmonary arterial hypertension (PAH), which is a disease characterized, in part, by vascular fibrosis and hyperaldosteronism (ALDO). We developed and analyzed a fibrosis protein-protein network (fibrosome) in silico, which predicted that the SMAD3 target neural precursor cell expressed developmentally down-regulated 9 (NEDD9) is a critical ALDO-regulated node underpinning pathogenic vascular fibrosis. Bioinformatics and microscale thermophoresis demonstrated that oxidation of Cys18 in the SMAD3 docking region of NEDD9 impairs SMAD3-NEDD9 protein-protein interactions in vitro. This effect was reproduced by ALDO-induced oxidant stress in cultured human pulmonary artery endothelial cells (HPAECs), resulting in impaired NEDD9 proteolytic degradation, increased NEDD9 complex formation with Nk2 homeobox 5 (NKX2-5), and increased NKX2-5 binding to COL3A1 Up-regulation of NEDD9-dependent collagen III expression corresponded to changes in cell stiffness measured by atomic force microscopy. HPAEC-derived exosomal signaling targeted NEDD9 to increase collagen I/III expression in human pulmonary artery smooth muscle cells, identifying a second endothelial mechanism regulating vascular fibrosis. ALDO-NEDD9 signaling was not affected by treatment with a TGF-β ligand trap and, thus, was not contingent on TGF-β signaling. Colocalization of NEDD9 with collagen III in HPAECs was observed in fibrotic pulmonary arterioles from PAH patients. Furthermore, NEDD9 ablation or inhibition prevented fibrotic vascular remodeling and pulmonary hypertension in animal models of PAH in vivo. These data identify a critical TGF-β-independent posttranslational modification that impairs SMAD3-NEDD9 binding in HPAECs to modulate vascular fibrosis and promote PAH.
Collapse
Affiliation(s)
- Andriy O Samokhin
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Thomas Stephens
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Bradley M Wertheim
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Rui-Sheng Wang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Sara O Vargas
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Lai-Ming Yung
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Minwei Cao
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Marcel Brown
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Elena Arons
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Paul B Dieffenbach
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | - Majed Matar
- Celsion Corporation, Lawrenceville, NJ 08648, USA
| | - Frederick P Bowman
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kathleen J Haley
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - George A Alba
- Department of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stefano M Marino
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Biotechnology, Akdeniz University, Konyaaltı, Antalya 07058, Turkey
| | - Rahul Kumar
- Program in Translational Lung Research, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Aaron B Waxman
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - William M Oldham
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Dinesh Khanna
- Division of Rheumatology, University of Michigan Scleroderma Program, Ann Arbor, MI 48109, USA
| | - Brian B Graham
- Program in Translational Lung Research, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sachiko Seo
- Department of Hematology and Oncology, National Cancer Research Center East, Kashiwa-shi, Chiba-ken 277-8577, Japan
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Paul B Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Laura E Fredenburgh
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jane A Leopold
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Global transcriptomic analysis suggests carbon dioxide as an environmental stressor in spaceflight: A systems biology GeneLab case study. Sci Rep 2018. [PMID: 29520055 PMCID: PMC5843582 DOI: 10.1038/s41598-018-22613-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Spaceflight introduces a combination of environmental stressors, including microgravity, ionizing radiation, changes in diet and altered atmospheric gas composition. In order to understand the impact of each environmental component on astronauts it is important to investigate potential influences in isolation. Rodent spaceflight experiments involve both standard vivarium cages and animal enclosure modules (AEMs), which are cages used to house rodents in spaceflight. Ground control AEMs are engineered to match the spaceflight environment. There are limited studies examining the biological response invariably due to the configuration of AEM and vivarium housing. To investigate the innate global transcriptomic patterns of rodents housed in spaceflight-matched AEM compared to standard vivarium cages we utilized publicly available data from the NASA GeneLab repository. Using a systems biology approach, we observed that AEM housing was associated with significant transcriptomic differences, including reduced metabolism, altered immune responses, and activation of possible tumorigenic pathways. Although we did not perform any functional studies, our findings revealed a mild hypoxic phenotype in AEM, possibly due to atmospheric carbon dioxide that was increased to match conditions in spaceflight. Our investigation illustrates the process of generating new hypotheses and informing future experimental research by repurposing multiple space-flown datasets.
Collapse
|
39
|
Boehm M, Arnold N, Braithwaite A, Pickworth J, Lu C, Novoyatleva T, Kiely DG, Grimminger F, Ghofrani HA, Weissmann N, Seeger W, Lawrie A, Schermuly RT, Kojonazarov B. Eplerenone attenuates pathological pulmonary vascular rather than right ventricular remodeling in pulmonary arterial hypertension. BMC Pulm Med 2018; 18:41. [PMID: 29499691 PMCID: PMC5833097 DOI: 10.1186/s12890-018-0604-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 02/23/2018] [Indexed: 12/11/2022] Open
Abstract
Background Aldosterone is a mineralocorticoid hormone critically involved in arterial blood pressure regulation. Although pharmacological aldosterone antagonism reduces mortality and morbidity among patients with severe left-sided heart failure, the contribution of aldosterone to the pathobiology of pulmonary arterial hypertension (PAH) and right ventricular (RV) heart failure is not fully understood. Methods The effects of Eplerenone (0.1% Inspra® mixed in chow) on pulmonary vascular and RV remodeling were evaluated in mice with pulmonary hypertension (PH) caused by Sugen5416 injection with concomitant chronic hypoxia (SuHx) and in a second animal model with established RV dysfunction independent from lung remodeling through surgical pulmonary artery banding. Results Preventive Eplerenone administration attenuated the development of PH and pathological remodeling of pulmonary arterioles. Therapeutic aldosterone antagonism – starting when RV dysfunction was established - normalized mineralocorticoid receptor gene expression in the right ventricle without direct effects on either RV structure (Cardiomyocyte hypertrophy, Fibrosis) or function (assessed by non-invasive echocardiography along with intra-cardiac pressure volume measurements), but significantly lowered systemic blood pressure. Conclusions Our data indicate that aldosterone antagonism with Eplerenone attenuates pulmonary vascular rather than RV remodeling in PAH.
Collapse
Affiliation(s)
- Mario Boehm
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Aulweg 130, 35392, Giessen, Germany
| | - Nadine Arnold
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Adam Braithwaite
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Josephine Pickworth
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Changwu Lu
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Aulweg 130, 35392, Giessen, Germany
| | - Tatyana Novoyatleva
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Aulweg 130, 35392, Giessen, Germany
| | - David G Kiely
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, UK
| | - Friedrich Grimminger
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Aulweg 130, 35392, Giessen, Germany
| | - Hossein A Ghofrani
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Aulweg 130, 35392, Giessen, Germany
| | - Norbert Weissmann
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Aulweg 130, 35392, Giessen, Germany
| | - Werner Seeger
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Aulweg 130, 35392, Giessen, Germany
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Ralph T Schermuly
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Aulweg 130, 35392, Giessen, Germany.
| | - Baktybek Kojonazarov
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Aulweg 130, 35392, Giessen, Germany
| |
Collapse
|
40
|
Karolczak K, Kubalczyk P, Glowacki R, Pietruszynski R, Watala C. Aldosterone modulates blood homocysteine and cholesterol in coronary artery disease patients - a possible impact on atherothrombosis? Physiol Res 2018; 67:197-207. [PMID: 29303611 DOI: 10.33549/physiolres.933668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Aldosterone plays a key role in maintaining the homeostasis of the whole organism. Under some circumstances, aldosterone can contribute to the progression of cardiovascular diseases, including coronary artery disease. This study demonstrates that aldosterone associates negatively with some lipidogram parameters and positively with the concentration of homocysteine. These associations are characteristic for coronary artery disease and are not present in control subjects. The findings also indicate that in vitro aldosterone stimulates homocysteine production by rat adrenal glands, which may explain the associations observed with coronary artery disease. Moreover, we have found that aldosterone significantly modulates in vitro platelet reactivity to arachidonate and collagen - aldosterone increases the pro-aggregatory action of collagen, but decreases the pro-aggregatory potential of arachidonate. Therefore, the findings of these in vitro and ex vivo experiments indicate the existence of new pathways by which aldosterone modulates lipid- homocysteine- and platelet-dependent atherogenesis.
Collapse
Affiliation(s)
- K Karolczak
- Department of Hemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Lodz, Poland. or
| | | | | | | | | |
Collapse
|
41
|
Peng M, Yang M, Ding Y, Yu L, Deng Y, Lai W, Hu Y. Mechanism of endogenous digitalis-like factor‑induced vascular endothelial cell damage in patients with severe preeclampsia. Int J Mol Med 2017; 41:985-994. [PMID: 29251320 DOI: 10.3892/ijmm.2017.3316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 11/23/2017] [Indexed: 11/06/2022] Open
Abstract
Although endogenous digitalis‑like factor (EDLF) is associated with the development of various physical disorders, the role in preeclampsia remains unclear. This study investigated the effects of EDLF on vascular endothelial cell damage in patients with preeclampsia and the potential mechanisms. From July 2014 to July 2015, 120 singleton pregnancy cases underwent a prenatal examination, inpatient delivery and had normal blood pressure were included in the study, either as patients with severe preeclampsia or the control patients. Serum EDLF levels were compared in these two groups, and an in vitro hypoxic trophocyte‑induced vascular endothelial cell damage model was established to explore the changes in hypoxic trophocyte EDLF level and the subsequent effects on human umbilical vein endothelial cells (HUVECs). Nuclear factor‑κB (NF‑κB) p65 gene expression was silenced in hypoxic trophocytes, and EDLF levels and HUVEC damage were subsequently assessed. Serum EDLF levels were significantly higher in the severe preeclampsia cases than in the controls at the same gestational week (P<0.001). EDLF levels in hypoxic trophocytes increased with the increasing co‑culture duration. Damage to the biofunctions of HUVECs co‑cultured with hypoxic trophocytes also increased with co‑culture duration. However, silencing of NF‑κB p65 in the hypoxic trophocytes reduced the EDLF levels. Annexin A2 was highly expressed in HUVECs, and no biofunctions were significantly damaged (P<0.05) compared with the group without receiving NF‑κB p65 silencing. Serum EDLF levels were significantly higher in patients with severe preeclampsia compared with the controls. The results of the current study indicate that NF‑κB p65 has a role in regulating EDLF production in hypoxic trophocytes.
Collapse
Affiliation(s)
- Mei Peng
- Department of Gynaecology and Obstetrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Mengyuan Yang
- Department of Gynaecology and Obstetrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yiling Ding
- Department of Gynaecology and Obstetrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Ling Yu
- Department of Gynaecology and Obstetrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yali Deng
- Department of Gynaecology and Obstetrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Weisi Lai
- Department of Gynaecology and Obstetrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yun Hu
- Department of Gynaecology and Obstetrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
42
|
Hypoxia-induced suppression of c-Myc by HIF-2α in human pulmonary endothelial cells attenuates TFAM expression. Cell Signal 2017; 38:230-237. [PMID: 28709643 DOI: 10.1016/j.cellsig.2017.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 07/03/2017] [Accepted: 07/10/2017] [Indexed: 01/26/2023]
Abstract
The adaptive response to hypoxia is mediated in large part by stabilization of the hypoxia-inducible factors, HIF-1α and HIF-2α. A hallmark of this response is the metabolic shift to decreased oxidative phosphorylation and increased glycolysis. We hypothesized that hypoxic responses would include a suppression of mitochondrial gene expression. We determined the effects of hypoxia on TFAM, a key mitochondrial transcription factor, in normal pulmonary artery endothelial cells. Hypoxia decreased gene expression of TFAM and that of its upstream regulator, the transcriptional co-activator PGC1β. Although HIF-1α and HIF-2α pathways both contributed to hypoxia-mediated PGC1β suppression, TFAM suppression was regulated solely by HIF-2α-dependent mechanisms. We found that HIF-2α suppresses TFAM by decreasing c-Myc expression. In addition, we show a role for c-Jun in this pathway, linking HIF-2α with attenuation of c-Jun activation. Taken together, these findings establish a new link between HIF-2α and MAPK-signaling that mediates the adaptive regulation of mitochondrial gene expression under low oxygen tension.
Collapse
|
43
|
DuPont JJ, Jaffe IZ. 30 YEARS OF THE MINERALOCORTICOID RECEPTOR: The role of the mineralocorticoid receptor in the vasculature. J Endocrinol 2017; 234. [PMID: 28634267 PMCID: PMC5518626 DOI: 10.1530/joe-17-0009] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since the mineralocorticoid receptor (MR) was cloned 30 years ago, it has become clear that MR is expressed in extra-renal tissues, including the cardiovascular system, where it is expressed in all cells of the vasculature. Understanding the role of MR in the vasculature has been of particular interest as clinical trials show that MR antagonism improves cardiovascular outcomes out of proportion to changes in blood pressure. The last 30 years of research have demonstrated that MR is a functional hormone-activated transcription factor in vascular smooth muscle cells and endothelial cells. This review summarizes advances in our understanding of the role of vascular MR in regulating blood pressure and vascular function, and its contribution to vascular disease. Specifically, vascular MR contributes directly to blood pressure control and to vascular dysfunction and remodeling in response to hypertension, obesity and vascular injury. The literature is summarized with respect to the role of vascular MR in conditions including: pulmonary hypertension; cerebral vascular remodeling and stroke; vascular inflammation, atherosclerosis and myocardial infarction; acute kidney injury; and vascular pathology in the eye. Considerations regarding the impact of age and sex on the function of vascular MR are also described. Further investigation of the precise molecular mechanisms by which MR contributes to these processes will aid in the identification of novel therapeutic targets to reduce cardiovascular disease (CVD)-related morbidity and mortality.
Collapse
Affiliation(s)
- Jennifer J DuPont
- Molecular Cardiology Research InstituteTufts Medical Center, Boston, MA, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research InstituteTufts Medical Center, Boston, MA, USA
| |
Collapse
|
44
|
Alesutan I, Voelkl J, Feger M, Kratschmar DV, Castor T, Mia S, Sacherer M, Viereck R, Borst O, Leibrock C, Gawaz M, Kuro-O M, Pilz S, Tomaschitz A, Odermatt A, Pieske B, Wagner CA, Lang F. Involvement Of Vascular Aldosterone Synthase In Phosphate-Induced Osteogenic Transformation Of Vascular Smooth Muscle Cells. Sci Rep 2017; 7:2059. [PMID: 28515448 PMCID: PMC5435689 DOI: 10.1038/s41598-017-01882-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 04/04/2017] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification resulting from hyperphosphatemia is a major determinant of mortality in chronic kidney disease (CKD). Vascular calcification is driven by aldosterone-sensitive osteogenic transformation of vascular smooth muscle cells (VSMCs). We show that even in absence of exogenous aldosterone, silencing and pharmacological inhibition (spironolactone, eplerenone) of the mineralocorticoid receptor (MR) ameliorated phosphate-induced osteo-/chondrogenic transformation of primary human aortic smooth muscle cells (HAoSMCs). High phosphate concentrations up-regulated aldosterone synthase (CYP11B2) expression in HAoSMCs. Silencing and deficiency of CYP11B2 in VSMCs ameliorated phosphate-induced osteogenic reprogramming and calcification. Phosphate treatment was followed by nuclear export of APEX1, a CYP11B2 transcriptional repressor. APEX1 silencing up-regulated CYP11B2 expression and stimulated osteo-/chondrogenic transformation. APEX1 overexpression blunted the phosphate-induced osteo-/chondrogenic transformation and calcification of HAoSMCs. Cyp11b2 expression was higher in aortic tissue of hyperphosphatemic klotho-hypomorphic (kl/kl) mice than in wild-type mice. In adrenalectomized kl/kl mice, spironolactone treatment still significantly ameliorated aortic osteoinductive reprogramming. Our findings suggest that VSMCs express aldosterone synthase, which is up-regulated by phosphate-induced disruption of APEX1-dependent gene suppression. Vascular CYP11B2 may contribute to stimulation of VSMCs osteo-/chondrogenic transformation during hyperphosphatemia.
Collapse
Affiliation(s)
- Ioana Alesutan
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Jakob Voelkl
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany
| | - Martina Feger
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Denise V Kratschmar
- Department of Pharmaceutical Sciences, and the National Center for Excellence in Research NCCR Kidney.CH, University of Basel, Basel, Switzerland
| | - Tatsiana Castor
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Sobuj Mia
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Michael Sacherer
- Div. of Cardiology, Medical University of Graz and Ludwig Boltzmann Institute for Translational Heart Failure Research, Graz, Austria
| | - Robert Viereck
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Oliver Borst
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, Germany
| | | | - Meinrad Gawaz
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, Germany
| | - Makoto Kuro-O
- Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Stefan Pilz
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Medical University of Graz, Graz, Austria
| | - Andreas Tomaschitz
- Div. of Cardiology, Medical University of Graz and Ludwig Boltzmann Institute for Translational Heart Failure Research, Graz, Austria
- Bad Gleichenberg Clinic, Bad Gleichenberg, Austria
| | - Alex Odermatt
- Department of Pharmaceutical Sciences, and the National Center for Excellence in Research NCCR Kidney.CH, University of Basel, Basel, Switzerland
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Department of Cardiology, University of Graz, Graz, Austria; Department of Internal Medicine and Cardiology, German Heart Center Berlin (DHZB), Berlin, Germany
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, and the National Center for Excellence in Research NCCR Kidney, Zurich, Switzerland
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
45
|
Pugliese SC, Kumar S, Janssen WJ, Graham BB, Frid MG, Riddle SR, El Kasmi KC, Stenmark KR. A Time- and Compartment-Specific Activation of Lung Macrophages in Hypoxic Pulmonary Hypertension. THE JOURNAL OF IMMUNOLOGY 2017; 198:4802-4812. [PMID: 28500078 DOI: 10.4049/jimmunol.1601692] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 04/12/2017] [Indexed: 01/10/2023]
Abstract
Studies in various animal models suggest an important role for pulmonary macrophages in the pathogenesis of pulmonary hypertension (PH). Yet, the molecular mechanisms characterizing the functional macrophage phenotype relative to time and pulmonary localization and compartmentalization remain largely unknown. In this study, we used a hypoxic murine model of PH in combination with FACS to quantify and isolate lung macrophages from two compartments over time and characterize their programing via RNA sequencing approaches. In response to hypoxia, we found an early increase in macrophage number that was restricted to the interstitial/perivascular compartment, without recruitment of macrophages to the alveolar compartment or changes in the number of resident alveolar macrophages. Principal component analysis demonstrated significant differences in overall gene expression between alveolar and interstitial macrophages (IMs) at baseline and after 4 and 14 d hypoxic exposure. Alveolar macrophages at both day 4 and 14 and IMs at day 4 shared a conserved hypoxia program characterized by mitochondrial dysfunction, proinflammatory gene activation, and mTORC1 signaling, whereas IMs at day 14 demonstrated a unique anti-inflammatory/proreparative programming state. We conclude that the pathogenesis of vascular remodeling in hypoxic PH involves an early compartment-independent activation of lung macrophages toward a conserved hypoxia program, with the development of compartment-specific programs later in the course of the disease. Thus, harnessing time- and compartment-specific differences in lung macrophage polarization needs to be considered in the therapeutic targeting of macrophages in hypoxic PH and potentially other inflammatory lung diseases.
Collapse
Affiliation(s)
- Steven C Pugliese
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO 80045
| | - Sushil Kumar
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - William J Janssen
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO 80045.,Department of Medicine, National Jewish Health, Denver, CO 80206
| | - Brian B Graham
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO 80045
| | - Maria G Frid
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Suzette R Riddle
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Karim C El Kasmi
- Division of Gastroenterology, Hepatology, and Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045;
| |
Collapse
|
46
|
Transcription factors regulate GPR91-mediated expression of VEGF in hypoxia-induced retinopathy. Sci Rep 2017; 7:45807. [PMID: 28374767 PMCID: PMC5379554 DOI: 10.1038/srep45807] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/03/2016] [Indexed: 12/28/2022] Open
Abstract
Hypoxia is the most important factor in the pathogenesis of diabetic retinopathy (DR). Our previous studies demonstrated that G protein-coupled receptor 91(GPR91) participated in the regulation of vascular endothelial growth factor (VEGF) secretion in DR. The present study induced OIR model in newborn rats using exposure to alternating 24-hour episodes of 50% and 12% oxygen for 14 days. Treatment with GPR91 shRNA attenuated the retinal avascular area, abnormal neovascularization and pericyte loss. Western blot and qRT-PCR demonstrated that CoCl2 exposure promoted VEGF expression and secretion, activated the ERK1/2 signaling pathways and upregulated C/EBP and AP-1. Knockdown of GPR91 inhibited ERK1/2 activity. GPR91 siRNA transduction and the ERK1/2 inhibitor U0126 inhibited the increases in C/EBP β, C/EBP δ, c-Fos and HIF-1α. Luciferase reporter assays and a chromatin immunoprecipitation (ChIP) assay demonstrated that C/EBP β and c-Fos bound the functional transcriptional factor binding site in the region of the VEGF promoter, but not C/EBP δ. Knockdown of C/EBP β and c-Fos using RNAi reduced VEGF expression. Our data suggest that activation of the GPR91-ERK1/2-C/EBP β (c-Fos, HIF-1α) signaling pathway plays a tonic role in regulating VEGF transcription in rat retinal ganglion cells.
Collapse
|
47
|
Riley CJ, Gavin M. Physiological Changes to the Cardiovascular System at High Altitude and Its Effects on Cardiovascular Disease. High Alt Med Biol 2017; 18:102-113. [PMID: 28294639 DOI: 10.1089/ham.2016.0112] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Riley, Callum James, and Matthew Gavin. Physiological changes to the cardiovascular system at high altitude and its effects on cardiovascular disease. High Alt Med Biol. 18:102-113, 2017.-The physiological changes to the cardiovascular system in response to the high altitude environment are well understood. More recently, we have begun to understand how these changes may affect and cause detriment to cardiovascular disease. In addition to this, the increasing availability of altitude simulation has dramatically improved our understanding of the physiology of high altitude. This has allowed further study on the effect of altitude in those with cardiovascular disease in a safe and controlled environment as well as in healthy individuals. Using a thorough PubMed search, this review aims to integrate recent advances in cardiovascular physiology at altitude with previous understanding, as well as its potential implications on cardiovascular disease. Altogether, it was found that the changes at altitude to cardiovascular physiology are profound enough to have a noteworthy effect on many forms of cardiovascular disease. While often asymptomatic, there is some risk in high altitude exposure for individuals with certain cardiovascular diseases. Although controlled research in patients with cardiovascular disease was largely lacking, meaning firm conclusions cannot be drawn, these risks should be a consideration to both the individual and their physician.
Collapse
Affiliation(s)
| | - Matthew Gavin
- 2 University of Leeds School of Biomedical Sciences , Leeds, United Kingdom
| |
Collapse
|
48
|
Maron BA, Machado RF, Shimoda L. Pulmonary vascular and ventricular dysfunction in the susceptible patient (2015 Grover Conference series). Pulm Circ 2016; 6:426-438. [PMID: 28090285 PMCID: PMC5210067 DOI: 10.1086/688315] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/21/2016] [Indexed: 12/14/2022] Open
Abstract
Pulmonary blood vessel structure and tone are maintained by a complex interplay between endogenous vasoactive factors and oxygen-sensing intermediaries. Under physiological conditions, these signaling networks function as an adaptive interface between the pulmonary circulation and environmental or acquired perturbations to preserve oxygenation and maintain systemic delivery of oxygen-rich hemoglobin. Chronic exposure to hypoxia, however, triggers a range of pathogenetic mechanisms that include hypoxia-inducible factor 1α (HIF-1α)-dependent upregulation of the vasoconstrictor peptide endothelin 1 in pulmonary endothelial cells. In pulmonary arterial smooth muscle cells, chronic hypoxia induces HIF-1α-mediated upregulation of canonical transient receptor potential proteins, as well as increased Rho kinase-Ca2+ signaling and pulmonary arteriole synthesis of the profibrotic hormone aldosterone. Collectively, these mechanisms contribute to a contractile or hypertrophic pulmonary vascular phenotype. Genetically inherited disorders in hemoglobin structure are also an important etiology of abnormal pulmonary vasoreactivity. In sickle cell anemia, for example, consumption of the vasodilator and antimitogenic molecule nitric oxide by cell-free hemoglobin is an important mechanism underpinning pulmonary hypertension. Contemporary genomic and transcriptomic analytic methods have also allowed for the discovery of novel risk factors relevant to sickle cell disease, including GALNT13 gene variants. In this report, we review cutting-edge observations characterizing these and other pathobiological mechanisms that contribute to pulmonary vascular and right ventricular vulnerability.
Collapse
Affiliation(s)
- Bradley A. Maron
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA; and Department of Cardiology, Boston Veterans Affairs Healthcare System, Boston, Massachusetts, USA
| | - Roberto F. Machado
- Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Larissa Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
49
|
Huetsch JC, Suresh K, Bernier M, Shimoda LA. Update on novel targets and potential treatment avenues in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2016; 311:L811-L831. [PMID: 27591245 PMCID: PMC5130539 DOI: 10.1152/ajplung.00302.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 08/29/2016] [Indexed: 02/08/2023] Open
Abstract
Pulmonary hypertension (PH) is a condition marked by a combination of constriction and remodeling within the pulmonary vasculature. It remains a disease without a cure, as current treatments were developed with a focus on vasodilatory properties but do not reverse the remodeling component. Numerous recent advances have been made in the understanding of cellular processes that drive pathologic remodeling in each layer of the vessel wall as well as the accompanying maladaptive changes in the right ventricle. In particular, the past few years have yielded much improved insight into the pathways that contribute to altered metabolism, mitochondrial function, and reactive oxygen species signaling and how these pathways promote the proproliferative, promigratory, and antiapoptotic phenotype of the vasculature during PH. Additionally, there have been significant advances in numerous other pathways linked to PH pathogenesis, such as sex hormones and perivascular inflammation. Novel insights into cellular pathology have suggested new avenues for the development of both biomarkers and therapies that will hopefully bring us closer to the elusive goal: a therapy leading to reversal of disease.
Collapse
Affiliation(s)
- John C Huetsch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; and
| | - Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; and
| | - Meghan Bernier
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; and
| |
Collapse
|
50
|
Jaisser F, Farman N. Emerging Roles of the Mineralocorticoid Receptor in Pathology: Toward New Paradigms in Clinical Pharmacology. Pharmacol Rev 2016; 68:49-75. [PMID: 26668301 DOI: 10.1124/pr.115.011106] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The mineralocorticoid receptor (MR) and its ligand aldosterone are the principal modulators of hormone-regulated renal sodium reabsorption. In addition to the kidney, there are several other cells and organs expressing MR, in which its activation mediates pathologic changes, indicating potential therapeutic applications of pharmacological MR antagonism. Steroidal MR antagonists have been used for decades to fight hypertension and more recently heart failure. New therapeutic indications are now arising, and nonsteroidal MR antagonists are currently under development. This review is focused on nonclassic MR targets in cardiac, vascular, renal, metabolic, ocular, and cutaneous diseases. The MR, associated with other risk factors, is involved in organ fibrosis, inflammation, oxidative stress, and aging; for example, in the kidney and heart MR mediates hormonal tissue-specific ion channel regulation. Genetic and epigenetic modifications of MR expression/activity that have been documented in hypertension may also present significant risk factors in other diseases and be susceptible to MR antagonism. Excess mineralocorticoid signaling, mediated by aldosterone or glucocorticoids binding, now appears deleterious in the progression of pathologies that may lead to end-stage organ failure and could therefore benefit from the repositioning of pharmacological MR antagonists.
Collapse
Affiliation(s)
- F Jaisser
- INSERM UMR 1138 Team 1, Cordeliers Research Center, Pierre et Marie Curie University, Paris, France (F.J., N.F); and University Paris-Est Creteil, Creteil, France (F.J.)
| | - N Farman
- INSERM UMR 1138 Team 1, Cordeliers Research Center, Pierre et Marie Curie University, Paris, France (F.J., N.F); and University Paris-Est Creteil, Creteil, France (F.J.)
| |
Collapse
|