51
|
Anjum H, Surani S. Pulmonary Hypertension in Pregnancy: A Review. ACTA ACUST UNITED AC 2021; 57:medicina57030259. [PMID: 33799910 PMCID: PMC8000005 DOI: 10.3390/medicina57030259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 02/05/2023]
Abstract
Pulmonary hypertension (PH) is a disease, which targets the pulmonary vasculature affecting the heart and the lungs, and is characterized by a vast array of signs and symptoms. These manifestations of PH in pregnancy are highly variable and non-specific hence, it is prudent to have a very keen and high index of suspicion while evaluating these patients. This rare disease can be extremely debilitating and can be associated with a poor overall prognosis. Pregnancy in women with PH puts them at an elevated risk because the physiological changes associated with pregnancy are not well endured leading to even higher morbidity and mortality in these patients. Although there are various modalities for evaluation and workup of PH, right heart catheterization (RHC) remains the gold standard. A mean pulmonary artery pressure (PAP) of more than 20 mm of Hg is considered diagnostic. It is indeed heartening to see that in the past decade many novel therapeutic modalities have emerged and along with a better understanding of the disease process have proved to be promising in terms of reducing the adverse outcomes and preventing death in this population of patients.
Collapse
Affiliation(s)
- Humayun Anjum
- Internal Medicine, University of North Texas, Fort Worth, TX 76107, USA
- Correspondence:
| | - Salim Surani
- Internal Medicine, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
52
|
Radchenko GD, Sirenko YM. Prognostic Significance of Systemic Arterial Stiffness Evaluated by Cardio-Ankle Vascular Index in Patients with Idiopathic Pulmonary Hypertension. Vasc Health Risk Manag 2021; 17:77-93. [PMID: 33731998 PMCID: PMC7957228 DOI: 10.2147/vhrm.s294767] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/27/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND In a previous study, the cardio-ankle vascular index (CAVI) was increased significantly in idiopathic pulmonary arterial hypertension (IPAH) patients compared to the healthy group and did not much differ from one in systemic hypertensives. In this study the relations between survival and CAVI was evaluated in patients with IPAH. PATIENTS AND METHODS We included 89 patients with new-diagnosed IPAH without concomitant diseases. Standard examinations, including right heart catheterization (RHC) and systemic arterial stiffness evaluation, were performed. All patients were divided according to CAVI value: the group with CAVI ≥ 8 (n = 18) and the group with CAVI < 8 (n = 71). The mean follow-up was 33.8 ± 23.7 months. Kaplan-Meier and Cox regression analysis were performed for the evaluation of our cohort survival and the predictors of death. RESULTS The group with CAVI≥8 was older and more severe compared to the group with CAVI< 8. Patients with CAVI≥8 had significantly reduced end-diastolic (73.79±18.94 vs 87.35±16.69 mL, P<0.009) and end-systolic (25.71±9.56 vs 33.55±10.33 mL, P<0.01) volumes of the left ventricle, the higher right ventricle thickness (0.77±0.12 vs 0.62±0.20 mm, P < 0.006), and the lower TAPSE (13.38±2.15 vs 15.98±4.4 mm, P<0.018). RHC data did not differ significantly between groups, except the higher level of the right atrial pressure in patients with CAVI≥ 8-11.38±7.1 vs 8.76±4.7 mmHg, P<0.08. The estimated overall survival rate was 61.2%. The CAVI≥8 increased the risk of mortality 2.34 times (CI 1.04-5.28, P = 0.041). The estimated Kaplan-Meier survival in the patients with CAVI ≥ 8 was only 46.7 ± 7.18% compared to patients with CAVI < 8 - 65.6 ± 4.2%, P = 0.035. At multifactorial regression analysis, the CAVI reduced but saved its relevance as death predictor - OR = 1.13, CI 1.001-1.871. SUMMARY We suggested the CAVI could be a new independent predictor of death in the IPAH population and could be used to better risk stratify this patient population if CAVI is validated as a marker in a larger multicenter trial.
Collapse
Affiliation(s)
- Ganna D Radchenko
- Department of Symptomatic Hypertension, “National Scientific Center “The M.D. Strazhesko Institute of Cardiology”” of National Academy of Medical Science, Kyiv, Ukraine
| | - Yuriy M Sirenko
- Department of Symptomatic Hypertension, “National Scientific Center “The M.D. Strazhesko Institute of Cardiology”” of National Academy of Medical Science, Kyiv, Ukraine
| |
Collapse
|
53
|
Type I interferon activation and endothelial dysfunction in caveolin-1 insufficiency-associated pulmonary arterial hypertension. Proc Natl Acad Sci U S A 2021; 118:2010206118. [PMID: 33836561 DOI: 10.1073/pnas.2010206118] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Interferonopathies, interferon (IFN)-α/β therapy, and caveolin-1 (CAV1) loss-of-function have all been associated with pulmonary arterial hypertension (PAH). Here, CAV1-silenced primary human pulmonary artery endothelial cells (PAECs) were proliferative and hypermigratory, with reduced cytoskeletal stress fibers. Signal transducers and activators of transcription (STAT) and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) were both constitutively activated in these cells, resulting in a type I IFN-biased inflammatory signature. Cav1 -/- mice that spontaneously develop pulmonary hypertension were found to have STAT1 and AKT activation in lung homogenates and increased circulating levels of CXCL10, a hallmark of IFN-mediated inflammation. PAH patients with CAV1 mutations also had elevated serum CXCL10 levels and their fibroblasts mirrored phenotypic and molecular features of CAV1-deficient PAECs. Moreover, immunofluorescence staining revealed endothelial CAV1 loss and STAT1 activation in the pulmonary arterioles of patients with idiopathic PAH, suggesting that this paradigm might not be limited to rare CAV1 frameshift mutations. While blocking JAK/STAT or AKT rescued aspects of CAV1 loss, only AKT inhibitors suppressed activation of both signaling pathways simultaneously. Silencing endothelial nitric oxide synthase (NOS3) prevented STAT1 and AKT activation induced by CAV1 loss, implicating CAV1/NOS3 uncoupling and NOS3 dysregulation in the inflammatory phenotype. Exogenous IFN reduced CAV1 expression, activated STAT1 and AKT, and altered the cytoskeleton of PAECs, implicating these mechanisms in PAH associated with autoimmune and autoinflammatory diseases, as well as IFN therapy. CAV1 insufficiency elicits an IFN inflammatory response that results in a dysfunctional endothelial cell phenotype and targeting this pathway may reduce pathologic vascular remodeling in PAH.
Collapse
|
54
|
Abstract
Pulmonary arterial hypertension (PAH) is characterized by impaired regulation of pulmonary hemodynamics and vascular growth. Alterations of metabolism and bioenergetics are increasingly recognized as universal hallmarks of PAH, as metabolic abnormalities are identified in lungs and hearts of patients, animal models of the disease, and cells derived from lungs of patients. Mitochondria are the primary organelle critically mediating the complex and integrative metabolic pathways in bioenergetics, biosynthetic pathways, and cell signaling. Here, we review the alterations in metabolic pathways that are linked to the pathologic vascular phenotype of PAH, including abnormalities in glycolysis and glucose oxidation, fatty acid oxidation, glutaminolysis, arginine metabolism, one-carbon metabolism, the reducing and oxidizing cell environment, and the tricarboxylic acid cycle, as well as the effects of PAH-associated nuclear and mitochondrial mutations on metabolism. Understanding of the metabolic mechanisms underlying PAH provides important knowledge for the design of new therapeutics for treatment of patients.
Collapse
Affiliation(s)
- Weiling Xu
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA;
| | - Allison J Janocha
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA;
| | - Serpil C Erzurum
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA; .,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
55
|
Abstract
PURPOSE OF REVIEW In 2013, the association between T-Box factor 4 (TBX4) variants and pulmonary arterial hypertension (PAH) has first been described. Now - in 2020 - growing evidence is emerging indicating that TBX4 variants associate with a wide spectrum of lung disorders. RECENT FINDINGS TBX4 variants are enriched in both children and adults with PAH. The clinical phenotype associated with a TBX4 variant seems to be milder than that in other PAH-associated gene mutations. Further, TBX4 variants have increasingly been associated with a variety of clinical and histopathological phenotypes, including lethal developmental parenchymal lung diseases such as not only acinar dysplasia in neonates, but also less outspoken parenchymal lung diseases in children and adults. SUMMARY The clinical phenotype of a TBX4 variant has recently been recognised to expand from bone disorders to different types of lung diseases. Recent data suggest that variants of TBX4, a transcription factor known to be an important regulator in embryonic development, are not rare in both children and adults with PAH and/or developmental parenchymal lung diseases.
Collapse
|
56
|
Wang XJ, Xu XQ, Sun K, Liu KQ, Li SQ, Jiang X, Zhao QH, Wang L, Peng FH, Ye J, Wu Y, Jiang R, Zhang J, Huang W, Wei WB, Yan Y, Li JH, Liu QQ, Li S, Wang Y, Zhang SY, Zhang X, Jing ZC. Association of Rare PTGIS Variants With Susceptibility and Pulmonary Vascular Response in Patients With Idiopathic Pulmonary Arterial Hypertension. JAMA Cardiol 2021; 5:677-684. [PMID: 32236489 PMCID: PMC7113838 DOI: 10.1001/jamacardio.2020.0479] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Question What is the novel susceptibility gene for idiopathic pulmonary arterial hypertension? Findings In this 2-stage genetic association study of 230 patients with idiopathic pulmonary arterial hypertension, heterozygous rare PTGIS variants were first found significantly overrepresented in 6.1%, conferring 7.8 higher odds of pulmonary arterial hypertension. In addition, patients carrying rare PTGIS variants were more responsive to iloprost stimulation than those without such variants. Meaning The rare variants of the PTGIS gene appear to contribute higher susceptibility to idiopathic pulmonary arterial hypertension, and screening of PTGIS variants may help improve personalized treatment of these patients. Importance Idiopathic pulmonary arterial hypertension (IPAH) is a fatal disease with high heritability; however, the bone morphogenetic protein receptor 2 (BMPR2) gene only accounts for 17% of IPAH. The genetic basis of IPAH needs further investigation. Objective To identify novel IPAH susceptibility genes other than BMPR2. Design, Setting, and Participants This 2-stage, case-control genetic association study enrolled 230 patients with IPAH from 2 referral pulmonary hypertension centers in China. Eligible patients had no BMPR2 variants and were compared with 968 healthy control participants. Data were collected from January 1, 2000, to July 31, 2015, and analyzed from August 1, 2015, to May 30, 2018. Exposures PTGIS rare variants. Main Outcomes and Measures Whole-genome sequencing was performed to identify putative IPAH genes in a discovery cohort, with validation in an independent referral cohort. Correlation of genotype and hemodynamic characteristics was then evaluated at baseline and after pulmonary vasodilator testing. Functional assessments were conducted to analyze the effects of identified genetic variants on transcript splicing, enzymatic activity, and endothelial cell phenotypes. Results Among 230 patients with IPAH (164 female [71.3%]; mean [SD] age, 34 [18] years), an enrichment of rare variants in a gene encoding prostacyclin synthase (PTGIS) was identified in the discovery cohort. The association of PTGIS rare variants with IPAH was confirmed in the replication cohort. In the combined data set, PTGIS rare variants were found in 14 of 230 cases (6.1%) and 8 of 968 controls (0.8%) (odds ratio, 7.8; 95% CI, 3.2-18.8; P = 5 × 10−6, logistic regression). Compared with patients without PTGIS variants, inhaled iloprost induced a more significant decrease of pulmonary vascular resistance (difference in the least square mean, −21.7%; 95% CI, −31.4% to −12.0%; P < .001, linear regression model) and an increase of cardiac index (difference in the least square mean, 18.3%; 95% CI, 8.8%-27.8%; P < .001, linear regression model) in patients with PTGIS variants. The minigene assay indicated that the c.521 + 1G>A variant resulted in aberrant messenger RNA transcripts. The functional studies showed that the 2 missense rare variants (R252Q and A447T) resulted in a decrease in prostacyclin production and increased cell death of pulmonary microvascular endothelial cells. Conclusions and Relevance This study identified 3 rare loss-of-function variants in the PTGIS gene from 2 independent cohorts with IPAH. The genetic variants of PTGIS predispose pulmonary vascular responses to the iloprost stimulation. These findings suggest that PTGIS variants may be involved in the pathogenesis of IPAH.
Collapse
Affiliation(s)
- Xiao-Jian Wang
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xi-Qi Xu
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
| | - Kai Sun
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
| | - Ke-Qiang Liu
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Su-Qi Li
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Jiang
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
| | - Qin-Hua Zhao
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lan Wang
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fu-Hua Peng
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jue Ye
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Wu
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Jiang
- Ministry of Education (MOE) Key Laboratory of Bioinformatics, Bioinformatics Division, Beijing National Research Center for Information Science and Technology, Department of Automation, Tsinghua University, Beijing, China
| | - Jin Zhang
- Cardiovascular Research Center, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, China
| | - Wei Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Wen-Bin Wei
- Cardiovascular Center, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yi Yan
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Hui Li
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian-Qian Liu
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sheng Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Yong Wang
- Department of Respiratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shu-Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
| | - Xue Zhang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhi-Cheng Jing
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
57
|
Abstract
Cellular senescence is a feature of most somatic cells. It is characterized by an irreversible cell cycle arrest and by the ability to secrete a plethora of mediators of inflammation and growth factors, which can alter the senescent cell's microenvironment. Senescent cells accumulate in tissues over time and contribute to both aging and the development of age-associated diseases. Senescent cells have antagonistic pleiotropic roles in cancer. Given the inability of senescent cells to proliferate, cellular senescence is a powerful tumor suppressor mechanism in young individuals. However, accumulation of senescent stromal cells during aging can fuel cancer cell growth in virtue of their capacity to release factors that stimulate cell proliferation. Caveolin-1 is a structural protein component of caveolae, invaginations of the plasma membrane involved in a variety of cellular processes, including signal transduction. Mounting evidence over the last 10-15 years has demonstrated a central role of caveolin-1 in the development of a senescent phenotype and the regulation of both the anti-tumorigenic and pro-tumorigenic properties of cellular senescence. In this review, we discuss the cellular mechanisms and functions of caveolin-1 in the context of cellular senescence and their relevance to the biology of cancer.
Collapse
|
58
|
Rai N, Shihan M, Seeger W, Schermuly RT, Novoyatleva T. Genetic Delivery and Gene Therapy in Pulmonary Hypertension. Int J Mol Sci 2021; 22:ijms22031179. [PMID: 33503992 PMCID: PMC7865388 DOI: 10.3390/ijms22031179] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive complex fatal disease of multiple etiologies. Hyperproliferation and resistance to apoptosis of vascular cells of intimal, medial, and adventitial layers of pulmonary vessels trigger excessive pulmonary vascular remodeling and vasoconstriction in the course of pulmonary arterial hypertension (PAH), a subgroup of PH. Multiple gene mutation/s or dysregulated gene expression contribute to the pathogenesis of PAH by endorsing the proliferation and promoting the resistance to apoptosis of pulmonary vascular cells. Given the vital role of these cells in PAH progression, the development of safe and efficient-gene therapeutic approaches that lead to restoration or down-regulation of gene expression, generally involved in the etiology of the disease is the need of the hour. Currently, none of the FDA-approved drugs provides a cure against PH, hence innovative tools may offer a novel treatment paradigm for this progressive and lethal disorder by silencing pathological genes, expressing therapeutic proteins, or through gene-editing applications. Here, we review the effectiveness and limitations of the presently available gene therapy approaches for PH. We provide a brief survey of commonly existing and currently applicable gene transfer methods for pulmonary vascular cells in vitro and describe some more recent developments for gene delivery existing in the field of PH in vivo.
Collapse
Affiliation(s)
- Nabham Rai
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Mazen Shihan
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ralph T. Schermuly
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Tatyana Novoyatleva
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Correspondence:
| |
Collapse
|
59
|
Abstract
Advances in high-throughput biotechnologies have facilitated omics profiling, a key component of precision phenotyping, in patients with pulmonary vascular disease. Omics provides comprehensive information pertaining to genes, transcripts, proteins, and metabolites. The resulting omics big datasets may be integrated for more robust results and are amenable to analysis using machine learning or newer analytical methodologies, such as network analysis. Results from fully integrated multi-omics datasets combined with clinical data are poised to provide novel insight into pulmonary vascular disease as well as diagnose the presence of disease and prognosticate outcomes.
Collapse
Affiliation(s)
- Jane A Leopold
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB0630K, Boston, MA 02115, USA.
| | - Anna R Hemnes
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, T1218 Medical Center North, 1161 21st Avenue South, Nashville, TN 37232, USA
| |
Collapse
|
60
|
Kurakula K, Smolders VFED, Tura-Ceide O, Jukema JW, Quax PHA, Goumans MJ. Endothelial Dysfunction in Pulmonary Hypertension: Cause or Consequence? Biomedicines 2021; 9:biomedicines9010057. [PMID: 33435311 PMCID: PMC7827874 DOI: 10.3390/biomedicines9010057] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/30/2020] [Accepted: 01/03/2021] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare, complex, and progressive disease that is characterized by the abnormal remodeling of the pulmonary arteries that leads to right ventricular failure and death. Although our understanding of the causes for abnormal vascular remodeling in PAH is limited, accumulating evidence indicates that endothelial cell (EC) dysfunction is one of the first triggers initiating this process. EC dysfunction leads to the activation of several cellular signalling pathways in the endothelium, resulting in the uncontrolled proliferation of ECs, pulmonary artery smooth muscle cells, and fibroblasts, and eventually leads to vascular remodelling and the occlusion of the pulmonary blood vessels. Other factors that are related to EC dysfunction in PAH are an increase in endothelial to mesenchymal transition, inflammation, apoptosis, and thrombus formation. In this review, we outline the latest advances on the role of EC dysfunction in PAH and other forms of pulmonary hypertension. We also elaborate on the molecular signals that orchestrate EC dysfunction in PAH. Understanding the role and mechanisms of EC dysfunction will unravel the therapeutic potential of targeting this process in PAH.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Cell and Chemical Biology, Laboratory for CardioVascular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Valérie F. E. D. Smolders
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.F.E.D.S.); (P.H.A.Q.)
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain;
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institut (IDIBGI), 17190 Girona, Catalonia, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Paul H. A. Quax
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.F.E.D.S.); (P.H.A.Q.)
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Laboratory for CardioVascular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Correspondence:
| |
Collapse
|
61
|
Swietlik EM, Greene D, Zhu N, Megy K, Cogliano M, Rajaram S, Pandya D, Tilly T, Lutz KA, Welch CC, Pauciulo MW, Southgate L, Martin JM, Treacy CM, Penkett CJ, Stephens JC, Bogaard HJ, Church C, Coghlan G, Coleman AW, Condliffe R, Eichstaedt CA, Eyries M, Gall H, Ghio S, Girerd B, Grünig E, Holden S, Howard L, Humbert M, Kiely DG, Kovacs G, Lordan J, Machado RD, MacKenzie Ross RV, McCabe C, Moledina S, Montani D, Olschewski H, Pepke-Zaba J, Price L, Rhodes CJ, Seeger W, Soubrier F, Suntharalingam J, Toshner MR, Vonk Noordegraaf A, Wharton J, Wild JM, Wort SJ, Lawrie A, Wilkins MR, Trembath RC, Shen Y, Chung WK, Swift AJ, Nichols WC, Morrell NW, Gräf S. Bayesian Inference Associates Rare KDR Variants with Specific Phenotypes in Pulmonary Arterial Hypertension. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2020; 14. [PMID: 33320693 PMCID: PMC7892262 DOI: 10.1161/circgen.120.003155] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 11/29/2020] [Indexed: 11/26/2022]
Abstract
Background - Approximately 25% of patients with pulmonary arterial hypertension (PAH) have been found to harbor rare mutations in disease-causing genes. To identify missing heritability in PAH we integrated deep phenotyping with whole-genome sequencing data using Bayesian statistics. Methods - We analyzed 13,037 participants enrolled in the NIHR BioResource - Rare Diseases (NBR) study, of which 1,148 were recruited to the PAH domain. To test for genetic associations between genes and selected phenotypes of pulmonary hypertension (PH), we used the Bayesian rare-variant association method BeviMed. Results - Heterozygous, high impact, likely loss-of-function variants in the Kinase Insert Domain Receptor (KDR) gene were strongly associated with significantly reduced transfer coefficient for carbon monoxide (KCO, posterior probability (PP)=0.989) and older age at diagnosis (PP=0.912). We also provide evidence for familial segregation of a rare nonsense KDR variant with these phenotypes. On computed tomographic imaging of the lungs, a range of parenchymal abnormalities were observed in the five patients harboring these predicted deleterious variants in KDR. Four additional PAH cases with rare likely loss-of-function variants in KDR were independently identified in the US PAH Biobank cohort with similar phenotypic characteristics. Conclusions - The Bayesian inference approach allowed us to independently validate KDR, which encodes for the Vascular Endothelial Growth Factor Receptor 2 (VEGFR2), as a novel PAH candidate gene. Furthermore, this approach specifically associated high impact likely loss-of-function variants in the genetically constrained gene with distinct phenotypes. These findings provide evidence for KDR being a clinically actionable PAH gene and further support the central role of the vascular endothelium in the pathobiology of PAH.
Collapse
Affiliation(s)
- Emilia M. Swietlik
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
| | - Daniel Greene
- Department of Haematology (D.G., K.M., C.J.P., J.C.S., S. Gräf), University of Cambridge
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
| | - Na Zhu
- Department of Pediatrics (N.Z., C.C.L.W.), Columbia University, NY
- Department of Systems Biology (N.Z., Y.S.), Columbia University, NY
| | - Karyn Megy
- Department of Haematology (D.G., K.M., C.J.P., J.C.S., S. Gräf), University of Cambridge
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
| | - Marcella Cogliano
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield (M.C., J.M.W., A.L., A.J.S.)
| | - Smitha Rajaram
- Sheffield Teaching Hospitals NHS Foundation Trust, United Kingdom (S.R.)
| | - Divya Pandya
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
| | - Tobias Tilly
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
| | - Katie A. Lutz
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center (K.A.L., M.W.P., A.W.C., W.C.N.)
| | | | - Michael W. Pauciulo
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center (K.A.L., M.W.P., A.W.C., W.C.N.)
- Department of Pediatrics, University of Cincinnati College of Medicine, OH (M.W.P., W.C.N.)
| | - Laura Southgate
- Molecular & Clinical Sciences Research Institute, St George’s, University of London, United Kingdom (L.S., R.D.M.)
| | - Jennifer M. Martin
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
| | - Carmen M. Treacy
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
| | - Christopher J. Penkett
- Department of Haematology (D.G., K.M., C.J.P., J.C.S., S. Gräf), University of Cambridge
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
| | - Jonathan C. Stephens
- Department of Haematology (D.G., K.M., C.J.P., J.C.S., S. Gräf), University of Cambridge
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
| | - Harm J. Bogaard
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands (H.J.B., A.V.N.)
| | - Colin Church
- Golden Jubilee National Hospital, Glasgow (C.C.)
| | | | - Anna W. Coleman
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center (K.A.L., M.W.P., A.W.C., W.C.N.)
| | - Robin Condliffe
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (R.C., D.G.K.)
| | - Christina A. Eichstaedt
- Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University (C.A.E.)
- Center for Pulmonary Hypertension, Thoraxklinik gGmbH Heidelberg at Heidelberg University Hospital (C.A.E., E.G.)
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany (C.A.E., E.G.)
| | - Mélanie Eyries
- Département de génétique, hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris & UMR_S 1166-ICAN, INSERM, UPMC Sorbonne Universités, Paris, France (M.E., F.S.)
| | - Henning Gall
- University of Giessen & Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL) and of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany (H.G., W.S.)
| | - Stefano Ghio
- Fondazione IRCCS Policlinico San Matteo, Pavia, Italy (S. Ghio)
| | - Barbara Girerd
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay (B.G., M.H., D.M.)
- AP-HP, Service de Pneumologie, Centre de référence de l’hypertension pulmonaire (B.G., M.H., D.M.)
- INSERM UMR_S 999, Hôpital Bicêtre, Le Kremlin-Bicêtre, Paris, France (B.G., M.H., D.M.)
| | - Ekkehard Grünig
- Center for Pulmonary Hypertension, Thoraxklinik gGmbH Heidelberg at Heidelberg University Hospital (C.A.E., E.G.)
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany (C.A.E., E.G.)
| | - Simon Holden
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge (S.H., N.W.M.)
| | - Luke Howard
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
| | - Marc Humbert
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay (B.G., M.H., D.M.)
- AP-HP, Service de Pneumologie, Centre de référence de l’hypertension pulmonaire (B.G., M.H., D.M.)
- INSERM UMR_S 999, Hôpital Bicêtre, Le Kremlin-Bicêtre, Paris, France (B.G., M.H., D.M.)
| | - David G. Kiely
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (R.C., D.G.K.)
| | - Gabor Kovacs
- Ludwig Boltzmann Institute for Lung Vascular Research (G.K., H.O.)
- Medical University of Graz, Austria (G.K., H.O.)
| | - Jim Lordan
- Freeman Hospital, Newcastle upon Tyne (J.L.)
| | - Rajiv D. Machado
- Molecular & Clinical Sciences Research Institute, St George’s, University of London, United Kingdom (L.S., R.D.M.)
| | | | - Colm McCabe
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
- Royal Brompton & Harefield NHS Foundation Trust (C.M., L.P., S.J.W.)
| | | | - David Montani
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay (B.G., M.H., D.M.)
- AP-HP, Service de Pneumologie, Centre de référence de l’hypertension pulmonaire (B.G., M.H., D.M.)
- INSERM UMR_S 999, Hôpital Bicêtre, Le Kremlin-Bicêtre, Paris, France (B.G., M.H., D.M.)
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research (G.K., H.O.)
- Medical University of Graz, Austria (G.K., H.O.)
| | - Joanna Pepke-Zaba
- Royal Papworth Hospital NHS Foundation Trust (J.P.-Z., M.R.T., N.W.M.)
| | - Laura Price
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
- Royal Brompton & Harefield NHS Foundation Trust (C.M., L.P., S.J.W.)
| | - Christopher J. Rhodes
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
| | - Werner Seeger
- University of Giessen & Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL) and of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany (H.G., W.S.)
| | - Florent Soubrier
- Département de génétique, hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris & UMR_S 1166-ICAN, INSERM, UPMC Sorbonne Universités, Paris, France (M.E., F.S.)
| | | | - Mark R. Toshner
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
- Royal Papworth Hospital NHS Foundation Trust (J.P.-Z., M.R.T., N.W.M.)
| | - Anton Vonk Noordegraaf
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands (H.J.B., A.V.N.)
| | - John Wharton
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
| | - James M. Wild
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield (M.C., J.M.W., A.L., A.J.S.)
| | - Stephen John Wort
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
- Royal Brompton & Harefield NHS Foundation Trust (C.M., L.P., S.J.W.)
| | | | | | | | - Allan Lawrie
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield (M.C., J.M.W., A.L., A.J.S.)
| | - Martin R. Wilkins
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
| | - Richard C. Trembath
- Department of Medical & Molecular Genetics, King’s College London, United Kingdom (R.C.T.)
| | - Yufeng Shen
- Department of Systems Biology (N.Z., Y.S.), Columbia University, NY
- Department of Biomedical Informatics (Y.S.), Columbia University, NY
| | | | - Andrew J. Swift
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield (M.C., J.M.W., A.L., A.J.S.)
| | - William C. Nichols
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center (K.A.L., M.W.P., A.W.C., W.C.N.)
- Department of Pediatrics, University of Cincinnati College of Medicine, OH (M.W.P., W.C.N.)
| | - Nicholas W. Morrell
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge (S.H., N.W.M.)
- Royal Papworth Hospital NHS Foundation Trust (J.P.-Z., M.R.T., N.W.M.)
| | - Stefan Gräf
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
- Department of Haematology (D.G., K.M., C.J.P., J.C.S., S. Gräf), University of Cambridge
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
| |
Collapse
|
62
|
Han B, Porta JC, Hanks JL, Peskova Y, Binshtein E, Dryden K, Claxton DP, Mchaourab HS, Karakas E, Ohi MD, Kenworthy AK. Structure and assembly of CAV1 8S complexes revealed by single particle electron microscopy. SCIENCE ADVANCES 2020; 6:6/49/eabc6185. [PMID: 33268374 PMCID: PMC7821874 DOI: 10.1126/sciadv.abc6185] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/16/2020] [Indexed: 05/21/2023]
Abstract
Highly stable oligomeric complexes of the monotopic membrane protein caveolin serve as fundamental building blocks of caveolae. Current evidence suggests these complexes are disc shaped, but the details of their structural organization and how they assemble are poorly understood. Here, we address these questions using single particle electron microscopy of negatively stained recombinant 8S complexes of human caveolin 1. We show that 8S complexes are toroidal structures ~15 nm in diameter that consist of an outer ring, an inner ring, and central protruding stalk. Moreover, we map the position of the N and C termini and determine their role in complex assembly, and visualize the 8S complexes in heterologous caveolae. Our findings provide critical insights into the structural features of 8S complexes and allow us to propose a model for how these highly stable membrane-embedded complexes are generated.
Collapse
Affiliation(s)
- Bing Han
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jason C Porta
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jessica L Hanks
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Yelena Peskova
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Elad Binshtein
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Kelly Dryden
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Derek P Claxton
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Hassane S Mchaourab
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Erkan Karakas
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Melanie D Ohi
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Anne K Kenworthy
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA.
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
63
|
Swietlik EM, Prapa M, Martin JM, Pandya D, Auckland K, Morrell NW, Gräf S. 'There and Back Again'-Forward Genetics and Reverse Phenotyping in Pulmonary Arterial Hypertension. Genes (Basel) 2020; 11:E1408. [PMID: 33256119 PMCID: PMC7760524 DOI: 10.3390/genes11121408] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Although the invention of right heart catheterisation in the 1950s enabled accurate clinical diagnosis of pulmonary arterial hypertension (PAH), it was not until 2000 when the landmark discovery of the causative role of bone morphogenetic protein receptor type II (BMPR2) mutations shed new light on the pathogenesis of PAH. Since then several genes have been discovered, which now account for around 25% of cases with the clinical diagnosis of idiopathic PAH. Despite the ongoing efforts, in the majority of patients the cause of the disease remains elusive, a phenomenon often referred to as "missing heritability". In this review, we discuss research approaches to uncover the genetic architecture of PAH starting with forward phenotyping, which in a research setting should focus on stable intermediate phenotypes, forward and reverse genetics, and finally reverse phenotyping. We then discuss potential sources of "missing heritability" and how functional genomics and multi-omics methods are employed to tackle this problem.
Collapse
Affiliation(s)
- Emilia M. Swietlik
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- Royal Papworth Hospital NHS Foundation Trust, Cambridge CB2 0AY, UK
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Matina Prapa
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Jennifer M. Martin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
| | - Divya Pandya
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
| | - Kathryn Auckland
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
| | - Nicholas W. Morrell
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- Royal Papworth Hospital NHS Foundation Trust, Cambridge CB2 0AY, UK
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
- NIHR BioResource for Translational Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- NIHR BioResource for Translational Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| |
Collapse
|
64
|
At the X-Roads of Sex and Genetics in Pulmonary Arterial Hypertension. Genes (Basel) 2020; 11:genes11111371. [PMID: 33233517 PMCID: PMC7699559 DOI: 10.3390/genes11111371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 11/16/2022] Open
Abstract
Group 1 pulmonary hypertension (pulmonary arterial hypertension; PAH) is a rare disease characterized by remodeling of the small pulmonary arteries leading to progressive elevation of pulmonary vascular resistance, ultimately leading to right ventricular failure and death. Deleterious mutations in the serine-threonine receptor bone morphogenetic protein receptor 2 (BMPR2; a central mediator of bone morphogenetic protein (BMP) signaling) and female sex are known risk factors for the development of PAH in humans. In this narrative review, we explore the complex interplay between the BMP and estrogen signaling pathways, and the potentially synergistic mechanisms by which these signaling cascades increase the risk of developing PAH. A comprehensive understanding of these tangled pathways may reveal therapeutic targets to prevent or slow the progression of PAH.
Collapse
|
65
|
Hemnes A, Rothman AMK, Swift AJ, Zisman LS. Role of biomarkers in evaluation, treatment and clinical studies of pulmonary arterial hypertension. Pulm Circ 2020; 10:2045894020957234. [PMID: 33282185 PMCID: PMC7682212 DOI: 10.1177/2045894020957234] [Citation(s) in RCA: 14] [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: 06/11/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022] Open
Abstract
Pulmonary arterial hypertension is a complex disease resulting from the interplay of myriad biological and environmental processes that lead to remodeling of the pulmonary vasculature with consequent pulmonary hypertension. Despite currently available therapies, there remains significant morbidity and mortality in this disease. There is great interest in identifying and applying biomarkers to help diagnose patients with pulmonary arterial hypertension, inform prognosis, guide therapy, and serve as surrogate endpoints. An extensive literature on potential biomarker candidates is available, but barriers to the implementation of biomarkers for clinical use in pulmonary arterial hypertension are substantial. Various omic strategies have been undertaken to identify key pathways regulated in pulmonary arterial hypertension that could serve as biomarkers including genomic, transcriptomic, proteomic, and metabolomic approaches. Other biologically relevant components such as circulating cells, microRNAs, exosomes, and cell-free DNA have recently been gaining attention. Because of the size of the datasets generated by these omic approaches and their complexity, artificial intelligence methods are being increasingly applied to decipher their meaning. There is growing interest in imaging the lung with various modalities to understand and visualize processes in the lung that lead to pulmonary vascular remodeling including high resolution computed tomography, Xenon magnetic resonance imaging, and positron emission tomography. Such imaging modalities have the potential to demonstrate disease modification resulting from therapeutic interventions. Because right ventricular function is a major determinant of prognosis, imaging of the right ventricle with echocardiography or cardiac magnetic resonance imaging plays an important role in the evaluation of patients and may also be useful in clinical studies of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Anna Hemnes
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Andrew J Swift
- University of Sheffield and Sheffield Teaching Hospitals NHS Trust, Sheffield, UK
| | | |
Collapse
|
66
|
Yaoita N, Satoh K, Satoh T, Shimizu T, Saito S, Sugimura K, Tatebe S, Yamamoto S, Aoki T, Kikuchi N, Kurosawa R, Miyata S, Nagasaki M, Yasuda J, Shimokawa H. Identification of the Novel Variants in Patients With Chronic Thromboembolic Pulmonary Hypertension. J Am Heart Assoc 2020; 9:e015902. [PMID: 33103541 PMCID: PMC7763425 DOI: 10.1161/jaha.120.015902] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 08/26/2020] [Indexed: 11/26/2022]
Abstract
Background Although chronic thromboembolic pulmonary hypertension (CTEPH) and acute pulmonary embolism (APE) share some clinical manifestations, a limited proportion of patients with CTEPH have a history of APE. Moreover, in histopathologic studies, it has been revealed that pulmonary vasculature lesions similar to pulmonary arterial hypertension existed in patients with CTEPH. Thus, it remains unknown whether these 3 disorders also share genetic backgrounds. Methods and Results Whole exome screening was performed with DNA isolated from 51 unrelated patients with CTEPH of Japanese ancestry. The frequency of genetic variants associated with pulmonary arterial hypertension or APE in patients with CTEPH was compared with those in the integrative Japanese Genome Variation Database 3.5KJPN. Whole exome screening analysis showed 17 049 nonsynonymous variants in patients with CTEPH. Although we found 6 nonsynonymous variants that are associated with APE in patients with CTEPH, there was no nonsynonymous variant associated with pulmonary arterial hypertension. Patients with CTEPH with a history of APE had nonsynonymous variants of F5, which encodes factor V. In contrast, patients with CTEPH without a history of APE had a nonsynonymous variant of THBD, which encodes thrombomodulin. Moreover, thrombin-activatable fibrinolysis inhibitor, which is one of the pathogenic proteins in CTEPH, was significantly more activated in those who had the variants of THBD compared with those without it. Conclusions These results provide the first evidence that patients with CTEPH have some variants associated with APE, regardless of the presence or absence of a history of APE. Furthermore, the variants might be different between patients with CTEPH with and without a history of APE.
Collapse
Affiliation(s)
- Nobuhiro Yaoita
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Kimio Satoh
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Taijyu Satoh
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Toru Shimizu
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Sakae Saito
- Department of Integrative GenomicsTohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Koichiro Sugimura
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Shunsuke Tatebe
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Saori Yamamoto
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Tatsuo Aoki
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Nobuhiro Kikuchi
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Ryo Kurosawa
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Satoshi Miyata
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Masao Nagasaki
- Department of Integrative GenomicsTohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Jun Yasuda
- Department of Integrative GenomicsTohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Hiroaki Shimokawa
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
67
|
Sharma S, Aldred MA. DNA Damage and Repair in Pulmonary Arterial Hypertension. Genes (Basel) 2020; 11:genes11101224. [PMID: 33086628 PMCID: PMC7603366 DOI: 10.3390/genes11101224] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex multifactorial disease with both genetic and environmental dynamics contributing to disease progression. Over the last decade, several studies have demonstrated the presence of genomic instability and increased levels of DNA damage in PAH lung vascular cells, which contribute to their pathogenic apoptosis-resistant and proliferating characteristics. In addition, the dysregulated DNA damage response pathways have been indicated as causal factors for the presence of persistent DNA damage. To understand the significant implications of DNA damage and repair in PAH pathogenesis, the current review summarizes the recent advances made in this field. This includes an overview of the observed DNA damage in the nuclear and mitochondrial genome of PAH patients. Next, the irregularities observed in various DNA damage response pathways and their role in accumulating DNA damage, escaping apoptosis, and proliferation under a DNA damaging environment are discussed. Although the current literature establishes the pertinence of DNA damage in PAH, additional studies are required to understand the temporal sequence of the above-mentioned events. Further, an exploration of different types of DNA damage in conjunction with associated impaired DNA damage response in PAH will potentially stimulate early diagnosis of the disease and development of novel therapeutic strategies.
Collapse
|
68
|
Haarman MG, Kerstjens-Frederikse WS, Vissia-Kazemier TR, Breeman KTN, Timens W, Vos YJ, Roofthooft MTR, Hillege HL, Berger RMF. The Genetic Epidemiology of Pediatric Pulmonary Arterial Hypertension. J Pediatr 2020; 225:65-73.e5. [PMID: 32502478 DOI: 10.1016/j.jpeds.2020.05.051] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/27/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To describe the prevalence of pulmonary arterial hypertension (PAH)-associated gene mutations, and other genetic characteristics in a national cohort of children with PAH from the Dutch National registry and to explore genotype-phenotype associations and outcomes. STUDY DESIGN Children (n = 70) diagnosed with idiopathic PAH, heritable PAH, PAH associated with congenital heart disease with coincidental shunt (PAH-congenital heart disease group 3), PAH after closure of a cardiac shunt (PAH-congenital heart disease group 4), or PAH associated with other noncardiac conditions were enrolled. Targeted next-generation sequencing was performed on PAH-associated genes (BMPR2, ACVRL1, EIF2AK4, CAV1, ENG, KCNK3, SMAD9, and TBX4). Also, children were tested for specific genetic disorders in case of clinical suspicion. Additionally, children were tested for copy number variations. RESULTS Nineteen children (27%) had a PAH-associated gene mutation/variant: BMPR2 n = 7, TBX4 n = 8, ACVRL1 n = 1, KCNK3 n = 1, and EIF2AK4 n = 2. Twelve children (17%) had a genetic disorder with an established association with PAH (including trisomy 21 and cobalamin C deficiency). In another 16 children (23%), genetic disorders without an established association with PAH were identified (including Noonan syndrome, Beals syndrome, and various copy number variations). Survival rates differed between groups and was most favorable in TBX4 variant carriers. CONCLUSIONS Children with PAH show a high prevalence of genetic disorders, not restricted to established PAH-associated genes. Genetic architecture could play a role in risk-stratified care management in pediatric PAH.
Collapse
Affiliation(s)
- Meindina G Haarman
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, the Netherlands.
| | | | - Theresia R Vissia-Kazemier
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, the Netherlands
| | - Karel T N Breeman
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, the Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
| | - Yvonne J Vos
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marc T R Roofthooft
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, the Netherlands
| | - Hans L Hillege
- Department of Epidemiology, University Medical Center Groningen, Groningen, the Netherlands; Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Rolf M F Berger
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
69
|
Customized Massive Parallel Sequencing Panel for Diagnosis of Pulmonary Arterial Hypertension. Genes (Basel) 2020; 11:genes11101158. [PMID: 33007923 PMCID: PMC7650688 DOI: 10.3390/genes11101158] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023] Open
Abstract
Pulmonary arterial hypertension is a very infrequent disease, with a variable etiology and clinical expressivity, making sometimes the clinical diagnosis a challenge. Current classification based on clinical features does not reflect the underlying molecular profiling of these groups. The advance in massive parallel sequencing in PAH has allowed for the describing of several new causative and susceptibility genes related to PAH, improving overall patient diagnosis. In order to address the molecular diagnosis of patients with PAH we designed, validated, and routinely applied a custom panel including 21 genes. Three hundred patients from the National Spanish PAH Registry (REHAP) were included in the analysis. A custom script was developed to annotate and filter the variants. Variant classification was performed according to the ACMG guidelines. Pathogenic and likely pathogenic variants have been found in 15% of the patients with 12% of variants of unknown significance (VUS). We have found variants in patients with connective tissue disease (CTD) and congenital heart disease (CHD). In addition, in a small proportion of patients (1.75%), we observed a possible digenic mode of inheritance. These results stand out the importance of the genetic testing of patients with associated forms of PAH (i.e., CHD and CTD) additionally to the classical IPAH and HPAH forms. Molecular confirmation of the clinical presumptive diagnosis is required in cases with a high clinical overlapping to carry out proper management and follow up of the individuals with the disease.
Collapse
|
70
|
Rathinasabapathy A, Copeland C, Crabtree A, Carrier EJ, Moore C, Shay S, Gladson S, Austin ED, Kenworthy AK, Loyd JE, Hemnes AR, West JD. Expression of a Human Caveolin-1 Mutation in Mice Drives Inflammatory and Metabolic Defect-Associated Pulmonary Arterial Hypertension. Front Med (Lausanne) 2020; 7:540. [PMID: 33015095 PMCID: PMC7516012 DOI: 10.3389/fmed.2020.00540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022] Open
Abstract
Background: In 2012, mutations in Cav1 were found to be the driving mutation in several cases of heritable pulmonary arterial hypertension (PAH). These mutations replaced the last 21 amino acids of Cav1 with a novel 22-amino-acid sequence. Because previously only Cav1 knockouts had been studied in the context of PAH, examining the in vivo effects of this novel mutation holds promise for new understanding of the role of Cav1 in disease etiology. Methods: The new 22 amino acids created by the human mutation were knocked into the native mouse Cav1 locus. The mice underwent hemodynamic, energy balance, and inflammatory measurements, both at baseline and after being stressed with either a metabolic or an inflammatory challenge [low-dose lipopolysaccharide (LPS)]. To metabolically challenge the mice, they were injected with streptozotocin (STZ) and fed a high-fat diet for 12 weeks. Results: Very little mutant protein was found in vivo (roughly 2% of wild-type by mass spectrometry), probably because of degradation after failure to traffic from the endoplasmic reticulum. The homozygous mutants developed a mild, low-penetrance PAH similar to that described previously in knockouts, and neither baseline nor metabolic nor inflammatory stress resulted in pressures above normal in heterozygous animals. The homozygous mutants had increased lean mass and worsened oral glucose tolerance, as previously described in knockouts. Novel findings include the preservation of Cav2 and accessory proteins in the liver and the kidney, while they are lost with homozygous Cav1 mutation in the lungs. We also found that the homozygous mutants had a significantly lower tolerance to voluntary spontaneous exercise than the wild-type mice, with the heterozygous mice at an intermediate level. The mutants also had higher circulating monocytes, with both heterozygous and homozygous animals having higher pulmonary MCP1 and MCP5 proteins. The heterozygous animals also lost weight at an LPS challenge level at which the wild-type mice continued to gain weight. Conclusions: The Cav1 mutation identified in human patients in 2012 is molecularly similar to a knockout of Cav1. It results in not only metabolic deficiencies and mild pulmonary hypertension, as expected, but also an inflammatory phenotype and reduced spontaneous exercise.
Collapse
Affiliation(s)
| | - Courtney Copeland
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Amber Crabtree
- Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Erica J Carrier
- Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Christy Moore
- Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sheila Shay
- Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Santhi Gladson
- Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Eric D Austin
- Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Anne K Kenworthy
- Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - James E Loyd
- Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Anna R Hemnes
- Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James D West
- Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
71
|
Mutgan AC, Jandl K, Kwapiszewska G. Endothelial Basement Membrane Components and Their Products, Matrikines: Active Drivers of Pulmonary Hypertension? Cells 2020; 9:cells9092029. [PMID: 32899187 PMCID: PMC7563239 DOI: 10.3390/cells9092029] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a vascular disease that is characterized by elevated pulmonary arterial pressure (PAP) due to progressive vascular remodeling. Extracellular matrix (ECM) deposition in pulmonary arteries (PA) is one of the key features of vascular remodeling. Emerging evidence indicates that the basement membrane (BM), a specialized cluster of ECM proteins underlying the endothelium, may be actively involved in the progression of vascular remodeling. The BM and its steady turnover are pivotal for maintaining appropriate vascular functions. However, the pathologically elevated turnover of BM components leads to an increased release of biologically active short fragments, which are called matrikines. Both BM components and their matrikines can interfere with pivotal biological processes, such as survival, proliferation, adhesion, and migration and thus may actively contribute to endothelial dysfunction. Therefore, in this review, we summarize the emerging role of the BM and its matrikines on the vascular endothelium and further discuss its implications on lung vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Ayse Ceren Mutgan
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, 8010 Graz, Austria;
| | - Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, 8010 Graz, Austria;
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
- Correspondence:
| |
Collapse
|
72
|
Badlam JB, Badesch DB, Austin ED, Benza RL, Chung WK, Farber HW, Feldkircher K, Frost AE, Poms AD, Lutz KA, Pauciulo MW, Yu C, Nichols WC, Elliott CG. United States Pulmonary Hypertension Scientific Registry: Baseline Characteristics. Chest 2020; 159:311-327. [PMID: 32858008 DOI: 10.1016/j.chest.2020.07.088] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The treatment, genotyping, and phenotyping of patients with World Health Organization Group 1 pulmonary arterial hypertension (PAH) have evolved dramatically in the last decade. RESEARCH QUESTION The United States Pulmonary Hypertension Scientific Registry was established as the first US PAH patient registry to investigate genetic information, reproductive histories, and environmental exposure data in a contemporary patient population. STUDY DESIGN AND METHODS Investigators at 15 US centers enrolled consecutively screened adults diagnosed with Group 1 PAH who had enrolled in the National Biological Sample and Data Repository for PAH (PAH Biobank) within 5 years of a cardiac catheterization demonstrating qualifying hemodynamic criteria. Exposure and reproductive histories were collected by using a structured interview and questionnaire. The biobank provided genetic data. RESULTS Between 2015 and 2018, a total of 499 of 979 eligible patients with clinical diagnoses of idiopathic PAH (IPAH) or familial PAH (n = 240 [48%]), associated PAH (APAH; n = 256 [51%]), or pulmonary venoocclusive disease/pulmonary capillary hemangiomatosis (n = 3 [1%]) enrolled. The mean age was 55.8 years, average BMI was 29.2 kg/m2, and 79% were women. Mean duration between symptom onset and diagnostic catheterization was 1.9 years. Sixty-six percent of patients were treated with more than one PAH medication at enrollment. Past use of prescription weight loss drugs (16%), recreational drugs (27%), and oral contraceptive pills (77%) was common. Women often reported miscarriage (37%), although PAH was rarely diagnosed within 6 months of pregnancy (1.9%). Results of genetic testing identified pathogenic or suspected pathogenic variants in 13% of patients, reclassifying 18% of IPAH patients and 5% of APAH patients to heritable PAH. INTERPRETATION Patients with Group 1 PAH remain predominately middle-aged women diagnosed with IPAH or APAH. Delays in diagnosis of PAH persist. Treatment with combinations of PAH-targeted medications is more common than in the past. Women often report pregnancy complications, as well as exposure to anorexigens, oral contraceptives, and/or recreational drugs. Results of genetic tests frequently identify unsuspected heritable PAH.
Collapse
Affiliation(s)
| | | | - Eric D Austin
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Raymond L Benza
- Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH
| | - Wendy K Chung
- Division of Clinical Genetics, Department of Pediatrics, Columbia University Medical Center, New York, NY
| | | | | | - Adaani E Frost
- Houston Methodist Hospital, Institute for Academic Medicine & Houston Methodist Research Institute, Houston, TX
| | - Abby D Poms
- E Squared Trials and Registries, Inc., Half Moon Bay, CA
| | - Katie A Lutz
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Chang Yu
- Department of Biostatistics, Vanderbilt University, Nashville, TN
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - C Gregory Elliott
- Intermountain Medical Center, Department of Medicine and the University of Utah, Pulmonary Division, Salt Lake City, UT
| | | |
Collapse
|
73
|
Filippini A, D’Alessio A. Caveolae and Lipid Rafts in Endothelium: Valuable Organelles for Multiple Functions. Biomolecules 2020; 10:biom10091218. [PMID: 32825713 PMCID: PMC7563503 DOI: 10.3390/biom10091218] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/22/2022] Open
Abstract
Caveolae are flask-shaped invaginations of the plasma membrane found in numerous cell types and are particularly abundant in endothelial cells and adipocytes. The lipid composition of caveolae largely matches that of lipid rafts microdomains that are particularly enriched in cholesterol, sphingomyelin, glycosphingolipids, and saturated fatty acids. Unlike lipid rafts, whose existence remains quite elusive in living cells, caveolae can be clearly distinguished by electron microscope. Despite their similar composition and the sharing of some functions, lipid rafts appear more heterogeneous in terms of size and are more dynamic than caveolae. Following the discovery of caveolin-1, the first molecular marker as well as the unique scaffolding protein of caveolae, we have witnessed a remarkable increase in studies aimed at investigating the role of these organelles in cell functions and human disease. The goal of this review is to discuss the most recent studies related to the role of caveolae and caveolins in endothelial cells. We first recapitulate the major embryological processes leading to the formation of the vascular tree. We next discuss the contribution of caveolins and cavins to membrane biogenesis and cell response to extracellular stimuli. We also address how caveolae and caveolins control endothelial cell metabolism, a central mechanism involved in migration proliferation and angiogenesis. Finally, as regards the emergency caused by COVID-19, we propose to study the caveolar platform as a potential target to block virus entry into endothelial cells.
Collapse
Affiliation(s)
- Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Roma, Italy;
| | - Alessio D’Alessio
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Roma, Italia
- Correspondence:
| |
Collapse
|
74
|
Ballash GA, Schober KE, Haw SR, Shilo K, Jennings RN. Idiopathic pulmonary arterial hypertension in a pot-bellied pig (Sus scrofa domesticus) with right-sided congestive heart failure. J Vet Cardiol 2020; 31:1-7. [PMID: 32836069 DOI: 10.1016/j.jvc.2020.07.001] [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: 08/16/2019] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 11/25/2022]
Abstract
Idiopathic pulmonary arterial hypertension is a rare disease reported in humans and dogs diagnosed as persistent elevation of pulmonary arterial blood pressure without predisposing or associated diseases. A four-month-old pot-bellied pig (Sus scrofa domesticus) was presented for decreased appetite, lethargy, respiratory distress, and occasional syncope. On physical examination, the pig was tachypneic with labored breathing, with a distended abdomen and a bilateral grade 4-5/6 parasternal systolic heart murmur. Systolic pulmonary arterial pressure was estimated at 95 mmHg by Doppler echocardiography, consistent with severe pulmonary hypertension. At autopsy, there was dilation of the main pulmonary artery and right ventricle. The lungs were diffusely rubbery, and there was tricavitary effusion. Microscopically, there was severe widespread pulmonary arterial concentric medial hypertrophy with rare plexiform lesions. The clinical history and gross and microscopic findings supported a diagnosis of idiopathic pulmonary arterial hypertension with subsequent right-sided congestive heart failure. Primary (idiopathic) pulmonary arterial hypertension should be considered as a differential diagnosis in young pigs with right-sided congestive heart failure.
Collapse
Affiliation(s)
- G A Ballash
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - K E Schober
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - S R Haw
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - K Shilo
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - R N Jennings
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
75
|
The BMP Receptor 2 in Pulmonary Arterial Hypertension: When and Where the Animal Model Matches the Patient. Cells 2020; 9:cells9061422. [PMID: 32521690 PMCID: PMC7348993 DOI: 10.3390/cells9061422] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/30/2020] [Accepted: 06/05/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Mutations in bone morphogenetic protein receptor type II (BMPR2) are leading to the development of hereditary pulmonary arterial hypertension (PAH). In non-hereditary forms of PAH, perturbations in the transforming growth factor-β (TGF-β)/BMP-axis are believed to cause deficient BMPR2 signaling by changes in receptor expression, the activity of the receptor and/or downstream signaling. To date, BMPR2 expression and its activity in the lungs of patients with non-hereditary PAH is poorly characterized. In recent decades, different animal models have been used to understand the role of BMPR2 signaling in PAH pathophysiology. Specifically, the monocrotaline (MCT) and Sugen-Hypoxia (SuHx) models are extensively used in interventional studies to examine if restoring BMPR2 signaling results in PAH disease reversal. While PAH is assumed to develop in patients over months or years, pulmonary hypertension in experimental animal models develops in days or weeks. It is therefore likely that modifications in BMP and TGF-β signaling in these models do not fully recapitulate those in patients. In order to determine the translational potential of the MCT and SuHx models, we analyzed the BMPR2 expression and activity in the lungs of rats with experimentally induced PAH and compared this to the BMPR2 expression and activity in the lungs of PAH patients. Methods: the BMPR2 expression was analyzed by Western blot analysis and immunofluorescence (IF) microscopy to determine the quantity and localization of the receptor in the lung tissue from normal control subjects and patients with hereditary or idiopathic PAH, as well as in the lungs of control rats and rats with MCT or SuHx-induced PAH. The activation of the BMP pathway was analyzed by determining the level and localization of phosphorylated Smad1/5/8 (pSmad 1/5/8), a downstream mediator of canonical BMPR2 signaling. Results: While BMPR2 and pSmad 1/5/8 expression levels were unaltered in whole lung lysates/homogenates from patients with hereditary and idiopathic PAH, IF analysis showed that BMPR2 and pSmad 1/5/8 levels were markedly decreased in the pulmonary vessels of both PAH patient groups. Whole lung BMPR2 expression was variable in the two PAH rat models, while in both experimental models the expression of BMPR2 in the lung vasculature was increased. However, in the human PAH lungs, the expression of pSmad 1/5/8 was downregulated in the lung vasculature of both experimental models. Conclusion: BMPR2 receptor expression and downstream signaling is reduced in the lung vasculature of patients with idiopathic and hereditary PAH, which cannot be appreciated when using human whole lung lysates. Despite increased BMPR2 expression in the lung vasculature, the MCT and SuHx rat models did develop PAH and impaired downstream BMPR2-Smad signaling similar to our findings in the human lung.
Collapse
|
76
|
Thomas CA, Anderson RJ, Condon DF, de Jesus Perez VA. Diagnosis and Management of Pulmonary Hypertension in the Modern Era: Insights from the 6th World Symposium. Pulm Ther 2020; 6:9-22. [PMID: 32048239 PMCID: PMC7229067 DOI: 10.1007/s41030-019-00105-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Indexed: 01/01/2023] Open
Abstract
The past 20 years have seen major advances in the diagnosis and management of pulmonary hypertension, a disease associated with significant morbidity and mortality. The 6th World Symposium in Pulmonary Hypertension (WSPH) took place in February 2018 and attempted to consolidate the current knowledge in the field into practical recommendations to help prioritize an action plan to improve patient outcomes and identify future research directions. In this review, we will summarize the highlights of the 6th WSPH proceedings, including revisions to the hemodynamic definitions and classification of the various types of pulmonary hypertension, genetic advances, approaches to risk stratification, and updated treatment algorithms.
Collapse
Affiliation(s)
- Christopher A Thomas
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Ryan J Anderson
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - David F Condon
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Vinicio A de Jesus Perez
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA.
- Vera Moulton Wall Center for Pulmonary Vascular Research, Stanford University, Stanford, CA, USA.
| |
Collapse
|
77
|
Novel Molecular Mechanisms of Pulmonary Hypertension: A Search for Biomarkers and Novel Drug Targets-From Bench to Bed Site. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7265487. [PMID: 32566097 PMCID: PMC7261339 DOI: 10.1155/2020/7265487] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/11/2020] [Indexed: 12/23/2022]
Abstract
Pulmonary hypertension (PH) is defined as increased mean pulmonary artery pressure (mPAP) above 25 mmHg, measured at rest by right heart catheterization. The exact global prevalence of PH is difficult to estimate, mainly due to the complex aetiology, and its spread may be underestimated. To date, numerous studies on the aetiology and pathophysiology of PH at molecular level were conducted. Simultaneously, some clinical studies have shown potential usefulness of well-known and widely recognized cardiovascular biomarkers, but their potential clinical usefulness in diagnosis and management of PH is poor due to their low specificity accompanied with numerous other cardiovascular comorbidities of PH subjects. On the other hand, a large body of basic research-based studies provides us with novel molecular pathomechanisms, biomarkers, and drug targets, according to the evidence-based medicine principles. Unfortunately, the simple implementation of these results to clinical practice is impossible due to a large heterogeneity of the PH pathophysiology, where the clinical symptoms constitute only a common denominator and a final result of numerous crosstalking metabolic pathways. Therefore, future studies, based mostly on translational medicine, are needed in order to both organize better the pathophysiological classification of various forms of PH and define precisely the optimal diagnostic markers and therapeutic targets in particular forms of PH. This review paper summarizes the current state of the art regarding the molecular background of PH with respect to its current classification. Novel therapeutic strategies and potential biomarkers are discussed with respect to their limitations in use in common clinical practice.
Collapse
|
78
|
Gorr MW, Sriram K, Muthusamy A, Insel PA. Transcriptomic analysis of pulmonary artery smooth muscle cells identifies new potential therapeutic targets for idiopathic pulmonary arterial hypertension. Br J Pharmacol 2020; 177:3505-3518. [PMID: 32337710 DOI: 10.1111/bph.15074] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/25/2020] [Accepted: 04/17/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Pulmonary arterial hypertension (PAH, type 1 pulmonary hypertension) has a 3-year survival of ~50% and is in need of new, effective therapies. In PAH, remodelling of the pulmonary artery (PA) increases pulmonary vascular resistance and can result in right heart dysfunction and failure. Genetic mutations can cause PAH but it can also be idiopathic (IPAH). Enhanced contractility and proliferation of PA smooth muscle cells (PASMCs) are key contributors to the pathophysiology of PAH, but the underlying mechanisms are not well understood. EXPERIMENTAL APPROACH We utilized RNA-sequencing (RNA-seq) of IPAH and control patient-derived PASMCs as an unbiased approach to define differentially expressed (DE) genes that may identify new biology and potential therapeutic targets. KEY RESULTS Analysis of DE genes for shared gene pathways revealed increases in genes involved in cell proliferation and mitosis and decreases in a variety of gene sets, including response to cytokine signalling. ADGRG6/GPR126, an adhesion G protein-coupled receptor (GPCR), was increased in IPAH-PASMCs compared to control-PASMCs. Increased expression of this GPCR in control-PASMCs decreased their proliferation; siRNA knockdown of ADGRG6/GPR126 in IPAH-PASMCs tended to increase proliferation. CONCLUSION AND IMPLICATIONS These data provide insights regarding the expression of current and experimental PAH drug targets, GPCRs and GPCR-related genes as potentially new therapeutic targets in PAH-PASMCs. Overall, the findings identify genes and pathways that may contribute to IPAH-PASMC function and suggest that ADGRG6/GPR126 is a novel therapeutic target for IPAH.
Collapse
Affiliation(s)
- Matthew W Gorr
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA.,Colleges of Nursing and Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Krishna Sriram
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Abinaya Muthusamy
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Paul A Insel
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
79
|
Swietlik EM, Gräf S, Morrell NW. The role of genomics and genetics in pulmonary arterial hypertension. Glob Cardiol Sci Pract 2020; 2020:e202013. [PMID: 33150157 PMCID: PMC7590931 DOI: 10.21542/gcsp.2020.13] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Emilia M Swietlik
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Addenbrooke's Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Royal Papworth Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.,NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Addenbrooke's Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Royal Papworth Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom.,NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
80
|
Eyries M, Montani D, Girerd B, Favrolt N, Riou M, Faivre L, Manaud G, Perros F, Gräf S, Morrell NW, Humbert M, Soubrier F. Familial pulmonary arterial hypertension by KDR heterozygous loss of function. Eur Respir J 2020; 55:13993003.02165-2019. [PMID: 31980491 DOI: 10.1183/13993003.02165-2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/02/2020] [Indexed: 11/05/2022]
Abstract
Beyond the major gene BMPR2, several new genes predisposing to PAH have been identified during the last decade. Recently, preliminary evidence of the involvement of the KDR gene was found in a large genetic association study.We prospectively analysed the KDR gene by targeted panel sequencing in a series of 311 PAH patients referred to a clinical molecular laboratory for genetic diagnosis of PAH.Two index cases with severe PAH from two different families were found to carry a loss-of-function mutation in the KDR gene. These two index cases were clinically characterised by low diffusing capacity for carbon monoxide adjusted for haemoglobin (D LCOc) and interstitial lung disease. In one family, segregation analysis revealed that variant carriers are either presenting with PAH associated with low D LCOc, or have only decreased D LCOc, whereas non-carrier relatives have normal D LCOc. In the second family, a single affected carrier was alive. His carrier mother was unaffected with normal D LCOc.We provided genetic evidence for considering KDR as a newly identified PAH-causing gene by describing the segregation of KDR mutations with PAH in two families. In our study, KDR mutations are associated with a particular form of PAH characterised by low D LCOc and radiological evidence of parenchymal lung disease including interstitial lung disease and emphysema.
Collapse
Affiliation(s)
- Mélanie Eyries
- Hôpital Pitié-Salpêtrière, Département de génétique, Assistance Publique-Hôpitaux de Paris, Paris, France.,UMR_S1166-ICAN, Sorbonne Université, INSERM, Paris, France.,Equally contributing authors
| | - David Montani
- Univ. Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France.,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France.,UMR_S 999, Univ. Paris-Sud, INSERM, Hôpital Marie Lannelongue, Le Plessis Robinson, France.,Equally contributing authors
| | - Barbara Girerd
- Univ. Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France.,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Nicolas Favrolt
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de référence constitutif des maladies pulmonaires rares de l'adulte, Centre de compétence de l'hypertension pulmonaire, CHU Dijon-Bourgogne, Dijon, France
| | - Marianne Riou
- Service de pneumologie, Nouvel hôpital civil, Strasbourg, France
| | - Laurence Faivre
- Centre de génétique, FHU TRANSLAD, Institut GIMI et UMR INSERM 1231, CHU de Dijon et Université de Bourgogne, Dijon, France
| | - Grégoire Manaud
- UMR_S 999, Univ. Paris-Sud, INSERM, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Frédéric Perros
- UMR_S 999, Univ. Paris-Sud, INSERM, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Stefan Gräf
- NIHR Bioresource - Rare Diseases, Cambridge Biomedical Campus, Cambridge, UK.,Dept of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.,Dept of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Nicholas W Morrell
- NIHR Bioresource - Rare Diseases, Cambridge Biomedical Campus, Cambridge, UK.,Dept of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Marc Humbert
- Univ. Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France.,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France.,UMR_S 999, Univ. Paris-Sud, INSERM, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Florent Soubrier
- Hôpital Pitié-Salpêtrière, Département de génétique, Assistance Publique-Hôpitaux de Paris, Paris, France .,UMR_S1166-ICAN, Sorbonne Université, INSERM, Paris, France
| |
Collapse
|
81
|
Liu B, Zhu L, Yuan P, Marsboom G, Hong Z, Liu J, Zhang P, Hu Q. Comprehensive identification of signaling pathways for idiopathic pulmonary arterial hypertension. Am J Physiol Cell Physiol 2020; 318:C913-C930. [PMID: 32159364 DOI: 10.1152/ajpcell.00382.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Whole exome sequencing (WES) was used in the research of familial pulmonary arterial hypertension (FPAH). CAV1 and KCNK3 were found as two novel candidate genes of FPAH. However, few pathogenic genes were identified in idiopathic pulmonary arterial hypertension (IPAH). We conducted WES in 20 unrelated IPAH patients who did not carry the known PAH-pathogenic variants among BMPR2, CAV1, KCNK3, SMAD9, ALK1, and ENG. We found a total of 4,950 variants in 3,534 genes, including 4,444 single-nucleotide polymorphisms and 506 insertions/deletions (InDels). Through the comprehensive and multilevel analysis, we disclosed several novel signaling cascades significantly connected to IPAH, including variants related to cadherin signaling pathway, dilated cardiomyopathy, glucose metabolism, immune response, mucin-type O-glycosylation, phospholipase C (PLC)-activating G protein-coupled receptor (GPCR) signaling pathway, vascular contraction and generation, and voltage-dependent Ca2+ channels. We also conducted validation studies in five mutant genes related to PLC-activating GPCR signaling pathway potentially involved in intracellular calcium regulation through Sanger sequencing for mutation accuracy, qRT-PCR for mRNA stability, immunofluorescence for subcellular localization, Western blotting for protein level, Fura-2 imaging for intracellular calcium, and proliferation analysis for cell function. The validation experiments showed that those variants in CCR5 and C3AR1 significantly increased the rise of intracellular calcium and the variant in CCR5 profoundly enhanced proliferative capacity of human pulmonary artery smooth muscle cells. Thus, our study suggests that multiple genetically affected signaling pathways take effect together to cause the formation of IPAH and the development of right heart failure and may further provide new therapy targets or putative clues for the present treatments such as limited therapeutic effectiveness of Ca2+ channel blockers.
Collapse
Affiliation(s)
- Bingxun Liu
- Department of Pathophysiology, School of Basic Medicine, and Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liping Zhu
- Department of Pathophysiology, School of Basic Medicine, and Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yuan
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Glenn Marsboom
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - Zhigang Hong
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - Jinming Liu
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qinghua Hu
- Department of Pathophysiology, School of Basic Medicine, and Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
82
|
Hodgson J, Swietlik EM, Salmon RM, Hadinnapola C, Nikolic I, Wharton J, Guo J, Liley J, Haimel M, Bleda M, Southgate L, Machado RD, Martin JM, Treacy CM, Yates K, Daugherty LC, Shamardina O, Whitehorn D, Holden S, Bogaard HJ, Church C, Coghlan G, Condliffe R, Corris PA, Danesino C, Eyries M, Gall H, Ghio S, Ghofrani HA, Gibbs JSR, Girerd B, Houweling AC, Howard L, Humbert M, Kiely DG, Kovacs G, Lawrie A, MacKenzie Ross RV, Moledina S, Montani D, Olschewski A, Olschewski H, Ouwehand WH, Peacock AJ, Pepke-Zaba J, Prokopenko I, Rhodes CJ, Scelsi L, Seeger W, Soubrier F, Suntharalingam J, Toshner MR, Trembath RC, Noordegraaf AV, Wort SJ, Wilkins MR, Yu PB, Li W, Gräf S, Upton PD, Morrell NW. Characterization of GDF2 Mutations and Levels of BMP9 and BMP10 in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2020; 201:575-585. [PMID: 31661308 PMCID: PMC7047445 DOI: 10.1164/rccm.201906-1141oc] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Rationale: Recently, rare heterozygous mutations in GDF2 were identified in patients with pulmonary arterial hypertension (PAH). GDF2 encodes the circulating BMP (bone morphogenetic protein) type 9, which is a ligand for the BMP2 receptor.Objectives: Here we determined the functional impact of GDF2 mutations and characterized plasma BMP9 and BMP10 levels in patients with idiopathic PAH.Methods: Missense BMP9 mutant proteins were expressed in vitro and the impact on BMP9 protein processing and secretion, endothelial signaling, and functional activity was assessed. Plasma BMP9 and BMP10 levels and activity were assayed in patients with PAH with GDF2 variants and in control subjects. Levels were also measured in a larger cohort of control subjects (n = 120) and patients with idiopathic PAH (n = 260).Measurements and Main Results: We identified a novel rare variation at the GDF2 and BMP10 loci, including copy number variation. In vitro, BMP9 missense proteins demonstrated impaired cellular processing and secretion. Patients with PAH who carried these mutations exhibited reduced plasma levels of BMP9 and reduced BMP activity. Unexpectedly, plasma BMP10 levels were also markedly reduced in these individuals. Although overall BMP9 and BMP10 levels did not differ between patients with PAH and control subjects, BMP10 levels were lower in PAH females. A subset of patients with PAH had markedly reduced plasma levels of BMP9 and BMP10 in the absence of GDF2 mutations.Conclusions: Our findings demonstrate that GDF2 mutations result in BMP9 loss of function and are likely causal. These mutations lead to reduced circulating levels of both BMP9 and BMP10. These findings support therapeutic strategies to enhance BMP9 or BMP10 signaling in PAH.
Collapse
Affiliation(s)
| | - Emilia M. Swietlik
- Department of Medicine and,Royal Papworth Hospital, Papworth, United Kingdom
| | | | | | - Ivana Nikolic
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | | | | | | | - Matthias Haimel
- Department of Medicine and,Department of Haematology, University of Cambridge, Cambridge, United Kingdom,National Institute for Health Research BioResource–Rare Diseases, Cambridge, United Kingdom
| | | | - Laura Southgate
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom,Molecular and Clinical Sciences Research Institute, St. George’s University of London, London, United Kingdom
| | - Rajiv D. Machado
- Molecular and Clinical Sciences Research Institute, St. George’s University of London, London, United Kingdom
| | - Jennifer M. Martin
- Department of Medicine and,Department of Haematology, University of Cambridge, Cambridge, United Kingdom,National Institute for Health Research BioResource–Rare Diseases, Cambridge, United Kingdom
| | - Carmen M. Treacy
- Department of Medicine and,Royal Papworth Hospital, Papworth, United Kingdom
| | - Katherine Yates
- Department of Medicine and,Department of Haematology, University of Cambridge, Cambridge, United Kingdom,National Institute for Health Research BioResource–Rare Diseases, Cambridge, United Kingdom
| | - Louise C. Daugherty
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom,National Institute for Health Research BioResource–Rare Diseases, Cambridge, United Kingdom
| | - Olga Shamardina
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom,National Institute for Health Research BioResource–Rare Diseases, Cambridge, United Kingdom
| | - Deborah Whitehorn
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom,National Institute for Health Research BioResource–Rare Diseases, Cambridge, United Kingdom
| | - Simon Holden
- Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Harm J. Bogaard
- Département de Génétique, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, and UMR_S 1166-ICAN, INSERM, UPMC Sorbonne Universités, Paris, France
| | - Colin Church
- Golden Jubilee National Hospital, Glasgow, United Kingdom
| | | | - Robin Condliffe
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | | | - Cesare Danesino
- Department of Molecular Medicine, University of Pavia, Pavia, Italy,Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mélanie Eyries
- Département de Génétique, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, and UMR_S 1166-ICAN, INSERM, UPMC Sorbonne Universités, Paris, France
| | - Henning Gall
- University of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL) and of the Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
| | - Stefano Ghio
- Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Hossein-Ardeschir Ghofrani
- Department of Medicine and,University of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL) and of the Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
| | - J. Simon R. Gibbs
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Barbara Girerd
- Faculté de Médecine, Université Paris-Saclay, Université Paris-Sud, Paris, France,Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire, Assistance Publique–Hôpitaux de Paris, Paris, France,Hôpital Bicêtre, Le Kremlin-Bicêtre, INSERM UMR_S 999, Paris, France
| | - Arjan C. Houweling
- Department of Clinical Genetics, Amsterdam Universitair Medische Centra, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Marc Humbert
- Faculté de Médecine, Université Paris-Saclay, Université Paris-Sud, Paris, France,Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire, Assistance Publique–Hôpitaux de Paris, Paris, France,Hôpital Bicêtre, Le Kremlin-Bicêtre, INSERM UMR_S 999, Paris, France
| | - David G. Kiely
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Gabor Kovacs
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria,Medical University of Graz, Graz, Austria
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | | | | | - David Montani
- Faculté de Médecine, Université Paris-Saclay, Université Paris-Sud, Paris, France,Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire, Assistance Publique–Hôpitaux de Paris, Paris, France,Hôpital Bicêtre, Le Kremlin-Bicêtre, INSERM UMR_S 999, Paris, France
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria,Medical University of Graz, Graz, Austria
| | - Willem H. Ouwehand
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom,National Institute for Health Research BioResource–Rare Diseases, Cambridge, United Kingdom
| | | | | | | | | | - Laura Scelsi
- Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Werner Seeger
- University of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL) and of the Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
| | - Florent Soubrier
- Département de Génétique, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, and UMR_S 1166-ICAN, INSERM, UPMC Sorbonne Universités, Paris, France
| | | | - Mark R. Toshner
- Department of Medicine and,Royal Papworth Hospital, Papworth, United Kingdom
| | - Richard C. Trembath
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom
| | - Anton Vonk Noordegraaf
- Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Stephen J. Wort
- National Heart and Lung Institute, Imperial College London, London, United Kingdom,Royal Brompton Hospital, London, United Kingdom
| | | | - Paul B. Yu
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Wei Li
- Department of Medicine and
| | - Stefan Gräf
- Department of Medicine and,Department of Haematology, University of Cambridge, Cambridge, United Kingdom,National Institute for Health Research BioResource–Rare Diseases, Cambridge, United Kingdom
| | | | - Nicholas W. Morrell
- Department of Medicine and,National Institute for Health Research BioResource–Rare Diseases, Cambridge, United Kingdom
| |
Collapse
|
83
|
Grimmer B, Kuebler WM. Cholesterol: A Novel Regulator of Vasoreactivity in Pulmonary Arteries. Am J Respir Cell Mol Biol 2020; 62:671-673. [PMID: 32011912 PMCID: PMC7258827 DOI: 10.1165/rcmb.2020-0020ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Benjamin Grimmer
- Institute of PhysiologyCharité-University Medicine BerlinBerlin, Germany
| | - Wolfgang M Kuebler
- Institute of PhysiologyCharité-University Medicine BerlinBerlin, Germany.,The Keenan Research Centre for Biomedical ScienceSt. Michael's HospitalToronto, Ontario, Canada.,Department of Surgeryand.,Department of PhysiologyUniversity of TorontoToronto, Ontario, Canada
| |
Collapse
|
84
|
Southgate L, Machado RD, Gräf S, Morrell NW. Molecular genetic framework underlying pulmonary arterial hypertension. Nat Rev Cardiol 2020; 17:85-95. [PMID: 31406341 DOI: 10.1038/s41569-019-0242-x] [Citation(s) in RCA: 172] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/12/2019] [Indexed: 02/02/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a rare, progressive disorder typified by occlusion of the pulmonary arterioles owing to endothelial dysfunction and uncontrolled proliferation of pulmonary artery smooth muscle cells and fibroblasts. Vascular occlusion can lead to increased pressure in the pulmonary arteries, often resulting in right ventricular failure with shortness of breath and syncope. Since the identification of BMPR2, which encodes a receptor in the transforming growth factor-β superfamily, the development of high-throughput sequencing approaches to identify novel causal genes has substantially advanced our understanding of the molecular genetics of PAH. In the past 6 years, additional pathways involved in PAH susceptibility have been described through the identification of deleterious genetic variants in potassium channels (KCNK3 and ABCC8) and transcription factors (TBX4 and SOX17), among others. Although familial PAH most often has an autosomal-dominant pattern of inheritance, cases of incomplete penetrance and evidence of genetic heterogeneity support a model of PAH as a Mendelian disorder with complex disease features. In this Review, we outline the latest advances in the detection of rare and common genetic variants underlying PAH susceptibility and disease progression. These findings have clinical implications for lung vascular function and can help to identify mechanistic pathways amenable to pharmacological intervention.
Collapse
Affiliation(s)
- Laura Southgate
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK.,Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Rajiv D Machado
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge, UK.,Department of Haematology, University of Cambridge, Cambridge, UK.,NIHR BioResource, Cambridge, UK
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge, UK. .,NIHR BioResource, Cambridge, UK.
| |
Collapse
|
85
|
Mathew R, Huang J, Iacobas S, Iacobas DA. Pulmonary Hypertension Remodels the Genomic Fabrics of Major Functional Pathways. Genes (Basel) 2020; 11:genes11020126. [PMID: 31979420 PMCID: PMC7074533 DOI: 10.3390/genes11020126] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/23/2022] Open
Abstract
Pulmonary hypertension (PH) is a serious disorder with high morbidity and mortality rate. We analyzed the right-ventricular systolic pressure (RVSP), right-ventricular hypertrophy (RVH), lung histology, and transcriptomes of six-week-old male rats with PH induced by (1) hypoxia (HO), (2) administration of monocrotaline (CM), or (3) administration of monocrotaline and exposure to hypoxia (HM). The results in PH rats were compared to those in control rats (CO). After four weeks exposure, increased RVSP and RVH, pulmonary arterial wall thickening, and alteration of the lung transcriptome were observed in all PH groups. The HM group exhibited the largest alterations, as well as neointimal lesions and obliteration of the lumen in small arteries. We found that PH increased the expression of caveolin1, matrix metallopeptidase 2, and numerous inflammatory and cell proliferation genes. The cell cycle, vascular smooth muscle contraction, and oxidative phosphorylation pathways, as well as their interplay, were largely perturbed. Our results also suggest that the upregulated Rhoa (Ras homolog family member A) mediates its action through expression coordination with several ATPases. The upregulation of antioxidant genes and the extensive mitochondrial damage observed, especially in the HM group, indicate metabolic shift toward aerobic glycolysis.
Collapse
Affiliation(s)
- Rajamma Mathew
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA; (R.M.); (J.H.)
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA
| | - Jing Huang
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA; (R.M.); (J.H.)
| | - Sanda Iacobas
- Department of Pathology, New York Medical College, Valhalla, NY 10595, USA
| | - Dumitru A. Iacobas
- Personalized Genomics Laboratory, Center for Computational Systems Biology, Roy G Perry College of Engineering, Prairie View A&M University, Prairie View, TX 77446, USA
- Correspondence: ; Tel.: +1-936-261-9926
| |
Collapse
|
86
|
Norton CE, Weise-Cross L, Ahmadian R, Yan S, Jernigan NL, Paffett ML, Naik JS, Walker BR, Resta TC. Altered Lipid Domains Facilitate Enhanced Pulmonary Vasoconstriction after Chronic Hypoxia. Am J Respir Cell Mol Biol 2020; 62:709-718. [PMID: 31945301 DOI: 10.1165/rcmb.2018-0318oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic hypoxia (CH) augments depolarization-induced pulmonary vasoconstriction through superoxide-dependent, Rho kinase-mediated Ca2+ sensitization. Nicotinamide adenine dinucleotide phosphate oxidase and EGFR (epidermal growth factor receptor) signaling contributes to this response. Caveolin-1 regulates the activity of a variety of proteins, including EGFR and nicotinamide adenine dinucleotide phosphate oxidase, and membrane cholesterol is an important regulator of caveolin-1 protein interactions. We hypothesized that derangement of these membrane lipid domain components augments depolarization-induced Ca2+ sensitization and resultant vasoconstriction after CH. Although exposure of rats to CH (4 wk, ∼380 mm Hg) did not alter caveolin-1 expression in intrapulmonary arteries or the incidence of caveolae in arterial smooth muscle, CH markedly reduced smooth muscle membrane cholesterol content as assessed by filipin fluorescence. Effects of CH on vasoreactivity and superoxide generation were examined using pressurized, Ca2+-permeabilized, endothelium-disrupted pulmonary arteries (∼150 μm inner diameter) from CH and control rats. Depolarizing concentrations of KCl evoked greater constriction in arteries from CH rats than in those obtained from control rats, and increased superoxide production as assessed by dihydroethidium fluorescence only in arteries from CH rats. Both cholesterol supplementation and the caveolin-1 scaffolding domain peptide antennapedia-Cav prevented these effects of CH, with each treatment restoring membrane cholesterol in CH arteries to control levels. Enhanced EGF-dependent vasoconstriction after CH similarly required reduced membrane cholesterol. However, these responses to CH were not associated with changes in EGFR expression or activity, suggesting that cholesterol regulates this signaling pathway downstream of EGFR. We conclude that alterations in membrane lipid domain signaling resulting from reduced cholesterol content facilitate enhanced depolarization- and EGF-induced pulmonary vasoconstriction after CH.
Collapse
Affiliation(s)
- Charles E Norton
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Laura Weise-Cross
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Rosstin Ahmadian
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Simin Yan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Michael L Paffett
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Jay S Naik
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
87
|
Affiliation(s)
- Jacqueline S Dron
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Robert A Hegele
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
88
|
Hester J, Ventetuolo C, Lahm T. Sex, Gender, and Sex Hormones in Pulmonary Hypertension and Right Ventricular Failure. Compr Physiol 2019; 10:125-170. [PMID: 31853950 DOI: 10.1002/cphy.c190011] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pulmonary hypertension (PH) encompasses a syndrome of diseases that are characterized by elevated pulmonary artery pressure and pulmonary vascular remodeling and that frequently lead to right ventricular (RV) failure and death. Several types of PH exhibit sexually dimorphic features in disease penetrance, presentation, and progression. Most sexually dimorphic features in PH have been described in pulmonary arterial hypertension (PAH), a devastating and progressive pulmonary vasculopathy with a 3-year survival rate <60%. While patient registries show that women are more susceptible to development of PAH, female PAH patients display better RV function and increased survival compared to their male counterparts, a phenomenon referred to as the "estrogen paradox" or "estrogen puzzle" of PAH. Recent advances in the field have demonstrated that multiple sex hormones, receptors, and metabolites play a role in the estrogen puzzle and that the effects of hormone signaling may be time and compartment specific. While the underlying physiological mechanisms are complex, unraveling the estrogen puzzle may reveal novel therapeutic strategies to treat and reverse the effects of PAH/PH. In this article, we (i) review PH classification and pathophysiology; (ii) discuss sex/gender differences observed in patients and animal models; (iii) review sex hormone synthesis and metabolism; (iv) review in detail the scientific literature of sex hormone signaling in PAH/PH, particularly estrogen-, testosterone-, progesterone-, and dehydroepiandrosterone (DHEA)-mediated effects in the pulmonary vasculature and RV; (v) discuss hormone-independent variables contributing to sexually dimorphic disease presentation; and (vi) identify knowledge gaps and pathways forward. © 2020 American Physiological Society. Compr Physiol 10:125-170, 2020.
Collapse
Affiliation(s)
- James Hester
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, Occupational and Sleep Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Corey Ventetuolo
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Department of Health Services, Policy and Practice, Brown University School of Public Health, Providence, Rhode Island, USA
| | - Tim Lahm
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, Occupational and Sleep Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
89
|
Zhu N, Pauciulo MW, Welch CL, Lutz KA, Coleman AW, Gonzaga-Jauregui C, Wang J, Grimes JM, Martin LJ, He H, Shen Y, Chung WK, Nichols WC. Novel risk genes and mechanisms implicated by exome sequencing of 2572 individuals with pulmonary arterial hypertension. Genome Med 2019; 11:69. [PMID: 31727138 PMCID: PMC6857288 DOI: 10.1186/s13073-019-0685-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/06/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Group 1 pulmonary arterial hypertension (PAH) is a rare disease with high mortality despite recent therapeutic advances. Pathogenic remodeling of pulmonary arterioles leads to increased pulmonary pressures, right ventricular hypertrophy, and heart failure. Mutations in bone morphogenetic protein receptor type 2 and other risk genes predispose to disease, but the vast majority of non-familial cases remain genetically undefined. METHODS To identify new risk genes, we performed exome sequencing in a large cohort from the National Biological Sample and Data Repository for PAH (PAH Biobank, n = 2572). We then carried out rare deleterious variant identification followed by case-control gene-based association analyses. To control for population structure, only unrelated European cases (n = 1832) and controls (n = 12,771) were used in association tests. Empirical p values were determined by permutation analyses, and the threshold for significance defined by Bonferroni's correction for multiple testing. RESULTS Tissue kallikrein 1 (KLK1) and gamma glutamyl carboxylase (GGCX) were identified as new candidate risk genes for idiopathic PAH (IPAH) with genome-wide significance. We note that variant carriers had later mean age of onset and relatively moderate disease phenotypes compared to bone morphogenetic receptor type 2 variant carriers. We also confirmed the genome-wide association of recently reported growth differentiation factor (GDF2) with IPAH and further implicate T-box 4 (TBX4) with child-onset PAH. CONCLUSIONS We report robust association of novel genes KLK1 and GGCX with IPAH, accounting for ~ 0.4% and 0.9% of PAH Biobank cases, respectively. Both genes play important roles in vascular hemodynamics and inflammation but have not been implicated in PAH previously. These data suggest new genes, pathogenic mechanisms, and therapeutic targets for this lethal vasculopathy.
Collapse
Affiliation(s)
- Na Zhu
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Carrie L Welch
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Katie A Lutz
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
| | - Anna W Coleman
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
| | | | - Jiayao Wang
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Joseph M Grimes
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Lisa J Martin
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Hua He
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA.
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
90
|
Chichger H, Rounds S, Harrington EO. Endosomes and Autophagy: Regulators of Pulmonary Endothelial Cell Homeostasis in Health and Disease. Antioxid Redox Signal 2019; 31:994-1008. [PMID: 31190562 PMCID: PMC6765061 DOI: 10.1089/ars.2019.7817] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/07/2019] [Indexed: 12/12/2022]
Abstract
Significance: Alterations in oxidant/antioxidant balance injure pulmonary endothelial cells and are important in the pathogenesis of lung diseases, such as Acute Respiratory Distress Syndrome (ARDS), ischemia/reperfusion injury, pulmonary arterial hypertension (PAH), and emphysema. Recent Advances: The endosomal and autophagic pathways regulate cell homeostasis. Both pathways support recycling or degradation of macromolecules or organelles, targeted to endosomes or lysosomes, respectively. Thus, both processes promote cell survival. However, with environmental stress or injury, imbalance in endosomal and autophagic pathways may enhance macromolecular or organelle degradation, diminish biosynthetic processes, and cause cell death. Critical Issues: While the role of autophagy in cellular homeostasis in pulmonary disease has been investigated, the role of the endosome in the lung vasculature is less known. Furthermore, autophagy can either decrease or exacerbate endothelial injury, depending upon inciting insult and disease process. Future Directions: Diseases affecting the pulmonary endothelium, such as emphysema, ARDS, and PAH, are linked to altered endosomal or autophagic processing, leading to enhanced degradation of macromolecules and potential cell death. Efforts to target this imbalance have yielded limited success as treatments for lung injuries, which may be due to the complexity of both processes. It is possible that endosomal trafficking proteins, such as Rab GTPases and late endosomal/lysosomal adaptor, MAPK and MTOR activator 1, may be novel therapeutic targets. While endocytosis or autophagy have been linked to improved function of the pulmonary endothelium in vitro and in vivo, further studies are needed to identify targets for modulating cellular homeostasis in the lung.
Collapse
Affiliation(s)
- Havovi Chichger
- Biomedical Research Group, Department of Biomedical and Forensic Sciences, Anglia Ruskin University, Cambridge, United Kingdom
| | - Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Elizabeth O. Harrington
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
91
|
Tielemans B, Stoian L, Gijsbers R, Michiels A, Wagenaar A, Farre Marti R, Belge C, Delcroix M, Quarck R. Cytokines trigger disruption of endothelium barrier function and p38 MAP kinase activation in BMPR2-silenced human lung microvascular endothelial cells. Pulm Circ 2019; 9:2045894019883607. [PMID: 31692724 PMCID: PMC6811766 DOI: 10.1177/2045894019883607] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/24/2019] [Indexed: 12/21/2022] Open
Abstract
The bone morphogenetic protein receptor II (BMPRII) signaling pathway is impaired
in pulmonary arterial hypertension and mutations in the BMPR2
gene have been observed in both heritable and idiopathic pulmonary arterial
hypertension. However, all BMPR2 mutation carriers do not
develop pulmonary arterial hypertension, and inflammation could trigger the
development of the disease in BMPR2 mutation carriers.
Circulating levels and/or lung tissue expression of cytokines such as tumor
necrosis factor-α or interleukin-18 are elevated in patients with pulmonary
arterial hypertension and could be involved in the pathogenesis of pulmonary
arterial hypertension. We consequently hypothesized that cytokines could trigger
endothelial dysfunction in addition to impaired BMPRII signaling. Our aim was to
determine whether impairment of BMPRII signaling might affect endothelium
barrier function and adhesiveness to monocytes, in response to cytokines.
BMPR2 was silenced in human lung microvascular endothelial
cells (HLMVECs) using lentiviral vectors encoding microRNA-based hairpins.
Effects of tumor necrosis factor-α and interleukin-18 on HLMVEC adhesiveness to
the human monocyte cell line THP-1, adhesion molecule expression, endothelial
barrier function and activation of P38MAPK were investigated in vitro. Stable
BMPR2 silencing in HLMVECs resulted in impaired endothelial
barrier function and constitutive activation of P38MAPK. Adhesiveness of
BMPR2-silenced HLMVECs to THP-1 cells was enhanced by tumor
necrosis factor-α and interleukin-18 through ICAM-1 adhesion molecule.
Interestingly, tumor necrosis factor-α induced activation of P38MAPK and
disrupted endothelial barrier function in BMPR2-silenced
HLMVECs. Altogether, our findings showed that stable BMPR2
silencing resulted in impaired endothelial barrier function and activation of
P38MAPK in HLMVECs. In BMPR2-silenced HLMVECs, cytokines
enhanced adhesiveness capacities, activation of P38MAPK and impaired endothelial
barrier function suggesting that cytokines could trigger the development of
pulmonary arterial hypertension in a context of impaired BMPRII signaling
pathway.
Collapse
Affiliation(s)
- Birger Tielemans
- Division of Respiratory Diseases, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Leanda Stoian
- Division of Respiratory Diseases, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Rik Gijsbers
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Leuven, Belgium.,Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven - University of Leuven, Leuven, Belgium
| | - Annelies Michiels
- Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven - University of Leuven, Leuven, Belgium.,Leuven Viral Vector Core, KU Leuven - University of Leuven, Leuven, Belgium
| | - Allard Wagenaar
- Division of Respiratory Diseases, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Ricard Farre Marti
- Translational Research in Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Catharina Belge
- Division of Respiratory Diseases, University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Marion Delcroix
- Division of Respiratory Diseases, University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Rozenn Quarck
- Division of Respiratory Diseases, University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| |
Collapse
|
92
|
Abstract
In early 2019, the 6th World Symposium on Pulmonary Hypertension (WSPH) released an updated document highlighting the advances in the last five years. During the quinquennial event many experts worked together to suggest new changes in the disease diagnosis and management. Since inception of the WSPH in 1973, this is the first time when the hemodynamic definition of pulmonary hypertension (PH) has been updated. These proceedings have re-defined the different hemodynamic types of PH that occur with the left heart disease along with introduction to the genetic testing as part of pulmonary arterial hypertension (PAH) evaluation. Objective of this review is to highlight the evaluation and diagnosis of PAH based on the proceedings of the 6th WSPH. Accurate early diagnosis and subsequent management of PH is necessary, as despite of treatment advances, survival remains suboptimal.
Collapse
Affiliation(s)
- Sandeep Sahay
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, Institute of Academic Medicine, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
93
|
Mercurio V, Bianco A, Campi G, Cuomo A, Diab N, Mancini A, Parrella P, Petretta M, Hassoun PM, Bonaduce D. New Drugs, Therapeutic Strategies, and Future Direction for the Treatment of Pulmonary Arterial Hypertension. Curr Med Chem 2019; 26:2844-2864. [DOI: 10.2174/0929867325666180201095743] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/21/2017] [Accepted: 12/21/2017] [Indexed: 12/20/2022]
Abstract
Despite recent advances in Pulmonary Arterial Hypertension (PAH) treatment, this condition is still characterized by an extremely poor prognosis. In this review, we discuss the use of newly-approved drugs for PAH treatment with already known mechanisms of action (macitentan), innovative targets (riociguat and selexipag), and novel therapeutic approaches with initial up-front combination therapy. Secondly, we describe new potential signaling pathways and investigational drugs with promising role in the treatment of PAH.
Collapse
Affiliation(s)
- Valentina Mercurio
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Anna Bianco
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Giacomo Campi
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Alessandra Cuomo
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Nermin Diab
- University of Ottawa, Department of Medicine, Ottawa, ON, Canada
| | - Angela Mancini
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Paolo Parrella
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Mario Petretta
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Paul M. Hassoun
- Johns Hopkins University, Division of Pulmonary and Critical Care Medicine, Baltimore, MD, United States
| | - Domenico Bonaduce
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| |
Collapse
|
94
|
Liu X, Mei M, Chen X, Lu Y, Dong X, Hu L, Hu X, Cheng G, Cao Y, Yang L, Zhou W. Identification of genetic factors underlying persistent pulmonary hypertension of newborns in a cohort of Chinese neonates. Respir Res 2019; 20:174. [PMID: 31382961 PMCID: PMC6683566 DOI: 10.1186/s12931-019-1148-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/29/2019] [Indexed: 01/02/2023] Open
Abstract
Background Persistent pulmonary hypertension of the newborn (PPHN) is a severe clinical problem among neonatal intensive care unit (NICU) patients. The genetic pathogenesis of PPHN is unclear. Only a few genetic polymorphisms have been identified in infants with PPHN. Our study aimed to investigate the potential genetic etiology of PPHN. Methods This study recruited PPHN patients admitted to the NICU of the Children’s Hospital of Fudan University from Jan 2016 to Dec 2017. Exome sequencing was performed for all patients. Variants in reported PPHN/pulmonary arterial hypertension (PAH)-related genes were assessed. Single nucleotide polymorphism (SNP) association and gene-level analyses were carried out in 74 PPHN cases and 115 non-PPHN controls with matched baseline characteristics. Results Among the patient cohort, 74 (64.3%) patients were late preterm and term infants (≥ 34 weeks gestation) and 41 (35.7%) were preterm infants (< 34 weeks gestation). Preterm infants with PPHN exhibited low birth weight and a high frequency of bronchopulmonary dysplasia, respiratory distress syndrome (RDS) and mortality. Nine patients (only one preterm infant) were identified as harboring genetic variants, including three with pathogenic/likely pathogenic variants in TBX4 and BMPR2 and six with variants of unknown significance in BMPR2, SMAD9, TGFB1, KCNA5 and TRPC6. Three SNPs (rs192759073, rs1047883 and rs2229589) in CPS1 and one SNP (rs1044008) in NOTCH3 were significantly associated with PPHN (p < 0.05). CPS1 and SMAD9 were identified as risk genes for PPHN (p < 0.05). Conclusions In this study, we identified genetic variants in PPHN patients, and we reported CPS1, NOTCH3 and SMAD9 as risk genes for late preterm and term PPHN in a single-center Chinese cohort. Our findings provide additional genetic evidence of the pathogenesis of PPHN and new insight into potential strategies for disease treatment. Electronic supplementary material The online version of this article (10.1186/s12931-019-1148-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xu Liu
- Clinical Genetic Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.,Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Mei Mei
- Department of Pulmonology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Xiang Chen
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Yulan Lu
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Xinran Dong
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Liyuan Hu
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Xiaojing Hu
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Guoqiang Cheng
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Yun Cao
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Lin Yang
- Clinical Genetic Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China. .,Key Laboratory of Birth Defects, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.
| | - Wenhao Zhou
- Clinical Genetic Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China. .,Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China. .,Key Laboratory of Birth Defects, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.
| |
Collapse
|
95
|
Abstract
Lipodystrophies are the result of a range of inherited and acquired causes, but all are characterized by perturbations in white adipose tissue function and, in many instances, its mass or distribution. Though patients are often nonobese, they typically manifest a severe form of the metabolic syndrome, highlighting the importance of white fat in the "safe" storage of surplus energy. Understanding the molecular pathophysiology of congenital lipodystrophies has yielded useful insights into the biology of adipocytes and informed therapeutic strategies. More recently, genome-wide association studies focused on insulin resistance have linked common variants to genes implicated in adipose biology and suggested that subtle forms of lipodystrophy contribute to cardiometabolic disease risk at a population level. These observations underpin the use of aligned treatment strategies in insulin-resistant obese and lipodystrophic patients, the major goal being to alleviate the energetic burden on adipose tissue.
Collapse
|
96
|
Kamely M, Karimi Torshizi MA, Wideman RF, West J. Upregulation of SERT and ADORA1 in broilers with acute right ventricular failure. Res Vet Sci 2019; 125:397-400. [DOI: 10.1016/j.rvsc.2017.09.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 08/10/2017] [Accepted: 09/30/2017] [Indexed: 12/30/2022]
|
97
|
Kozawa S, Nata M. Sudden death of a preschool child diagnosed by postmortem examination. J Forensic Leg Med 2019; 66:144-146. [PMID: 31302445 DOI: 10.1016/j.jflm.2019.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/28/2019] [Accepted: 07/10/2019] [Indexed: 11/16/2022]
Abstract
An autopsy case of sudden death due to pulmonary arterial hypertension (PAH) in a 5-year-old boy whose cause of death was not determined during autopsy, but was later determined by postmortem examination, is presented. The boy developed convulsions that subsequently stopped, but remained unconscious. He was transported to hospital by ambulance, but died soon after. The boy had been found to have right ventricular overload on ECG 2 weeks earlier. A plan had been made to consult a doctor for a specialist visit 2 months later. During autopsy, significant abnormalities or injuries were not observed on the body's external surface. Internal examination showed congested organs, and the blood remaining in the body was dark red with fluidity. The heart was significantly enlarged (146 g), with nearly equivalent thickness of the left and right ventricles, showing right ventricular hypertrophy. Obvious macroscopic abnormalities were not observed at the origin and main trunk of the pulmonary artery. The lungs were slightly swollen (right lung 100 g, left lung 95 g), severely congested, and edematous. A postmortem CT scan displayed some patchy shadows in both lungs; however, no significant abnormalities were detected. Histopathological examination suggested a diagnosis of PAH. Three genes (BMPR2, ALK1, and ENG) were tested, revealing a heterozygous insertion of five nucleotides, TTTCC, between nucleotides 2677 and 2678 within exon 12 of the BMPR2 gene. Therefore, the subject was considered to have had heritable PAH due to a BMPR2 gene mutation.
Collapse
Affiliation(s)
- Shuji Kozawa
- Department of Forensic Medicine and Sciences, Mie University Graduate School of Medicine, Japan.
| | - Masayuki Nata
- Department of Forensic Medicine and Sciences, Mie University Graduate School of Medicine, Japan
| |
Collapse
|
98
|
Condon DF, Nickel NP, Anderson R, Mirza S, de Jesus Perez VA. The 6th World Symposium on Pulmonary Hypertension: what's old is new. F1000Res 2019; 8. [PMID: 31249672 PMCID: PMC6584967 DOI: 10.12688/f1000research.18811.1] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/11/2019] [Indexed: 12/24/2022] Open
Abstract
In February 2018, the 6th World Symposium on Pulmonary Hypertension (WSPH) brought together experts from various disciplines to review the most relevant clinical and scientific advances in the field of PH over the last 5 years. Based on careful review and discussions by members of the different task forces, major revisions were made on the hemodynamic definition for various forms of PH and new genes were added to the list of genetic markers associated with pulmonary arterial hypertension (PAH) and pulmonary veno-occlusive disease. In addition, the use of risk stratification tools was encouraged as a strategy to reduce one-year mortality risk in PAH patients through early implementation of PAH therapies. While members of the medical community are still debating some of the proposed changes, the new WSPH guidelines advocate early diagnosis and initiation of combination therapy to reduce mortality and improve quality of life in patients with PH.
Collapse
Affiliation(s)
- David F Condon
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, USA
| | - Nils P Nickel
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, USA
| | - Ryan Anderson
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, USA
| | - Shireen Mirza
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, USA.,Vera Moulton Wall Center for Pulmonary Vascular Research, Stanford University, Stanford, USA
| | - Vinicio A de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, USA.,Vera Moulton Wall Center for Pulmonary Vascular Research, Stanford University, Stanford, USA
| |
Collapse
|
99
|
Nickel NP, de Jesus Perez VA, Zamanian RT, Fessel JP, Cogan JD, Hamid R, West JD, de Caestecker MP, Yang H, Austin ED. Low-grade albuminuria in pulmonary arterial hypertension. Pulm Circ 2019; 9:2045894018824564. [PMID: 30632900 PMCID: PMC6557031 DOI: 10.1177/2045894018824564] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Low-grade albuminuria, determined by the urinary albumin to creatinine ratio, has been linked to systemic vascular dysfunction and is associated with cardiovascular mortality. Pulmonary arterial hypertension is related to mutations in the bone morphogenetic protein receptor type 2, pulmonary vascular dysfunction and is increasingly recognized as a systemic disease. In a total of 283 patients (two independent cohorts) diagnosed with pulmonary arterial hypertension, 18 unaffected BMPR2 mutation carriers and 68 healthy controls, spot urinary albumin to creatinine ratio and its relationship to demographic, functional, hemodynamic and outcome data were analyzed. Pulmonary arterial hypertension patients and unaffected BMPR2 mutation carriers had significantly elevated urinary albumin to creatinine ratios compared with healthy controls ( P < 0.01; P = 0.04). In pulmonary arterial hypertension patients, the urinary albumin to creatinine ratio was associated with older age, lower six-minute walking distance, elevated levels of C-reactive protein and hemoglobin A1c, but there was no correlation between the urinary albumin to creatinine ratio and hemodynamic variables. Pulmonary arterial hypertension patients with a urinary albumin to creatinine ratio above 10 µg/mg had significantly higher rates of poor outcome ( P < 0.001). This study shows that low-grade albuminuria is prevalent in pulmonary arterial hypertension patients and is associated with poor outcome. This study shows that albuminuria in pulmonary arterial hypertension is associated with systemic inflammation and insulin resistance.
Collapse
Affiliation(s)
- Nils P Nickel
- 1 Stanford University School of Medicine, Stanford University, USA.,2 Vanderbilt University Medical Center, USA
| | | | - Roham T Zamanian
- 1 Stanford University School of Medicine, Stanford University, USA
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Huang K, Dang Y, Zhang P, Shen C, Sui X, Xia G, Qin Y, Jiao X, Wang C, Huo R, Chen ZJ. CAV1 regulates primordial follicle formation via the Notch2 signalling pathway and is associated with premature ovarian insufficiency in humans. Hum Reprod 2019; 33:2087-2095. [PMID: 30304446 DOI: 10.1093/humrep/dey299] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/19/2018] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION What is the function of CAV1 in folliculogenesis and female reproduction? SUMMARY ANSWER CAV1 regulates germline cyst breakdown and primordial follicle (PF) formation in mice, and CAV1 mutation may be related to premature ovarian insufficiency (POI). WHAT IS KNOWN ALREADY Pre-granulosa cells are essential for the establishment of the PF pool, which determines female fertility and reproductive lifespan. Cav1 participates in vascularization in fetal mouse ovaries. However, the role of CAV1 in early folliculogenesis and POI pathogenesis remains unclear. STUDY DESIGN, SIZE, DURATION Cav1 function was investigated in mice and Human Embryonic Kidney 293 cells. Ovaries (six per group) were randomly assigned to Cav1-vivo-morpholino, control and control-morpholino groups, and all experiments were repeated at least three times. To investigate CAV1 mutations in women, 200 Chinese women with POI and 200 control individuals with regular menstrual cycles and normal endocrine profiles were recruited from the Center for Reproductive Medicine of Shandong University between September 2012 and December 2013. PARTICIPANTS/MATERIALS, SETTING, METHODS Wild-type CD1 mice, Lgr5-EGFP-ires-CreERT2 (Lgr5-KI) reporter mice and Human Embryonic Kidney 293 cells were used for these experiments. Protein expression was detected by Western blot, and quantitative RT-PCR was used to detect gene expression. The expression pattern of CAV1 in mouse ovaries and the phenotype of Cav1 deficiency in mice were detected by immunofluorescence. Pre-granulosa cell proliferation in ovaries was detected by bromodeoxyuridine (BrdU) assay and immunofluorescence. The coding region of the CAV1 gene was sequenced in 200 women with POI and 200 controls. The functional effect of the novel mutation c.142 G > C (p.Glu48Gln) was investigated by Cell Counting Kit-8 (CCK8) assays and Western blot. MAIN RESULTS AND THE ROLE OF CHANCE We confirmed that Cav1 deficiency in mouse ovary induced by CAV1-vivo-morpholino resulted in more multi-oocyte follicles than in the control and control-morpholino groups (P < 0.01). Suppression of Cav1 decreased Leucine rich repeat containing G protein coupled receptor 5 (Lgr5)-positive cell proliferation (P < 0.01) and reduced the number of Lgr5 and Forkhead box L2 (Foxl2) double-positive cells (P < 0.01). Furthermore, suppression of Cav1 inhibited ovarian epithelial Lgr5-positive cell proliferation and differentiation through the Notch2 signalling pathway. Two of the POI women carried novel CAV1 mutations (c.45 C > G synonymous and c.142 G > C [Glu48Gln]). The deleterious effect of p.Glu48Gln was corroborated by showing that it adversely affected the function of CAV1 in cell proliferation and NOTCH2 expression in HEK293FT cells. LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The novel Glu48Gln mutation was only detected in one of 200 POI patients and we were unable to investigate its effects in the ovary. WIDER IMPLICATIONS OF THE FINDINGS The identification of CAV1 as a potentially causative gene for POI provides a theoretical basis to devise treatments for POI in women. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Basic Research Program of China (973 Programs: 2012CB944700; 2013CB945501; 2013CB911400; 2014CB943202), the National Key Research and Development Program of China (2016YFC1000604, 2017YFC1001301), the State Key Program of National Natural Science Foundation of China (81430029), and the National Natural Science Foundation of China (31571540, 81522018, 81471509, 81601245, 81701406, 81571406). The authors declare no competing financial interests.
Collapse
Affiliation(s)
- Kun Huang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yujie Dang
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics; The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education; Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China.,Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Pan Zhang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Cong Shen
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Xuesong Sui
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yingying Qin
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics; The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education; Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
| | - Xue Jiao
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics; The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education; Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Ran Huo
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Zi-Jiang Chen
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics; The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education; Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China.,Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|