51
|
Bonnet D, Szezepanski I, Delacourt C, Malkezadeh-Milani S, Lévy M. Multifactorial pulmonary hypertension in infantile scimitar syndrome. Arch Cardiovasc Dis 2022; 115:142-150. [DOI: 10.1016/j.acvd.2022.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/29/2022]
|
52
|
Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Ferguson JF, Generoso G, Ho JE, Kalani R, Khan SS, Kissela BM, Knutson KL, Levine DA, Lewis TT, Liu J, Loop MS, Ma J, Mussolino ME, Navaneethan SD, Perak AM, Poudel R, Rezk-Hanna M, Roth GA, Schroeder EB, Shah SH, Thacker EL, VanWagner LB, Virani SS, Voecks JH, Wang NY, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation 2022; 145:e153-e639. [PMID: 35078371 DOI: 10.1161/cir.0000000000001052] [Citation(s) in RCA: 2913] [Impact Index Per Article: 971.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2022 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population and an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, and the global burden of cardiovascular disease and healthy life expectancy. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
53
|
Jeong EM, Pereira M, So EY, Wu KQ, Del Tatto M, Wen S, Dooner MS, Dubielecka PM, Reginato AM, Ventetuolo CE, Quesenberry PJ, Klinger JR, Liang OD. Targeting RUNX1 as a novel treatment modality for pulmonary arterial hypertension. Cardiovasc Res 2022; 118:3211-3224. [PMID: 35018410 PMCID: PMC9799056 DOI: 10.1093/cvr/cvac001] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 01/06/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS Pulmonary arterial hypertension (PAH) is a fatal disease without a cure. Previously, we found that transcription factor RUNX1-dependent haematopoietic transformation of endothelial progenitor cells may contribute to the pathogenesis of PAH. However, the therapeutic potential of RUNX1 inhibition to reverse established PAH remains unknown. In the current study, we aimed to determine whether RUNX1 inhibition was sufficient to reverse Sugen/hypoxia (SuHx)-induced pulmonary hypertension (PH) in rats. We also aimed to demonstrate possible mechanisms involved. METHODS AND RESULTS We administered a small molecule specific RUNX1 inhibitor Ro5-3335 before, during, and after the development of SuHx-PH in rats to investigate its therapeutic potential. We quantified lung macrophage recruitment and activation in vivo and in vitro in the presence or absence of the RUNX1 inhibitor. We generated conditional VE-cadherin-CreERT2; ZsGreen mice for labelling adult endothelium and lineage tracing in the SuHx-PH model. We also generated conditional Cdh5-CreERT2; Runx1(flox/flox) mice to delete Runx1 gene in adult endothelium and LysM-Cre; Runx1(flox/flox) mice to delete Runx1 gene in cells of myeloid lineage, and then subjected these mice to SuHx-PH induction. RUNX1 inhibition in vivo effectively prevented the development, blocked the progression, and reversed established SuHx-induced PH in rats. RUNX1 inhibition significantly dampened lung macrophage recruitment and activation. Furthermore, lineage tracing with the inducible VE-cadherin-CreERT2; ZsGreen mice demonstrated that a RUNX1-dependent endothelial to haematopoietic transformation occurred during the development of SuHx-PH. Finally, tissue-specific deletion of Runx1 gene either in adult endothelium or in cells of myeloid lineage prevented the mice from developing SuHx-PH, suggesting that RUNX1 is required for the development of PH. CONCLUSION By blocking RUNX1-dependent endothelial to haematopoietic transformation and pulmonary macrophage recruitment and activation, targeting RUNX1 may be as a novel treatment modality for pulmonary arterial hypertension.
Collapse
Affiliation(s)
| | | | - Eui-Young So
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Keith Q Wu
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Michael Del Tatto
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Sicheng Wen
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Mark S Dooner
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Patrycja M Dubielecka
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Anthony M Reginato
- Division of Rheumatology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Corey E Ventetuolo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Peter J Quesenberry
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - James R Klinger
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Olin D Liang
- Corresponding author. Tel: 617-816-8885; fax: 401-444-2486, E-mail:
| |
Collapse
|
54
|
Affiliation(s)
- Paul M Hassoun
- From the Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore
| |
Collapse
|
55
|
Epstein R, Krishnan US. Management of Pulmonary Hypertension in the Pediatric Patient. Cardiol Clin 2021; 40:115-127. [PMID: 34809912 DOI: 10.1016/j.ccl.2021.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pediatric pulmonary hypertension (PH) is a rare disease with historically very high morbidity and mortality. In the past 20 years, there has been a growing recognition that pediatric PH, although having similarities to adult PH, is a unique entity with its own particular pathogeneses, presentation, and management. With better understanding and earlier diagnosis of pediatric PH, and as more medications have become available, survival of children with PH has also significantly improved. This article reviews the various forms of PH in childhood, with a focus on both established and investigational therapies that are available for children with PH.
Collapse
Affiliation(s)
- Rebecca Epstein
- Pediatric Cardiology, Columbia University Irving Medical Center, New York Presbyterian Hospital, CHN 2N, #255, 3959 Broadway, New York, NY 10032, USA
| | - Usha S Krishnan
- Pediatric Cardiology, Columbia University Irving Medical Center, New York Presbyterian Hospital, CHN 2N, #255, 3959 Broadway, New York, NY 10032, USA.
| |
Collapse
|
56
|
Lu C, Yang D, Lei C, Wang R, Guo T, Luo H. Identification of Two Novel DNAAF2 Variants in Two Consanguineous Families with Primary Ciliary Dyskinesia. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2021; 14:1415-1423. [PMID: 34785929 PMCID: PMC8591118 DOI: 10.2147/pgpm.s338981] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/26/2021] [Indexed: 01/16/2023]
Abstract
Background Dynein axonemal assembly factor 2 (DNAAF2) is involved in the early preassembly of dynein in the cytoplasm, which is essential for motile cilia function. Primary ciliary dyskinesia (PCD) associated with DNAAF2 variants has rarely been reported in females with infertility. Moreover, there is no report linking DNAAF2 to scoliosis in human. Materials and Methods We recruited patients from two consanguineous families with a clinical diagnosis of PCD and collected their clinical history, laboratory tests, and radiographic data. Sequencing and bioinformatics analysis were then performed. Immunofluorescence and high-speed microscope analysis were used to support the pathogenicity of the variant. Results Proband 1, a 26-year-old female from family I, exhibited scoliosis, bronchiectasis, sinusitis, situs inversus, and infertility. We found a novel homozygous missense variant in DNAAF2, c.491T>C, p.(Leu164Pro) in this patient. Subsequent immunofluorescence indicated the absence of outer dynein arm and inner dynein arm of cilia, and high-speed microscopy analysis showed that the most of the cilia are static, which support the pathogenicity of this variant. Proband 2, a 53-year-old female, presented with bronchiectasis, sinusitis, and infertility. In this patient, a new homozygous frameshift variant DNAAF2, c.822del, p.(Ala275Profs*10) was identified. The disease-causing variants mentioned above are not included in the current authorized genetic databases. Conclusion Our findings expand the spectrum of DNAAF2 variants and link DNAAF2 to female infertility and likely scoliosis in patients with PCD.
Collapse
Affiliation(s)
- Chenyang Lu
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Research Unit of Respiratory Disease, Central South University, Changsha, People's Republic of China.,Hunan Diagnosis and Treatment Center of Respiratory Disease, Changsha, People's Republic of China
| | - Danhui Yang
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Research Unit of Respiratory Disease, Central South University, Changsha, People's Republic of China.,Hunan Diagnosis and Treatment Center of Respiratory Disease, Changsha, People's Republic of China
| | - Cheng Lei
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Research Unit of Respiratory Disease, Central South University, Changsha, People's Republic of China.,Hunan Diagnosis and Treatment Center of Respiratory Disease, Changsha, People's Republic of China
| | - Rongchun Wang
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Research Unit of Respiratory Disease, Central South University, Changsha, People's Republic of China.,Hunan Diagnosis and Treatment Center of Respiratory Disease, Changsha, People's Republic of China
| | - Ting Guo
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Research Unit of Respiratory Disease, Central South University, Changsha, People's Republic of China.,Hunan Diagnosis and Treatment Center of Respiratory Disease, Changsha, People's Republic of China
| | - Hong Luo
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Research Unit of Respiratory Disease, Central South University, Changsha, People's Republic of China.,Hunan Diagnosis and Treatment Center of Respiratory Disease, Changsha, People's Republic of China
| |
Collapse
|
57
|
Jansen K, Constantine A, Condliffe R, Tulloh R, Clift P, Moledina S, Wort SJ, Dimopoulos K. Pulmonary arterial hypertension in adults with congenital heart disease: markers of disease severity, management of advanced heart failure and transplantation. Expert Rev Cardiovasc Ther 2021; 19:837-855. [PMID: 34511015 DOI: 10.1080/14779072.2021.1977124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Pulmonary arterial hypertension associated with congenital heart disease (PAH-CHD) is a progressive, life-limiting disease. AREAS COVERED In this paper, we review the classification and pathophysiology of PAH-CHD, including the mechanisms of disease progression and multisystem effects of disease. We evaluate current strategies of risk stratification and the use of biological markers of disease severity, and review principles of management of PAH-CHD. The indications, timing, and the content of advanced heart failure assessment and transplant listing are discussed, along with a review of the types of transplant and other forms of available circulatory support in this group of patients. Finally, the integral role of advance care planning and palliative care is discussed. EXPERT OPINION/COMMENTARY All patients with PAH-CHD should be followed up in expert centers, where they can receive appropriate risk assessment, PAH therapy, and supportive care. Referral for transplant assessment should be considered if there continue to be clinical high-risk features, persistent symptoms, or acute heart failure decompensation despite appropriate PAH specific therapy. Expert management of PAH-CHD patients, therefore, requires vigilance for these features, along with a close relationship with local advanced heart failure services and a working knowledge of listing criteria, which may disadvantage congenital heart disease patients.
Collapse
Affiliation(s)
- Katrijn Jansen
- Adult Congenital and Paediatric Heart Unit, Freeman Hospital, Newcastle upon Tyne Hospitals Nhs Foundation Trust, Newcastle upon Tyne, UK.,Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew Constantine
- Adult Congenital Heart Centre and Centre for Pulmonary Hypertension, Royal Brompton Hospital, London, UK.,National Heart and Lung Institute, Imperial College London, UK
| | - Robin Condliffe
- Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, UK
| | - Robert Tulloh
- Department of Congenital Heart Disease, Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, UK
| | - Paul Clift
- Department of Cardiology, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Shahin Moledina
- National Paediatric Pulmonary Hypertension Service Uk, Great Ormond Street Hospital for Children Nhs Foundation Trust, London, UK.,Institute of Cardiovascular Science, University College London, UK
| | - S John Wort
- Adult Congenital Heart Centre and Centre for Pulmonary Hypertension, Royal Brompton Hospital, London, UK.,National Heart and Lung Institute, Imperial College London, UK
| | - Konstantinos Dimopoulos
- Adult Congenital Heart Centre and Centre for Pulmonary Hypertension, Royal Brompton Hospital, London, UK.,National Heart and Lung Institute, Imperial College London, UK
| |
Collapse
|
58
|
Bajolle F, Malekzadeh-Milani S, Lévy M, Bonnet D. Multifactorial origin of pulmonary hypertension in a child with congenital heart disease, Down syndrome, and BMPR-2 mutation. Pulm Circ 2021; 11:20458940211027433. [PMID: 34285797 PMCID: PMC8264736 DOI: 10.1177/20458940211027433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/05/2021] [Indexed: 11/25/2022] Open
Abstract
A late preterm infant had pulmonary hypertension caused by a variety of mechanisms leading to complex management. This child had complete atrioventricular septal defect associated with mild left ventricular hypoplasia and Down syndrome diagnosed prenatally. The mother had been treated by antiretroviral HIV treatment during pregnancy. Aortic coarctation was diagnosed and rapidly repaired. After surgery, he required noninvasive ventilation for persisting elevated PCO2. Pulmonary CT scan showed normal bronchial tree, lung parenchymal abnormalities with mosaic aspect and hyperlucent zones, and indirect signs of lung hypoplasia with peripheral microbubbles. During follow-up, severe pulmonary hypertension was diagnosed on echocardiography without recoarctation, significant intracardiac shunting or diastolic dysfunction. The patient died after four months unable to be weaned from noninvasive ventilation. Post mortem lung biopsy showed abnormally muscularized arterioles with intimal fibrosis and pulmonary immaturity. Gentetic screening identified a BMPR-2 mutation. This patient illustrates the multifactorial origin of pulmonary hypertension in the neonatal period. The respective contribution of left-to-right shunt, post-capillary obstruction, and abnormally elevated pulmonary vascular resistances led to perform right heart catheterization to exclude excessive shunting and restrictive physiology of the left heart. Subjects with Down syndrome are also highly susceptible to decreased lung vascular and alveolar growth, which may increase the risk for pulmonary hypertension and lung hypoplasia. This case highlights two issues. The first one is that right heart catheterization should be discussed in neonates with unexplained pulmonary hypertension and the second is to extend indications of genetic testing for pulmonary hypertension genes in neonates who have unusual course of neonatal pulmonary hypertension, particularly in the setting of associated congenital heart disease (CHD).
Collapse
Affiliation(s)
- Fanny Bajolle
- M3C-Necker, Centre de Référence Malformations Cardiaques Congénitales Complexes, Hôpital Universitaire Necker-Enfants malades, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - S Malekzadeh-Milani
- M3C-Necker, Centre de Référence Malformations Cardiaques Congénitales Complexes, Hôpital Universitaire Necker-Enfants malades, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - M Lévy
- M3C-Necker, Centre de Référence Malformations Cardiaques Congénitales Complexes, Hôpital Universitaire Necker-Enfants malades, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - D Bonnet
- M3C-Necker, Centre de Référence Malformations Cardiaques Congénitales Complexes, Hôpital Universitaire Necker-Enfants malades, Assistance Publique - Hôpitaux de Paris, Paris, France.,Université de Paris, Paris, France
| |
Collapse
|
59
|
van der Feen DE, Bossers GPL, Hagdorn QAJ, Moonen JR, Kurakula K, Szulcek R, Chappell J, Vallania F, Donato M, Kok K, Kohli JS, Petersen AH, van Leusden T, Demaria M, Goumans MJTH, De Boer RA, Khatri P, Rabinovitch M, Berger RMF, Bartelds B. Cellular senescence impairs the reversibility of pulmonary arterial hypertension. Sci Transl Med 2021; 12:12/554/eaaw4974. [PMID: 32727916 DOI: 10.1126/scitranslmed.aaw4974] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 10/26/2019] [Accepted: 06/04/2020] [Indexed: 12/24/2022]
Abstract
Pulmonary arterial hypertension (PAH) in congenital cardiac shunts can be reversed by hemodynamic unloading (HU) through shunt closure. However, this reversibility potential is lost beyond a certain point in time. The reason why PAH becomes irreversible is unknown. In this study, we used MCT+shunt-induced PAH in rats to identify a dichotomous reversibility response to HU, similar to the human situation. We compared vascular profiles of reversible and irreversible PAH using RNA sequencing. Cumulatively, we report that loss of reversibility is associated with a switch from a proliferative to a senescent vascular phenotype and confirmed markers of senescence in human PAH-CHD tissue. In vitro, we showed that human pulmonary endothelial cells of patients with PAH are more vulnerable to senescence than controls in response to shear stress and confirmed that the senolytic ABT263 induces apoptosis in senescent, but not in normal, endothelial cells. To support the concept that vascular cell senescence is causal to the irreversible nature of end-stage PAH, we targeted senescence using ABT263 and induced reversal of the hemodynamic and structural changes associated with severe PAH refractory to HU. The factors that drive the transition from a reversible to irreversible pulmonary vascular phenotype could also explain the irreversible nature of other PAH etiologies and provide new leads for pharmacological reversal of end-stage PAH.
Collapse
Affiliation(s)
- Diederik E van der Feen
- Center for Congenital Heart Diseases, University Medical Center Groningen, 9713 GZ Groningen, Netherlands.
| | - Guido P L Bossers
- Center for Congenital Heart Diseases, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Quint A J Hagdorn
- Center for Congenital Heart Diseases, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Jan-Renier Moonen
- Department of Pediatrics, Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Kondababu Kurakula
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Robert Szulcek
- Department of Pulmonology, VU University Medical Center, 1081 HV Amsterdam, Netherlands
| | - James Chappell
- Department of Pediatrics, Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Francesco Vallania
- Institute for Immunity, Transplantation and Infection, Stanford School of Medicine, Stanford, CA 94305, USA.,Stanford Center of Biomedical Informatics Research, Department of Medicine, Stanford, CA 94305, USA
| | - Michele Donato
- Institute for Immunity, Transplantation and Infection, Stanford School of Medicine, Stanford, CA 94305, USA.,Stanford Center of Biomedical Informatics Research, Department of Medicine, Stanford, CA 94305, USA
| | - Klaas Kok
- Department of Genetics, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Jaskaren S Kohli
- European Research Institute for the Biology of Ageing, 9700 AD Groningen, Netherlands
| | - Arjen H Petersen
- Department of Medical Biology, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Tom van Leusden
- Department of Experimental Cardiology, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Marco Demaria
- European Research Institute for the Biology of Ageing, 9700 AD Groningen, Netherlands
| | - Marie-José T H Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Rudolf A De Boer
- Department of Experimental Cardiology, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Stanford School of Medicine, Stanford, CA 94305, USA.,Stanford Center of Biomedical Informatics Research, Department of Medicine, Stanford, CA 94305, USA
| | - Marlene Rabinovitch
- Department of Pediatrics, Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Rolf M F Berger
- Center for Congenital Heart Diseases, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Beatrijs Bartelds
- Center for Congenital Heart Diseases, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| |
Collapse
|
60
|
Breidbart E, Deng L, Lanzano P, Fan X, Guo J, Leibel RL, LeDuc CA, Chung WK. Frequency and characterization of mutations in genes in a large cohort of patients referred to MODY registry. J Pediatr Endocrinol Metab 2021; 34:633-638. [PMID: 33852230 PMCID: PMC8970616 DOI: 10.1515/jpem-2020-0501] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 02/12/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES There have been few large-scale studies utilizing exome sequencing for genetically undiagnosed maturity onset diabetes of the young (MODY), a monogenic form of diabetes that is under-recognized. We describe a cohort of 160 individuals with suspected monogenic diabetes who were genetically assessed for mutations in genes known to cause MODY. METHODS We used a tiered testing approach focusing initially on GCK and HNF1A and then expanding to exome sequencing for those individuals without identified mutations in GCK or HNF1A. The average age of onset of hyperglycemia or diabetes diagnosis was 19 years (median 14 years) with an average HbA1C of 7.1%. RESULTS Sixty (37.5%) probands had heterozygous likely pathogenic/pathogenic variants in one of the MODY genes, 90% of which were in GCK or HNF1A. Less frequently, mutations were identified in PDX1, HNF4A, HNF1B, and KCNJ11. For those probands with available family members, 100% of the variants segregated with diabetes in the family. Cascade genetic testing in families identified 75 additional family members with a familial MODY mutation. CONCLUSIONS Our study is one of the largest and most ethnically diverse studies using exome sequencing to assess MODY genes. Tiered testing is an effective strategy to genetically diagnose atypical diabetes, and familial cascade genetic testing identified on average one additional family member with monogenic diabetes for each mutation identified in a proband.
Collapse
Affiliation(s)
- Emily Breidbart
- Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes, NYU School of Medicine, NY, NY USA
| | - Liyong Deng
- Department of Pediatrics, Division of Molecular Genetics Columbia University Medical Center, NY, NY USA
| | - Patricia Lanzano
- Department of Pediatrics, Division of Molecular Genetics Columbia University Medical Center, NY, NY USA
| | - Xiao Fan
- Department of Pediatrics, Division of Molecular Genetics Columbia University Medical Center, NY, NY USA
| | - Jiancheng Guo
- Department of Pediatrics, Division of Molecular Genetics Columbia University Medical Center, NY, NY USA
| | - Rudolph L. Leibel
- Department of Pediatrics, Division of Molecular Genetics Columbia University Medical Center, NY, NY USA
| | - Charles A. LeDuc
- Department of Pediatrics, Division of Molecular Genetics Columbia University Medical Center, NY, NY USA
| | - Wendy K. Chung
- Department of Pediatrics, Division of Molecular Genetics Columbia University Medical Center, NY, NY USA
| |
Collapse
|
61
|
Zhu N, Swietlik EM, Welch CL, Pauciulo MW, Hagen JJ, Zhou X, Guo Y, Karten J, Pandya D, Tilly T, Lutz KA, Martin JM, Treacy CM, Rosenzweig EB, Krishnan U, Coleman AW, Gonzaga-Jauregui C, Lawrie A, Trembath RC, Wilkins MR, Morrell NW, Shen Y, Gräf S, Nichols WC, Chung WK. Rare variant analysis of 4241 pulmonary arterial hypertension cases from an international consortium implicates FBLN2, PDGFD, and rare de novo variants in PAH. Genome Med 2021; 13:80. [PMID: 33971972 PMCID: PMC8112021 DOI: 10.1186/s13073-021-00891-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 04/19/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a lethal vasculopathy characterized by pathogenic remodeling of pulmonary arterioles leading to increased pulmonary pressures, right ventricular hypertrophy, and heart failure. PAH can be associated with other diseases (APAH: connective tissue diseases, congenital heart disease, and others) but often the etiology is idiopathic (IPAH). Mutations in bone morphogenetic protein receptor 2 (BMPR2) are the cause of most heritable cases but the vast majority of other cases are genetically undefined. METHODS To identify new risk genes, we utilized an international consortium of 4241 PAH cases with exome or genome sequencing data from the National Biological Sample and Data Repository for PAH, Columbia University Irving Medical Center, and the UK NIHR BioResource - Rare Diseases Study. The strength of this combined cohort is a doubling of the number of IPAH cases compared to either national cohort alone. We identified protein-coding variants and performed rare variant association analyses in unrelated participants of European ancestry, including 1647 IPAH cases and 18,819 controls. We also analyzed de novo variants in 124 pediatric trios enriched for IPAH and APAH-CHD. RESULTS Seven genes with rare deleterious variants were associated with IPAH with false discovery rate smaller than 0.1: three known genes (BMPR2, GDF2, and TBX4), two recently identified candidate genes (SOX17, KDR), and two new candidate genes (fibulin 2, FBLN2; platelet-derived growth factor D, PDGFD). The new genes were identified based solely on rare deleterious missense variants, a variant type that could not be adequately assessed in either cohort alone. The candidate genes exhibit expression patterns in lung and heart similar to that of known PAH risk genes, and most variants occur in conserved protein domains. For pediatric PAH, predicted deleterious de novo variants exhibited a significant burden compared to the background mutation rate (2.45×, p = 2.5e-5). At least eight novel pediatric candidate genes carrying de novo variants have plausible roles in lung/heart development. CONCLUSIONS Rare variant analysis of a large international consortium identified two new candidate genes-FBLN2 and PDGFD. The new genes have known functions in vasculogenesis and remodeling. Trio analysis predicted that ~ 15% of pediatric IPAH may be explained by de novo variants.
Collapse
Affiliation(s)
- Na Zhu
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Emilia M Swietlik
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Carrie L Welch
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jacob J Hagen
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Xueya Zhou
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Yicheng Guo
- Department of Systems Biology, Columbia University, New York, NY, USA
| | | | - Divya Pandya
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Tobias Tilly
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Katie A Lutz
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jennifer M Martin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, UK
| | - Carmen M Treacy
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Erika B Rosenzweig
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
| | - Usha Krishnan
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
| | - Anna W Coleman
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Richard C Trembath
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Martin R Wilkins
- National Heart & Lung Institute, Imperial College London, London, UK
| | | | | | | | | | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, UK
- Addenbrooke's Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
- Royal Papworth Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - Yufeng Shen
- Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
62
|
Barradas-Pires A, Constantine A, Dimopoulos K. Preventing disease progression in Eisenmenger syndrome. Expert Rev Cardiovasc Ther 2021; 19:501-518. [PMID: 33853494 DOI: 10.1080/14779072.2021.1917995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Eisenmenger syndrome describes a condition in which a congenital heart defect has caused severe pulmonary vascular disease, resulting in reversed (right-left) or bidirectional shunting and chronic cyanosis.Areas covered: In this paper, the progression of congenital heart defects to Eisenmenger syndrome, including early screening, diagnosis and operability are covered. The mechanisms of disease progression in Eisenmenger syndrome and management strategies to combat this, including the role of pulmonary arterial hypertension therapies, are also discussed.Expert opinion/commentary: Patients with congenital heart disease (CHD) are at increased risk of developing pulmonary arterial hypertension with Eisenmenger syndrome being its extreme manifestation. All CHD patients should be regularly assessed for pulmonary hypertension. Once Eisenmenger syndrome develops, shunt closure should be avoided. The clinical manifestations of Eisenmenger syndrome are driven by the systemic effects of the pulmonary hypertension, congenital defect and long-standing cyanosis. Expert care is essential for avoiding pitfalls and preventing disease progression in this severe chronic condition, which is associated with significant morbidity and mortality. Pulmonary arterial hypertension therapies have been used alongside supportive care to improve the quality of life, exercise tolerance and the outcome of these patients, although the optimal timing for their introduction and escalation remains uncertain.
Collapse
Affiliation(s)
- Ana Barradas-Pires
- Department of Cardiology, Adult Congenital Heart Centre and National Centre for Pulmonary Hypertension, Royal Brompton Hospital, London, UK
| | - Andrew Constantine
- Department of Cardiology, Adult Congenital Heart Centre and National Centre for Pulmonary Hypertension, Royal Brompton Hospital, London, UK.,Biomedical Research Unit, National Heart & Lung Institute, Imperial College London, UK
| | - Konstantinos Dimopoulos
- Department of Cardiology, Adult Congenital Heart Centre and National Centre for Pulmonary Hypertension, Royal Brompton Hospital, London, UK.,Biomedical Research Unit, National Heart & Lung Institute, Imperial College London, UK
| |
Collapse
|
63
|
Wang TM, Wang SS, Xu YJ, Zhao CM, Qiao XH, Yang CX, Liu XY, Yang YQ. SOX17 Loss-of-Function Mutation Underlying Familial Pulmonary Arterial Hypertension. Int Heart J 2021; 62:566-574. [PMID: 33952808 DOI: 10.1536/ihj.20-711] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Pulmonary arterial hypertension (PAH) refers to a rare, progressive disorder that is characterized by occlusive pulmonary vascular remodeling, resulting in increased pulmonary arterial pressure, right-sided heart failure, and eventual death. Emerging evidence from genetic investigations of pediatric-onset PAH highlights the strong genetic basis underpinning PAH, and deleterious variants in multiple genes have been found to cause PAH. Nevertheless, PAH is of substantial genetic heterogeneity, and the genetic defects underlying PAH in the overwhelming majority of cases remain elusive. In this investigation, a consanguineous family suffering from PAH transmitted as an autosomal-dominant trait was identified. Through whole-exome sequencing and bioinformatic analyses as well as Sanger sequencing analyses of the PAH family, a novel heterozygous SOX17 mutation, NM_022454.4: c.379C>T; p. (Gln127*), was found to co-segregate with the disease in the family, with complete penetrance. The nonsense mutation was neither observed in 612 unrelated healthy volunteers nor retrieved in the population genetic databases encompassing the Genome Aggregation Database, the Exome Aggregation Consortium database, and the Single Nucleotide Polymorphism database. Biological analyses using a dual-luciferase reporter assay system revealed that the Gln127*-mutant SOX17 protein lost the ability to transcriptionally activate its target gene NOTCH1. Moreover, the Gln127*-mutant SOX17 protein exhibited no inhibitory effect on the function of CTNNB1-encode β-catenin, which is a key player in vascular morphogenesis. This research firstly links SOX17 loss-of-function mutation to familial PAH, which provides novel insight into the molecular pathogenesis of PAH, suggesting potential implications for genetic and prognostic risk evaluation as well as personalized prophylaxis of the family members affected with PAH.
Collapse
Affiliation(s)
- Tian-Ming Wang
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine
| | - Shan-Shan Wang
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University
| | - Cui-Mei Zhao
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine
| | - Xiao-Hui Qiao
- Department of Pediatric Internal Medicine, Ningbo Women & Children's Hospital
| | - Chen-Xi Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University
| | - Xing-Yuan Liu
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University.,Cardiovascular Research Laboratory, Shanghai Fifth People's Hospital, Fudan University.,Central Laboratory, Shanghai Fifth People's Hospital, Fudan University
| |
Collapse
|
64
|
Pienkos S, Gallego N, Condon DF, Cruz-Utrilla A, Ochoa N, Nevado J, Arias P, Agarwal S, Patel H, Chakraborty A, Lapunzina P, Escribano P, Tenorio-Castaño J, de Jesús Pérez VA. Novel TNIP2 and TRAF2 Variants Are Implicated in the Pathogenesis of Pulmonary Arterial Hypertension. Front Med (Lausanne) 2021; 8:625763. [PMID: 33996849 PMCID: PMC8119639 DOI: 10.3389/fmed.2021.625763] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Pulmonary arterial hypertension (PAH) is a rare disease characterized by pulmonary vascular remodeling and right heart failure. Specific genetic variants increase the incidence of PAH in carriers with a family history of PAH, those who suffer from certain medical conditions, and even those with no apparent risk factors. Inflammation and immune dysregulation are related to vascular remodeling in PAH, but whether genetic susceptibility modifies the PAH immune response is unclear. TNIP2 and TRAF2 encode for immunomodulatory proteins that regulate NF-κB activation, a transcription factor complex associated with inflammation and vascular remodeling in PAH. Methods: Two unrelated families with PAH cases underwent whole-exome sequencing (WES). A custom pipeline for variant prioritization was carried out to obtain candidate variants. To determine the impact of TNIP2 and TRAF2 in cell proliferation, we performed an MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] assay on healthy lung pericytes transfected with siRNA specific for each gene. To measure the effect of loss of TNIP2 and TRAF2 on NF-kappa-beta (NF-κB) activity, we measured levels of Phospho-p65-NF-κB in siRNA-transfected pericytes using western immunoblotting. Results: We discovered a novel missense variant in the TNIP2 gene in two affected individuals from the same family. The two patients had a complex form of PAH with interatrial communication and scleroderma. In the second family, WES of the proband with PAH and primary biliary cirrhosis revealed a de novo protein-truncating variant in the TRAF2. The knockdown of TNIP2 and TRAF2 increased NF-κB activity in healthy lung pericytes, which correlated with a significant increase in proliferation over 24 h. Conclusions: We have identified two rare novel variants in TNIP2 and TRAF2 using WES. We speculate that loss of function in these genes promotes pulmonary vascular remodeling by allowing overactivation of the NF-κB signaling activity. Our findings support a role for WES in helping identify novel genetic variants associated with dysfunctional immune response in PAH.
Collapse
Affiliation(s)
- Shaun Pienkos
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Natalia Gallego
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - David F. Condon
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Alejandro Cruz-Utrilla
- Pulmonary Hypertension Unit, Department of Cardiology, Hospital Universitario Doce de Octubre, Madrid, Spain
- Centro de Investigación Biomedica en Red en Enfermedades Cardiovasculares, Instituto de Salud Carlos III (CIBERCV), Madrid, Spain
| | - Nuria Ochoa
- Pulmonary Hypertension Unit, Department of Cardiology, Hospital Universitario Doce de Octubre, Madrid, Spain
- Centro de Investigación Biomedica en Red en Enfermedades Cardiovasculares, Instituto de Salud Carlos III (CIBERCV), Madrid, Spain
| | - Julián Nevado
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Intellectual Disability, TeleHealth, Autism and Congenital Anomalies (ITHACA), European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Brussels, Belgium
| | - Pedro Arias
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Intellectual Disability, TeleHealth, Autism and Congenital Anomalies (ITHACA), European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Brussels, Belgium
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Hiral Patel
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Ananya Chakraborty
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Pablo Lapunzina
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Intellectual Disability, TeleHealth, Autism and Congenital Anomalies (ITHACA), European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Brussels, Belgium
| | - Pilar Escribano
- Pulmonary Hypertension Unit, Department of Cardiology, Hospital Universitario Doce de Octubre, Madrid, Spain
- Centro de Investigación Biomedica en Red en Enfermedades Cardiovasculares, Instituto de Salud Carlos III (CIBERCV), Madrid, Spain
| | - Jair Tenorio-Castaño
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Intellectual Disability, TeleHealth, Autism and Congenital Anomalies (ITHACA), European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Brussels, Belgium
| | - Vinicio A. de Jesús Pérez
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
65
|
Massadeh S, Albeladi M, Albesher N, Alhabshan F, Kampe KD, Chaikhouni F, Kabbani MS, Beetz C, Alaamery M. Novel Autosomal Recessive Splice-Altering Variant in PRKD1 Is Associated with Congenital Heart Disease. Genes (Basel) 2021; 12:genes12050612. [PMID: 33919081 PMCID: PMC8143129 DOI: 10.3390/genes12050612] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/14/2021] [Accepted: 04/18/2021] [Indexed: 12/13/2022] Open
Abstract
Congenital heart defects (CHDs) are the most common types of birth defects, and global incidence of CHDs is on the rise. Despite the prevalence of CHDs, the genetic determinants of the defects are still in the process of being identified. Herein, we report a consanguineous Saudi family with three CHD affected daughters. We used whole exome sequencing (WES) to investigate the genetic cause of CHDs in the affected daughters. We found that all affected individuals were homozygous for a novel splice-altering variant (NM_001330069.1: c.265-1G>T) of PRKD1, which encodes a calcium/calmodulin-dependent protein kinase in the heart. The homozygous variant was found in the affected patients with Pulmonary Stenosis (PS), Truncus Arteriosis (TA), and Atrial Septal Defect (ASD). Based on the family’s pedigree, the variant acts in an autosomal recessive manner, which makes it the second autosomal recessive variant of PRKD1 to be identified with a link to CHDs, while all other previously described variants act dominantly. Interestingly, the father of the affected daughters was also homozygous for the variant, though he was asymptomatic of CHDs himself. Since both of his sisters had CHDs as well, this raises the possibility that the novel PRKD1 variant may undergo autosomal recessive inheritance mode with gender limitation. This finding confirms that CHD can be associated with both dominant and recessive mutations of the PRKD1 gene, and it provides a new insight to genotype–phenotype association between PRKD1 and CHDs. To our knowledge, this is the first report of this specific PRKD1 mutation associated with CHDs.
Collapse
Affiliation(s)
- Salam Massadeh
- Developmental Medicine Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard- Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia; (S.M.); (M.A.); (N.A.)
- KACST-BWH Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
- Saudi Human Genome Project (SHGP), King Abdulaziz City for Science and Technology (KACST), Satellite Lab at King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
| | - Maha Albeladi
- Developmental Medicine Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard- Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia; (S.M.); (M.A.); (N.A.)
- KACST-BWH Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Nour Albesher
- Developmental Medicine Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard- Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia; (S.M.); (M.A.); (N.A.)
- KACST-BWH Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Fahad Alhabshan
- Department of Cardiac Sciences, Ministry of the National Guard—Health Affairs, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia; (F.A.); (F.C.); (M.S.K.)
| | | | - Farah Chaikhouni
- Department of Cardiac Sciences, Ministry of the National Guard—Health Affairs, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia; (F.A.); (F.C.); (M.S.K.)
| | - Mohamed S. Kabbani
- Department of Cardiac Sciences, Ministry of the National Guard—Health Affairs, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia; (F.A.); (F.C.); (M.S.K.)
| | | | - Manal Alaamery
- Developmental Medicine Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard- Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia; (S.M.); (M.A.); (N.A.)
- KACST-BWH Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
- Saudi Human Genome Project (SHGP), King Abdulaziz City for Science and Technology (KACST), Satellite Lab at King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
- Correspondence:
| |
Collapse
|
66
|
Zhao L, Jiang WF, Yang CX, Qiao Q, Xu YJ, Shi HY, Qiu XB, Wu SH, Yang YQ. SOX17 loss-of-function variation underlying familial congenital heart disease. Eur J Med Genet 2021; 64:104211. [PMID: 33794346 DOI: 10.1016/j.ejmg.2021.104211] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/11/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
As the most prevalent form of human birth defect, congenital heart disease (CHD) contributes to substantial morbidity, mortality and socioeconomic burden worldwide. Aggregating evidence has convincingly demonstrated that genetic defects exert a pivotal role in the pathogenesis of CHD, and causative mutations in multiple genes have been causally linked to CHD. Nevertheless, CHD is of pronounced genetic heterogeneity, and the genetic components underpinning CHD in the overwhelming majority of patients remain obscure. In this research, a four-generation consanguineous family suffering from CHD transmitted in an autosomal dominant mode was recruited. By whole-exome sequencing and bioinformatics analyses as well as Sanger sequencing analyses of the family members, a new heterozygous SOX17 variation, NM_022454.4: c.553G > T; p.(Glu185*), was identified to co-segregate with CHD in the family, with complete penetrance. The nonsense variation was neither detected in 310 unrelated healthy volunteers used as controls nor retrieved in such population genetics databases as the Exome Aggregation Consortium database, Genome Aggregation Database, and the Single Nucleotide Polymorphism database. Functional assays by utilizing a dual-luciferase reporter assay system unveiled that the Glu185*-mutant SOX17 protein had no transcriptional activity on its two target genes NOTCH1 and GATA4, which have been reported to cause CHD. Furthermore, the mutation abrogated the synergistic transactivation between SOX17 and NKX2.5, another established CHD-causing transcription factor. These findings firstly indicate SOX17 loss-of-function mutation predisposes to familial CHD, which adds novel insight to the molecular mechanism of CHD, implying potential implications for genetic risk appraisal and individualized prophylaxis of the family members affected with CHD.
Collapse
Affiliation(s)
- Lan Zhao
- Department of Cardiology, Yantaishan Hospital, Yantai, 264003, Shandong Province, China
| | - Wei-Feng Jiang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Chen-Xi Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Qi Qiao
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Hong-Yu Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Shao-Hui Wu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Cardiovascular Research Laboratory, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Central Laboratory, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
67
|
Macias D, Moore S, Crosby A, Southwood M, Du X, Tan H, Xie S, Vassallo A, Wood AJT, Wallace EM, Cowburn AS. Targeting HIF2α-ARNT hetero-dimerisation as a novel therapeutic strategy for pulmonary arterial hypertension. Eur Respir J 2021; 57:13993003.02061-2019. [PMID: 32972983 PMCID: PMC7930471 DOI: 10.1183/13993003.02061-2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 08/26/2020] [Indexed: 12/13/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a destructive disease of the pulmonary vasculature often leading to right heart failure and death. Current therapeutic intervention strategies only slow disease progression. The role of aberrant hypoxia-inducible factor (HIF)2α stability and function in the initiation and development of pulmonary hypertension (PH) has been an area of intense interest for nearly two decades.Here we determine the effect of a novel HIF2α inhibitor (PT2567) on PH disease initiation and progression, using two pre-clinical models of PH. Haemodynamic measurements were performed, followed by collection of heart, lung and blood for pathological, gene expression and biochemical analysis. Blood outgrowth endothelial cells from idiopathic PAH patients were used to determine the impact of HIF2α-inhibition on endothelial function.Global inhibition of HIF2a reduced pulmonary vascular haemodynamics and pulmonary vascular remodelling in both su5416/hypoxia prevention and intervention models. PT2567 intervention reduced the expression of PH-associated target genes in both lung and cardiac tissues and restored plasma nitrite concentration. Treatment of monocrotaline-exposed rodents with PT2567 reduced the impact on cardiovascular haemodynamics and promoted a survival advantage. In vitro, loss of HIF2α signalling in human pulmonary arterial endothelial cells suppresses target genes associated with inflammation, and PT2567 reduced the hyperproliferative phenotype and overactive arginase activity in blood outgrowth endothelial cells from idiopathic PAH patients. These data suggest that targeting HIF2α hetero-dimerisation with an orally bioavailable compound could offer a new therapeutic approach for PAH. Future studies are required to determine the role of HIF in the heterogeneous PAH population.
Collapse
Affiliation(s)
- David Macias
- CRUK Cambridge Centre Early Detection Programme, Dept of Oncology, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK.,Both authors contributed equally
| | - Stephen Moore
- Dept of Medicine, University of Cambridge, Cambridge, UK.,Both authors contributed equally
| | - Alexi Crosby
- Dept of Medicine, University of Cambridge, Cambridge, UK
| | - Mark Southwood
- Dept of Pathology, Papworth Hospital National Health Service Foundation Trust, Cambridge, UK
| | - Xinlin Du
- Peloton Therapeutics Inc. (a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA), Dallas, TX, USA
| | - Huiling Tan
- Peloton Therapeutics Inc. (a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA), Dallas, TX, USA
| | - Shanhai Xie
- Peloton Therapeutics Inc. (a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA), Dallas, TX, USA
| | | | | | - Eli M Wallace
- Peloton Therapeutics Inc. (a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA), Dallas, TX, USA
| | - Andrew S Cowburn
- Dept of Medicine, University of Cambridge, Cambridge, UK .,National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
68
|
Welch CL, Austin ED, Chung WK. Genes that drive the pathobiology of pediatric pulmonary arterial hypertension. Pediatr Pulmonol 2021; 56:614-620. [PMID: 31917901 PMCID: PMC7343584 DOI: 10.1002/ppul.24637] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/27/2019] [Indexed: 12/15/2022]
Abstract
Emerging data from studies of pediatric-onset pulmonary arterial hypertension (PAH) indicate that the genomics of pediatric PAH is different than that of adults. There is a greater genetic burden in children, with rare genetic factors contributing to at least 35% of pediatric-onset idiopathic PAH (IPAH) compared with ~11% of adult-onset IPAH. De novo variants are the most frequent genetic cause of PAH in children, likely contributing to ~15% of all cases. Rare deleterious variants in bone morphogenetic protein receptor 2 (BMPR2) contribute to pediatric-onset familial PAH and IPAH with similar frequency as adult-onset. While likely gene-disrupting (LGD) variants in BMPR2 contribute across the lifespan, damaging missense variants are more frequent in early-onset PAH. Rare deleterious variants in T-box 4-containing protein (TBX4) are more common in pediatric-compared with adult-onset PAH, explaining ~8% of pediatric IPAH. PAH associated with congenital heart disease (APAH-CHD) and other developmental disorders account for a large proportion of pediatric PAH. SRY-related HMG box transcription factor (SOX17) was recently identified as an APAH-CHD risk gene, contributing less frequently to IPAH, with a greater prevalence of rare deleterious variants in children compared with adults. The differences in genetic burden and genes underlying pediatric- vs adult-onset PAH indicate that genetic information relevant to pediatric PAH cannot be extrapolated from adult studies. Large cohorts of pediatric-onset PAH are necessary to identify the unique etiological differences of PAH in children, as well as the natural history and response to therapy.
Collapse
Affiliation(s)
- Carrie L Welch
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Eric D Austin
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York.,Department of Medicine, Columbia University Medical Center, New York, New York
| |
Collapse
|
69
|
Wu Y, Wharton J, Walters R, Vasilaki E, Aman J, Zhao L, Wilkins MR, Rhodes CJ. The pathophysiological role of novel pulmonary arterial hypertension gene SOX17. Eur Respir J 2021; 58:13993003.04172-2020. [PMID: 33632800 DOI: 10.1183/13993003.04172-2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/08/2021] [Indexed: 11/05/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease predominantly targeting pre-capillary blood vessels. Adverse structural remodelling and increased pulmonary vascular resistance result in cardiac hypertrophy and ultimately failure of the right ventricle. Recent whole-genome and whole-exome sequencing studies have identified SOX17 as a novel risk gene in PAH, with a dominant mode of inheritance and incomplete penetrance. Rare deleterious variants in the gene and more common variants in upstream enhancer sites have both been associated with the disease, and a deficiency of SOX17 expression may predispose to PAH. This review aims to consolidate the evidence linking genetic variants in SOX17 to PAH, and explores the numerous targets and effects of the transcription factor, focusing on the pulmonary vasculature and the pathobiology of PAH.
Collapse
Affiliation(s)
- Yukyee Wu
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Rachel Walters
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Eleni Vasilaki
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Jurjan Aman
- National Heart and Lung Institute, Imperial College London, London, UK.,VUmc, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Lan Zhao
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
70
|
Virani SS, Alonso A, Aparicio HJ, Benjamin EJ, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Cheng S, Delling FN, Elkind MSV, Evenson KR, Ferguson JF, Gupta DK, Khan SS, Kissela BM, Knutson KL, Lee CD, Lewis TT, Liu J, Loop MS, Lutsey PL, Ma J, Mackey J, Martin SS, Matchar DB, Mussolino ME, Navaneethan SD, Perak AM, Roth GA, Samad Z, Satou GM, Schroeder EB, Shah SH, Shay CM, Stokes A, VanWagner LB, Wang NY, Tsao CW. Heart Disease and Stroke Statistics-2021 Update: A Report From the American Heart Association. Circulation 2021; 143:e254-e743. [PMID: 33501848 DOI: 10.1161/cir.0000000000000950] [Citation(s) in RCA: 3345] [Impact Index Per Article: 836.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2021 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population, an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, the global burden of cardiovascular disease, and further evidence-based approaches to changing behaviors related to cardiovascular disease. RESULTS Each of the 27 chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policy makers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
71
|
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: 30] [Impact Index Per Article: 7.5] [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
|
72
|
Varshney A, Chahal G, Santos L, Stolper J, Hallab JC, Nim HT, Nikolov M, Yip A, Ramialison M. Human Cardiac Transcription Factor Networks. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11597-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
73
|
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: 4.2] [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
|
74
|
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: 12] [Impact Index Per Article: 2.4] [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
|
75
|
Gelinas SM, Benson CE, Khan MA, Berger RMF, Trembath RC, Machado RD, Southgate L. Whole Exome Sequence Analysis Provides Novel Insights into the Genetic Framework of Childhood-Onset Pulmonary Arterial Hypertension. Genes (Basel) 2020; 11:E1328. [PMID: 33187088 PMCID: PMC7696319 DOI: 10.3390/genes11111328] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) describes a rare, progressive vascular disease caused by the obstruction of pulmonary arterioles, typically resulting in right heart failure. Whilst PAH most often manifests in adulthood, paediatric disease is considered to be a distinct entity with increased morbidity and often an unexplained resistance to current therapies. Recent genetic studies have substantially increased our understanding of PAH pathogenesis, providing opportunities for molecular diagnosis and presymptomatic genetic testing in families. However, the genetic architecture of childhood-onset PAH remains relatively poorly characterised. We sought to investigate a previously unsolved paediatric cohort (n = 18) using whole exome sequencing to improve the molecular diagnosis of childhood-onset PAH. Through a targeted investigation of 26 candidate genes, we applied a rigorous variant filtering methodology to enrich for rare, likely pathogenic variants. This analysis led to the detection of novel PAH risk alleles in five genes, including the first identification of a heterozygous ATP13A3 mutation in childhood-onset disease. In addition, we provide the first independent validation of BMP10 and PDGFD as genetic risk factors for PAH. These data provide a molecular diagnosis in 28% of paediatric cases, reflecting the increased genetic burden in childhood-onset disease and highlighting the importance of next-generation sequencing approaches to diagnostic surveillance.
Collapse
Affiliation(s)
- Simone M. Gelinas
- Genetics Research Centre, Molecular and Clinical Sciences Research Institute, St George’s University of London, London SW17 0RE, UK; (S.M.G.); (C.E.B.); (M.A.K.)
| | - Clare E. Benson
- Genetics Research Centre, Molecular and Clinical Sciences Research Institute, St George’s University of London, London SW17 0RE, UK; (S.M.G.); (C.E.B.); (M.A.K.)
| | - Mohammed A. Khan
- Genetics Research Centre, Molecular and Clinical Sciences Research Institute, St George’s University of London, London SW17 0RE, UK; (S.M.G.); (C.E.B.); (M.A.K.)
| | - Rolf M. F. Berger
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children’s Hospital, University Medical Center Groningen, 9700 RB Groningen, The Netherlands;
| | - Richard C. Trembath
- Department of Medical & Molecular Genetics, Faculty of Life Sciences & Medicine, King’s College London, London SE1 9RT, UK;
| | - Rajiv D. Machado
- Genetics Research Centre, Molecular and Clinical Sciences Research Institute, St George’s University of London, London SW17 0RE, UK; (S.M.G.); (C.E.B.); (M.A.K.)
- Institute of Medical and Biomedical Education, St George’s University of London, London SW17 0RE, UK
| | - Laura Southgate
- Genetics Research Centre, Molecular and Clinical Sciences Research Institute, St George’s University of London, London SW17 0RE, UK; (S.M.G.); (C.E.B.); (M.A.K.)
- Department of Medical & Molecular Genetics, Faculty of Life Sciences & Medicine, King’s College London, London SE1 9RT, UK;
| |
Collapse
|
76
|
Welch CL, Chung WK. Genetics and Genomics of Pediatric Pulmonary Arterial Hypertension. Genes (Basel) 2020; 11:E1213. [PMID: 33081265 PMCID: PMC7603012 DOI: 10.3390/genes11101213] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/02/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease with high mortality despite recent therapeutic advances. The disease is caused by both genetic and environmental factors and likely gene-environment interactions. While PAH can manifest across the lifespan, pediatric-onset disease is particularly challenging because it is frequently associated with a more severe clinical course and comorbidities including lung/heart developmental anomalies. In light of these differences, it is perhaps not surprising that emerging data from genetic studies of pediatric-onset PAH indicate that the genetic basis is different than that of adults. There is a greater genetic burden in children, with rare genetic factors contributing to ~42% of pediatric-onset PAH compared to ~12.5% of adult-onset PAH. De novo variants are frequently associated with PAH in children and contribute to at least 15% of all pediatric cases. The standard of medical care for pediatric PAH patients is based on extrapolations from adult data. However, increased etiologic heterogeneity, poorer prognosis, and increased genetic burden for pediatric-onset PAH calls for a dedicated pediatric research agenda to improve molecular diagnosis and clinical management. A genomics-first approach will improve the understanding of pediatric PAH and how it is related to other rare pediatric genetic disorders.
Collapse
Affiliation(s)
- Carrie L Welch
- Department of Pediatrics, Irving Medical Center, Columbia University, 1150 St. Nicholas Avenue, New York, NY 10032, USA
| | - Wendy K Chung
- Department of Pediatrics, Irving Medical Center, Columbia University, 1150 St. Nicholas Avenue, New York, NY 10032, USA
- Department of Medicine, Irving Medical Center, Columbia University, 622 W 168th St, New York, NY 10032, USA
| |
Collapse
|
77
|
Xin J, Zhang H, He Y, Duren Z, Bai C, Chen L, Luo X, Yan DS, Zhang C, Zhu X, Yuan Q, Feng Z, Cui C, Qi X, Ouzhuluobu, Wong WH, Wang Y, Su B. Chromatin accessibility landscape and regulatory network of high-altitude hypoxia adaptation. Nat Commun 2020; 11:4928. [PMID: 33004791 PMCID: PMC7529806 DOI: 10.1038/s41467-020-18638-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/03/2020] [Indexed: 12/27/2022] Open
Abstract
High-altitude adaptation of Tibetans represents a remarkable case of natural selection during recent human evolution. Previous genome-wide scans found many non-coding variants under selection, suggesting a pressing need to understand the functional role of non-coding regulatory elements (REs). Here, we generate time courses of paired ATAC-seq and RNA-seq data on cultured HUVECs under hypoxic and normoxic conditions. We further develop a variant interpretation methodology (vPECA) to identify active selected REs (ASREs) and associated regulatory network. We discover three causal SNPs of EPAS1, the key adaptive gene for Tibetans. These SNPs decrease the accessibility of ASREs with weakened binding strength of relevant TFs, and cooperatively down-regulate EPAS1 expression. We further construct the downstream network of EPAS1, elucidating its roles in hypoxic response and angiogenesis. Collectively, we provide a systematic approach to interpret phenotype-associated noncoding variants in proper cell types and relevant dynamic conditions, to model their impact on gene regulation.
Collapse
Affiliation(s)
- Jingxue Xin
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
- Bio-X Program, Stanford University, Stanford, CA, 94305, USA
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Hui Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
| | - Yaoxi He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Zhana Duren
- Departments of Statistics, Stanford University, Stanford, CA, 94305, USA
- Center for Human Genetics and Department of Genetics and Biochemistry, Clemson University, Greenwood, SC, 29646, USA
| | - Caijuan Bai
- High Altitude Medical Research Center, School of Medicine, Tibetan University, 850000, Lhasa, China
| | - Lang Chen
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Xin Luo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Dong-Sheng Yan
- School of Mathematical Science, Inner Mongolia University, 010021, Huhhot, China
| | - Chaoyu Zhang
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Xiang Zhu
- Departments of Statistics, Stanford University, Stanford, CA, 94305, USA
| | - Qiuyue Yuan
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Zhanying Feng
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Chaoying Cui
- High Altitude Medical Research Center, School of Medicine, Tibetan University, 850000, Lhasa, China
| | - Xuebin Qi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
| | - Ouzhuluobu
- High Altitude Medical Research Center, School of Medicine, Tibetan University, 850000, Lhasa, China
| | - Wing Hung Wong
- Bio-X Program, Stanford University, Stanford, CA, 94305, USA.
- Departments of Statistics, Stanford University, Stanford, CA, 94305, USA.
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Yong Wang
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China.
- University of Chinese Academy of Sciences, 100101, Beijing, China.
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 330106, Hangzhou, China.
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China.
| |
Collapse
|
78
|
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: 31] [Impact Index Per Article: 6.2] [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
|
79
|
Nees SN, Chung WK. Genetic Basis of Human Congenital Heart Disease. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a036749. [PMID: 31818857 DOI: 10.1101/cshperspect.a036749] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Congenital heart disease (CHD) is the most common major congenital anomaly with an incidence of ∼1% of live births and is a significant cause of birth defect-related mortality. The genetic mechanisms underlying the development of CHD are complex and remain incompletely understood. Known genetic causes include all classes of genetic variation including chromosomal aneuploidies, copy number variants, and rare and common single-nucleotide variants, which can be either de novo or inherited. Among patients with CHD, ∼8%-12% have a chromosomal abnormality or aneuploidy, between 3% and 25% have a copy number variation, and 3%-5% have a single-gene defect in an established CHD gene with higher likelihood of identifying a genetic cause in patients with nonisolated CHD. These genetic variants disrupt or alter genes that play an important role in normal cardiac development and in some cases have pleiotropic effects on other organs. This work reviews some of the most common genetic causes of CHD as well as what is currently known about the underlying mechanisms.
Collapse
Affiliation(s)
| | - Wendy K Chung
- Department of Pediatrics.,Department of Medicine, Columbia University Irving Medical Center, New York, New York 10032, USA
| |
Collapse
|
80
|
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: 5.0] [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
|
81
|
|
82
|
Sun X, Sun BL, Babicheva A, Vanderpool R, Oita RC, Casanova N, Tang H, Gupta A, Lynn H, Gupta G, Rischard F, Sammani S, Kempf CL, Moreno-Vinasco L, Ahmed M, Camp SM, Wang J, Desai AA, Yuan JXJ, Garcia JGN. Direct Extracellular NAMPT Involvement in Pulmonary Hypertension and Vascular Remodeling. Transcriptional Regulation by SOX and HIF-2α. Am J Respir Cell Mol Biol 2020; 63:92-103. [PMID: 32142369 PMCID: PMC7328254 DOI: 10.1165/rcmb.2019-0164oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 03/05/2020] [Indexed: 12/21/2022] Open
Abstract
We previously demonstrated involvement of NAMPT (nicotinamide phosphoribosyltransferase) in pulmonary arterial hypertension (PAH) and now examine NAMPT regulation and extracellular NAMPT's (eNAMPT's) role in PAH vascular remodeling. NAMPT transcription and protein expression in human lung endothelial cells were assessed in response to PAH-relevant stimuli (PDGF [platelet-derived growth factor], VEGF [vascular endothelial growth factor], TGF-β1 [transforming growth factor-β1], and hypoxia). Endothelial-to-mesenchymal transition was detected by SNAI1 (snail family transcriptional repressor 1) and PECAM1 (platelet endothelial cell adhesion molecule 1) immunofluorescence. An eNAMPT-neutralizing polyclonal antibody was tested in a PAH model of monocrotaline challenge in rats. Plasma eNAMPT concentrations, significantly increased in patients with idiopathic pulmonary arterial hypertension, were highly correlated with indices of PAH severity. eNAMPT increased endothelial-to-mesenchymal transition, and each PAH stimulus significantly increased endothelial cell NAMPT promoter activity involving transcription factors STAT5 (signal transducer and activator of transcription 5), SOX18 (SRY-box transcription factor 18), and SOX17 (SRY-box transcription factor 17), a PAH candidate gene newly defined by genome-wide association study. The hypoxia-induced transcription factor HIF-2α (hypoxia-inducible factor-2α) also potently regulated NAMPT promoter activity, and HIF-2α binding sites were identified between -628 bp and -328 bp. The PHD2 (prolyl hydroxylase domain-containing protein 2) inhibitor FG-4592 significantly increased NAMPT promoter activity and protein expression in an HIF-2α-dependent manner. Finally, the eNAMPT-neutralizing polyclonal antibody significantly reduced monocrotaline-induced vascular remodeling, PAH hemodynamic alterations, and NF-κB activation. eNAMPT is a novel and attractive therapeutic target essential to PAH vascular remodeling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Mohamed Ahmed
- Department of Pediatrics, University of Arizona Health Sciences, Tucson, Arizona
| | | | | | | | | | | |
Collapse
|
83
|
Affiliation(s)
- Florent Soubrier
- Genetics Department, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, UMR_S 1166-ICAN, Sorbonne-Université, INSERM, Paris, France
| |
Collapse
|
84
|
Tolchin D, Yeager JP, Prasad P, Dorrani N, Russi AS, Martinez-Agosto JA, Haseeb A, Angelozzi M, Santen GWE, Ruivenkamp C, Mercimek-Andrews S, Depienne C, Kuechler A, Mikat B, Ludecke HJ, Bilan F, Le Guyader G, Gilbert-Dussardier B, Keren B, Heide S, Haye D, Van Esch H, Keldermans L, Ortiz D, Lancaster E, Krantz ID, Krock BL, Pechter KB, Arkader A, Medne L, DeChene ET, Calpena E, Melistaccio G, Wilkie AOM, Suri M, Foulds N, Begtrup A, Henderson LB, Forster C, Reed P, McDonald MT, McConkie-Rosell A, Thevenon J, Le Tanno P, Coutton C, Tsai ACH, Stewart S, Maver A, Gorazd R, Pichon O, Nizon M, Cogné B, Isidor B, Martin-Coignard D, Stoeva R, Lefebvre V, Le Caignec C. De Novo SOX6 Variants Cause a Neurodevelopmental Syndrome Associated with ADHD, Craniosynostosis, and Osteochondromas. Am J Hum Genet 2020; 106:830-845. [PMID: 32442410 PMCID: PMC7273536 DOI: 10.1016/j.ajhg.2020.04.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/24/2020] [Indexed: 12/21/2022] Open
Abstract
SOX6 belongs to a family of 20 SRY-related HMG-box-containing (SOX) genes that encode transcription factors controlling cell fate and differentiation in many developmental and adult processes. For SOX6, these processes include, but are not limited to, neurogenesis and skeletogenesis. Variants in half of the SOX genes have been shown to cause severe developmental and adult syndromes, referred to as SOXopathies. We here provide evidence that SOX6 variants also cause a SOXopathy. Using clinical and genetic data, we identify 19 individuals harboring various types of SOX6 alterations and exhibiting developmental delay and/or intellectual disability; the individuals are from 17 unrelated families. Additional, inconstant features include attention-deficit/hyperactivity disorder (ADHD), autism, mild facial dysmorphism, craniosynostosis, and multiple osteochondromas. All variants are heterozygous. Fourteen are de novo, one is inherited from a mosaic father, and four offspring from two families have a paternally inherited variant. Intragenic microdeletions, balanced structural rearrangements, frameshifts, and nonsense variants are predicted to inactivate the SOX6 variant allele. Four missense variants occur in residues and protein regions highly conserved evolutionarily. These variants are not detected in the gnomAD control cohort, and the amino acid substitutions are predicted to be damaging. Two of these variants are located in the HMG domain and abolish SOX6 transcriptional activity in vitro. No clear genotype-phenotype correlations are found. Taken together, these findings concur that SOX6 haploinsufficiency leads to a neurodevelopmental SOXopathy that often includes ADHD and abnormal skeletal and other features.
Collapse
Affiliation(s)
- Dara Tolchin
- Department of Surgery, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jessica P Yeager
- Department of Surgery, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Priya Prasad
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Naghmeh Dorrani
- Division of Medical Genetics, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alvaro Serrano Russi
- Division of Medical Genetics, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Julian A Martinez-Agosto
- Division of Medical Genetics, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Abdul Haseeb
- Department of Surgery, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Marco Angelozzi
- Department of Surgery, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - G W E Santen
- Department of Clinical Genetics, Leiden University Medical Centre, 2300 LC Leiden, the Netherlands
| | - Claudia Ruivenkamp
- Department of Clinical Genetics, Leiden University Medical Centre, 2300 LC Leiden, the Netherlands
| | - Saadet Mercimek-Andrews
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Christel Depienne
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Alma Kuechler
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Barbara Mikat
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Hermann-Josef Ludecke
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | - Frederic Bilan
- Neurovascular Unit and Cognitive Disorders (EA-3808 NEUVACOD), Université de Poitiers, 86073 Poitiers, France; Service de Génétique Clinique, Centre Hospitalier Universitaire de Poitiers, 86021 Poitiers, France
| | - Gwenael Le Guyader
- Neurovascular Unit and Cognitive Disorders (EA-3808 NEUVACOD), Université de Poitiers, 86073 Poitiers, France; Service de Génétique Clinique, Centre Hospitalier Universitaire de Poitiers, 86021 Poitiers, France
| | - Brigitte Gilbert-Dussardier
- Neurovascular Unit and Cognitive Disorders (EA-3808 NEUVACOD), Université de Poitiers, 86073 Poitiers, France; Service de Génétique Clinique, Centre Hospitalier Universitaire de Poitiers, 86021 Poitiers, France
| | - Boris Keren
- Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique, 75013 Paris, France
| | - Solveig Heide
- Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique, 75013 Paris, France
| | - Damien Haye
- Service de Génétique, Centre Hospitalier Universitaire de Nice Hôpital de l'Archet 2,151 route Saint Antoine de la Ginestière, 062002 Nice, France
| | - Hilde Van Esch
- Center for Human Genetics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Liesbeth Keldermans
- Laboratory for Molecular Diagnosis, Center for Human Genetics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Damara Ortiz
- University of Pittsburgh Medical Center, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Emily Lancaster
- University of Pittsburgh Medical Center, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Ian D Krantz
- Roberts Individualized Medical Genetics Center, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bryan L Krock
- Division of Genomic Diagnostics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kieran B Pechter
- Division of Genomic Diagnostics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexandre Arkader
- Department of Surgery, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Livija Medne
- Roberts Individualized Medical Genetics Center, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Elizabeth T DeChene
- Division of Genomic Diagnostics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eduardo Calpena
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Giada Melistaccio
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Andrew O M Wilkie
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK; Clinical Genetics Service, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham NG5 1PB, UK
| | - Mohnish Suri
- Clinical Genetics Service, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham NG5 1PB, UK
| | - Nicola Foulds
- Wessex Clinical Genetics Services, University Hospital Southampton NHS Foundation Trust, Southampton SO16 5YA, UK
| | | | | | | | | | - Marie T McDonald
- Division of Medical Genetics, Department of Pediatrics, Duke University, Durham, NC 27707, USA
| | - Allyn McConkie-Rosell
- Division of Medical Genetics, Department of Pediatrics, Duke University, Durham, NC 27707, USA
| | - Julien Thevenon
- Service de Génétique, Génomique, et Procréation, Centre Hospitalier Universitaire Grenoble Alpes, 38700 La Tronche, France; INSERM 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Université Grenoble Alpes, 38706 Grenoble, France
| | - Pauline Le Tanno
- Service de Génétique, Génomique, et Procréation, Centre Hospitalier Universitaire Grenoble Alpes, 38700 La Tronche, France; INSERM 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Université Grenoble Alpes, 38706 Grenoble, France
| | - Charles Coutton
- Service de Génétique, Génomique, et Procréation, Centre Hospitalier Universitaire Grenoble Alpes, 38700 La Tronche, France; INSERM 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Université Grenoble Alpes, 38706 Grenoble, France
| | - Anne C H Tsai
- Section of Genetics, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Sarah Stewart
- Section of Genetics, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Ales Maver
- Clinical Institute of Medical Genetics, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Rudolf Gorazd
- Clinical Institute of Medical Genetics, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Olivier Pichon
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, 44000 Nantes, France
| | - Mathilde Nizon
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, 44000 Nantes, France; Université de Nantes, CNRS, INSERM, L'Institut du Thorax, 44000 Nantes, France
| | - Benjamin Cogné
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, 44000 Nantes, France; Université de Nantes, CNRS, INSERM, L'Institut du Thorax, 44000 Nantes, France
| | - Bertrand Isidor
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, 44000 Nantes, France; Université de Nantes, CNRS, INSERM, L'Institut du Thorax, 44000 Nantes, France
| | | | - Radka Stoeva
- Service de Cytogénétique, Centre Hospitalier Universitaire de Le Mans, 72037 Le Mans, France
| | - Véronique Lefebvre
- Department of Surgery, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Cédric Le Caignec
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, 44000 Nantes, France; Centre Hospitalier Universitaire Toulouse, Service de Génétique Médicale, 31000 Toulouse, France.
| |
Collapse
|
85
|
Opotowsky AR, Cedars A, Kutty S. Atrial septal defects and pulmonary hemodynamics: a time for holey reflection. Am J Physiol Heart Circ Physiol 2020; 318:H1159-H1161. [DOI: 10.1152/ajpheart.00226.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Alexander R. Opotowsky
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ari Cedars
- The Helen B. Taussig Heart Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shelby Kutty
- The Helen B. Taussig Heart Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
86
|
Eichstaedt CA, Benjamin N, Grünig E. Genetics of pulmonary hypertension and high-altitude pulmonary edema. J Appl Physiol (1985) 2020; 128:1432-1438. [PMID: 32324476 DOI: 10.1152/japplphysiol.00113.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Heritable pulmonary arterial hypertension (PAH) is an autosomal dominantly inherited disease caused by mutations in the bone morphogenetic protein receptor 2 (BMPR2) gene and/or genes of its signaling pathway in ~85% of patients. A genetic predisposition to high-altitude pulmonary edema (HAPE) has long been suspected because of familial HAPE cases, but very few possibly disease-causing mutations have been identified to date. This minireview provides an overview of genetic analyses investigating common polymorphisms in HAPE-susceptible patients and the directed identification of disease-causing mutations in PAH patients. Increased pulmonary artery pressure is highlighted as an overlapping clinical feature of the two diseases. Moreover, studies showing increased pulmonary artery pressures in HAPE-susceptible patients during exercise or hypoxia as well as in healthy BMPR2 mutation carriers are illustrated. Finally, high-altitude pulmonary hypertension is introduced and future research perspectives outlined.
Collapse
Affiliation(s)
- Christina A Eichstaedt
- Centre for Pulmonary Hypertension, Thoraxklinik Heidelberg gGmbH at Heidelberg University Hospital, Heidelberg Germany.,Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Nicola Benjamin
- Centre for Pulmonary Hypertension, Thoraxklinik Heidelberg gGmbH at Heidelberg University Hospital, Heidelberg Germany.,Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Ekkehard Grünig
- Centre for Pulmonary Hypertension, Thoraxklinik Heidelberg gGmbH at Heidelberg University Hospital, Heidelberg Germany.,Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
87
|
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: 0.8] [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
|
88
|
Harbaum L, Rhodes CJ, Otero-Núñez P, Wharton J, Wilkins MR. The application of 'omics' to pulmonary arterial hypertension. Br J Pharmacol 2020; 178:108-120. [PMID: 32201940 DOI: 10.1111/bph.15056] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/03/2020] [Accepted: 02/18/2020] [Indexed: 12/14/2022] Open
Abstract
Recent genome-wide analyses of rare and common sequence variations have brought greater clarity to the genetic architecture of pulmonary arterial hypertension and implicated novel genes in disease development. Transcriptional signatures have been reported in whole lung tissue, pulmonary vascular cells and peripheral circulating cells. High-throughput platforms for plasma proteomics and metabolomics have identified novel biomarkers associated with clinical outcomes and provided molecular instruments for risk assessment. There are methodological challenges to integrating these datasets, coupled to statistical power limitations inherent to the study of a rare disease, but the expectation is that this approach will reveal novel druggable targets and biomarkers that will open the way to personalized medicine. Here, we review the current state-of-the-art and future promise of 'omics' in the field of translational medicine in pulmonary arterial hypertension. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.1/issuetoc.
Collapse
Affiliation(s)
- Lars Harbaum
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Pablo Otero-Núñez
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
89
|
Nees SN, Chung WK. The genetics of isolated congenital heart disease. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2020; 184:97-106. [PMID: 31876989 PMCID: PMC8211463 DOI: 10.1002/ajmg.c.31763] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/06/2019] [Accepted: 12/12/2019] [Indexed: 12/12/2022]
Abstract
The genetic mechanisms underlying congenital heart disease (CHD) are complex and remain incompletely understood. The majority of patients with CHD have an isolated heart defect without other organ system involvement, but the genetic basis of isolated CHD has been even more difficult to elucidate compared to syndromic CHD. Our understanding of the genetics of isolated CHD is advancing in large part due to advances in next generation sequencing, and the list of genes associated with CHD is rapidly expanding. Variants in hundreds of genes have been identified that may cause or contribute to CHD, but a genetic cause can still only be identified in about 20-30% of patients. Identifying a genetic cause for CHD can have an impact on clinical outcomes and prognosis and thus it is important for clinicians to understand when and what to test in patients with isolated CHD. This chapter reviews some of the known genetic mechanisms that contribute to isolated inherited and sporadic CHD as well as recommendations for evaluation and genetic testing in patients with isolated CHD.
Collapse
Affiliation(s)
- Shannon N Nees
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
- Department of Medicine, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
90
|
Genetics and Other Omics in Pediatric Pulmonary Arterial Hypertension. Chest 2020; 157:1287-1295. [PMID: 32006592 DOI: 10.1016/j.chest.2020.01.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/09/2019] [Accepted: 01/07/2020] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease with high mortality despite therapeutic advances. Clinical management of children with PAH is particularly challenging because of increased complexity of disease etiology and clinical presentation, and the lack of data from pediatric-specific clinical trials. In children, PAH often develops in association with congenital heart disease and other developmental disorders. Emerging data from genetic studies of pediatric-onset PAH indicate that the genetic basis is different than that of adults. There is a greater genetic burden in children, with rare genetic factors contributing to at least 35% of pediatric-onset idiopathic PAH (IPAH) compared with approximately 11% of adult-onset IPAH. De novo variants are the most frequent monogenetic cause of PAH in children, likely contributing to approximately 15% of all cases. Rare deleterious variants in BMPR2 contribute to pediatric-onset IPAH and familial PAH with similar frequency as adult-onset disease but rarely explain cases of PAH associated with other diseases. Rare deleterious variants in developmental genes-including TBX4, SOX17, and other genes requiring confirmation in larger cohorts-are emerging as important contributors to pediatric-onset disease. Because each causal gene contributes to only a small number of cases, large cohorts of pediatric-onset PAH are needed to further identify the unique etiologic differences of PAH in children. We propose a genetics-first approach followed by focused phenotyping of pediatric patients grouped by genetic diagnosis to define endophenotypes that can be used to improve risk stratification and treatment.
Collapse
|
91
|
Virani SS, Alonso A, Benjamin EJ, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Chang AR, Cheng S, Delling FN, Djousse L, Elkind MSV, Ferguson JF, Fornage M, Khan SS, Kissela BM, Knutson KL, Kwan TW, Lackland DT, Lewis TT, Lichtman JH, Longenecker CT, Loop MS, Lutsey PL, Martin SS, Matsushita K, Moran AE, Mussolino ME, Perak AM, Rosamond WD, Roth GA, Sampson UKA, Satou GM, Schroeder EB, Shah SH, Shay CM, Spartano NL, Stokes A, Tirschwell DL, VanWagner LB, Tsao CW. Heart Disease and Stroke Statistics-2020 Update: A Report From the American Heart Association. Circulation 2020; 141:e139-e596. [PMID: 31992061 DOI: 10.1161/cir.0000000000000757] [Citation(s) in RCA: 5186] [Impact Index Per Article: 1037.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports on the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2020 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population, metrics to assess and monitor healthy diets, an enhanced focus on social determinants of health, a focus on the global burden of cardiovascular disease, and further evidence-based approaches to changing behaviors, implementation strategies, and implications of the American Heart Association's 2020 Impact Goals. RESULTS Each of the 26 chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policy makers, media professionals, clinicians, healthcare administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
92
|
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: 76] [Impact Index Per Article: 12.7] [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
|
93
|
Kumar P, Mistri TK. Transcription factors in SOX family: Potent regulators for cancer initiation and development in the human body. Semin Cancer Biol 2019; 67:105-113. [PMID: 31288067 DOI: 10.1016/j.semcancer.2019.06.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 06/17/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022]
Abstract
Transcription factors (TFs) have a key role in controlling the gene regulatory network that sustains explicit cell states in humans. However, an uncontrolled regulation of these genes potentially results in a wide range of diseases, including cancer. Genes of the SOX family are indeed crucial as deregulation of SOX family TFs can potentially lead to changes in cell fate as well as irregular cell growth. SOX TFs are a conserved group of transcriptional regulators that mediate DNA binding through a highly conserved high-mobility group (HMG) domain. Accumulating evidence demonstrates that cell fate and differentiation in major developmental processes are controlled by SOX TFs. Besides; numerous reports indicate that both up- and down-regulation of SOX TFs may induce cancer progression. In this review, we discuss the involvement of key TFs of SOX family in human cancers.
Collapse
Affiliation(s)
- Prasann Kumar
- The Division of Research and Development, Lovely Professional University, Jalandhar, Punjab, 144411, India; The Department of Agronomy, Lovely Professional University, Jalandhar, Punjab, 144411, India
| | - Tapan Kumar Mistri
- The Division of Research and Development, Lovely Professional University, Jalandhar, Punjab, 144411, India; The Department of Chemistry, Lovely Professional University, Jalandhar, Punjab, 144411, India.
| |
Collapse
|
94
|
Angelozzi M, Lefebvre V. SOXopathies: Growing Family of Developmental Disorders Due to SOX Mutations. Trends Genet 2019; 35:658-671. [PMID: 31288943 DOI: 10.1016/j.tig.2019.06.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022]
Abstract
The SRY-related (SOX) transcription factor family pivotally contributes to determining cell fate and identity in many lineages. Since the original discovery that SRY deletions cause sex reversal, mutations in half of the 20 human SOX genes have been associated with rare congenital disorders, henceforward called SOXopathies. Mutations are generally de novo, heterozygous, and inactivating, revealing gene haploinsufficiency, but other types, including duplications, have been reported too. Missense variants primarily target the HMG domain, the SOX hallmark that mediates DNA binding and bending, nuclear trafficking, and protein-protein interactions. We here review key clinical and molecular features of SOXopathies and discuss the prospect that the disease family likely involves more SOX genes and larger clinical and genetic spectrums than currently appreciated.
Collapse
Affiliation(s)
- Marco Angelozzi
- Department of Surgery/Division of Orthopaedic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Véronique Lefebvre
- Department of Surgery/Division of Orthopaedic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
95
|
Elliott CG, Austin ED, Badesch D, Badlam J, Benza RL, Chung WK, Farber HW, Feldkircher K, Frost AE, Poms AD, Lutz KA, Pauciulo MW, Yu C, Nichols WC. United States Pulmonary Hypertension Scientific Registry (USPHSR): rationale, design, and clinical implications. Pulm Circ 2019; 9:2045894019851696. [PMID: 31099303 PMCID: PMC6540712 DOI: 10.1177/2045894019851696] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Diagnostic World Health Organization (WHO) Group 1 pulmonary arterial hypertension (PAH) and Diagnostic Group 1' pulmonary veno-occlusive disease (PVOD) and/or pulmonary capillary hemangiomatosis (PCH) are progressive and fatal disorders. Past registries provided important insights into these disorders, but did not include hormonal exposures or genomic data. The United States Pulmonary Hypertension Scientific Registry (USPHSR) will provide demographic, physiologic, anorexigen and hormone exposure, genomic, and survival data in the current therapeutic era for 499 patients diagnosed with PAH, PVOD, or PCH. The USPHSR also will explore the relationship between pharmacologic, non-pharmacologic, and dietary hormonal exposures and the increased risk for women to develop idiopathic or heritable PAH.
Collapse
Affiliation(s)
- C Gregory Elliott
- 1 Intermountain Medical Center Department of Medicine and the University of Utah, Pulmonary Division, Salt Lake City, UT, USA
| | - Eric D Austin
- 2 Vanderbilt University Medical Center Department of Pediatrics, Nashville, TN, USA
| | | | | | | | - Wendy K Chung
- 6 Columbia University Medical Center, New York, NY, USA
| | | | | | - Adaani E Frost
- 9 Houston Methodist Hospital Lung Center, Houston, TX, USA
| | - Abby D Poms
- 9 Houston Methodist Hospital Lung Center, Houston, TX, USA
| | - Katie A Lutz
- 10 Division of Human Genetics, Cincinnati Children's Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael W Pauciulo
- 10 Division of Human Genetics, Cincinnati Children's Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Chang Yu
- 11 Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - William C Nichols
- 10 Division of Human Genetics, Cincinnati Children's Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
96
|
Genetic determinants of risk in pulmonary arterial hypertension: international genome-wide association studies and meta-analysis. THE LANCET. RESPIRATORY MEDICINE 2019; 7:227-238. [PMID: 30527956 PMCID: PMC6391516 DOI: 10.1016/s2213-2600(18)30409-0] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/20/2018] [Accepted: 09/26/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Rare genetic variants cause pulmonary arterial hypertension, but the contribution of common genetic variation to disease risk and natural history is poorly characterised. We tested for genome-wide association for pulmonary arterial hypertension in large international cohorts and assessed the contribution of associated regions to outcomes. METHODS We did two separate genome-wide association studies (GWAS) and a meta-analysis of pulmonary arterial hypertension. These GWAS used data from four international case-control studies across 11 744 individuals with European ancestry (including 2085 patients). One GWAS used genotypes from 5895 whole-genome sequences and the other GWAS used genotyping array data from an additional 5849 individuals. Cross-validation of loci reaching genome-wide significance was sought by meta-analysis. Conditional analysis corrected for the most significant variants at each locus was used to resolve signals for multiple associations. We functionally annotated associated variants and tested associations with duration of survival. All-cause mortality was the primary endpoint in survival analyses. FINDINGS A locus near SOX17 (rs10103692, odds ratio 1·80 [95% CI 1·55-2·08], p=5·13 × 10-15) and a second locus in HLA-DPA1 and HLA-DPB1 (collectively referred to as HLA-DPA1/DPB1 here; rs2856830, 1·56 [1·42-1·71], p=7·65 × 10-20) within the class II MHC region were associated with pulmonary arterial hypertension. The SOX17 locus had two independent signals associated with pulmonary arterial hypertension (rs13266183, 1·36 [1·25-1·48], p=1·69 × 10-12; and rs10103692). Functional and epigenomic data indicate that the risk variants near SOX17 alter gene regulation via an enhancer active in endothelial cells. Pulmonary arterial hypertension risk variants determined haplotype-specific enhancer activity, and CRISPR-mediated inhibition of the enhancer reduced SOX17 expression. The HLA-DPA1/DPB1 rs2856830 genotype was strongly associated with survival. Median survival from diagnosis in patients with pulmonary arterial hypertension with the C/C homozygous genotype was double (13·50 years [95% CI 12·07 to >13·50]) that of those with the T/T genotype (6·97 years [6·02-8·05]), despite similar baseline disease severity. INTERPRETATION This is the first study to report that common genetic variation at loci in an enhancer near SOX17 and in HLA-DPA1/DPB1 is associated with pulmonary arterial hypertension. Impairment of SOX17 function might be more common in pulmonary arterial hypertension than suggested by rare mutations in SOX17. Further studies are needed to confirm the association between HLA typing or rs2856830 genotyping and survival, and to determine whether HLA typing or rs2856830 genotyping improves risk stratification in clinical practice or trials. FUNDING UK NIHR, BHF, UK MRC, Dinosaur Trust, NIH/NHLBI, ERS, EMBO, Wellcome Trust, EU, AHA, ACClinPharm, Netherlands CVRI, Dutch Heart Foundation, Dutch Federation of UMC, Netherlands OHRD and RNAS, German DFG, German BMBF, APH Paris, INSERM, Université Paris-Sud, and French ANR.
Collapse
|
97
|
Rosenzweig EB, Abman SH, Adatia I, Beghetti M, Bonnet D, Haworth S, Ivy DD, Berger RMF. Paediatric pulmonary arterial hypertension: updates on definition, classification, diagnostics and management. Eur Respir J 2019; 53:1801916. [PMID: 30545978 PMCID: PMC6351335 DOI: 10.1183/13993003.01916-2018] [Citation(s) in RCA: 359] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 10/09/2018] [Indexed: 12/16/2022]
Abstract
Paediatric pulmonary arterial hypertension (PAH) shares common features of adult disease, but is associated with several additional disorders and challenges that require unique approaches. This article discusses recent advances, ongoing challenges and distinct approaches for the care of children with PAH, as presented by the Paediatric Task Force of the 6th World Symposium on Pulmonary Hypertension. We provide updates of the current definition, epidemiology, classification, diagnostics and treatment of paediatric PAH, and identify critical knowledge gaps. Several features of paediatric PAH including the prominence of neonatal PAH, especially in pre-term infants with developmental lung diseases, and novel genetic causes of paediatric PAH are highlighted. The use of cardiac catheterisation as a diagnostic modality and haemodynamic definitions of PAH, including acute vasoreactivity, are addressed. Updates are provided on issues related to utility of the previous classification system to reflect paediatric-specific aetiologies and approaches to medical and interventional management of PAH, including the Potts shunt. Although a lack of clinical trial data for the use of PAH-targeted therapy persists, emerging data are improving the identification of appropriate targets for goal-oriented therapy in children. Such data will likely improve future clinical trial design to enhance outcomes in paediatric PAH.
Collapse
Affiliation(s)
- Erika B Rosenzweig
- Columbia University Medical Center, New York Presbyterian Hospital, New York, NY, USA
| | - Steven H Abman
- University of Colorado, Children's Hospital Colorado, Denver, CO, USA
| | - Ian Adatia
- Glenwood Children's Heart Clinic, Edmonton, AB, Canada
| | - Maurice Beghetti
- Pediatric Cardiology Unit and Centre Universitaire de Cardiologie et Chirurgie Cardiaque Pédiatrique, University Hospitals of Geneva and Lausanne, Lausanne, Switzerland
| | - Damien Bonnet
- Hôpital Necker Enfants Malades, AP-HP, Université Paris Descartes, Paris, France
| | | | - D Dunbar Ivy
- University of Colorado, Children's Hospital Colorado, Denver, CO, USA
| | - Rolf M F Berger
- Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
98
|
van der Feen DE, Bartelds B, de Boer RA, Berger RMF. Assessment of reversibility in pulmonary arterial hypertension and congenital heart disease. Heart 2018; 105:276-282. [DOI: 10.1136/heartjnl-2018-314025] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/08/2018] [Accepted: 11/03/2018] [Indexed: 12/31/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) in congenital heart disease (CHD) can be reversed by early shunt closure, but this potential is lost beyond a certain point of no return. Therefore, it is crucial to accurately assess the reversibility of this progressive pulmonary arteriopathy in an early stage. Reversibility assessment is currently based on a combination of clinical symptoms and haemodynamic variables such as pulmonary vascular resistance. These measures, however, are of limited predictive value and leave many patients in the grey zone. This review provides a concise overview of the mechanisms involved in flow-dependent progression of PAH in CHD and evaluates existing and future alternatives to more directly investigate the stage of the pulmonary arteriopathy. Structural quantification of the pulmonary arterial tree using fractal branching algorithms, functional imaging with intravascular ultrasound, nuclear imaging, putative new blood biomarkers, genetic testing and the potential for transcriptomic analysis of circulating endothelial cells and educated platelets are being reviewed.
Collapse
|
99
|
Hopper RK, Mullen MP. Genotypes and Phenotypes: Making Progress Toward a Precision Medicine Approach in Pediatric Pulmonary Hypertension. ACTA ACUST UNITED AC 2018. [DOI: 10.21693/1933-088x-17.4.153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Pediatric pulmonary hypertension (PH) is a heterogeneous disease that includes etiologies related to growth and development that are unique to children. Recent pediatric registry studies have characterized diverse phenotypes even within recognized PH subtypes, including PH associated with congenital heart disease and developmental lung disease. Advances in genetics are resulting in increased understanding of the genetic basis for PH, with recent discoveries such as TBX4 mutations specific for pediatric-onset pulmonary arterial hypertension (PAH) and SOX17 related to congenital heart disease–associated PAH. In addition to potential genetic underpinnings, PAH risk and clinical presentation in children with congenital heart disease may vary by cardiac condition, such as single-ventricle physiology or transposition of the great arteries. Growth and development of the pulmonary vasculature likely plays a role in all pediatric PH, which is highlighted by the disruption of normal lung growth in patients with PH related to prematurity and developmental lung disease. These diverse pediatric genotypes and phenotypes underscore a need for an individualized approach to diagnose and treat the complex pediatric PH population. Magnetic resonance imaging (MRI) is increasingly being used to improve clinical characterization of PH in children, with recent identification of specific MRI biomarkers associated with PH severity and outcomes. While much progress has been made, additional understanding of the important genetic causes and developmental concepts in pediatric PH is needed to develop a precision medicine approach to diagnosis and treatment of children with PH.
Collapse
Affiliation(s)
- Rachel K. Hopper
- Division of Cardiology, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital Stanford, Palo Alto, CA
| | - Mary P. Mullen
- Department of Cardiology, Boston Children's Hospital, Boston, MA
| |
Collapse
|