1
|
Shen YH, Ding D, Lian TY, Qiu BC, Yan Y, Wang PW, Zhang WH, Jing ZC. Panorama of artery endothelial cell dysfunction in pulmonary arterial hypertension. J Mol Cell Cardiol 2024; 197:61-77. [PMID: 39437884 DOI: 10.1016/j.yjmcc.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal lung disease characterized by progressive pulmonary vascular remodeling. The initial cause of pulmonary vascular remodeling is the dysfunction of pulmonary arterial endothelial cells (PAECs), manifested by changes in the categorization of cell subtypes, endothelial programmed cell death, such as apoptosis, necroptosis, pyroptosis, ferroptosis, et al., overproliferation, senescence, metabolic reprogramming, endothelial-to-mesenchymal transition, mechanosensitivity, and regulation ability of peripheral cells. Therefore, it is essential to explore the mechanism of endothelial dysfunction in the context of PAH. This review aims to provide a comprehensive understanding of the molecular mechanisms underlying endothelial dysfunction in PAH. We highlight the developmental process of PAECs and changes in PAH and summarise the latest classification of endothelial dysfunction. Our review could offer valuable insights into potential novel EC-specific targets for preventing and treating PAH.
Collapse
Affiliation(s)
- Ying-Huizi Shen
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Dong Ding
- National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tian-Yu Lian
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences, Southern Medical University, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bao-Chen Qiu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Yan
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei-Wen Wang
- National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei-Hua Zhang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China.
| | - Zhi-Cheng Jing
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences, Southern Medical University, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Ejikeme C, Safdar Z. Exploring the pathogenesis of pulmonary vascular disease. Front Med (Lausanne) 2024; 11:1402639. [PMID: 39050536 PMCID: PMC11267418 DOI: 10.3389/fmed.2024.1402639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Pulmonary hypertension (PH) is a complex cardiopulmonary disorder impacting the lung vasculature, resulting in increased pulmonary vascular resistance that leads to right ventricular dysfunction. Pulmonary hypertension comprises of 5 groups (PH group 1 to 5) where group 1 pulmonary arterial hypertension (PAH), results from alterations that directly affect the pulmonary arteries. Although PAH has a complex pathophysiology that is not completely understood, it is known to be a multifactorial disease that results from a combination of genetic, epigenetic and environmental factors, leading to a varied range of symptoms in PAH patients. PAH does not have a cure, its incidence and prevalence continue to increase every year, resulting in higher morbidity and mortality rates. In this review, we discuss the different pathologic mechanisms with a focus on epigenetic modifications and their roles in the development and progression of PAH. These modifications include DNA methylation, histone modifications, and microRNA dysregulation. Understanding these epigenetic modifications will improve our understanding of PAH and unveil novel therapeutic targets, thus steering research toward innovative treatment strategies.
Collapse
Affiliation(s)
| | - Zeenat Safdar
- Department of Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
3
|
Chida-Nagai A, Masaki N, Maeda K, Sasaki K, Sato H, Muneuchi J, Ochiai Y, Murayama H, Tahara M, Shiono A, Shinozuka A, Kono F, Machida D, Toyooka S, Sugimoto S, Nakamura K, Akagi S, Kondo M, Kasahara S, Kotani Y, Koizumi J, Oda K, Harada M, Nakajima D, Murata A, Nagata H, Yatsunami K, Kobayashi T, Matsunaga Y, Inoue T, Yamagishi H, Nakagawa N, Ohtani K, Yamamoto M, Ito Y, Hokosaki T, Kuwahara Y, Masutani S, Nomura K, Wada T, Sawada H, Abiko M, Takahashi T, Ishikawa Y, Okada S, Naitoh A, Toda T, Ando T, Masuzawa A, Hoshino S, Kawada M, Nomura Y, Ueno K, Ohashi N, Tachibana T, Cao Y, Ueda H, Yanagi S, Koide M, Mitsushita N, Higashi K, Minosaki Y, Hayashi T, Okamoto T, Kuraishi K, Ehara E, Ishida H, Horigome H, Murakami T, Takei K, Ishii T, Harada G, Hirata Y, Maeda J, Tatebe S, Ota C, Hayabuchi Y, Sakazaki H, Sasaki T, Hirono K, Suzuki S, Yasuda M, Takeda A, Sawada M, Miyaji K, Kitagawa A, Nakai Y, Kakimoto N, Agematsu K, Manabe A, Saiki Y. Use of the index of pulmonary vascular disease for predicting long-term outcome of pulmonary arterial hypertension associated with congenital heart disease. Front Cardiovasc Med 2023; 10:1212882. [PMID: 37731527 PMCID: PMC10507182 DOI: 10.3389/fcvm.2023.1212882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
Aims Limited data exist on risk factors for the long-term outcome of pulmonary arterial hypertension (PAH) associated with congenital heart disease (CHD-PAH). We focused on the index of pulmonary vascular disease (IPVD), an assessment system for pulmonary artery pathology specimens. The IPVD classifies pulmonary vascular lesions into four categories based on severity: (1) no intimal thickening, (2) cellular thickening of the intima, (3) fibrous thickening of the intima, and (4) destruction of the tunica media, with the overall grade expressed as an additive mean of these scores. This study aimed to investigate the relationship between IPVD and the long-term outcome of CHD-PAH. Methods This retrospective study examined lung pathology images of 764 patients with CHD-PAH aged <20 years whose lung specimens were submitted to the Japanese Research Institute of Pulmonary Vasculature for pulmonary pathological review between 2001 and 2020. Clinical information was collected retrospectively by each attending physician. The primary endpoint was cardiovascular death. Results The 5-year, 10-year, 15-year, and 20-year cardiovascular death-free survival rates for all patients were 92.0%, 90.4%, 87.3%, and 86.1%, respectively. The group with an IPVD of ≥2.0 had significantly poorer survival than the group with an IPVD <2.0 (P = .037). The Cox proportional hazards model adjusted for the presence of congenital anomaly syndromes associated with pulmonary hypertension, and age at lung biopsy showed similar results (hazard ratio 4.46; 95% confidence interval: 1.45-13.73; P = .009). Conclusions The IPVD scoring system is useful for predicting the long-term outcome of CHD-PAH. For patients with an IPVD of ≥2.0, treatment strategies, including choosing palliative procedures such as pulmonary artery banding to restrict pulmonary blood flow and postponement of intracardiac repair, should be more carefully considered.
Collapse
Affiliation(s)
- Ayako Chida-Nagai
- Department of Pediatrics, Hokkaido University, Sapporo, Japan
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Naoki Masaki
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kay Maeda
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Konosuke Sasaki
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroki Sato
- Department of Cardiology and Clinical Examination, Oita University, Yufu, Japan
- Advanced Trauma, Emergency and Critical Care Center, Oita University Hospital, Yufu, Japan
| | - Jun Muneuchi
- Department of Pediatrics, Kyushu Hospital, Japan Community Healthcare Organization, Kitakyushu, Japan
| | - Yoshie Ochiai
- Department of Cardiovascular Surgery, Kyushu Hospital, Japan Community Healthcare Organization, Kitakyushu, Japan
| | - Hiroomi Murayama
- Department of Cardiovascular Surgery, Aichi Children’s Health and Medical Center, Obu, Japan
| | - Masahiro Tahara
- Department of Pediatrics, Tsuchiya General Hospital, Hiroshima, Japan
| | - Atsuko Shiono
- Department of Pediatric Cardiology, Ibaraki Children’s Hospital, Mito, Japan
| | - Atsushi Shinozuka
- Department of Pediatrics, Uji-Tokushukai Medical Center, Kyoto, Japan
| | - Fumihiko Kono
- Department of Diagnostic Pathology, Uji-Tokushukai Medical Center, Kyoto, Japan
| | - Daisuke Machida
- Cardiovascular Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Seiichiro Sugimoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Maiko Kondo
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan
| | - Shingo Kasahara
- Department of Cardiovascular Surgery, Okayama University, Okayama, Japan
| | - Yasuhiro Kotani
- Department of Cardiovascular Surgery, Okayama University, Okayama, Japan
| | - Junichi Koizumi
- Department of Cardiovascular Surgery, Iwate Medical University Hospital, Yahaba, Japan
| | - Katsuhiko Oda
- Department of Cardiovascular Surgery, Iwate Prefectural Central Hospital, Morioka, Japan
| | - Masako Harada
- Division of Pediatrics, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Daisuke Nakajima
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akira Murata
- Department of Cardiovascular Surgery, Kanazawa University, Kanazawa, Japan
| | - Hazumu Nagata
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Yatsunami
- Department of Pediatric Cardiology, Kumamoto City Hospital, Kumamoto, Japan
| | - Tomio Kobayashi
- Division of Cardiology, Gunma Children’s Medical Center, Shibukawa, Japan
| | - Yoshikiyo Matsunaga
- Department of Cardiovascular Surgery,Gunma Children’s Medical Center, Shibukawa, Japan
| | - Takahiro Inoue
- Department of Pediatrics, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hiroyuki Yamagishi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Naomi Nakagawa
- Department of Pediatric Cardiology, Hiroshima City Hospital, Hiroshima, Japan
| | - Katsuki Ohtani
- Department of Pediatric Cardiology, Hirosaki University School of Medicine, Hirosaki, Japan
| | - Masaki Yamamoto
- Department of Pediatrics, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Yushi Ito
- Division of Neonatology, Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Tatsunori Hokosaki
- Department of Pediatric Cardiology, Yokohama City University Hospital, Yokohama, Japan
| | - Yuta Kuwahara
- Department of Pediatric Cardiovascular Surgery, Sakakibara Heart Institute, Tokyo, Japan
| | - Satoshi Masutani
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
- Department of Pediatric Cardiology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Koji Nomura
- Department of Pediatric Cardiovascular Surgery, Saitama Children’s Medical Center, Saitama, Japan
| | - Tsutomu Wada
- Department of Pediatrics, School of Medicine Sapporo Medical University, Sapporo, Japan
| | - Hirofumi Sawada
- Department of Pediatrics, Mie University Graduate School of Medicine, Mie, Japan
| | - Masayuki Abiko
- Department of Pediatrics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Tatsunori Takahashi
- Department of Pediatrics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yuichi Ishikawa
- Department of Pediatrics, Saiseikai Shimonoseki General Hospital, Shimonoseki, Japan
| | - Seigo Okada
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Atsushi Naitoh
- Department of Neonatology, Yamanashi Prefectural Central Hospital, Kofu, Japan
| | - Takako Toda
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Tatsuya Ando
- Department of Paediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Akihiro Masuzawa
- Department of Cardiac Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Shinsuke Hoshino
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Masaaki Kawada
- Department of Cardiac Surgery, Section of Pediatric and Congenital Cardiovascular Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Yuichi Nomura
- Department of Pediatrics, Kagoshima City Hospital, Kagoshima, Japan
| | - Kentaro Ueno
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Naoki Ohashi
- Department of Pediatric Cardiology, Chukyo Children Heart Centre, Community Health Care Organization Chukyo Hospital, Nagoya, Japan
| | - Tsuyoshi Tachibana
- Department of Cardiovascular Surgery, Kanagawa Children’s Medical Center, Yokohama, Japan
| | - Yuchen Cao
- Department of Cardiovascular Surgery, Kanagawa Children’s Medical Center, Yokohama, Japan
| | - Hideaki Ueda
- Department of Pediatric Cardiology, Kanagawa Children’s Medical Center, Yokohama, Japan
| | - Sadamitsu Yanagi
- Department of Pediatric Cardiology, Kanagawa Children’s Medical Center, Yokohama, Japan
| | - Masaaki Koide
- Department of Cardiovascular Surgery, Seirei Hamamatsu General Hospital, Shizuoka, Japan
| | - Norie Mitsushita
- Department of Cardiology, Shizuoka Children’s Hospital, Shizuoka, Japan
| | - Kouji Higashi
- Division of Cardiology, Chiba Children's Hospital, Chiba, Japan
| | - Yoshihiro Minosaki
- Neonatal Intensive Care Unit, Kawaguchi Municipal Medical Center, Kawaguchi, Japan
| | - Tomohiro Hayashi
- Department of Pediatrics, Kurashiki Central Hospital, Okayama, Japan
| | - Takashi Okamoto
- Department of Cardiac Surgery, Daiyukai General Hospital, Ichinomiya, Japan
| | - Kenji Kuraishi
- Department of Pediatric Cardiology and Neonatology, Ogaki Municipal Hospital, Gifu, Japan
| | - Eiji Ehara
- Department of Pediatric Cardiology, Osaka City General Hospital, Osaka, Japan
| | - Hidekazu Ishida
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hitoshi Horigome
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takashi Murakami
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kohta Takei
- Department of Pediatric Cardiology, Nagano Children’s Hospital, Azumino, Japan
| | - Taku Ishii
- Department of Global Health Promotion, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Gen Harada
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Yasutaka Hirata
- Department of Cardiac Surgery, The University of Tokyo Hospital, Tokyo, Japan
| | - Jun Maeda
- Department of Cardiology, Tokyo Metropolitan Children’s Medical Center, Fuchu, Japan
| | - Shunsuke Tatebe
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chiharu Ota
- Department of Pediatrics, Tohoku University Hospital, Sendai, Japan
| | - Yasunobu Hayabuchi
- Department of Pediatrics, School of Medicine, University of Tokushima, Tokushima, Japan
| | - Hisanori Sakazaki
- Department of Pediatric Cardiology, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Takashi Sasaki
- Department of Cardiovascular Surgery, Nippon Medical School, Tokyo, Japan
| | - Keiichi Hirono
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Sayo Suzuki
- Department of Cardiology, Fukuoka Children’s Hospital, Fukuoka, Japan
| | - Masahiro Yasuda
- Department of Pediatrics, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Atsuhito Takeda
- Department of Pediatrics, Hokkaido University, Sapporo, Japan
| | - Madoka Sawada
- Department of Pediatric Cardiology, Hokkaido Medical Center for Child Health and Rehabilitation, Sapporo, Japan
| | - Kagami Miyaji
- Department of Cardiovascular Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Atsushi Kitagawa
- Department of Pediatrics, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yosuke Nakai
- Department of Cardiovascular Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Nobuyuki Kakimoto
- Department of Pediatrics, Wakayama Medical University, Wakayama, Japan
| | - Kouta Agematsu
- Departmentof Thoracic and Cardiovascular Surgery, Wakayama Medical University, Wakayama, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University, Sapporo, Japan
| | - Yoshikatsu Saiki
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
4
|
Postma AV, Rapp CK, Knoflach K, Volk AE, Lemke JR, Ackermann M, Regamey N, Latzin P, Celant L, Jansen SM, Bogaard HJ, Ilgun A, Alders M, van Spaendonck-Zwarts KY, Jonigk D, Klein C, Gräf S, Kubisch C, Houweling AC, Griese M. Biallelic variants in the calpain regulatory subunit CAPNS1 cause pulmonary arterial hypertension. GENETICS IN MEDICINE OPEN 2023; 1:100811. [PMID: 38230350 PMCID: PMC10790724 DOI: 10.1016/j.gimo.2023.100811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 01/18/2024]
Abstract
Purpose The aim of this study was to identify the monogenic cause of pulmonary arterial hypertension (PAH), a multifactorial and often fatal disease, in 2 unrelated consanguine families. Methods We performed exome sequencing and validated variant pathogenicity by whole-blood RNA and protein expression analysis in both families. Further RNA sequencing of preserved lung tissue was performed to investigate the consequences on selected genes that are involved in angiogenesis, proliferation, and apoptosis. Results We identified 2 rare biallelic variants in CAPNS1, encoding the regulatory subunit of calpain. The variants cosegregated with PAH in the families. Both variants lead to loss of function (LoF), which is demonstrated by aberrant splicing resulting in the complete absence of the CAPNS1 protein in affected patients. No other LoF CAPNS1 variant was identified in the genome data of more than 1000 patients with unresolved PAH. Conclusion The calpain holoenzyme was previously linked to pulmonary vascular development and progression of PAH in patients. We demonstrated that biallelic LoF variants in CAPNS1 can cause idiopathic PAH by the complete absence of CAPNS1 protein. Screening of this gene in patients who are affected by PAH, especially with suspected autosomal recessive inheritance, should be considered.
Collapse
Affiliation(s)
- Alex V. Postma
- Department of Medical Biology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Christina K. Rapp
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, LMU Klinikum, Ludwig Maximilians University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Katrin Knoflach
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, LMU Klinikum, Ludwig Maximilians University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Alexander E. Volk
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes R. Lemke
- Institute of Human Genetics, Leipzig University Medical Center, Leipzig, Germany
- Center for Rare Diseases, Leipzig University Medical Center, Leipzig, Germany
| | - Maximilian Ackermann
- Institute of Functional and Clinical Anatomy, University Medical Centre, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nicolas Regamey
- Division of Paediatric Pulmonology, Children’s Hospital, Lucerne Cantonal Hospital, Lucerne, Switzerland
| | - Philipp Latzin
- Division of Paediatric Respiratory Medicine and Allergology, Department of Pediatrics, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Lucas Celant
- Department of Pulmonary Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Samara M.A. Jansen
- Department of Pulmonary Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Harm J. Bogaard
- Department of Pulmonary Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Aho Ilgun
- Department of Human Genetics, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Mariëlle Alders
- Department of Human Genetics, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | | | - Danny Jonigk
- Institute of Pathology, Medizinische Hochschule Hannover, Hanover, Germany
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, LMU Klinikum, Ludwig Maximilians University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, United Kingdom
- NIHR BioResource for Translational Research–Rare Diseases, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Christian Kubisch
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Arjan C. Houweling
- Department of Human Genetics, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Matthias Griese
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, LMU Klinikum, Ludwig Maximilians University of Munich, German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
5
|
Suh JW, Lee KM, Ko EA, Yoon DS, Park KH, Kim HS, Yook JI, Kim NH, Lee JW. Promoting angiogenesis and diabetic wound healing through delivery of protein transduction domain-BMP2 formulated nanoparticles with hydrogel. J Tissue Eng 2023; 14:20417314231190641. [PMID: 37601810 PMCID: PMC10434183 DOI: 10.1177/20417314231190641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/12/2023] [Indexed: 08/22/2023] Open
Abstract
Decreased angiogenesis contributes to delayed wound healing in diabetic patients. Recombinant human bone morphogenetic protein-2 (rhBMP2) has also been demonstrated to promote angiogenesis. However, the short half-lives of soluble growth factors, including rhBMP2, limit their use in wound-healing applications. To address this limitation, we propose a novel delivery model using a protein transduction domain (PTD) formulated in a lipid nanoparticle (LNP). We aimed to determine whether a gelatin hydrogel dressing loaded with LNP-formulated PTD-BMP2 (LNP-PTD-BMP2) could enhance the angiogenic function of BMP2 and improve diabetic wound healing. In vitro, compared to the control and rhBMP2, LNP-PTD-BMP2 induced greater tube formation in human umbilical vein endothelial cells and increased the cell recruitment capacity of HaCaT cells. We inflicted large, full-thickness back skin wounds on streptozotocin-induced diabetic mice and applied gelatin hydrogel (GH) cross-linked by microbial transglutaminase containing rhBMP2, LNP-PTD-BMP2, or a control to these wounds. Wounds treated with LNP-PTD-BMP2-loaded GH exhibited enhanced wound closure, increased re-epithelialization rates, and higher collagen deposition than those with other treatments. Moreover, LNP-PTD-BMP2-loaded GH treatment resulted in more CD31- and α-SMA-positive cells, indicating greater neovascularization capacity than rhBMP2-loaded GH or GH treatments alone. Furthermore, in vivo near-infrared fluorescence revealed that LNP-PTD-BMP2 has a longer half-life than rhBMP2 and that BMP2 localizes around wounds. In conclusion, LNP-PTD-BMP2-loaded GH is a viable treatment option for diabetic wounds.
Collapse
Affiliation(s)
- Jae Wan Suh
- Department of Orthopaedic Surgery, Dankook University College of Medicine, Cheonan, South Korea
| | - Kyoung-Mi Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Ae Ko
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Dong Suk Yoon
- Department of Biomedical Science, Hwasung Medi-Science University, Hwaseong-Si, Gyeonggi-Do, South Korea
| | - Kwang Hwan Park
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Sil Kim
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, South Korea
| | - Jong In Yook
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, South Korea
| | - Nam Hee Kim
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, South Korea
| | - Jin Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
6
|
Solinc J, Ribot J, Soubrier F, Pavoine C, Dierick F, Nadaud S. The Platelet-Derived Growth Factor Pathway in Pulmonary Arterial Hypertension: Still an Interesting Target? Life (Basel) 2022; 12:life12050658. [PMID: 35629326 PMCID: PMC9143262 DOI: 10.3390/life12050658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 12/03/2022] Open
Abstract
The lack of curative options for pulmonary arterial hypertension drives important research to understand the mechanisms underlying this devastating disease. Among the main identified pathways, the platelet-derived growth factor (PDGF) pathway was established to control vascular remodeling and anti-PDGF receptor (PDGFR) drugs were shown to reverse the disease in experimental models. Four different isoforms of PDGF are produced by various cell types in the lung. PDGFs control vascular cells migration, proliferation and survival through binding to their receptors PDGFRα and β. They elicit multiple intracellular signaling pathways which have been particularly studied in pulmonary smooth muscle cells. Activation of the PDGF pathway has been demonstrated both in patients and in pulmonary hypertension (PH) experimental models. Tyrosine kinase inhibitors (TKI) are numerous but without real specificity and Imatinib, one of the most specific, resulted in beneficial effects. However, adverse events and treatment discontinuation discouraged to pursue this therapy. Novel therapeutic strategies are currently under experimental evaluation. For TKI, they include intratracheal drug administration, low dosage or nanoparticles delivery. Specific anti-PDGF and anti-PDGFR molecules can also be designed such as new TKI, soluble receptors, aptamers or oligonucleotides.
Collapse
Affiliation(s)
- Julien Solinc
- INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, UMR_S1166, F-75013 Paris, France; (J.S.); (J.R.); (F.S.); (C.P.)
| | - Jonathan Ribot
- INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, UMR_S1166, F-75013 Paris, France; (J.S.); (J.R.); (F.S.); (C.P.)
| | - Florent Soubrier
- INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, UMR_S1166, F-75013 Paris, France; (J.S.); (J.R.); (F.S.); (C.P.)
| | - Catherine Pavoine
- INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, UMR_S1166, F-75013 Paris, France; (J.S.); (J.R.); (F.S.); (C.P.)
| | - France Dierick
- Lady Davis Institute for Medical Research, McGill University, Montreal, QC H3T 1E2, Canada;
| | - Sophie Nadaud
- INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, UMR_S1166, F-75013 Paris, France; (J.S.); (J.R.); (F.S.); (C.P.)
- Correspondence: ; Tel.: +33-14077-9681
| |
Collapse
|
7
|
Chida-Nagai A, Sato H, Sato I, Shiraishi M, Sasaki D, Izumi G, Yamazawa H, Cho K, Manabe A, Takeda A. Risk factors for hospitalisation due to respiratory syncytial virus infection in children receiving prophylactic palivizumab. Eur J Pediatr 2022; 181:539-547. [PMID: 34417646 DOI: 10.1007/s00431-021-04216-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/18/2021] [Accepted: 07/20/2021] [Indexed: 12/01/2022]
Abstract
Respiratory syncytial virus (RSV) is a common pathogen that causes extremely severe respiratory symptoms in the first few weeks and months of life. In infants with cardiopulmonary diseases, RSV infections have a significant clinical impact. Palivizumab, a humanised monoclonal antibody for RSV, has been shown to significantly reduce the rate of hospitalisation of high-risk infants diagnosed with RSV. However, we have experienced a significant number of RSV infections in our institution that required hospitalisation or intensive care, despite the administration of palivizumab. This study aimed to analyse the risk factors associated with severe RSV despite the use of palivizumab. We retrospectively reviewed the medical records of 688 patients who visited or were admitted to our hospital and received palivizumab. Thirty-seven (5.4%) patients required hospitalisation for RSV, despite receiving palivizumab. In addition, 31 of these patients (83.8%) required hospitalisation out of season for palivizumab injection. Preterm birth (≤ 28-week gestation), bronchopulmonary dysplasia (BPD), and trisomy 21 were risk factors for RSV-related hospitalisation in infected patients, despite receiving palivizumab. Furthermore, subgroup analysis of 69 patients with RSV revealed that hemodynamically significant congenital heart disease (CHD) was also a risk factor for RSV-related hospitalisation.Conclusion: Preterm birth (≤ 28 weeks of gestation), BPD, trisomy 21, hemodynamically significant CHD, and CHD requiring surgery or cardiac catheterisation/intervention during infancy could be considered when determining whether year-round administration of palivizumab is appropriate. What is Known: • Respiratory syncytial virus causes severe respiratory symptoms in infants, particularly those with cardiopulmonary diseases. • The use of palivizumab has reduced the rate of hospitalisation of infants diagnosed with RSV. Despite this, the rate of hospitalisation is still high. What is New: • We identified that preterm birth (≤ 28-week gestation), bronchopulmonary dysplasia, trisomy 21, and hemodynamically significant congenital heart disease were risk factors for RSV-related hospitalisation, even after receiving palivizumab treatment. • High-risk infants should be closely monitored and the prolonged use of palivizumab should be considered.
Collapse
Affiliation(s)
- Ayako Chida-Nagai
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Hiroki Sato
- Department of Cardiology and Clinical Examination, Oita University, Oita, Japan.,Department of Preventive Medicine and Public Health, Tokyo Medical University, Shinjuku, Tokyo, Japan
| | - Itsumi Sato
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Masahiro Shiraishi
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Daisuke Sasaki
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Gaku Izumi
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Hirokuni Yamazawa
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Kazutoshi Cho
- Maternity and Perinatal Care Center, Hokkaido University Hospital, Sapporo, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Atsuhito Takeda
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan.
| |
Collapse
|
8
|
Hatai E, Muneuchi J, Sugitani Y, Doi H, Furuta T, Ezaki H, Kobayashi M, Sato R, Watanabe M. Pulmonary vascular resistance and compliance in individuals with trisomy 18. Am J Med Genet A 2021; 188:534-539. [PMID: 34729911 DOI: 10.1002/ajmg.a.62550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/14/2021] [Accepted: 10/16/2021] [Indexed: 11/10/2022]
Abstract
Individuals with trisomy 18 (T18) usually have congenital heart disease, often with pulmonary hypertension, which is associated with poor outcomes. This study aimed to explore the characteristics of pulmonary circulation including pulmonary vascular resistance (Rp) and compliance (Cp) among them. We retrospectively reviewed cardiac catheterization data in subjects with T18, trisomy 21 (T21), and without chromosomal anomaly (control group) who were referred due to heart failure associated with ventricular septal defect between 2000 and 2020. Pulmonary hemodynamic parameters including Rp and Cp were compared between these groups. We studied 20 subjects with T18, 88 subjects with T21, and 240 control subjects. There was no significant difference in age (T18: 4.6 [3.0-6. 9] vs. T21: 2.8 [1.9-4.0] vs. control: 2.9 [1.6-3.2] months, p = 0.06) and mean pulmonary arterial pressure (T18: 41 [33-49] vs. T21: 35 [30-41] vs. control: 36 [28-43] mmHg, p = 0.121) between the groups. The pulmonary to systemic blood flow ratio (Qp/Qs) (p = 0.983), Rp (p = 0.449), and Cp (p = 0.195) did not differ between T18 and control groups. However, Qp/Qs and Cp in T18 group were significantly greater than that in T21 group (T18: Qp/Qs: 3.4 [2.3-5.2] vs. T: 21 2.3 [1.7-3.7], p = 0.001. Cp: 3.5 [2.3-5.5] vs. 2.3 [1.6-3.1] mmHg/mL/m2 , p = 0.007), while Rp was identical between the groups (T18: 2.0 [1.6-3.3] vs. T21: 2.3 [1.7-3.7], p = 0.386). The pulmonary circulation in T18 subjects differed from that observed in T21 subjects, and identical to that observed in control subjects. Pulmonary hypertension is expected to be normalized after reasonable corrective surgery in T18 patients with congenital heart disease.
Collapse
Affiliation(s)
- Eriko Hatai
- Department of Pediatrics, Kyushu Hospital, Japan Community Healthcare Organization, Fukuoka, Japan
| | - Jun Muneuchi
- Department of Pediatrics, Kyushu Hospital, Japan Community Healthcare Organization, Fukuoka, Japan
| | - Yuichiro Sugitani
- Department of Pediatrics, Kyushu Hospital, Japan Community Healthcare Organization, Fukuoka, Japan
| | - Hirohito Doi
- Department of Pediatrics, Kyushu Hospital, Japan Community Healthcare Organization, Fukuoka, Japan
| | - Takashi Furuta
- Department of Pediatrics, Kyushu Hospital, Japan Community Healthcare Organization, Fukuoka, Japan
| | - Hiroki Ezaki
- Department of Pediatrics, Kyushu Hospital, Japan Community Healthcare Organization, Fukuoka, Japan
| | - Masaru Kobayashi
- Department of Pediatrics, Kyushu Hospital, Japan Community Healthcare Organization, Fukuoka, Japan
| | - Rie Sato
- Department of Pediatrics, Kyushu Hospital, Japan Community Healthcare Organization, Fukuoka, Japan
| | - Mamie Watanabe
- Department of Pediatrics, Kyushu Hospital, Japan Community Healthcare Organization, Fukuoka, Japan
| |
Collapse
|
9
|
Murthy S, Benza R. The Evolution of Risk Assessment in Pulmonary Arterial Hypertension. Methodist Debakey Cardiovasc J 2021; 17:134-144. [PMID: 34326933 PMCID: PMC8298117 DOI: 10.14797/lrpr7655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2020] [Indexed: 11/08/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a chronic debilitating disease that
carries an unacceptably high morbidity and mortality rate despite improved
survival with modern therapies. The combination of several modifiable and
nonmodifiable variables yields a robust risk assessment across various available
clinical calculators. The role of risk calculation is integral to managing PAH
and aids in the timely referral to expert centers and potentially lung
transplantation. Studies are ongoing to determine the role of risk calculators
in the framework of clinical trials and to elucidate novel markers of high risk
in PAH.
Collapse
Affiliation(s)
| | - Raymond Benza
- Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
10
|
Guihaire J, Deuse T, Wang D, Spin JM, Blankenberg FG, Fadel E, Reichenspurner H, Schrepfer S. Immunomodulation Therapy Using Tolerogenic Macrophages in a Rodent Model of Pulmonary Hypertension. Stem Cells Dev 2021; 30:515-525. [PMID: 33726521 DOI: 10.1089/scd.2021.0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Inflammation plays a major role in the pathogenesis of pulmonary hypertension (PH). We sought to investigate the effects of a cell-based immunomodulation in a dysimmune model of PH. PH was induced in athymic nude rats using semaxinib (Su group, n = 6). Tolerogenic macrophages (toM) were generated from monocyte isolation and then injected either the day before semaxinib injection (Prevention group, n = 6) or 3 weeks after (Reversion group, n = 6). Six athymic nude rats were used as controls. In vivo trafficking of toM was investigated with bioluminescence imaging showing that toM were mainly located into the lungs until 48 h after injection. Right ventricular (RV) end-systolic pressure and RV systolic function were assessed at 4 weeks using echocardiography. Morphometric analysis and RNA sequencing of the lungs were realized at 4 weeks. Rats treated with toM (Prevention and Reversion groups) had a significantly lower RV end-systolic pressure at 4 weeks (respectively, 25 ± 8 and 30 ± 6 mmHg vs. 67 ± 9 mmHg, P < 0.001), while RV systolic dysfunction was observed in Su and Reversion groups. Mean medial wall thickness of small arterioles was lower in Prevention and Reversion groups compared with the Su group (respectively, 10.9% ± 0.8% and 16.4% ± 1.3% vs. 28.2% ± 2.1%, P < 0.001). Similarly, cardiomyocyte area was decreased in rats treated with toM (150 ± 18 and 160 ± 86 μm2 vs. 279 ± 50 μm2, P < 0.001). A trend toward upregulation of genes involved in pulmonary arterial hypertension pathobiology was found in Su rats, while KCNK3 was significantly downregulated (fold-change = 9.8, P < 0.001). Injection of toM was associated with a less severe phenotype of PH in rats exposed to angioproliferative stress. Preserved expression of KCNK3 may explain the protective effect of toM.
Collapse
Affiliation(s)
- Julien Guihaire
- Transplant and Stem Cells Immunobiology (TSI) Lab, University Heart Center of Hamburg, Hamburg, Germany
- Department of Cardiac Surgery, Inserm UMR_S 999, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Groupe Hospitalier Paris Saint Joseph, University of Paris-Saclay School of Medicine, Le Plessis Robinson, France
| | - Tobias Deuse
- Transplant and Stem Cells Immunobiology (TSI) Lab, University Heart Center of Hamburg, Hamburg, Germany
- Transplant and Stem Cells Immunobiology (TSI) Lab, Department of Surgery, University of California San Francisco, San Francisco, California, USA
| | - Dong Wang
- Transplant and Stem Cells Immunobiology (TSI) Lab, University Heart Center of Hamburg, Hamburg, Germany
- Transplant and Stem Cells Immunobiology (TSI) Lab, Department of Surgery, University of California San Francisco, San Francisco, California, USA
- Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Joshua M Spin
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, California, USA
| | - Francis G Blankenberg
- Division of Pediatric Radiology, Department of Radiology/MIPS, Lucile Salter Packard Children's Hospital, Stanford University, Palo Alto, California, USA
| | - Elie Fadel
- Thoracic and Vascular Surgery, Heart and Lung Transplantation, Marie Lannelongue Hospital, Groupe Hospitalier Paris Saint Joseph, University of Paris-Saclay School of Medicine, Le Plessis Robinson, France
| | - Hermann Reichenspurner
- Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Department of Cardiovascular Surgery, University Heart Center of Hamburg, Hamburg, Germany
| | - Sonja Schrepfer
- Transplant and Stem Cells Immunobiology (TSI) Lab, University Heart Center of Hamburg, Hamburg, Germany
- Transplant and Stem Cells Immunobiology (TSI) Lab, Department of Surgery, University of California San Francisco, San Francisco, California, USA
- Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
| |
Collapse
|
11
|
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: 2.0] [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
|
12
|
Yaoita N, Satoh K, Satoh T, Shimizu T, Saito S, Sugimura K, Tatebe S, Yamamoto S, Aoki T, Kikuchi N, Kurosawa R, Miyata S, Nagasaki M, Yasuda J, Shimokawa H. Identification of the Novel Variants in Patients With Chronic Thromboembolic Pulmonary Hypertension. J Am Heart Assoc 2020; 9:e015902. [PMID: 33103541 PMCID: PMC7763425 DOI: 10.1161/jaha.120.015902] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 08/26/2020] [Indexed: 11/26/2022]
Abstract
Background Although chronic thromboembolic pulmonary hypertension (CTEPH) and acute pulmonary embolism (APE) share some clinical manifestations, a limited proportion of patients with CTEPH have a history of APE. Moreover, in histopathologic studies, it has been revealed that pulmonary vasculature lesions similar to pulmonary arterial hypertension existed in patients with CTEPH. Thus, it remains unknown whether these 3 disorders also share genetic backgrounds. Methods and Results Whole exome screening was performed with DNA isolated from 51 unrelated patients with CTEPH of Japanese ancestry. The frequency of genetic variants associated with pulmonary arterial hypertension or APE in patients with CTEPH was compared with those in the integrative Japanese Genome Variation Database 3.5KJPN. Whole exome screening analysis showed 17 049 nonsynonymous variants in patients with CTEPH. Although we found 6 nonsynonymous variants that are associated with APE in patients with CTEPH, there was no nonsynonymous variant associated with pulmonary arterial hypertension. Patients with CTEPH with a history of APE had nonsynonymous variants of F5, which encodes factor V. In contrast, patients with CTEPH without a history of APE had a nonsynonymous variant of THBD, which encodes thrombomodulin. Moreover, thrombin-activatable fibrinolysis inhibitor, which is one of the pathogenic proteins in CTEPH, was significantly more activated in those who had the variants of THBD compared with those without it. Conclusions These results provide the first evidence that patients with CTEPH have some variants associated with APE, regardless of the presence or absence of a history of APE. Furthermore, the variants might be different between patients with CTEPH with and without a history of APE.
Collapse
Affiliation(s)
- Nobuhiro Yaoita
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Kimio Satoh
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Taijyu Satoh
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Toru Shimizu
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Sakae Saito
- Department of Integrative GenomicsTohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Koichiro Sugimura
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Shunsuke Tatebe
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Saori Yamamoto
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Tatsuo Aoki
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Nobuhiro Kikuchi
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Ryo Kurosawa
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Satoshi Miyata
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Masao Nagasaki
- Department of Integrative GenomicsTohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Jun Yasuda
- Department of Integrative GenomicsTohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Hiroaki Shimokawa
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
13
|
Manaud G, Nossent EJ, Lambert M, Ghigna MR, Boët A, Vinhas MC, Ranchoux B, Dumas SJ, Courboulin A, Girerd B, Soubrier F, Bignard J, Claude O, Lecerf F, Hautefort A, Florio M, Sun B, Nadaud S, Verleden SE, Remy S, Anegon I, Bogaard HJ, Mercier O, Fadel E, Simonneau G, Vonk Noordegraaf A, Grünberg K, Humbert M, Montani D, Dorfmüller P, Antigny F, Perros F. Comparison of Human and Experimental Pulmonary Veno-Occlusive Disease. Am J Respir Cell Mol Biol 2020; 63:118-131. [PMID: 32209028 DOI: 10.1165/rcmb.2019-0015oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Pulmonary veno-occlusive disease (PVOD) occurs in humans either as a heritable form (hPVOD) due to biallelic inactivating mutations of EIF2AK4 (encoding GCN2) or as a sporadic form in older age (sPVOD). The chemotherapeutic agent mitomycin C (MMC) is a potent inducer of PVOD in humans and in rats (MMC-PVOD). Here, we compared human hPVOD and sPVOD, and MMC-PVOD pathophysiology at the histological, cellular, and molecular levels to unravel common altered pathomechanisms. MMC exposure in rats was associated primarily with arterial and microvessel remodeling, and secondarily by venous remodeling, when PVOD became symptomatic. In all forms of PVOD tested, there was convergent GCN2-dependent but eIF2α-independent pulmonary protein overexpression of HO-1 (heme oxygenase 1) and CHOP (CCAAT-enhancer-binding protein [C/EBP] homologous protein), two downstream effectors of GCN2 signaling and endoplasmic reticulum stress. In human PVOD samples, CHOP immunohistochemical staining mainly labeled endothelial cells in remodeled veins and arteries. Strong HO-1 staining was observed only within capillary hemangiomatosis foci, where intense microvascular proliferation occurs. HO-1 and CHOP stainings were not observed in control and pulmonary arterial hypertension lung tissues, supporting the specificity for CHOP and HO-1 involvement in PVOD pathobiology. In vivo loss of GCN2 (EIF2AK4 mutations carriers and Eif2ak4-/- rats) or in vitro GCN2 inhibition in cultured pulmonary artery endothelial cells using pharmacological and siRNA approaches demonstrated that GCN2 loss of function negatively regulates BMP (bone morphogenetic protein)-dependent SMAD1/5/9 signaling. Exogenous BMP9 was still able to reverse GCN2 inhibition-induced proliferation of pulmonary artery endothelial cells. In conclusion, we identified CHOP and HO-1 inhibition, and BMP9, as potential therapeutic options for PVOD.
Collapse
Affiliation(s)
- Grégoire Manaud
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| | - Esther J Nossent
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Mélanie Lambert
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| | | | - Angèle Boët
- Department of Research, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | | | - Benoit Ranchoux
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| | - Sébastien J Dumas
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| | - Audrey Courboulin
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| | - Barbara Girerd
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| | - Florent Soubrier
- INSERM UMR_S 956, Pierre and Marie Curie Université (Paris 06), Paris, France
| | - Juliette Bignard
- INSERM UMR_S 956, Pierre and Marie Curie Université (Paris 06), Paris, France
| | - Olivier Claude
- INSERM UMR_S 956, Pierre and Marie Curie Université (Paris 06), Paris, France
| | - Florence Lecerf
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| | - Aurélie Hautefort
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| | - Monica Florio
- Cardio-Metabolic Disorders, Amgen Research, Amgen Inc., Thousand Oaks, California
| | - Banghua Sun
- Cardio-Metabolic Disorders, Amgen Research, Amgen Inc., Thousand Oaks, California
| | - Sophie Nadaud
- INSERM UMR_S 956, Pierre and Marie Curie Université (Paris 06), Paris, France
| | - Stijn E Verleden
- Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases, Metabolism and Ageing KU Leuven, Leuven, Belgium
| | - Séverine Remy
- INSERM UMR 1064, Center for Research in Transplantation and Immunology-ITUN et Transgenic Rats and Immunophenomic Platform, Nantes, France; and
| | - Ignacio Anegon
- INSERM UMR 1064, Center for Research in Transplantation and Immunology-ITUN et Transgenic Rats and Immunophenomic Platform, Nantes, France; and
| | - Harm Jan Bogaard
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Olaf Mercier
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and.,Service de Chirurgie Thoracique, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Elie Fadel
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and.,Service de Chirurgie Thoracique, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Gérald Simonneau
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| | - Anton Vonk Noordegraaf
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Katrien Grünberg
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Marc Humbert
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| | - David Montani
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| | - Peter Dorfmüller
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and.,Department of Pathology and.,Department of Pathology, University of Giessen and Marburg Lung Center, Justus-Liebig University Giessen, German Center for Lung Research, Giessen, Germany
| | - Fabrice Antigny
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| | - Frédéric Perros
- Université Paris-Saclay-Faculté de Médecine, Le Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, and
| |
Collapse
|
14
|
Soleha U, Qomaruddin MB. Saltwater fish consumption pattern and incidence of hypertension in adults: A study on the population of Gresik coast, Indonesia. J Public Health Res 2020; 9:1846. [PMID: 32728585 PMCID: PMC7376448 DOI: 10.4081/jphr.2020.1846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/13/2020] [Indexed: 11/29/2022] Open
Abstract
Background: Hypertension is one of the main causes of morbidity and mortality in the Indonesian population, characterized by an increasing number of sufferers each year. Curtailing saltwater fish consumption pattern is an optional effort to prevent hypertension, as most users tend to attribute little or no attention to this possibility. Design and Methods: The purpose of this study, therefore, was to analyze the correlation between saltwater fish consumption pattern and the incidence of hypertension in adults, using analytic research design with cross sectional approach. Results: The analysis showed the dominance of female respondents (63%), aged 36-45 years (60%), graduated from senior high school (33.8%), working as housewives (32.5%), monthly income above the average (63.6%), absence of hypertension history (80.5%), eat sea fish often as a side dish (59.7%), and pre hypertension (59.1%). Furthermore, data analysis showed a Pvalue of 0.000, indicating the presence of a significant relationship between the consumption pattern of sea fish and the incidence of hypertension. Conclusions: The results of this study show the high consumption of saltwater fish by most people living in Kebungson, Gresik Village. This correlates with the incidence of hypertension in indigenous adults, as the majority was classified with prehypertension levels.
Collapse
Affiliation(s)
| | - Mochammad Bagus Qomaruddin
- Department of Health Promotion and Behavioral Sciences, Faculty of Public Health, Universitas Airlangga, Mulyorejo, Surabaya, Indonesia
| |
Collapse
|
15
|
Chida-Nagai A, Sagawa K, Tsujioka T, Fujimoto T, Taniguchi K, Sasaki O, Izumi G, Yamazawa H, Masaki N, Manabe A, Takeda A. Pulmonary vasodilators can lead to various complications in pulmonary "arterial" hypertension associated with congenital heart disease. Heart Vessels 2020; 35:1307-1315. [PMID: 32285188 PMCID: PMC7152743 DOI: 10.1007/s00380-020-01604-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/03/2020] [Indexed: 11/25/2022]
Abstract
Congenital heart disease-associated pulmonary arterial hypertension (CHD-PAH) is one of the major complications in patients with CHD. A timely closure of the left-to-right shunt will generally result in the normalization of the pulmonary hemodynamics, but a few patients have severe prognosis in their early childhood. We hypothesized that wide-ranging pathological mechanism in PAH could elucidate the clinical state of severe CHD-PAH. Using electronic medical records, we retrospectively analyzed six infants with severe CHD-PAH who had treatment-resistant PH. All patients were born with congenital malformation syndrome. After starting on a pulmonary vasodilator, five of the six patients developed complications including pulmonary edema and interstitial lung disease (ILD), and four patients had alveolar hemorrhage. After steroid therapy, the clinical condition improved in four patients, but two patients died. The autopsy findings in one of the deceased patients indicated the presence of recurrent alveolar hemorrhage, pulmonary venous hypertension, ILD, and PAH. Based on the clinical course of these CHD-PAH in patients and the literature, CHD-PAH can occur with pulmonary vascular obstructive disease (PVOD)/pulmonary capillary hemangiomatosis (PCH), ILD, and/or alveolar hemorrhage. The severity of CHD-PAH may depend on a genetic disorder, respiratory infection, and upper airway stenosis. Additionally, pulmonary vasodilators may be involved in the development of PVOD/PCH and ILD. When patients with CHD-PAH show unexpected deterioration, clinicians should consider complications associated with PVOD/PCH and/or pulmonary disease. In addition, the choice of upfront combination therapy for pediatric patients with CHD-PAH should be selected carefully.
Collapse
Affiliation(s)
- Ayako Chida-Nagai
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Koichi Sagawa
- Department of Pediatric Cardiology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Takao Tsujioka
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Takanori Fujimoto
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Kota Taniguchi
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Osamu Sasaki
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Gaku Izumi
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Hirokuni Yamazawa
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Naoki Masaki
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Atsuhito Takeda
- Department of Pediatrics, Hokkaido University, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan.
| |
Collapse
|
16
|
Idiopathic, heritable and veno-occlusive pulmonary arterial hypertension in childhood: computed tomography angiography features in the initial assessment of the disease. Pediatr Radiol 2019; 49:575-585. [PMID: 30652195 DOI: 10.1007/s00247-018-04331-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 10/22/2018] [Accepted: 12/12/2018] [Indexed: 01/19/2023]
Abstract
BACKGROUND In children, idiopathic and heritable pulmonary arterial hypertension present echocardiographic and heart catheterization findings similar to findings in pulmonary veno-occlusive disease. OBJECTIVE To provide a systematic analysis of CT angiography anomalies in children with idiopathic or heritable pulmonary arterial hypertension, or pulmonary veno-occlusive disease. We also sought to identify correlations between CT findings and patients' baseline characteristics. MATERIALS AND METHODS We retrospectively analyzed CT features of children with idiopathic and heritable pulmonary arterial hypertension or pulmonary veno-occlusive disease and 30 age-matched controls between 2008 and 2014. We compared CT findings and patient characteristics, including gene mutation type, and disease outcome until 2017. RESULTS The pulmonary arterial hypertension group included idiopathic (n=15) and heritable pulmonary arterial hypertension (n=11) and pulmonary veno-occlusive disease (n=4). Median age was 6.5 years. Children with pulmonary arterial hypertension showed enlargement of pulmonary artery and right cardiac chambers. A threshold for the ratio between the pulmonary artery and the ascending aorta of ≥1.2 had a sensitivity of 90% and a specificity of 100% for pulmonary arterial hypertension. All children with pulmonary veno-occlusive disease had thickened interlobular septa, centrilobular ground-glass opacities, and lymphadenopathy. In children with idiopathic and heritable pulmonary arterial hypertension, presence of intrapulmonary neovessels and enlargement of the right atrium were correlated with higher mean pulmonary artery pressure (P=0.011) and pulmonary vascular resistance (P=0.038), respectively. Mediastinal lymphadenopathy was associated with disease worsening within the first 2 years of follow-up (P=0.024). CONCLUSION CT angiography could contribute to early diagnosis and prediction of severity in children with pulmonary arterial hypertension.
Collapse
|
17
|
Bai P, Lyu L, Yu T, Zuo C, Fu J, He Y, Wan Q, Wan N, Jia D, Lyu A. Macrophage-Derived Legumain Promotes Pulmonary Hypertension by Activating the MMP (Matrix Metalloproteinase)-2/TGF (Transforming Growth Factor)-β1 Signaling. Arterioscler Thromb Vasc Biol 2019; 39:e130-e145. [PMID: 30676070 DOI: 10.1161/atvbaha.118.312254] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Objective—
Macrophages participate in the pathogenesis of pulmonary arterial hypertension (PAH). Lgmn (Legumain), a newly discovered cysteine proteinase belonging to the C13 peptidase family, is primarily expressed in macrophages; however, its roles in PAH remain unknown.
Approach and Results—
Herein, Lgmn was upregulated in lung tissues of PAH mice subjected to hypoxia plus SU5416 and PAH rats challenged with monocrotaline. Global Lgmn ablation and macrophage-specific ablation alleviated PAH compared with wild-type mice, evident from a reduction in right ventricular systolic pressure, the ratio of the right ventricular wall to the left ventricular wall plus the septum, the pulmonary vascular media thickness, and pulmonary vascular muscularization. Increased expression of ECM (extracellular matrix) proteins was correlated with MMP (matrix metalloproteinase)-2 activation and TGF (transforming growth factor)-β1 signaling in the PAs. Although Lgmn did not affect inflammatory cell infiltration and PA smooth muscle cell proliferation, it drove increased the synthesis of ECM proteins via MMP-2 activation. MMP-2 hydrolyzed the TGF-β1 precursor to the active form. An Lgmn-specific inhibitor markedly ameliorated PAH. Clinically, serum Lgmn levels were closely associated with the severity of idiopathic PAH.
Conclusions—
Our results indicate that Lgmn inhibition could be an effective strategy for preventing or delaying PAH.
Collapse
Affiliation(s)
- Peiyuan Bai
- From the Department of Cardiology, Ruijin Hospital (P.B., N.W., A.L.), Shanghai Jiaotong University School of Medicine, China
| | - Luheng Lyu
- Biology Major, School of Arts and Science, University of Miami, Coral Gables, FL (L.L.)
| | - Tingting Yu
- Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital (T.Y.), Shanghai Jiaotong University School of Medicine, China
| | - Caojian Zuo
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, China (C.Z., Q.W.)
| | - Jie Fu
- Department of Pediatrics, Renmin Hospital of Wuhan University, China (J.F.)
| | - Yuhu He
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China (Y.H.)
| | - Qiangyou Wan
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, China (C.Z., Q.W.)
| | - Naifu Wan
- From the Department of Cardiology, Ruijin Hospital (P.B., N.W., A.L.), Shanghai Jiaotong University School of Medicine, China
| | - Daile Jia
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China (D.J.)
| | - Ankang Lyu
- From the Department of Cardiology, Ruijin Hospital (P.B., N.W., A.L.), Shanghai Jiaotong University School of Medicine, China
| |
Collapse
|
18
|
Zhang CF, Zhao FY, Xu SL, Liu J, Xing XQ, Yang J. Autophagy in pulmonary hypertension: Emerging roles and therapeutic implications. J Cell Physiol 2019; 234:16755-16767. [PMID: 30932199 DOI: 10.1002/jcp.28531] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/21/2019] [Accepted: 03/06/2019] [Indexed: 02/05/2023]
Abstract
Autophagy is an important mechanism for cellular self-digestion and basal homeostasis. This gene- and modulator-regulated pathway is conserved in cells. Recently, several studies have shown that autophagic dysfunction is associated with pulmonary hypertension (PH). However, the relationship between autophagy and PH remains controversial. In this review, we mainly introduce the effects of autophagy-related genes and some regulatory molecules on PH and the relationship between autophagy and PH under the conditions of hypoxia, monocrotaline injection, thromboembolic stress, oxidative stress, and other drugs and toxins. The effects of other autophagy-related drugs, such as chloroquine, 3-methyladenine, rapamycin, and other potential therapeutic drugs and targets, in PH are also described.
Collapse
Affiliation(s)
- Chun-Fang Zhang
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of Kunming Medical University, The Second People's Hospital of Yunnan, Kunming, Yunnan, China
| | - Fang-Yun Zhao
- Department of Pharmacy, Yan'An Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Shuang-Lan Xu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of Kunming Medical University, The Second People's Hospital of Yunnan, Kunming, Yunnan, China
| | - Jie Liu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of Kunming Medical University, The Second People's Hospital of Yunnan, Kunming, Yunnan, China
| | - Xi-Qian Xing
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of Kunming Medical University, The Second People's Hospital of Yunnan, Kunming, Yunnan, China
| | - Jiao Yang
- First Department of Respiratory Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
19
|
Zhang Q, Xiao M, Gu S, Xu Y, Liu T, Li H, Yu Y, Qin L, Zhu Y, Chen F, Wang Y, Ding C, Wu H, Ji H, Chen Z, Zu Y, Malkoski S, Li Y, Liang T, Ji J, Qin J, Xu P, Zhao B, Shen L, Lin X, Feng XH. ALK phosphorylates SMAD4 on tyrosine to disable TGF-β tumour suppressor functions. Nat Cell Biol 2019; 21:179-189. [PMID: 30664791 DOI: 10.1038/s41556-018-0264-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/10/2018] [Indexed: 12/14/2022]
Abstract
Loss of TGF-β tumour suppressive response is a hallmark of human cancers. As a central player in TGF-β signal transduction, SMAD4 (also known as DPC4) is frequently mutated or deleted in gastrointestinal and pancreatic cancer. However, such genetic alterations are rare in most cancer types and the underlying mechanism for TGF-β resistance is not understood. Here we describe a mechanism of TGF-β resistance in ALK-positive tumours, including lymphoma, lung cancer and neuroblastoma. We demonstrate that, in ALK-positive tumours, ALK directly phosphorylates SMAD4 at Tyr 95. Phosphorylated SMAD4 is unable to bind to DNA and fails to elicit TGF-β gene responses and tumour suppressing responses. Chemical or genetic interference of the oncogenic ALK restores TGF-β responses in ALK-positive tumour cells. These findings reveal that SMAD4 is tyrosine-phosphorylated by an oncogenic tyrosine kinase during tumorigenesis. This suggests a mechanism by which SMAD4 is inactivated in cancers and provides guidance for targeted therapies in ALK-positive cancers.
Collapse
Affiliation(s)
- Qianting Zhang
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Mu Xiao
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Shuchen Gu
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yongxian Xu
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Ting Liu
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Hao Li
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yi Yu
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Lan Qin
- DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yezhang Zhu
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Fenfang Chen
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yulong Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chen Ding
- Beijing Proteome Research Center, National Center for Protein Sciences, Beijing, China.,College of Life Sciences, Fudan University, Shanghai, China
| | - Hongxing Wu
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Hongbin Ji
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Zhe Chen
- Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Youli Zu
- The Methodist Hospital Research Institute, Houston, TX, USA
| | - Stephen Malkoski
- Department of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Yi Li
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery and the Key Laboratory of Cancer Prevention and Intervention, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Junfang Ji
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Jun Qin
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Beijing Proteome Research Center, National Center for Protein Sciences, Beijing, China.,Department of Biochemistry & Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Pinglong Xu
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Bin Zhao
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Li Shen
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xia Lin
- DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Xin-Hua Feng
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China. .,DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
20
|
Ceylan AC, Citli S, Erdem HB, Sahin I, Acar Arslan E, Erdogan M. Importance and usage of chromosomal microarray analysis in diagnosing intellectual disability, global developmental delay, and autism; and discovering new loci for these disorders. Mol Cytogenet 2018; 11:54. [PMID: 30258496 PMCID: PMC6154794 DOI: 10.1186/s13039-018-0402-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023] Open
Abstract
Background Chromosomal microarray analysis is a first-stage test that is used for the diagnosis of intellectual disability and global developmental delay. Chromosomal microarray analysis can detect well-known microdeletion syndromes. It also contributes to the identification of genes that are responsible for the phenotypes in the new copy number variations. Results Chromosomal microarray analysis was conducted on 124 patients with intellectual disability and global developmental delay. Multiplex ligation-dependent probe amplification was used for the confirmation of chromosome 22q11.2 deletion/duplication. 26 pathogenic and likely pathogenic copy number variations were detected in 23 patients (18.55%) in a group of 124 Turkish patients with intellectual disability and global developmental delay. Chromosomal microarray analysis revealed pathogenic de novo Copy number variations, such as a novel 2.9-Mb de novo deletion at 18q22 region with intellectual disability and autism spectrum disorder, and a 22q11.2 region homozygote duplication with new clinical features. Conclusion Our data expand the spectrum of 22q11.2 region mutations, reveal new loci responsible from autism spectrum disorder and provide new insights into the genotype–phenotype correlations of intellectual disability and global developmental delay.
Collapse
Affiliation(s)
- Ahmet Cevdet Ceylan
- Trabzon Kanuni Training and Research Hospital, Medical Genetics Unit, Trabzon, Turkey.,2Ankara Yıldırım Beyazıt University, Ankara Atatürk Training and Research Hospital, Department of Medical Genetics, Ankara, Turkey
| | - Senol Citli
- Trabzon Kanuni Training and Research Hospital, Medical Genetics Unit, Trabzon, Turkey
| | - Haktan Bagis Erdem
- Ankara Diskapi Yildirim Beyazit Training and Research Hospital, Medical Genetics Unit, Ankara, Turkey
| | - Ibrahim Sahin
- Ankara Diskapi Yildirim Beyazit Training and Research Hospital, Medical Genetics Unit, Ankara, Turkey
| | - Elif Acar Arslan
- 4Karadeniz Technical University, School of Medicine, Department of Child Neurology, Trabzon, Turkey
| | - Murat Erdogan
- 5Kayseri Training and Research Hospital, Department of Medical Genetics, Kayseri, Turkey
| |
Collapse
|
21
|
Abbasi Y, Jabbari J, Jabbari R, Glinge C, Izadyar S, Spiekerkoetter E, Zamanian RT, Carlsen J, Tfelt‐Hansen J. Exome data clouds the pathogenicity of genetic variants in Pulmonary Arterial Hypertension. Mol Genet Genomic Med 2018; 6:835-844. [PMID: 30084161 PMCID: PMC6160702 DOI: 10.1002/mgg3.452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 04/25/2018] [Accepted: 06/03/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND We aimed to provide a set of previously reported PAH-associated missense and nonsense variants, and evaluate the pathogenicity of those variants. METHODS The Human Gene Mutation Database, PubMed, and Google Scholar were searched for previously reported PAH-associated genes and variants. Thereafter, both exome sequencing project and exome aggregation consortium as background population searched for previously reported PAH-associated missense and nonsense variants. The pathogenicity of previously reported PAH-associated missense variants evaluated by using four in silico prediction tools. RESULTS In total, 14 PAH-associated genes and 180 missense and nonsense variants were gathered. The BMPR2, the most frequent reported gene, encompasses 135 of 180 missense and nonsense variants. The exome sequencing project comprised 9, and the exome aggregation consortium counted 25 of 180 PAH-associated missense and nonsense variants. The TOPBP1 and ENG genes are unlikely to be the monogenic cause of PAH pathogenesis based on allele frequency in background population and prediction analysis. CONCLUSION This is the first evaluation of previously reported PAH-associated missense and nonsense variants. The BMPR2 identified as the major gene out of 14 PAH-associated genes. Based on findings, the ENG and TOPBP1 gene are not likely to be the monogenic cause of PAH.
Collapse
Affiliation(s)
- Yeganeh Abbasi
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of CardiologySection for Pulmonary Hypertension and Right Heart FailureCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | | | - Reza Jabbari
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of CardiologySection for Pulmonary Hypertension and Right Heart FailureCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Charlotte Glinge
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Seyed Bahador Izadyar
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Edda Spiekerkoetter
- Division of Pulmonary and Critical CareStanford University School of MedicineCalifornia
| | - Roham T. Zamanian
- Division of Pulmonary and Critical CareStanford University School of MedicineCalifornia
| | - Jørn Carlsen
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of CardiologySection for Pulmonary Hypertension and Right Heart FailureCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Jacob Tfelt‐Hansen
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of CardiologySection for Pulmonary Hypertension and Right Heart FailureCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of Forensic MedicineFaculty of Medical SciencesUniversity of CopenhagenDenmark
| |
Collapse
|
22
|
Htet M, Nally JE, Shaw A, Foote BE, Martin PE, Dempsie Y. Connexin 43 Plays a Role in Pulmonary Vascular Reactivity in Mice. Int J Mol Sci 2018; 19:E1891. [PMID: 29954114 PMCID: PMC6073802 DOI: 10.3390/ijms19071891] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/07/2018] [Accepted: 06/20/2018] [Indexed: 11/25/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a chronic condition characterized by vascular remodeling and increased vaso-reactivity. PAH is more common in females than in males (~3:1). Connexin (Cx)43 has been shown to be involved in cellular communication within the pulmonary vasculature. Therefore, we investigated the role of Cx43 in pulmonary vascular reactivity using Cx43 heterozygous (Cx43+/−) mice and 37,43Gap27, which is a pharmacological inhibitor of Cx37 and Cx43. Contraction and relaxation responses were studied in intra-lobar pulmonary arteries (IPAs) derived from normoxic mice and hypoxic mice using wire myography. IPAs from male Cx43+/− mice displayed a small but significant increase in the contractile response to endothelin-1 (but not 5-hydroxytryptamine) under both normoxic and hypoxic conditions. There was no difference in the contractile response to endothelin-1 (ET-1) or 5-hydroxytryptamine (5-HT) in IPAs derived from female Cx43+/−mice compared to wildtype mice. Relaxation responses to methacholine (MCh) were attenuated in IPAs from male and female Cx43+/− mice or by pre-incubation of IPAs with 37,43Gap27. Nω-Nitro-L-arginine methyl ester (l-NAME) fully inhibited MCh-induced relaxation. In conclusion, Cx43 is involved in nitric oxide (NO)-induced pulmonary vascular relaxation and plays a gender-specific and agonist-specific role in pulmonary vascular contractility. Therefore, reduced Cx43 signaling may contribute to pulmonary vascular dysfunction.
Collapse
Affiliation(s)
- Myo Htet
- Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK.
| | - Jane E Nally
- Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK.
| | - Andrew Shaw
- Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK.
| | - Bradley E Foote
- Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK.
| | - Patricia E Martin
- Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK.
| | - Yvonne Dempsie
- Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK.
| |
Collapse
|
23
|
Frank BS, Ivy DD. Diagnosis, Evaluation and Treatment of Pulmonary Arterial Hypertension in Children. CHILDREN (BASEL, SWITZERLAND) 2018; 5:E44. [PMID: 29570688 PMCID: PMC5920390 DOI: 10.3390/children5040044] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 02/28/2018] [Accepted: 03/16/2018] [Indexed: 12/23/2022]
Abstract
Pulmonary Hypertension (PH), the syndrome of elevated pressure in the pulmonary arteries, is associated with significant morbidity and mortality for affected children. PH is associated with a wide variety of potential underlying causes, including cardiac, pulmonary, hematologic and rheumatologic abnormalities. Regardless of the cause, for many patients the natural history of PH involves progressive elevation in pulmonary arterial resistance and pressure, right ventricular dysfunction, and eventually heart failure. In recent years, a number of pulmonary arterial hypertension (PAH)-targeted therapies have become available to reduce pulmonary artery pressure and improve outcome. A growing body of evidence in both the adult and pediatric literature demonstrates enhanced quality of life, functional status, and survival among treated patients. This review provides a description of select etiologies of PH seen in pediatrics and an update on the most recent data pertaining to evaluation and management of children with PH/PAH. The available evidence for specific classes of PAH-targeted therapies in pediatrics is additionally discussed.
Collapse
Affiliation(s)
- Benjamin S Frank
- Section of Cardiology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO 80045, USA.
| | - D Dunbar Ivy
- Section of Cardiology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO 80045, USA.
| |
Collapse
|
24
|
Wang GK, Li SH, Zhao ZM, Liu SX, Zhang GX, Yang F, Wang Y, Wu F, Zhao XX, Xu ZY. Inhibition of heat shock protein 90 improves pulmonary arteriole remodeling in pulmonary arterial hypertension. Oncotarget 2018; 7:54263-54273. [PMID: 27472464 PMCID: PMC5342340 DOI: 10.18632/oncotarget.10855] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 07/17/2016] [Indexed: 01/25/2023] Open
Abstract
While the molecular chaperone heat shock protein 90 (HSP90) is involved in a multitude of physiological and pathological processes, its role relating to pulmonary arterial hypertension (PAH) remains unclear. In the present study, we investigated the effect in which HSP90 improves pulmonary arteriole remodeling, and explored the therapeutic utility of targeting HSP90 as therapeutic drug for PAH. By Elisa and immunohistochemistry, HSP90 was found to be increased in both plasma and membrane walls of pulmonary arterioles from PAH patients. Moreover, plasma HSP90 levels positively correlated with mean pulmonary arterial pressure and C-reactive protein. In a monocrotaline-induced rat model of PH, we found that 17-AAG, a HSP90-inhibitor, alleviated the progress of PH, demonstrated by lower pulmonary arterial pressure and absence of right ventricular hypertrophy. Immunohistochemical staining demonstrated that 17-AAG improved pulmonary arteriole remodeling on the basis of reduced wall thickness and wall area. The inflammatory response attributed to PH could be attenuated by 17-AAG through reduction of NF-κB signaling. Moreover, 17-AAG was found to suppress PDGF-stimulated proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) through induction of cell cycle arrest in the G1 phase. In conclusion, HSP90 inhibitor 17-AAG could improve pulmonary arteriole remodeling via inhibiting the excessive proliferation of PASMCs, and inhibition of HSP90 may represent a therapeutic avenue for the treatment of PAH.
Collapse
Affiliation(s)
- Guo-Kun Wang
- Institution of Cardiac Surgery, Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Song-Hua Li
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhi-Min Zhao
- Institution of Cardiac Surgery, Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Su-Xuan Liu
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Guan-Xin Zhang
- Institution of Cardiac Surgery, Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Fan Yang
- Institution of Cardiac Surgery, Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yang Wang
- Institution of Cardiac Surgery, Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Feng Wu
- Department of Cardiology, 98th Military Hospital, Huzhou, Zhejiang, China
| | - Xian-Xian Zhao
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhi-Yun Xu
- Institution of Cardiac Surgery, Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
25
|
Chiu JS, Ma L, Wynn J, Krishnan U, Rosenzweig EB, Aspelund G, Arkovitz M, Warner BW, Lim FY, Mychaliska GB, Azarow K, Cusick RA, Chung DH, Chung WK. Mutations in BMPR2 are not present in patients with pulmonary hypertension associated with congenital diaphragmatic hernia. J Pediatr Surg 2017; 52:1747-1750. [PMID: 28162765 DOI: 10.1016/j.jpedsurg.2017.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/03/2017] [Accepted: 01/14/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND Congenital diaphragmatic hernia (CDH) is a prevalent major congenital anomaly with significant morbidity and mortality. Thirty to 40% mortality in CDH is largely attributed to pulmonary hypoplasia and pulmonary hypertension (PH). We hypothesized that the underlying genetic risk factors for hereditary PH are shared with CDH associated PH. METHODS Participants were recruited as part of the Diaphragmatic Hernia Research & Exploration; Advancing Molecular Science (DHREAMS) study, a prospective cohort of neonates with a diaphragmatic defect enrolled from 2005 to 2012. PH affected patients with available DNA for sequencing had one of the following: moderate or severe PH on echocardiography at 3months of age; moderate of severe PH at 1month of age with death occurring prior to the 3month echocardiogram; or on PH medications at 1month of age. We sequenced the coding regions of the hereditary PH genes bone morphogenetic protein receptor type II (BMPR2), caveolin 1 (CAV1) and potassium channel subfamily K, member 3 (KCNK3) to screen for mutations. RESULTS There were 29 CDH patients with PH including 16 males and 13 females. Sequencing of BMPR2, CAV1, and KCNK3 coding regions did not identify any pathogenic variants in these genes. TYPE OF STUDY Prognosis study LEVEL OF EVIDENCE: Level IV.
Collapse
Affiliation(s)
- Joanne S Chiu
- Department of Pediatrics, Division of Cardiology, Columbia University Medical Center, 3959 Broadway, 2-Babies North, New York, NY 10032, USA.
| | - Lijiang Ma
- Department of Pediatrics, Division of Molecular Genetics, Columbia University Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY 10032, USA
| | - Julia Wynn
- Department of Pediatrics, Division of Molecular Genetics, Columbia University Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY 10032, USA
| | - Usha Krishnan
- Department of Pediatrics, Division of Cardiology, Columbia University Medical Center, 3959 Broadway, 2-Babies North, New York, NY 10032, USA
| | - Erika B Rosenzweig
- Department of Pediatrics, Division of Cardiology, Columbia University Medical Center, 3959 Broadway, 2-Babies North, New York, NY 10032, USA
| | - Gudrun Aspelund
- Division of Pediatric Surgery, Columbia University Medical Center, 3959 Broadway, New York, NY 10032, USA
| | - Marc Arkovitz
- Department of Pediatric Surgery, Tel Hashomer Medical Center, Emek HaEla St 1, Ramat Gan, Israel
| | - Brad W Warner
- Division of Pediatric Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Foong-Yen Lim
- Division of Pediatric General, Thoracic, and Fetal Surgery, Center for Molecular Fetal Therapy, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229, USA
| | - George B Mychaliska
- Department of Surgery, University of Michigan Health System, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Kenneth Azarow
- Division of Pediatric Surgery, Oregon Health & Sciences University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
| | - Robert A Cusick
- Division of Pediatric Surgery, University of Nebraska Medical Center, College of Medicine, 42nd St and Emile St., Omaha, NE 68198, USA
| | - Dai H Chung
- Department of Pediatric Surgery, Vanderbilt University Medical Center, Monroe Carell Jr. Children's Hospital at Vanderbilt, 2200 Children's Way, Doctors' Office Tower, Suite 7100, Nashville, TN 37232-9780, USA
| | - Wendy K Chung
- Department of Pediatrics, Division of Molecular Genetics, Columbia University Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY 10032, USA
| |
Collapse
|
26
|
Ghataorhe P, Rhodes CJ, Harbaum L, Attard M, Wharton J, Wilkins MR. Pulmonary arterial hypertension - progress in understanding the disease and prioritizing strategies for drug development. J Intern Med 2017; 282:129-141. [PMID: 28524624 DOI: 10.1111/joim.12623] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pulmonary arterial hypertension (PAH), at one time a largely overlooked disease, is now the subject of intense study in many academic and biotech groups. The availability of new treatments has increased awareness of the condition. This in turn has driven a change in the demographics of PAH, with an increase in the mean age at diagnosis. The diagnosis of PAH in more elderly patients has highlighted the need for careful phenotyping of patients and for further studies to understand how best to manage pulmonary hypertension associated with, for example, left heart disease. The breadth and depth of expertise focused on unravelling the molecular pathology of PAH has yielded novel insights, including the role of growth factors, inflammation and metabolic remodelling. The description of the genetic architecture of PAH is accelerating in parallel, with novel variants, such as those reported in potassium two-pore domain channel subfamily K member 3 (KCNK3), adding to the list of more established mutations in genes associated with bone morphogenetic protein receptor type 2 (BMPR2) signalling. These insights have supported a paradigm shift in treatment strategies away from simply addressing the imbalance of vasoactive mediators observed in PAH towards tackling more directly the structural remodelling of the pulmonary vasculature. Here, we summarize the changing clinical and molecular landscape of PAH. We highlight novel drug therapies that are in various stages of clinical development, targeting for example cell proliferation, metabolic, inflammatory/immune and BMPR2 dysfunction, and the challenges around developing these treatments. We argue that advances in the treatment of PAH will come through deep molecular phenotyping with the integration of clinical, genomic, transcriptomic, proteomic and metabolomic information in large populations of patients through international collaboration. This approach provides the best opportunity for identifying key signalling pathways, both as potential drug targets and as biomarkers for patient selection. The expectation is that together these will enable the prioritization of potential therapies in development and the evolution of personalized medicine for PAH.
Collapse
Affiliation(s)
- P Ghataorhe
- Department of Medicine, Imperial College London, London, UK
| | - C J Rhodes
- Department of Medicine, Imperial College London, London, UK
| | - L Harbaum
- Department of Medicine, Imperial College London, London, UK
| | - M Attard
- Department of Medicine, Imperial College London, London, UK
| | - J Wharton
- Department of Medicine, Imperial College London, London, UK
| | - M R Wilkins
- Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
27
|
Pulmonary Arterial Hypertension in Glycogen Storage Disease Type I. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2017. [DOI: 10.1177/2326409817707773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
28
|
Higasa K, Ogawa A, Terao C, Shimizu M, Kosugi S, Yamada R, Date H, Matsubara H, Matsuda F. A burden of rare variants in BMPR2 and KCNK3 contributes to a risk of familial pulmonary arterial hypertension. BMC Pulm Med 2017; 17:57. [PMID: 28388887 PMCID: PMC5383973 DOI: 10.1186/s12890-017-0400-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/24/2017] [Indexed: 12/13/2022] Open
Abstract
Background Pulmonary arterial hypertension (PAH) is a severe lung disease with only few effective treatments available. Familial cases of PAH are usually recognized as an autosomal dominant disease, but incomplete penetrance of the disease makes it difficult to identify pathogenic variants in accordance with a Mendelian pattern of inheritance. Methods To elucidate the complex genetic basis of PAH, we obtained whole exome- or genome-sequencing data of 17 subjects from 9 families with heritable PAH and applied gene-based association analysis with 9 index patients and 300 PAH-free controls. Results A burden of rare variants in BMPR2 significantly contributed to the risk of the disease (p = 6.0 × 10−8). Eight of nine families carried four previously reported single nucleotide variants and four novel insertion/deletion variants in the gene. One of the novel variants was a large 6.5 kilobase-deletion. In the remaining one family, the patient carried a pathogenic variant in a member of potassium channels, KCNK3, which was the first replicative finding of channelopathy in an Asian population. Conclusions The variety of rare pathogenic variants suggests that gene-based association analysis using genome-wide sequencing data from increased number of samples is essential to tracing the genetic heterogeneity and developing an appropriate panel for genetic testing. Electronic supplementary material The online version of this article (doi:10.1186/s12890-017-0400-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Koichiro Higasa
- Human Disease Genomics, Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kawara-cho 53, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Aiko Ogawa
- Department of Clinical Science, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Chikashi Terao
- Human Disease Genomics, Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kawara-cho 53, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masakazu Shimizu
- Human Disease Genomics, Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kawara-cho 53, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shinji Kosugi
- Department of Medical Ethics/Medical Genetics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryo Yamada
- Statistical Genetics, Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiromi Matsubara
- Department of Clinical Science, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Fumihiko Matsuda
- Human Disease Genomics, Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kawara-cho 53, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
29
|
Idiopathic Pulmonary Arterial Hypertension in Children: A Review. Pulm Ther 2017. [DOI: 10.1007/s41030-017-0035-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
30
|
Yanai S, Wakayama M, Nakayama H, Shinozaki M, Tsukuma H, Tochigi N, Nemoto T, Saji T, Shibuya K. Implication of overexpression of dishevelled-associated activator of morphogenesis 1 (Daam-1) for the pathogenesis of human Idiopathic Pulmonary Arterial Hypertension (IPAH). Diagn Pathol 2017; 12:25. [PMID: 28288669 PMCID: PMC5348773 DOI: 10.1186/s13000-017-0614-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/20/2017] [Indexed: 01/31/2023] Open
Abstract
Background Idiopathic pulmonary arterial hypertension (IPAH) is a rare, fatal disease of unknown pathogenesis. Evidence from our recent study suggests that IPAH pathogenesis is related to upregulation of the Wnt/planar cell polarity (Wnt/PCP) pathway. We used microscopic observation and immunohistochemical techniques to identify expression patterns of cascading proteins—namely Wnt-11, dishevelled-2 (Dvl-2), and dishevelled-associated activator of morphogenesis 1 (Daam-1)—in pulmonary arteries. Methods We analyzed sections of formalin-fixed and paraffin-embedded autopsied lung tissues obtained from 9 IPAH cases, 7 associated pulmonary arterial hypertension cases, and 16 age-matched controls without pulmonary arterial abnormalities. Results of microscopic observation were analyzed in relation to the cellular components and size of pulmonary arteries. Results Varying rates of positive reactivity to Dvl-2 and Daam-1 were confirmed in all cellular components of pulmonary arteries, namely, endothelial cells, myofibroblasts, and medial smooth muscle cells. In contrast, none of these components was reactive to Wnt-11. No specific expression patterns were observed for endothelial cells or myofibroblasts under any experimental conditions. However, marked expression of Dvl-2 and Daam-1 was confirmed in smooth muscle cells. In addition, Dvl-2 was depleted while Daam-1 expression was elevated in IPAH, in contrast with specimens from associated pulmonary arterial hypertension cases and controls. Conclusions High Daam-1 expression may upregulate the Wnt/PCP pathway and cause IPAH.
Collapse
Affiliation(s)
- Shun Yanai
- Department of Pediatrics, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Megumi Wakayama
- Department of Surgical Pathology, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan.
| | - Haruo Nakayama
- Department of Neurosurgery, Toho University Ohashi Medical Center, 2-17-6 Ohashi, Meguro-ku, Tokyo, 153-8515, Japan
| | - Minoru Shinozaki
- Department of Surgical Pathology, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Hisayuki Tsukuma
- Toho University School of Medicine, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Naobumi Tochigi
- Department of Surgical Pathology, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Tetsuo Nemoto
- Department of Surgical Pathology, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Tsutomu Saji
- Advanced and Integrated Cardiovascular Research Course in the Young and Adolescence, Toho University School of Medicine, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Kazutoshi Shibuya
- Department of Surgical Pathology, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| |
Collapse
|
31
|
Prins KW, Thenappan T. World Health Organization Group I Pulmonary Hypertension: Epidemiology and Pathophysiology. Cardiol Clin 2017; 34:363-74. [PMID: 27443134 DOI: 10.1016/j.ccl.2016.04.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a debilitating disease characterized by pathologic remodeling of the resistance pulmonary arteries, ultimately leading to right ventricular (RV) failure and death. In this article we discuss the definition of PAH, the initial epidemiology based on the National Institutes of Health Registry, and the updated epidemiology gleaned from contemporary registries, pathogenesis of pulmonary vascular dysfunction and proliferation, and RV failure in PAH.
Collapse
Affiliation(s)
- Kurt W Prins
- Cardiovascular Division, University of Minnesota Medical School, 420 Delaware Street Southeast, Minneapolis, MN 55455, USA
| | - Thenappan Thenappan
- Section of Advanced Heart Failure and Pulmonary Hypertension, Cardiovascular Division, University of Minnesota Medical School, 420 Delaware Street Southeast, Minneapolis, MN 55455, USA.
| |
Collapse
|
32
|
Ma L, Chung WK. The role of genetics in pulmonary arterial hypertension. J Pathol 2016; 241:273-280. [PMID: 27770446 DOI: 10.1002/path.4833] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 09/30/2016] [Accepted: 10/17/2016] [Indexed: 12/14/2022]
Abstract
Group 1 pulmonary hypertension or pulmonary arterial hypertension (PAH) is a rare disease characterized by proliferation and occlusion of small pulmonary arterioles, leading to progressive elevation of pulmonary artery pressure and pulmonary vascular resistance, and right ventricular failure. Historically, it has been associated with a high mortality rate, although, over the last decade, treatment has improved survival. PAH includes idiopathic PAH (IPAH), heritable PAH (HPAH), and PAH associated with certain medical conditions. The aetiology of PAH is heterogeneous, and genetics play an important role in some cases. Mutations in BMPR2, encoding bone morphogenetic protein receptor 2, a member of the transforming growth factor-β superfamily of receptors, have been identified in 70% of cases of HPAH, and in 10-40% of cases of IPAH. Other genetic causes of PAH include mutations in the genes encoding activin receptor-like type 1, endoglin, SMAD9, caveolin 1, and potassium two-pore-domain channel subfamily K member 3. Mutations in the gene encoding T-box 4 have been identified in 10-30% of paediatric PAH patients, but rarely in adults with PAH. PAH in children is much more heterogeneous than in adults, and can be associated with several genetic syndromes, congenital heart disease, pulmonary disease, and vascular disease. In addition to rare mutations as a monogenic cause of HPAH, common variants in the gene encoding cerebellin 2 increase the risk of PAH by approximately two-fold. A PAH panel of genes is available for clinical testing, and should be considered for use in clinical management, especially for patients with a family history of PAH. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Lijiang Ma
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA
| |
Collapse
|
33
|
Rhodes CJ, Ghataorhe P, Wharton J, Rue-Albrecht KC, Hadinnapola C, Watson G, Bleda M, Haimel M, Coghlan G, Corris PA, Howard LS, Kiely DG, Peacock AJ, Pepke-Zaba J, Toshner MR, Wort SJ, Gibbs JSR, Lawrie A, Gräf S, Morrell NW, Wilkins MR. Plasma Metabolomics Implicates Modified Transfer RNAs and Altered Bioenergetics in the Outcomes of Pulmonary Arterial Hypertension. Circulation 2016; 135:460-475. [PMID: 27881557 PMCID: PMC5287439 DOI: 10.1161/circulationaha.116.024602] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/09/2016] [Indexed: 11/27/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Pulmonary arterial hypertension (PAH) is a heterogeneous disorder with high mortality. Methods: We conducted a comprehensive study of plasma metabolites using ultraperformance liquid chromatography mass spectrometry to identify patients at high risk of early death, to identify patients who respond well to treatment, and to provide novel molecular insights into disease pathogenesis. Results: Fifty-three circulating metabolites distinguished well-phenotyped patients with idiopathic or heritable PAH (n=365) from healthy control subjects (n=121) after correction for multiple testing (P<7.3e-5) and confounding factors, including drug therapy, and renal and hepatic impairment. A subset of 20 of 53 metabolites also discriminated patients with PAH from disease control subjects (symptomatic patients without pulmonary hypertension, n=139). Sixty-two metabolites were prognostic in PAH, with 36 of 62 independent of established prognostic markers. Increased levels of tRNA-specific modified nucleosides (N2,N2-dimethylguanosine, N1-methylinosine), tricarboxylic acid cycle intermediates (malate, fumarate), glutamate, fatty acid acylcarnitines, tryptophan, and polyamine metabolites and decreased levels of steroids, sphingomyelins, and phosphatidylcholines distinguished patients from control subjects. The largest differences correlated with increased risk of death, and correction of several metabolites over time was associated with a better outcome. Patients who responded to calcium channel blocker therapy had metabolic profiles similar to those of healthy control subjects. Conclusions: Metabolic profiles in PAH are strongly related to survival and should be considered part of the deep phenotypic characterization of this disease. Our results support the investigation of targeted therapeutic strategies that seek to address the alterations in translational regulation and energy metabolism that characterize these patients.
Collapse
Affiliation(s)
- Christopher J Rhodes
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Pavandeep Ghataorhe
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - John Wharton
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Kevin C Rue-Albrecht
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Charaka Hadinnapola
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Geoffrey Watson
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Marta Bleda
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Matthias Haimel
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Gerry Coghlan
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Paul A Corris
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Luke S Howard
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - David G Kiely
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Andrew J Peacock
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Joanna Pepke-Zaba
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Mark R Toshner
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - S John Wort
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - J Simon R Gibbs
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Allan Lawrie
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Stefan Gräf
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Nicholas W Morrell
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.)
| | - Martin R Wilkins
- From the Department of Medicine, Imperial College London, Hammersmith Campus, United Kingdom (C.J.R., P.G., J.W., K.C.R.-A., G.W., M.R.W.); Department of Medicine, University of Cambridge School of Clinical Medicine, United Kingdom (C.H., M.B., M.H., M.R.T., S.G., N.W.M.); Cardiology Department, Royal Free Hospital, London, United Kingdom (G.C.); Institute of Cellular Medicine, Newcastle University and the Newcastle Upon Tyne Hospitals NHS Foundation Trust, United Kingdom (P.A.C.); National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom (L.S.H., J.S.R.G.); National Heart and Lung Institute, Imperial College London, Hammersmith Campus, United Kingdom (L.S.H., J.S.R.G.); Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (D.G.K.); Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, United Kingdom (D.G.K., A.L.); Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, United Kingdom (A.J.P.); Pulmonary Vascular Disease Unit, Papworth Hospital, Cambridge, United Kingdom (J.P.Z., M.R.T.); Pulmonary Hypertension Service, Royal Brompton Hospital, London, United Kingdom (S.J.W.); and Department of Haematology, University of Cambridge, United Kingdom (S.G.).
| |
Collapse
|
34
|
Tsujino K, Reed NI, Atakilit A, Ren X, Sheppard D. Transforming growth factor-β plays divergent roles in modulating vascular remodeling, inflammation, and pulmonary fibrosis in a murine model of scleroderma. Am J Physiol Lung Cell Mol Physiol 2016; 312:L22-L31. [PMID: 27864286 DOI: 10.1152/ajplung.00428.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 11/13/2016] [Indexed: 12/25/2022] Open
Abstract
The efficacy and feasibility of targeting transforming growth factor-β (TGFβ) in pulmonary fibrosis and lung vascular remodeling in systemic sclerosis (SSc) have not been well elucidated. In this study we analyzed how blocking TGFβ signaling affects pulmonary abnormalities in Fos-related antigen 2 (Fra-2) transgenic (Tg) mice, a murine model that manifests three important lung pathological features of SSc: fibrosis, inflammation, and vascular remodeling. To interrupt TGFβ signaling in the Fra-2 Tg mice, we used a pan-TGFβ-blocking antibody, 1D11, and Tg mice in which TGFβ receptor type 2 (Tgfbr2) is deleted from smooth muscle cells and myofibroblasts (α-SMA-CreER;Tgfbr2flox/flox). Global inhibition of TGFβ by 1D11 did not ameliorate lung fibrosis histologically or biochemically, whereas it resulted in a significant increase in the number of immune cells infiltrating the lungs. In contrast, 1D11 treatment ameliorated the severity of pulmonary vascular remodeling in Fra-2 Tg mice. Similarly, genetic deletion of Tgfbr2 from smooth muscle cells resulted in improvement of pulmonary vascular remodeling in the Fra-2 Tg mice, as well as a decrease in the number of Ki67-positive vascular smooth muscle cells, suggesting that TGFβ signaling contributes to development of pulmonary vascular remodeling by promoting the proliferation of vascular smooth muscle cells. Deletion of Tgfbr2 from α-smooth muscle actin-expressing cells had no effect on fibrosis or inflammation in this model. These results suggest that efforts to target TGFβ in SSc will likely require more precision than simply global inhibition of TGFβ function.
Collapse
Affiliation(s)
- Kazuyuki Tsujino
- Department of Medicine, University of California, San Francisco, California
| | - Nilgun Isik Reed
- Department of Medicine, University of California, San Francisco, California
| | - Amha Atakilit
- Department of Medicine, University of California, San Francisco, California
| | - Xin Ren
- Department of Medicine, University of California, San Francisco, California
| | - Dean Sheppard
- Department of Medicine, University of California, San Francisco, California
| |
Collapse
|
35
|
Abstract
The prevalence of PH is increasing in the pediatric population, because of improved recognition and increased survival of patients, and remains a significant cause of morbidity and mortality. Recent studies have improved the understanding of pediatric PH, but management remains challenging because of a lack of evidence-based clinical trials. The growing contribution of developmental lung disease requires dedicated research to explore the use of existing therapies as well as the creation of novel therapies. Adequate study of pediatric PH will require multicenter collaboration due to the small numbers of patients, multifactorial disease causes, and practice variability.
Collapse
Affiliation(s)
- Dunbar Ivy
- Section of Pediatric Cardiology, Children's Hospital Colorado, University of Colorado School of Medicine, 13123 East 16th Avenue, B100, Aurora, CO 80045, USA.
| |
Collapse
|
36
|
Affiliation(s)
- Jianhua Xiong
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
37
|
Rameh V, Kossaify A. Role of Biomarkers in the Diagnosis, Risk Assessment, and Management of Pulmonary Hypertension. Biomark Insights 2016; 11:85-9. [PMID: 27385910 PMCID: PMC4920202 DOI: 10.4137/bmi.s38323] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/15/2016] [Accepted: 05/19/2016] [Indexed: 12/03/2022] Open
Abstract
Pulmonary hypertension is a severe and debilitating disease with no definite cure, and the domain of targeted therapies is a promising field for better management of this severe condition. The disease comprises pulmonary arterial remodeling, hypoxia, endothelial dysfunction, and inflammation, with subsequent organ damage including right heart and liver dysfunction. Biomarkers have a valuable role at different levels of the disease, from diagnosis to risk assessment and management, in order to decrease the burden of the disease in terms of both morbidity and mortality.
Collapse
Affiliation(s)
- Vanessa Rameh
- Echocardiography Unit, Cardiology division, University Hospital Notre Dame des Secours, Byblos, Lebanon
| | - Antoine Kossaify
- Echocardiography Unit, Cardiology division, University Hospital Notre Dame des Secours, Byblos, Lebanon
| |
Collapse
|
38
|
Molecular Background of miRNA Role in Asthma and COPD: An Updated Insight. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7802521. [PMID: 27376086 PMCID: PMC4916273 DOI: 10.1155/2016/7802521] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/29/2016] [Accepted: 05/15/2016] [Indexed: 12/14/2022]
Abstract
Inflammatory airway diseases are a significant health problems requiring new approaches to the existing therapies and addressing fundamental issues. Difficulties in developing effective therapeutic strategies might be caused by lack of understanding of their exact molecular mechanism. MicroRNAs (miRNAs) are a class of regulators that already revolutionized the view of gene expression regulation. A cumulating number of investigations show a pivotal role of miRNAs in the pathogenesis of asthma, chronic obstructive pulmonary disease (COPD), or airway remodeling through the regulation of many pathways involved in their pathogenesis. Expression changes of several miRNAs have also been found to play a role in the development and/or improvement in asthma or COPD. Still, relatively little is known about the role of miRNAs in inflammatory disorders. The microRNA profiles may differ depending on the cell type or antigen-presenting cell. Based on the newest literature, this review discusses the current knowledge concerning miRNA contribution and influence on lung inflammation and chosen inflammatory airway diseases: asthma and COPD.
Collapse
|
39
|
Johansen AKZ, Dean A, Morecroft I, Hood K, Nilsen M, Loughlin L, Anagnostopoulou A, Touyz RM, White K, MacLean MR. The serotonin transporter promotes a pathological estrogen metabolic pathway in pulmonary hypertension via cytochrome P450 1B1. Pulm Circ 2016; 6:82-92. [PMID: 27162617 PMCID: PMC4860551 DOI: 10.1086/685023] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating vasculopathy that predominates in women and has been associated with dysregulated estrogen and serotonin signaling. Overexpression of the serotonin transporter (SERT(+)) in mice results in an estrogen-dependent development of pulmonary hypertension (PH). Estrogen metabolism by cytochrome P450 1B1 (CYP1B1) contributes to the pathogenesis of PAH, and serotonin can increase CYP1B1 expression in human pulmonary arterial smooth muscle cells (hPASMCs). We hypothesized that an increase in intracellular serotonin via increased SERT expression may dysregulate estrogen metabolism via CYP1B1 to facilitate PAH. Consistent with this hypothesis, we found elevated lung CYP1B1 protein expression in female SERT(+) mice accompanied by PH, which was attenuated by the CYP1B1 inhibitor 2,3',4,5'-tetramethoxystilbene (TMS). Lungs from female SERT(+) mice demonstrated an increase in oxidative stress that was marked by the expression of 8-hydroxyguanosine; however, this was unaffected by CYP1B1 inhibition. SERT expression was increased in monocrotaline-induced PH in female rats; however, TMS did not reverse PH in monocrotaline-treated rats but prolonged survival. Stimulation of hPASMCs with the CYP1B1 metabolite 16α-hydroxyestrone increased cellular proliferation, which was attenuated by an inhibitor (MPP) of estrogen receptor alpha (ERα) and a specific ERα antibody. Thus, increased intracellular serotonin caused by increased SERT expression may contribute to PAH pathobiology by dysregulation of estrogen metabolic pathways via increased CYP1B1 activity. This promotes PASMC proliferation by the formation of pathogenic metabolites of estrogen that mediate their effects via ERα. Our studies indicate that targeting this pathway in PAH may provide a promising antiproliferative therapeutic strategy.
Collapse
Affiliation(s)
- Anne Katrine Z Johansen
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Afshan Dean
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ian Morecroft
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Katie Hood
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Margaret Nilsen
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Lynn Loughlin
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Aikaterini Anagnostopoulou
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Kevin White
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Margaret R MacLean
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
40
|
Abstract
The circulation of the lung is unique both in volume and function. For example, it is the only organ with two circulations: the pulmonary circulation, the main function of which is gas exchange, and the bronchial circulation, a systemic vascular supply that provides oxygenated blood to the walls of the conducting airways, pulmonary arteries and veins. The pulmonary circulation accommodates the entire cardiac output, maintaining high blood flow at low intravascular arterial pressure. As compared with the systemic circulation, pulmonary arteries have thinner walls with much less vascular smooth muscle and a relative lack of basal tone. Factors controlling pulmonary blood flow include vascular structure, gravity, mechanical effects of breathing, and the influence of neural and humoral factors. Pulmonary vascular tone is also altered by hypoxia, which causes pulmonary vasoconstriction. If the hypoxic stimulus persists for a prolonged period, contraction is accompanied by remodeling of the vasculature, resulting in pulmonary hypertension. In addition, genetic and environmental factors can also confer susceptibility to development of pulmonary hypertension. Under normal conditions, the endothelium forms a tight barrier, actively regulating interstitial fluid homeostasis. Infection and inflammation compromise normal barrier homeostasis, resulting in increased permeability and edema formation. This article focuses on reviewing the basics of the lung circulation (pulmonary and bronchial), normal development and transition at birth and vasoregulation. Mechanisms contributing to pathological conditions in the pulmonary circulation, in particular when barrier function is disrupted and during development of pulmonary hypertension, will also be discussed.
Collapse
Affiliation(s)
- Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Larissa A. Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
41
|
Signal Mechanisms of Vascular Remodeling in the Development of Pulmonary Arterial Hypertension. J Cardiovasc Pharmacol 2016; 67:182-90. [DOI: 10.1097/fjc.0000000000000328] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
42
|
Xi Q, Liu Z, Zhao Z, Luo Q, Huang Z. High Frequency of Pulmonary Hypertension-Causing Gene Mutation in Chinese Patients with Chronic Thromboembolic Pulmonary Hypertension. PLoS One 2016; 11:e0147396. [PMID: 26820968 PMCID: PMC4731080 DOI: 10.1371/journal.pone.0147396] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 01/04/2016] [Indexed: 12/31/2022] Open
Abstract
The pathogenesis of chronic thromboembolic pulmonary hypertension (CTEPH) is unknown. Histopathologic studies revealed that pulmonary vasculature lesions similar to idiopathic pulmonary arterial hypertension (PAH) existed in CTEPH patients as well. It’s well-known that genetic predisposition plays an important role in the mechanism of PAH. So we hypothesized that PAH-causing gene mutation might exist in some CTEPH patients and act as a background to facilitate the development of CTEPH. In this study, we analyzed 7 PAH-causing genes including BMPR2, ACVRL1, ENG, SMAD9, CAV1, KCNK3, and CBLN2 in 49 CTEPH patients and 17 patients recovered from pulmonary embolism (PE) but without pulmonary hypertension(PH). The results showed that the nonsynonymous mutation rate in CTEPH patients is significantly higher than that in PE without PH patients (25 out of 49 (51%) CTEPH patients vs. 3 out of 17 PE without PH patients (18%); p = 0.022). Four CTEPH patients had the same point mutation in ACVRL1 exon 10 (c.1450C>G), a mutation approved to be associated with PH in a previous study. In addition, we identified two CTEPH associated SNPs (rs3739817 and rs55805125). Our results suggest that PAH-causing gene mutation might play an important role in the development of CTEPH.
Collapse
Affiliation(s)
- Qunying Xi
- 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
- Center for Pulmonary Vascular Diseases, 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
| | - Zhihong Liu
- Center for Pulmonary Vascular Diseases, 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
- * E-mail:
| | - Zhihui Zhao
- Center for Pulmonary Vascular Diseases, 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
| | - Qin Luo
- Center for Pulmonary Vascular Diseases, 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
| | - Zhiwei Huang
- 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
- Center for Pulmonary Vascular Diseases, 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
| |
Collapse
|
43
|
Hounkpe BW, Fiusa MML, Colella MP, da Costa LNG, Benatti RDO, Saad STO, Costa FF, dos Santos MNN, De Paula EV. Role of innate immunity-triggered pathways in the pathogenesis of Sickle Cell Disease: a meta-analysis of gene expression studies. Sci Rep 2015; 5:17822. [PMID: 26648000 PMCID: PMC4673434 DOI: 10.1038/srep17822] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 11/06/2015] [Indexed: 12/19/2022] Open
Abstract
Despite the detailed characterization of the inflammatory and endothelial changes observed in Sickle Cell Disease (SCD), the hierarchical relationship between elements involved in the pathogenesis of this complex disease is yet to be described. Meta-analyses of gene expression studies from public repositories represent a novel strategy, capable to identify key mediators in complex diseases. We performed several meta-analyses of gene expression studies involving SCD, including studies with patient samples, as well as in-vitro models of the disease. Meta-analyses were performed with the Inmex bioinformatics tool, based on the RankProd package, using raw gene expression data. Functional gene set analysis was performed using more than 60 gene-set libraries. Our results demonstrate that the well-characterized association between innate immunity, hemostasis, angiogenesis and heme metabolism with SCD is also consistently observed at the transcriptomic level, across independent studies. The enrichment of genes and pathways associated with innate immunity and damage repair-associated pathways supports the model of erythroid danger-associated molecular patterns (DAMPs) as key mediators of the pathogenesis of SCD. Our study also generated a novel database of candidate genes, pathways and transcription factors not previously associated with the pathogenesis of SCD that warrant further investigation in models and patients of SCD.
Collapse
Affiliation(s)
| | - Maiara Marx Luz Fiusa
- Faculty of Medical Sciences, University of Campinas/Hematology and Hemotherapy Center, Campinas, SP, Brazil
| | - Marina Pereira Colella
- Faculty of Medical Sciences, University of Campinas/Hematology and Hemotherapy Center, Campinas, SP, Brazil
| | | | | | - Sara T Olalla Saad
- Faculty of Medical Sciences, University of Campinas/Hematology and Hemotherapy Center, Campinas, SP, Brazil
| | - Fernando Ferreira Costa
- Faculty of Medical Sciences, University of Campinas/Hematology and Hemotherapy Center, Campinas, SP, Brazil
| | | | - Erich Vinicius De Paula
- Faculty of Medical Sciences, University of Campinas/Hematology and Hemotherapy Center, Campinas, SP, Brazil
| |
Collapse
|
44
|
Li X, Wang G, An Y, Li H, Li Y, Wu C. Association Between Sequence Variations in RCAN1 Promoter and the Risk of Sporadic Congenital Heart Disease in a Chinese Population. Pediatr Cardiol 2015; 36:1393-9. [PMID: 25863471 DOI: 10.1007/s00246-015-1172-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/02/2015] [Indexed: 12/22/2022]
Abstract
The pathogenesis of congenital heart disease (CHD) is unclear. There is a high incidence of CHD in Down syndrome, in which RCAN1 (regulator of calcineurin 1) overexpression is observed. However, whether RCAN1 plays an important role in non-syndromic CHD is unknown. This study investigates the relationship between sequence variations in the RCAN1 promoter and sporadic CHD. This was a case-control study in which the RCAN1 promoter was cloned and sequenced in 128 CHD patients (median age 1.1 year) and 150 normal controls (median age 3.0 year). No mutation sites had been identified in this research. Three single-nucleotide (C to T) polymorphisms were detected: rs193289374, rs149048873 and rs143081213. The polymorphisms were not associated with CHD risk according to a logistic regression analysis. Functional assays in vitro showed that compared with the wild-type genotype, the rs149048873 polymorphism decreased, and the rs143081213 increased, the RCAN1 promoter activity, though the rs193289374 polymorphism had no effect. In conclusion, the sequence variations in RCAN1 promoter are not major genetic factors involved in sporadic CHD, at least in the current research population.
Collapse
Affiliation(s)
- Xiaoyong Li
- Department of Thoracic and Cardiovascular Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Gang Wang
- Department of Thoracic and Cardiovascular Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yong An
- Department of Thoracic and Cardiovascular Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Hongbo Li
- Department of Thoracic and Cardiovascular Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yonggang Li
- Department of Thoracic and Cardiovascular Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Chun Wu
- Department of Thoracic and Cardiovascular Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
45
|
Li F, Zhang A, Shi Y, Ma Y, Du Y. 1α,25-Dihydroxyvitamin D3 prevents the differentiation of human lung fibroblasts via microRNA-27b targeting the vitamin D receptor. Int J Mol Med 2015; 36:967-74. [PMID: 26311239 PMCID: PMC4564074 DOI: 10.3892/ijmm.2015.2318] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 08/14/2015] [Indexed: 01/11/2023] Open
Abstract
Pulmonary fibroblasts have key roles in the formation and maintenance of lung structure and function, and are involved in tissue repair and remodeling. Transforming growth factor-β1 (TGF-β1) induces differentiation of fibroblasts into myofibroblasts, the key effector cells in fibrotic states, which are characterized by the expression of α-smooth muscle actin (α-SMA) markers. 1α,25-Dihydroxyvitamin D3 [1,25(OH)2D3] has been implicated in regulating differentiation, and the vitamin D receptor (VDR) may be a regulator of TGF-β signaling. In addition, there is presently only limited information regarding microRNA (miRNA) regulation of lung fibroblast differentiation. To determine the role of 1,25(OH)2D3 in regulating the differentiation of fibroblasts induced by TGF-β1 and the functional importance of miR-27b, cell culture systems, cell transfection and the 3′ untranslated region (3′UTR) luciferase assay were employed. 1,25(OH)2D3 inhibited differentiation and downregulated miR-27b expression in human lung fibroblasts induced by TGF-β1. In addition, human lung fibroblasts were transfected with miR-27b mimic or miR-27b inhibitor, and demonstrated that the overexpression of miR-27b decreased the VDR protein expression and increased the expression of α-SMA, while reducing levels of miR-27b had opposing effects. Finally, the luciferase reporter assays were performed to confirm that miR-27b directly targeted VDR 3′UTR. Taken together, these results suggest that 1,25(OH)2D3 inhibits lung fibroblast differentiation induced by TGF-β1 via miR-27b targeting VDR 3′UTR, which may be used as a novel treatment strategy in differentiation pathways.
Collapse
Affiliation(s)
- Fei Li
- Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Aizhen Zhang
- Department of Respiratory Diseases, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Yiwei Shi
- Department of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yuehong Ma
- Central Laboratory, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Yongcheng Du
- Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
46
|
Ma L, Bao R. Pulmonary capillary hemangiomatosis: a focus on the EIF2AK4 mutation in onset and pathogenesis. APPLICATION OF CLINICAL GENETICS 2015; 8:181-8. [PMID: 26300654 PMCID: PMC4536836 DOI: 10.2147/tacg.s68635] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pulmonary capillary hemangiomatosis (PCH) is a pulmonary vascular disease that mainly affects small capillaries in the lung, and is often misdiagnosed as pulmonary arterial hypertension or pulmonary veno-occlusive disease due to similarities in their clinical presentations, prognosis, and management. In patients who are symptomatic, there is a high mortality rate with median survival of 3 years after diagnosis. Both idiopathic and familial PCH cases are being reported, indicating there is genetic component in disease etiology. Mutations in the eukaryotic translation initiation factor 2α kinase 4 (EIF2AK4) gene were identified in familial and idiopathic PCH cases, suggesting EIF2AK4 is a genetic risk factor for PCH. EIF2AK4 mutations were identified in 100% (6/6) of autosomal recessively inherited familial PCH and 20% (2/10) of sporadic PCH cases. EIF2AK4 is a member of serine/threonine kinases. It downregulates protein synthesis in response to a variety of cellular stress such as hypoxia, viral infection, and amino acid deprivation. Bone morphogenetic protein receptor 2 (BMPR2) is a major genetic risk factor in pulmonary arterial hypertension and EIF2AK4 potentially connects with BMPR2 to cause PCH. L-Arginine is substrate of nitric oxide synthase, and L-arginine is depleted during the production of nitric oxide, which may activate EIF2AK4 to inhibit protein synthesis and negatively regulate vasculogenesis. Mammalian target of rapamycin and EIF2α kinase are two major pathways for translational regulation. Mutant EIF2AK4 could promote proliferation of small pulmonary arteries by crosstalk with mammalian targets of the rapamycin signaling pathway. EIF2AK4 may regulate angiogenesis by modulating the immune system in PCH pathogenesis. The mechanisms of abnormal capillary angiogenesis are suggested to be similar to that of tumor vascularization. Specific therapies were developed according to pathogenesis and are proved to be effective in reported cases. Targeting the EIF2AK4 pathway may provide a novel therapy for PCH.
Collapse
Affiliation(s)
- Lijiang Ma
- Department of Pediatrics and Medicine, Division of Molecular Genetics, Columbia University Medical Center, New York, NY, USA
| | - Ruijun Bao
- The Children's IBD Center, Mount Sinai Hospital, New York, NY, USA
| |
Collapse
|
47
|
Kim JD, Lee A, Choi J, Park Y, Kang H, Chang W, Lee MS, Kim J. Epigenetic modulation as a therapeutic approach for pulmonary arterial hypertension. Exp Mol Med 2015; 47:e175. [PMID: 26228095 PMCID: PMC4525299 DOI: 10.1038/emm.2015.45] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare but progressive and currently incurable disease, which is characterized by vascular remodeling in association with muscularization of the arterioles, medial thickening and plexiform lesion formation. Despite our advanced understanding of the pathogenesis of PAH and the recent therapeutic advances, PAH still remains a fatal disease. In addition, the susceptibility to PAH has not yet been adequately explained. Much evidence points to the involvement of epigenetic changes in the pathogenesis of a number of human diseases including cancer, peripheral hypertension and asthma. The knowledge gained from the epigenetic study of various human diseases can also be applied to PAH. Thus, the pursuit of novel therapeutic targets via understanding the epigenetic alterations involved in the pathogenesis of PAH, such as DNA methylation, histone modification and microRNA, might be an attractive therapeutic avenue for the development of a novel and more effective treatment. This review provides a general overview of the current advances in epigenetics associated with PAH, and discusses the potential for improved treatment through understanding the role of epigenetics in the development of PAH.
Collapse
Affiliation(s)
- Jun-Dae Kim
- Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Aram Lee
- Department of Life Systems, Sookmyung Women's University, Seoul, Korea
| | - Jihea Choi
- Department of Life Systems, Sookmyung Women's University, Seoul, Korea
| | - Youngsook Park
- Department of Life Systems, Sookmyung Women's University, Seoul, Korea
| | - Hyesoo Kang
- Department of Life Systems, Sookmyung Women's University, Seoul, Korea
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan, Korea
| | - Myeong-Sok Lee
- Department of Life Systems, Sookmyung Women's University, Seoul, Korea
| | - Jongmin Kim
- Department of Life Systems, Sookmyung Women's University, Seoul, Korea
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW The identification of the genetic basis for heritable predisposition to pulmonary arterial hypertension (PAH) has altered the clinical and research landscape for PAH patients and their care providers. This review aims to describe the genetic discoveries and their impact on clinical medicine. RECENT FINDINGS Since the landmark discovery that bone morphogenetic protein receptor type II (BMPR2) mutations cause the majority of cases of familial PAH, investigators have discovered mutations in genes that cause PAH in families without BMPR2 mutations, including the type I receptor ACVRL1 and the type III receptor ENG (both associated with hereditary hemorrhagic telangiectasia), caveolin-1 (CAV1), and a gene (KCNK3) encoding a two-pore potassium channel. Mutations in these genes cause an autosomal-dominant predisposition to PAH in which a fraction of mutation carriers develop PAH (incomplete penetrance). In 2014, scientists discovered mutations in eukaryotic initiation factor 2 alpha kinase 4 (EIF2AK4) that cause pulmonary capillary hemangiomatosis and pulmonary veno-occlusive disease, an autosomal recessively inherited disorder. SUMMARY The discovery that some forms of pulmonary hypertension are heritable and can be genetically defined adds important opportunities for physicians to educate their patients and their families to understand the potential risks and benefits of genetic testing.
Collapse
Affiliation(s)
- D Hunter Best
- aDepartment of Pathology, University of Utah School of Medicine bARUP Laboratories, ARUP Institute for Clinical and Experimental Pathology, Salt Lake City, Utah cDepartment of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee dDepartments of Pediatrics and Medicine, Columbia University Medical Center, New York, New York eDepartment of Medicine, Intermountain Medical Center, Murray fDepartment of Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | | | | | | |
Collapse
|
49
|
Fonseca DJ, Patiño LC, Suárez YC, de Jesús Rodríguez A, Mateus HE, Jiménez KM, Ortega-Recalde O, Díaz-Yamal I, Laissue P. Next generation sequencing in women affected by nonsyndromic premature ovarian failure displays new potential causative genes and mutations. Fertil Steril 2015; 104:154-62.e2. [PMID: 25989972 DOI: 10.1016/j.fertnstert.2015.04.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 04/07/2015] [Accepted: 04/12/2015] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To identify new molecular actors involved in nonsyndromic premature ovarian failure (POF) etiology. DESIGN This is a retrospective case-control cohort study. SETTING University research group and IVF medical center. PATIENT(S) Twelve women affected by nonsyndromic POF. The control group included 176 women whose menopause had occurred after age 50 and had no antecedents regarding gynecological disease. A further 345 women from the same ethnic origin (general population group) were also recruited to assess allele frequency for potentially deleterious sequence variants. INTERVENTION(S) Next generation sequencing (NGS), Sanger sequencing, and bioinformatics analysis. MAIN OUTCOME MEASURE(S) The complete coding regions of 70 candidate genes were massively sequenced, via NGS, in POF patients. Bioinformatics and genetics were used to confirm NGS results and to identify potential sequence variants related to the disease pathogenesis. RESULT(S) We have identified mutations in two novel genes, ADAMTS19 and BMPR2, that are potentially related to POF origin. LHCGR mutations, which might have contributed to the phenotype, were also detected. CONCLUSION(S) We thus recommend NGS as a powerful tool for identifying new molecular actors in POF and for future diagnostic/prognostic purposes.
Collapse
Affiliation(s)
- Dora Janeth Fonseca
- Unidad de Genética, Grupo GENIUROS, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogota, Colombia
| | - Liliana Catherine Patiño
- Unidad de Genética, Grupo GENIUROS, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogota, Colombia
| | - Yohjana Carolina Suárez
- Unidad de Genética, Grupo GENIUROS, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogota, Colombia
| | - Asid de Jesús Rodríguez
- Unidad de Genética, Grupo GENIUROS, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogota, Colombia
| | - Heidi Eliana Mateus
- Unidad de Genética, Grupo GENIUROS, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogota, Colombia
| | - Karen Marcela Jiménez
- Unidad de Genética, Grupo GENIUROS, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogota, Colombia
| | - Oscar Ortega-Recalde
- Unidad de Genética, Grupo GENIUROS, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogota, Colombia
| | | | - Paul Laissue
- Unidad de Genética, Grupo GENIUROS, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogota, Colombia.
| |
Collapse
|
50
|
Vattulainen S, Aho J, Salmenperä P, Bruce S, Tallila J, Gentile M, Sankelo M, Laitinen T, Koskenvuo JW, Alastalo TP, Myllykangas S. Accurate genetic diagnosis of Finnish pulmonary arterial hypertension patients using oligonucleotide-selective sequencing. Mol Genet Genomic Med 2015; 3:354-62. [PMID: 26247051 PMCID: PMC4521970 DOI: 10.1002/mgg3.147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/24/2015] [Indexed: 01/09/2023] Open
Abstract
The genetic basis of pulmonary arterial hypertension (PAH) among Finnish PAH patients is poorly understood. We adopted a novel-targeted next-generation sequencing (NGS) approach called Oligonucleotide-Selective Sequencing (OS-Seq) and developed a custom data analysis and interpretation pipeline to identify pathogenic base substitutions, insertions, and deletions in seven genes associated with PAH (BMPR2, BMPR1B, ACVRL1, ENG, SMAD9, CAV1, and KCNK3) from Finnish PAH patients. This study represents the first clinical study with OS-Seq technology on patients suffering from a rare genetic disorder. We analyzed DNA samples from 21 Finnish PAH patients, whose BMPR2 and ACVRL1 mutation status had been previously studied using Sanger sequencing. Our sequencing panel covered 100% of the targeted base pairs with >15× sequencing depth. Pathogenic base substitutions were identified in the BMPR2 gene in 29% of the Finnish PAH cases. Two of the pathogenic variant-positive patients had been previously tested negative using Sanger sequencing. No clinically significant variants were identified in the six other PAH genes. Our study validates the use of targeted OS-Seq for genetic diagnostics of PAH and revealed pathogenic variants that had been previously missed using Sanger sequencing.
Collapse
Affiliation(s)
- Sanna Vattulainen
- Pediatric Cardiology, Children's Hospital Helsinki, University of Helsinki and Helsinki University Central Hospital Helsinki, Finland
| | - Joonas Aho
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku Turku, Finland
| | | | | | | | | | - Marja Sankelo
- Department of Internal Medicine, Tampere University Hospital Tampere, Finland
| | - Tarja Laitinen
- Department of Pulmonary Diseases and Allergology, Turku University Hospital and University of Turku Turku, Finland
| | - Juha W Koskenvuo
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku Turku, Finland ; Blueprint Genetics Helsinki, Finland ; Department of Clinical Physiology and Nuclear Medicine, HUS Medical Imaging Center, Helsinki University of Central Hospital and University of Helsinki Helsinki, Finland
| | - Tero-Pekka Alastalo
- Pediatric Cardiology, Children's Hospital Helsinki, University of Helsinki and Helsinki University Central Hospital Helsinki, Finland ; Blueprint Genetics Helsinki, Finland
| | - Samuel Myllykangas
- Blueprint Genetics Helsinki, Finland ; Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki Helsinki, Finland
| |
Collapse
|