1
|
Garry DJ, Garry MG, Nakauchi H, Masaki H, Sachs DH, Weiner JI, Reichart D, Wolf E. Allogeneic, Xenogeneic, and Exogenic Hearts for Transplantation. Methodist Debakey Cardiovasc J 2025; 21:92-99. [PMID: 40384731 PMCID: PMC12082467 DOI: 10.14797/mdcvj.1590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/07/2025] [Indexed: 05/20/2025] Open
Abstract
The only curative therapy for end-stage heart failure is orthotopic allogeneic heart transplantation. This therapy has extended the survival of patients worldwide but is limited due to the scarcity of donor organs. Potential alternative donor sources of organs for transplantation include genetically-modified (GM) large animal donors (ie, xenografts) and human organs developed in large animal hosts. These strategies utilize gene editing and somatic cell nuclear transfer technologies to engineer partially or completely humanized organs. Preclinical xenotransplantation studies of GM pig hearts into baboons have already provided an important clinical foundation, as two patients have received cardiac xenografts from GM pigs and have survived for up to 2 months. Additional issues need to be addressed in order for patients to survive more than 1 year, which would make these strategies clinically applicable. Thus, in combination with immunosuppression agents, xenogeneic and exogenic organ sources hold tremendous promise for an unlimited and transformative supply of organs for transplantation.
Collapse
Affiliation(s)
- Daniel J. Garry
- Stem Cell Institute, IN
- Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, US
| | - Mary G. Garry
- Stem Cell Institute, IN
- Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, US
| | - Hiromitsu Nakauchi
- University of Tokyo, Tokyo, JP
- Institute of Science Tokyo (formerly Tokyo Medical and Dental University), Tokyo, JP
- Stanford University School of Medicine, Stanford, California, US
| | - Hideki Masaki
- University of Tokyo, Tokyo, JP
- Institute of Science Tokyo (formerly Tokyo Medical and Dental University) Tokyo, JP
| | - David H. Sachs
- Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, US
- Massachusetts General Hospital, Boston, Massachusetts, US
| | - Joshua I. Weiner
- Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, US
| | - Daniel Reichart
- University Hospital, LMU Munich, Munich, DE
- Gene Center and Center for Innovative Medical Models (CiMM), DE
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, DE
| | - Eckhard Wolf
- Gene Center and Center for Innovative Medical Models (CiMM), DE
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, DE
| |
Collapse
|
2
|
Ge K, Aierken D, Deng D, Jiang Z, Feng T, Meng J, Zhang H, Cao J, Liu C. High-Fat Diet during Mouse Pregnancy Impairs Fetal Heart Development. Int Heart J 2025; 66:144-156. [PMID: 39894543 DOI: 10.1536/ihj.24-245] [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] [Indexed: 02/04/2025]
Abstract
Maternal overnutrition correlates with detrimental outcomes in offspring. However, the specific effects of gestational exposure to a high-fat diet (HFD) on fetal development remain unclear. This study aimed to elucidate the developmental phenotypes of neonatal organs and cardiomyocytes of mice exposed to gestational HFD, revealing growth retardation and a notable reduction in cardiomyocyte cell cycle activity. In this study, an HFD model was used to investigate the effects of maternal HFD on offspring development. Defective development was observed in the offspring, and severe restriction of cell proliferation was noted in the neonatal organs as a result of maternal HFD. Based on this evidence, we detected a reduction in cardiomyocyte proliferation in offspring exposed to maternal HFD. Moreover, RNA sequencing analysis revealed that HFD diminished fatty acid metabolism, enhanced the inflammatory response, and upregulated the transcription of genes involved in Tp53-regulated cell cycle arrest in postnatal day 0 (P0) cardiomyocytes. Furthermore, our results showed that the effects of the maternal diet during gestation are profound and normal lactation and feeding after delivery cannot help adult offspring recover from defective heart development. These findings highlight the diverse pathways affected by maternal HFD, particularly implicating a potential TP53-dependent mechanism contributing to cardiac defects in offspring.
Collapse
Affiliation(s)
- Kaixin Ge
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University
| | | | - Defang Deng
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University
| | - Zhen Jiang
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University
| | - Teng Feng
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University
| | - Jufeng Meng
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University
| | - Hui Zhang
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University
| | - Jinjun Cao
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Shanghai Jiao Tong University
| | - Chen Liu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University
| |
Collapse
|
3
|
Yang W, Yin D, Zhang K, Xiang P, Zhou X, Zheng M, Li M, Cheng Z. Non-compaction of the ventricular myocardium associated with large patent ductus arteriosus: primary or secondary? BMC Cardiovasc Disord 2024; 24:671. [PMID: 39581959 PMCID: PMC11587748 DOI: 10.1186/s12872-024-04334-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND The absence of other structural heart disease is a prerequisite for the diagnosis of non-compaction of the ventricular myocardium (NVM). We also observed that the phenomenon of non-compaction in ventricular muscle in some large patent ductus arteriosus (PDA) patients in children. This study was aimed to explore the prognosis of NVM associated with large PDA in children and provide a better understanding of the interplay between genetic and hemodynamic factors that lead to the phenotype of NVM. METHODS We retrospectively analyzed the clinical data of the patients with the diagnosis of NVM with large PDA from January 2015 to January 2022 who underwent the interventional occlusion. We collected the data of the non-compacted myocardium/compacted myocardium (N/C) ratio, the size of the heart, cardiac function measured on color Doppler echocardiography and electrocardiograph (ECG) before and after interventional occlusion. RESULTS From a total of 504 patients with large PDA underwent occlusion, 20 patients (3.94%; mean age 1.72 ± 1.44 years, 6males and 14 females) were also diagnosed with NVM. The mean diameter of the pulmonary artery end of the PDA was 7.15 ± 1.19 mm. The N/C ratio of all children significantly improved after PDA interventional occlusion with the decreased trend of LVDd and LVDs. The phenomenon of NVM regressed during the follow-up period: 9 cases(45%) after 1 M, 2 cases(10%) after 3 Ms, 4 cases(20%) after 6Ms, 1 case(5%) after 9Ms, 3 cases(15%) after 12Ms, and 1 case(5%) after 24Ms. However, there was no significant difference in the ejection fraction(EF) at all time points (P > 0.05). During the follow-up, the typical complications of NVM, include congestive heart failure, ventricular arrhythmias and thromboembolic events weren't observed in these patients. CONCLUSION The phenomenon of NVM in this group maybe secondary to a large PDA. After the primary factors have been removed, hemodynamic changes and a decrease in the left heart's preload favor the complete regression of the NVM.
Collapse
Affiliation(s)
- Wenyi Yang
- Department of Cardiovascular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Dan Yin
- Department of Cardiovascular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Kaijun Zhang
- Department of Cardiovascular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Ping Xiang
- Department of Cardiovascular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xue Zhou
- Department of Cardiovascular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Min Zheng
- Department of Cardiovascular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Mi Li
- Department of Cardiovascular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zhenli Cheng
- Department of Cardiovascular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.
- National Clinical Research Center for Child Health and Disorders, Chongqing, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.
- Chongqing Key Laboratory of Pediatrics, Chongqing, China.
| |
Collapse
|
4
|
Hirono K, Hata Y, Imamura T, Tsuboi K, Takarada S, Okabe M, Nakaoka H, Ibuki K, Ozawa S, Ichimata S, Nishida N, Iwasaki H, Urata S, Okada S, Hiratsuji T, Sakaguchi H, Takigiku K, Nakazawa M, Nishihara E, Harada M, Matsuo O, Yasuda K, Yoshida Y, Namiki H, Yasuda K, Ifuku T, Urayama K, Oka H, Ogino K, Kato A, Kan N, Seki S, Seki M, Odanaka Y, Iwashima S, Yoshida S, Miyata T, Miyamoto T, Watanabe K, Kuwabara N, Inuzuka R, Takahashi Y, Sakazaki H, Muneuchi J, Kogaki S, Numano F, Kido S, Nii M, Hoshino S, Ishida H, Maeda J, Hayabuchi Y, Otsubo Y, Ikeda K, Tsukano S, Watanabe M, Momoi N, Fujii T, Fujioka T, Fujino M, Uchiyama H, Baba S, Horigome H, Honda T, Suzuki K, Ichida F. Determination of Genotype and Phenotypes in Pediatric Patients With Biventricular Noncompaction. J Am Heart Assoc 2024; 13:e035614. [PMID: 39494597 PMCID: PMC11935668 DOI: 10.1161/jaha.124.035614] [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: 03/21/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Left ventricular noncompaction (LVNC) is a hereditary type of cardiomyopathy characterized by prominent trabeculations. Detailed characteristics of biventricular noncompaction (BiVNC) remain unknown. This study aimed to elucidate the clinical characteristics and genetic landscape of BiVNC. METHODS AND RESULTS We recruited children with left ventricular noncompaction from Japanese multi-institutional centers from 2013 to 2021. Left ventricular noncompaction was classified as BiVNC, congenital heart disease, arrhythmia, dilated cardiomyopathy, or normal function. In these patients, cardiomyopathy-associated genes were screened. A total of 234 patients (127 male; mean age, 4 months [range, 0-6.6 years]) were enrolled in this study, of whom 25 had BiVNC; 55, normal function; 84, dilated cardiomyopathy; 38, congenital heart disease; and 32, arrhythmia. BiVNC was diagnosed during the perinatal period in 10 patients, in whom the prevalence was higher than that in other patients. A total of 14 patients in the group with BiVNC had congenital heart disease, but not necessarily right heart lesions. Left ventricular dyskinesis was frequently observed in the lateral wall (24%) and apex (28%). Eleven pathogenic variants were found in 11 patients with BiVNC (44.0%). The group with BiVNC had a higher ratio of mitochondrial and developmental gene variants than the other groups. Among all groups, the group with BiVNC had the worst survival rate (P=0.0009). CONCLUSIONS Pediatric patients with BiVNC had a high rate of ventricular dyskinesis and poor outcome. A comprehensive and careful screening for disease-causing genes and phenotype may help identify specific patients with left ventricular noncompaction and mortality-related cardiac phenotypes.
Collapse
Affiliation(s)
- Keiichi Hirono
- Department of Pediatrics, Faculty of MedicineUniversity of ToyamaJapan
| | - Yukiko Hata
- Legal Medicine, Faculty of MedicineUniversity of ToyamaJapan
| | - Teruhiko Imamura
- 2nd Department of Internal Medicine, Faculty of MedicineUniversity of ToyamaJapan
| | - Kaori Tsuboi
- Department of Pediatrics, Faculty of MedicineUniversity of ToyamaJapan
| | - Shinya Takarada
- Department of Pediatrics, Faculty of MedicineUniversity of ToyamaJapan
| | - Mako Okabe
- Department of Pediatrics, Faculty of MedicineUniversity of ToyamaJapan
| | - Hideyuki Nakaoka
- Department of Pediatrics, Faculty of MedicineUniversity of ToyamaJapan
| | - Keijiro Ibuki
- Department of Pediatrics, Faculty of MedicineUniversity of ToyamaJapan
| | - Sayaka Ozawa
- Department of Pediatrics, Faculty of MedicineUniversity of ToyamaJapan
| | | | - Naoki Nishida
- Legal Medicine, Faculty of MedicineUniversity of ToyamaJapan
| | - Hidenori Iwasaki
- Department of PediatricsKanazawa University HospitalIshikawaJapan
| | - Susumu Urata
- Division of CardiologyNational Center for Child Health and DevelopmentTokyoJapan
| | - Seigo Okada
- Department of PediatricsYamaguchi University Graduate School of MedicineYamaguchiJapan
| | - Tomoya Hiratsuji
- Department of PediatricsOkinawa Prefectural Hokubu HospitalOkinawaJapan
| | - Heima Sakaguchi
- Department of Pediatric CardiologyNational Cerebral and Cardiovascular CenterOsakaJapan
| | - Kiyohiro Takigiku
- Department of Pediatric CardiologyNagano Children’s HospitalNaganoJapan
| | - Makoto Nakazawa
- Department of PediatricsSouthern Tohoku Research Institute for NeuroscienceFukushimaJapan
| | - Eiki Nishihara
- Department of Pediatric Cardiology and NeonatologyOgaki Municipal HospitalGifuJapan
| | - Masako Harada
- Department of PediatricsMiyazaki University HospitalMiyazakiJapan
| | - Osamu Matsuo
- Department of PediatricsKumamoto UniversityKumamotoJapan
| | - Kenji Yasuda
- Department of PediatricsShimane University HospitalShimaneJapan
| | - Yoko Yoshida
- Decision of Pediatric ElectrophysiologyOsaka City General HospitalOsakaJapan
| | - Hidemasa Namiki
- Department of Pediatrics and Child HealthNihon University School of Medicine & Itabashi HospitalTokyoJapan
| | - Kazushi Yasuda
- Department of Pediatric CardiologyAichi Children’s Health and Medical CenterAichiJapan
| | - Toshinobu Ifuku
- Department of PediatricsMiyazaki Prefectural Miyazaki HospitalMiyazakiJapan
| | - Kotaro Urayama
- Department of PediatricsTsuchiya General HospitalHiroshimaJapan
| | - Hideharu Oka
- Department of PediatricsAsahikawa Medical University HospitalAsahikawaHokkaidoJapan
| | - Kayo Ogino
- Department of PediatricsKurashiki Central HospitalOkayamaJapan
| | - Akio Kato
- Department of Pediatric CardiologyOkinawa Prefectural Nanbu Medical Center and Children’s Medical CenterOkinawaJapan
| | - Nobuhiko Kan
- Department of Fetal and Neonatal CardiologyFukuoka Children’s HospitalFukuokaJapan
| | - Shunji Seki
- Department of PediatricsIbusuki Medical CenterKagoshimaJapan
| | - Mitsuru Seki
- Department of PediatricsJichi Medical UniversityTochigiJapan
| | - Yutaka Odanaka
- Department of PediatricsOsaka Medical and Pharmaceutical UniversityOsakaJapan
| | - Satoru Iwashima
- Department of PediatricsChutoen General Medical CenterShizuokaJapan
| | - Shuichiro Yoshida
- Department of Pediatrics Cardiology, Chukyo HospitalJapan Community Healthcare OrganizationAichiJapan
| | | | | | - Ken Watanabe
- Department of PediatricsKitano Hospital Tazuke Kofukai Medical Research InstituteOsakaJapan
| | - Naoki Kuwabara
- Department of Pediatric CardiologyGifu Prefectural General Medical CenterGifuJapan
| | - Ryo Inuzuka
- Department of PediatricsTokyo University HospitalTokyoJapan
| | | | - Hisanori Sakazaki
- Department of Pediatric CardiologyHyogo Prefectural Amagasaki General Medical CenterHyogoJapan
| | - Jun Muneuchi
- Department of Pediatrics, Kyushu HospitalJapan Community Healthcare OrganizationFukuokaJapan
| | - Shigetoyo Kogaki
- Department of Pediatrics and NeonatologyOsaka General Medical CenterOsakaJapan
| | - Fujito Numano
- Department of PediatricsNiigata University Medical and Dental HospitalNiigataJapan
| | - Sachiko Kido
- Department of CardiologyHyogo Prefectural Children’s HospitalHyogoJapan
| | - Masaki Nii
- Department of CardiologyShizuoka Children’s HospitalShizuokaJapan
| | - Shinsuke Hoshino
- Department of PediatricsShiga University of Medical ScienceOtsuShigaJapan
| | - Hidekazu Ishida
- Department of PediatricsOsaka University Graduate School of MedicineOsakaJapan
| | - Jun Maeda
- Division of CardiologyTokyo Metropolitan Children’s Medical CenterTokyoJapan
| | | | - Yoshikazu Otsubo
- Department of PediatricsSasebo City General HospitalNagasakiJapan
| | - Kazuyuki Ikeda
- Department of PediatricsKyoto Prefectural University of MedicineKyotoJapan
| | - Shinya Tsukano
- Department of PediatricsNiigata City General HospitalNiigataJapan
| | - Makoto Watanabe
- Department of PediatricsNippon Medical School HospitalTokyoJapan
| | - Nobuo Momoi
- Department of PediatricsFukushima Medical University HospitalFukushimaJapan
| | - Takanari Fujii
- Pediatric Heart Disease and Adult Congenital Heart Disease CenterShowa University HospitalTokyoJapan
| | - Tao Fujioka
- Department of PediatricsJapanese Red Cross Medical CenterTokyoJapan
| | - Mitsuhiro Fujino
- Department of Pediatric CardiologyOsaka City General HospitalOsakaJapan
| | - Hiroki Uchiyama
- Department of PediatricsHamamatsu University HospitalShizuokaJapan
| | - Shigehito Baba
- Department of PediatricsNiigata University Medical and Dental HospitalNiigataJapan
| | - Hitoshi Horigome
- Department of PediatricsUniversity of Tsukuba HospitalIbarakiJapan
| | - Takashi Honda
- Department of PediatricsKitasato University School of MedicineKanagawaJapan
| | - Kazutaka Suzuki
- Department of PediatricsNagoya City University HospitalAichiJapan
| | - Fukiko Ichida
- Department of PediatricsInternational University of Health and WelfareTokyoJapan
| |
Collapse
|
5
|
Li Y, Ma K, Dong Z, Gao S, Zhang J, Huang S, Yang J, Fang G, Li Y, Li X, Welch C, Griffin EL, Ramaswamy P, Valivullah Z, Liu X, Dong J, Wang DW, Du, Chung WK, Li Y. Frameshift variants in C10orf71 cause dilated cardiomyopathy in human, mouse, and organoid models. J Clin Invest 2024; 134:e177172. [PMID: 38950288 PMCID: PMC11178530 DOI: 10.1172/jci177172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/25/2024] [Indexed: 07/03/2024] Open
Abstract
Research advances over the past 30 years have confirmed a critical role for genetics in the etiology of dilated cardiomyopathies (DCMs). However, full knowledge of the genetic architecture of DCM remains incomplete. We identified candidate DCM causal gene, C10orf71, in a large family with 8 patients with DCM by whole-exome sequencing. Four loss-of-function variants of C10orf71 were subsequently identified in an additional group of492 patients with sporadic DCM from 2 independent cohorts. C10orf71 was found to be an intrinsically disordered protein specifically expressed in cardiomyocytes. C10orf71-KO mice had abnormal heart morphogenesis during embryonic development and cardiac dysfunction as adults with altered expression and splicing of contractile cardiac genes. C10orf71-null cardiomyocytes exhibited impaired contractile function with unaffected sarcomere structure. Cardiomyocytes and heart organoids derived from human induced pluripotent stem cells with C10orf71 frameshift variants also had contractile defects with normal electrophysiological activity. A rescue study using a cardiac myosin activator, omecamtiv mecarbil, restored contractile function in C10orf71-KO mice. These data support C10orf71 as a causal gene for DCM by contributing to the contractile function of cardiomyocytes. Mutation-specific pathophysiology may suggest therapeutic targets and more individualized therapy.
Collapse
Affiliation(s)
- Yang Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Ke Ma
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Zhujun Dong
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Shijuan Gao
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Jing Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Shan Huang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Jie Yang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Guangming Fang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Yujie Li
- Novogene Co. Ltd., Beijing, China
| | - Xiaowei Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Carrie Welch
- Department of Pediatrics, Columbia University, New York, New York, USA
| | - Emily L. Griffin
- Department of Pediatrics, Columbia University, New York, New York, USA
| | | | | | | | - Jianzeng Dong
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Wendy K. Chung
- Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| |
Collapse
|
6
|
Jensen B, Chang YH, Bamforth SD, Mohun T, Sedmera D, Bartos M, Anderson RH. The changing morphology of the ventricular walls of mouse and human with increasing gestation. J Anat 2024; 244:1040-1053. [PMID: 38284175 PMCID: PMC11095311 DOI: 10.1111/joa.14017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 01/30/2024] Open
Abstract
That the highly trabeculated ventricular walls of the developing embryos transform to the arrangement during the fetal stages, when the mural architecture is dominated by the thickness of the compact myocardium, has been explained by the coalescence of trabeculations, often erroneously described as 'compaction'. Recent data, however, support differential rates of growth of the trabecular and compact layers as the major driver of change. Here, these processes were assessed quantitatively and visualized in standardized views. We used a larger dataset than has previously been available of mouse hearts, covering the period from embryonic day 10.5 to postnatal day 3, supported by images from human hearts. The volume of the trabecular layer increased throughout development, in contrast to what would be expected had there been 'compaction'. During the transition from embryonic to fetal life, the rapid growth of the compact layer diminished the proportion of trabeculations. Similarly, great expansion of the central cavity reduced the proportion of the total cavity made up of intertrabecular recesses. Illustrations of the hearts with the median value of left ventricular trabeculation confirm a pronounced growth of the compact wall, with prominence of the central cavity. This corresponds, in morphological terms, to a reduction in the extent of the trabecular layer. Similar observations were made in the human hearts. We conclude that it is a period of comparatively slow growth of the trabecular layer, rather than so-called compaction, that is the major determinant of the changing morphology of the ventricular walls of both mouse and human hearts.
Collapse
Affiliation(s)
- Bjarke Jensen
- Department of Medical Biology, Amsterdam Cardiovascular SciencesUniversity of Amsterdam, Amsterdam UMCAmsterdamthe Netherlands
| | - Yun Hee Chang
- Department of Medical Biology, Amsterdam Cardiovascular SciencesUniversity of Amsterdam, Amsterdam UMCAmsterdamthe Netherlands
| | - Simon D. Bamforth
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastleUK
| | | | - David Sedmera
- Institute of Anatomy, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Martin Bartos
- Institute of Anatomy, First Faculty of MedicineCharles UniversityPragueCzech Republic
- Institute of Dental Medicine, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Robert H. Anderson
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastleUK
| |
Collapse
|
7
|
Shi L, Wei X, Luo J, Tu L. SGLT2 inhibition, venous thrombolism, and death due to cardiac causes: a mediation Mendelian randomization study. Front Cardiovasc Med 2024; 11:1339094. [PMID: 38803667 PMCID: PMC11128626 DOI: 10.3389/fcvm.2024.1339094] [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: 11/16/2023] [Accepted: 05/01/2024] [Indexed: 05/29/2024] Open
Abstract
Objective To investigate the causal role of venous thrombolism mediating sodium-glucose cotransporter 2 (SGLT2) inhibition in death due to cardiac causes using Mendelian randomization (MR). Methods A two-sample two-step MR was used to determine (1) the causal effects of SGLT2 inhibition on death due to cardiac causes; (2) the causal effects of venous thrombolism on death due to cardiac causes; and (3) the mediation effects of venous thrombolism. Genetic proxies for SGLT2 inhibition were identified as variants in the SLC5A2 gene that were associated with both levels of gene expression and hemoglobin A1c. Additionally, employing MR to investigate the causal association between SGLT2 inhibition and cardiac arrest as well as coronary heart disease (CHD). Results SGLT2 inhibition was associated with a lower risk of death due to cardiac causes (odds ratio [OR] = 0.983, [95% CI = 0.972, 0.993], P = 0.0016). Venous thrombolism was associated with death due to cardiac causes ([OR] = 1.031, [95% CI = 1.005, 1.057], P = 0.0199). Mediation analysis showed evidence of indirect effect of SGLT2 inhibition on death due to cardiac causes through venous thrombolism [β = -0.0015, (95% CI = -0.0032 -0.0002), P = 0.042], with a mediated proportion of 8.9% (95% CI = 1.2%, 18.7%) of the total. Furthermore, SGLT2 inhibition was linked to a lower risk of cardiac arrest ([OR] = 0.097, [95% CI = 0.013, 0.742], P = 0.025). SGLT2 inhibition was linked to a lower risk of CHD ([OR] = 0.957, [95% CI = 0.932, 0.982], P = 0.0009). Conclusions Our study identified the causal roles of SGLT2 inhibition in venous thrombolism. SGLT2 inhibition may influence death due to cardiac causes through venous thrombolism. Additionally, SGLT2 inhibition was associated with reduced risk of cardiac arrest and CHD.
Collapse
Affiliation(s)
- Lili Shi
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xiupan Wei
- Department of Rehabilitation Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jinlan Luo
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Ling Tu
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
8
|
Jensen B, Salvatori D, Schouten J, Meijborg VMF, Lauridsen H, Agger P. Trabeculations of the porcine and human cardiac ventricles are different in number but similar in total volume. Clin Anat 2024; 37:440-454. [PMID: 38217386 DOI: 10.1002/ca.24135] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/21/2023] [Indexed: 01/15/2024]
Abstract
An intricate meshwork of trabeculations lines the luminal side of cardiac ventricles. Compaction, a developmental process, is thought to reduce trabeculations by adding them to the neighboring compact wall which is then enlarged. When pig, a plausible cardiac donor for xenotransplantation, is compared to human, the ventricular walls appear to have fewer trabeculations. We hypothesized the trabecular volume is proportionally smaller in pig than in human. Macroscopically, we observed in 16 pig hearts that the ventricular walls harbor few but large trabeculations. Close inspection revealed a high number of tiny trabeculations, a few hundred, within the recesses of the large trabeculations. While tiny, these were still larger than embryonic trabeculations and even when considering their number, the total tally of trabeculations in pig was much fewer than in human. Volumetrics based on high-resolution MRI of additional six pig hearts compared to six human hearts, revealed the left ventricles were not significantly differently trabeculated (21.5 versus 22.8%, respectively), and the porcine right ventricles were only slightly less trabeculated (42.1 vs 49.3%, respectively). We then analyzed volumetrically 10 pig embryonic hearts from gestational day 14-35. The trabecular and compact layer always grew, as did the intertrabecular recesses, in contrast to what compaction predicts. The proportions of the trabecular and compact layers changed substantially, nonetheless, due to differences in their growth rate rather than compaction. In conclusion, processes that affect the trabecular morphology do not necessarily affect the proportion of trabecular-to-compact myocardium and they are then distinct from compaction.
Collapse
Affiliation(s)
- Bjarke Jensen
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Daniela Salvatori
- Department of Clinical Sciences, Anatomy and Physiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jacobine Schouten
- Department of Clinical Sciences, Anatomy and Physiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Veronique M F Meijborg
- Department of Experimental Cardiology, University of Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Henrik Lauridsen
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Peter Agger
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| |
Collapse
|
9
|
Sebastian WA, Inoue M, Shimizu N, Sato R, Oguri S, Itonaga T, Kishimoto S, Shiraishi H, Hanada T, Ihara K. Cardiac manifestations of human ACTA2 variants recapitulated in a zebrafish model. J Hum Genet 2024; 69:133-138. [PMID: 38316882 DOI: 10.1038/s10038-024-01221-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/31/2023] [Accepted: 01/19/2024] [Indexed: 02/07/2024]
Abstract
The ACTA2 gene encodes actin α2, a major smooth muscle protein in vascular smooth muscle cells. Missense variants in the ACTA2 gene can cause inherited thoracic aortic diseases with characteristic symptoms, such as dysfunction of smooth muscle cells in the lungs, brain vessels, intestines, pupils, bladder, or heart. We identified a heterozygous missense variant of Gly148Arg (G148R) in a patient with a thoracic aortic aneurysm, dissection, and left ventricular non-compaction. We used zebrafish as an in vivo model to investigate whether or not the variants might cause functional or histopathological abnormalities in the heart. Following the fertilization of one-cell stage embryos, we injected in vitro synthesized ACTA2 mRNA of wild-type, novel variant G148R, or the previously known pathogenic variant Arg179His (R179H). The embryos were maintained and raised for 72 h post-fertilization for a heart analysis. Shortening fractions of heart were significantly reduced in both pathogenic variants. A histopathological evaluation showed that the myocardial wall of ACTA2 pathogenic variants was thinner than that of the wild type, and the total cell number within the myocardium was markedly decreased in all zebrafish with pathogenic variants mRNAs. Proliferating cell numbers were also significantly decreased in the endothelial and myocardial regions of zebrafish with ACTA2 variants compared to the wild type. These results demonstrate the effects of ACTA2 G148R and R179H on the development of left ventricle non-compaction and cardiac morphological abnormalities. Our study highlights the previously unknown significance of the ACTA2 gene in several aspects of cardiovascular development.
Collapse
Affiliation(s)
| | - Masanori Inoue
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Nobuyuki Shimizu
- Department of Cell Biology, Oita University, Faculty of Medicine, Oita, Japan
| | - Ryosuke Sato
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Saori Oguri
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Tomoyo Itonaga
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Shintaro Kishimoto
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Hiroshi Shiraishi
- Department of Cell Biology, Oita University, Faculty of Medicine, Oita, Japan
| | - Toshikatsu Hanada
- Department of Cell Biology, Oita University, Faculty of Medicine, Oita, Japan.
| | - Kenji Ihara
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan.
| |
Collapse
|
10
|
Jensen B, Andelfinger GU, Postma AV. Molecular Pathways and Animal Models of Ebstein's Anomaly. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:915-928. [PMID: 38884760 DOI: 10.1007/978-3-031-44087-8_58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Ebstein's anomaly is a congenital malformation of the tricuspid valve characterized by abnormal attachment of the valve leaflets, resulting in varying degrees of valve dysfunction. The anatomic hallmarks of this entity are the downward displacement of the attachment of the septal and posterior leaflets of the tricuspid valve. Additional intracardiac malformations are common. From an embryological point of view, the cavity of the future right atrium does not have a direct orifice connected to the developing right ventricle. This chapter provides an overview of current insight into how this connection is formed and how malformations of the tricuspid valve arise from dysregulation of molecular and morphological events involved in this process. Furthermore, mouse models that show features of Ebstein's anomaly and the naturally occurring model of canine tricuspid valve malformation are described and compared to the human model. Although Ebstein's anomaly remains one of the least understood cardiac malformations to date, the studies summarized here provide, in aggregate, evidence for monogenic and oligogenic factors driving pathogenesis.
Collapse
Affiliation(s)
- Bjarke Jensen
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Gregor U Andelfinger
- Cardiovascular Genetics, Department of Pediatrics, CHU Sainte Justine, Université de Montréal, Montréal, QC, Canada
| | - Alex V Postma
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centres, Amsterdam, The Netherlands.
- Department of Human Genetics, Amsterdam University Medical Centres, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Garry DJ, Zhang J(J, Larson TA, Sadek HA, Garry MG. Networks that Govern Cardiomyocyte Proliferation to Facilitate Repair of the Injured Mammalian Heart. Methodist Debakey Cardiovasc J 2023; 19:16-25. [PMID: 38028968 PMCID: PMC10655759 DOI: 10.14797/mdcvj.1300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Cardiovascular diseases are the number one cause of death worldwide and in the United States (US). Cardiovascular diseases frequently progress to end-stage heart failure, and curative therapies are extremely limited. Intense interest has focused on deciphering the cascades and networks that govern cardiomyocyte proliferation and regeneration of the injured heart. For example, studies have shown that lower organisms such as the adult newt and adult zebrafish have the capacity to completely regenerate their injured heart with restoration of function. Similarly, the neonatal mouse and pig are also able to completely regenerate injured myocardium due to cardiomyocyte proliferation from preexisting cardiomyocytes. Using these animal models and transcriptome analyses, efforts have focused on the definition of factors and signaling pathways that can reactivate and induce cardiomyocyte proliferation in the adult mammalian injured heart. These studies and discoveries have the potential to define novel therapies to promote cardiomyocyte proliferation and repair of the injured, mammalian heart.
Collapse
Affiliation(s)
- Daniel J. Garry
- University of Minnesota, Minneapolis, Minnesota, US
- NorthStar Genomics, Eagan, Minnesota, US
| | | | | | | | - Mary G. Garry
- NorthStar Genomics, Eagan, Minnesota, US
- University of Minnesota, Minneapolis, MN
| |
Collapse
|
12
|
Hayal TB, Doğan A, Şenkal S, Bulut E, Şişli HB, Şahin F. Evaluation of the effect of boron derivatives on cardiac differentiation of mouse pluripotent stem cells. J Trace Elem Med Biol 2023; 79:127258. [PMID: 37451093 DOI: 10.1016/j.jtemb.2023.127258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 06/06/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND The heart is one of the first organs to form during embryonic development and has a very important place. So much that the formation of a functional heart is completed on the 55th day of human development and the 15th day of mouse development. Myocardial, endocardial and epicardial cells, which are derived from the mesoderm layer, are the cells that form the basis of the heart. Cardiac development, like other embryonic developments, is tightly controlled and regulated by various signaling pathways. The WNT signaling pathway is the most studied of these signaling pathways and the one with the clearest relationship with heart development. It is known that boron compounds and the Wnt/β-catenin pathway are highly correlated. Therefore, this study aimed to investigate the role of boron compounds in heart development as well as its effect on pluripotency of mouse embryonic stem cells for the first time in the literature. METHODS Toxicity of boron compounds was evaluated by using MTS analysis and obtained results were supported by morphological pictures, Trypan Blue staining and Annexin V staining. Additionally, the possible boron-related change in pluripotency of embryonic stem cells were analyzed with alkaline phosphatase activity and immunocytochemical staining of Oct4 protein as well as gene expression levels of pluripotency related OCT4, SOX2 and KLF4 genes. The alterations in the embryonic body formation capacity of mouse embryonic stem cells due to the application boron derivatives were also evaluated. Three linage differentiation was conducted to clarify the real impact of boron compounds on embryonic development. Lastly, cardiac differentiation of mESCs was investigated by using morphological pictures, cytosolic calcium measurement, gene expression and immunocytochemical analysis of cardiac differentiation related genes and in the presence of boron compounds. RESULTS Obtained results show that boron treatment maintains the pluripotency of embryonic stem cells at non-toxic concentrations. Additionally, endodermal, and mesodermal fate was found to be triggered after boron treatment. Also, initiation of cardiomyocyte differentiation by boron derivative treatments caused an increased gene expression levels of cardiac differentiation related TNNT2, Nkx2.5 and ISL-1 gene expression levels. CONCLUSION This study indicates that boron application, which is responsible for maintaining pluripotency of mESCs, can be used for increased cardiomyocyte differentiation of mESCs.
Collapse
Affiliation(s)
- Taha Bartu Hayal
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey; Current affiliation: Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States.
| | - Ayşegül Doğan
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Selinay Şenkal
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Ezgi Bulut
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Hatice Burcu Şişli
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
13
|
Yang H, Song S, Li J, Li Y, Feng J, Sun Q, Qiu X, Chen Z, Bai X, Liu X, Lian H, Liu L, Bai Y, Zhang G, Nie Y. Omentin-1 drives cardiomyocyte cell cycle arrest and metabolic maturation by interacting with BMP7. Cell Mol Life Sci 2023; 80:186. [PMID: 37344704 PMCID: PMC11071824 DOI: 10.1007/s00018-023-04829-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 04/05/2023] [Accepted: 06/04/2023] [Indexed: 06/23/2023]
Abstract
Mammalian cardiomyocytes (CMs) undergo maturation during postnatal heart development to meet the increased demands of growth. Here, we found that omentin-1, an adipokine, facilitates CM cell cycle arrest and metabolic maturation. Deletion of omentin-1 causes mouse heart enlargement and dysfunction in adulthood and CM maturation retardation in juveniles, including delayed cell cycle arrest and reduced fatty acid oxidation. Through RNA sequencing, molecular docking analysis, and proximity ligation assays, we found that omentin-1 regulates CM maturation by interacting directly with bone morphogenetic protein 7 (BMP7). Omentin-1 prevents BMP7 from binding to activin type II receptor B (ActRIIB), subsequently decreasing the downstream pathways mothers against DPP homolog 1 (SMAD1)/Yes-associated protein (YAP) and p38 mitogen-activated protein kinase (p38 MAPK). In addition, omentin-1 is required and sufficient for the maturation of human embryonic stem cell-derived CMs. Together, our findings reveal that omentin-1 is a pro-maturation factor for CMs that is essential for postnatal heart development and cardiac function maintenance.
Collapse
Affiliation(s)
- Huijun Yang
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Street, Beilishi Road, Xicheng District, Beijing, 100037, People's Republic of China
- Department of Cardiovascular Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Shen Song
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Street, Beilishi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Jiacheng Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
| | - Yandong Li
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Street, Beilishi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Jie Feng
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Street, Beilishi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Quan Sun
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Geriatric Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Street, Xiangya Road, Kaifu District, Changsha, 410008, People's Republic of China
| | - Xueting Qiu
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Geriatric Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Street, Xiangya Road, Kaifu District, Changsha, 410008, People's Republic of China
| | - Ziwei Chen
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Street, Beilishi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Xue Bai
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Street, Beilishi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Xinchang Liu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Street, Beilishi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Hong Lian
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Street, Beilishi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Lihui Liu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Street, Beilishi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Yongping Bai
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Geriatric Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Street, Xiangya Road, Kaifu District, Changsha, 410008, People's Republic of China.
| | - Guogang Zhang
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Geriatric Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Street, Xiangya Road, Kaifu District, Changsha, 410008, People's Republic of China.
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Street, Beilishi Road, Xicheng District, Beijing, 100037, People's Republic of China.
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China.
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou, 450046, China.
| |
Collapse
|
14
|
Lei YQ, Ye ZJ, Wei YL, Zhu LP, Zhuang XD, Wang XR, Cao H. Nono deficiency impedes the proliferation and adhesion of H9c2 cardiomyocytes through Pi3k/Akt signaling pathway. Sci Rep 2023; 13:7134. [PMID: 37130848 PMCID: PMC10154399 DOI: 10.1038/s41598-023-32572-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/29/2023] [Indexed: 05/04/2023] Open
Abstract
Congenital heart disease (CHD) is the most common type of birth defect and the main noninfectious cause of death during the neonatal stage. The non-POU domain containing, octamer-binding gene, NONO, performs a variety of roles involved in DNA repair, RNA synthesis, transcriptional and post-transcriptional regulation. Currently, hemizygous loss-of-function mutation of NONO have been described as the genetic origin of CHD. However, essential effects of NONO during cardiac development have not been fully elucidated. In this study, we aim to understand role of Nono in cardiomyocytes during development by utilizing the CRISPR/Cas9 gene editing system to deplete Nono in the rat cardiomyocytes H9c2. Functional comparison of H9c2 control and knockout cells showed that Nono deficiency suppressed cell proliferation and adhesion. Furthermore, Nono depletion significantly affected the mitochondrial oxidative phosphorylation (OXPHOS) and glycolysis, resulting in H9c2 overall metabolic deficits. Mechanistically we demonstrated that the Nono knockout impeded the cardiomyocyte function by attenuating phosphatidyl inositol 3 kinase-serine/threonine kinase (Pi3k/Akt) signaling via the assay for transposase-accessible chromatin using sequencing in combination with RNA sequencing. From these results we propose a novel molecular mechanism of Nono to influence cardiomyocytes differentiation and proliferation during the development of embryonic heart. We conclude that NONO may represent an emerging possible biomarkers and targets for the diagnosis and treatment of human cardiac development defects.
Collapse
Affiliation(s)
- Yu-Qing Lei
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350011, China
| | - Zhou-Jie Ye
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China
| | - Ya-Lan Wei
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China
| | - Li-Ping Zhu
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China
| | - Xu-Dong Zhuang
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China
| | - Xin-Rui Wang
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China.
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China.
| | - Hua Cao
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China.
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China.
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350011, China.
| |
Collapse
|
15
|
Hao L, Ma J, Wu F, Ma X, Qian M, Sheng W, Yan T, Tang N, Jiang X, Zhang B, Xiao D, Qian Y, Zhang J, Jiang N, Zhou W, Chen W, Ma D, Huang G. WDR62 variants contribute to congenital heart disease by inhibiting cardiomyocyte proliferation. Clin Transl Med 2022; 12:e941. [PMID: 35808830 PMCID: PMC9270576 DOI: 10.1002/ctm2.941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 12/02/2022] Open
Abstract
Background Congenital heart disease (CHD) is the most common birth defect and has high heritability. Although some susceptibility genes have been identified, the genetic basis underlying the majority of CHD cases is still undefined. Methods A total of 1320 unrelated CHD patients were enrolled in our study. Exome‐wide association analysis between 37 tetralogy of Fallot (TOF) patients and 208 Han Chinese controls from the 1000 Genomes Project was performed to identify the novel candidate gene WD repeat‐containing protein 62 (WDR62). WDR62 variants were searched in another expanded set of 200 TOF patients by Sanger sequencing. Rescue experiments in zebrafish were conducted to observe the effects of WDR62 variants. The roles of WDR62 in heart development were examined in mouse models with Wdr62 deficiency. WDR62 variants were investigated in an additional 1083 CHD patients with similar heart phenotypes to knockout mice by multiplex PCR‐targeting sequencing. The cellular phenotypes of WDR62 deficiency and variants were tested in cardiomyocytes, and the molecular mechanisms were preliminarily explored by RNA‐seq and co‐immunoprecipitation. Results Seven WDR62 coding variants were identified in the 237 TOF patients and all were indicated to be loss of function variants. A total of 25 coding and 22 non‐coding WDR62 variants were identified in 80 (6%) of the 1320 CHD cases sequenced, with a higher proportion of WDR62 variation (8%) found in the ventricular septal defect (VSD) cohort. WDR62 deficiency resulted in a series of heart defects affecting the outflow tract and right ventricle in mouse models, including VSD as the major abnormality. Cell cycle arrest and an increased number of cells with multipolar spindles that inhibited proliferation were observed in cardiomyocytes with variants or knockdown of WDR62. WDR62 deficiency weakened the association between WDR62 and the cell cycle‐regulated kinase AURKA on spindle poles, reduced the phosphorylation of AURKA, and decreased expression of target genes related to cell cycle and spindle assembly shared by WDR62 and AURKA. Conclusions WDR62 was identified as a novel susceptibility gene for CHD with high variant frequency. WDR62 was shown to participate in the cardiac development by affecting spindle assembly and cell cycle pathway in cardiomyocytes.
Collapse
Affiliation(s)
- Lili Hao
- Shanghai Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, China.,Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jing Ma
- ENT institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Feizhen Wu
- Laboratory of Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiaojing Ma
- Shanghai Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, China
| | - Maoxiang Qian
- Shanghai Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, China
| | - Wei Sheng
- Shanghai Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, China
| | - Tizhen Yan
- Department of Medical Genetics, Department of Clinical Laboratory, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Ning Tang
- Department of Medical Genetics, Department of Clinical Laboratory, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Xin Jiang
- Medical Laboratory of Nantong ZhongKe, Nantong, Jiangsu
| | - Bowen Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Deyong Xiao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yanyan Qian
- Shanghai Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, China
| | - Jin Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Nan Jiang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wenhao Zhou
- Shanghai Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, China
| | - Weicheng Chen
- Shanghai Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, China
| | - Duan Ma
- Shanghai Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, China.,Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Guoying Huang
- Shanghai Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
16
|
Klaassen S, Kühnisch J, Schultze-Berndt A, Seidel F. Left Ventricular Noncompaction in Children: The Role of Genetics, Morphology, and Function for Outcome. J Cardiovasc Dev Dis 2022; 9:jcdd9070206. [PMID: 35877568 PMCID: PMC9320003 DOI: 10.3390/jcdd9070206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/05/2023] Open
Abstract
Left ventricular noncompaction (LVNC) is a ventricular wall anomaly morphologically characterized by numerous, excessively prominent trabeculations and deep intertrabecular recesses. Accumulating data now suggest that LVNC is a distinct phenotype but must not constitute a pathological phenotype. Some individuals fulfill the morphologic criteria of LVNC and are without clinical manifestations. Most importantly, morphologic criteria for LVNC are insufficient to diagnose patients with an associated cardiomyopathy (CMP). Genetic testing has become relevant to establish a diagnosis associated with CMP, congenital heart disease, neuromuscular disease, inborn error of metabolism, or syndromic disorder. Genetic factors play a more decisive role in children than in adults and severe courses of LVNC tend to occur in childhood. We reviewed the current literature and highlight the difficulties in establishing the correct diagnosis for children with LVNC. Novel insights show that the interplay of genetics, morphology, and function determine the outcome in pediatric LVNC.
Collapse
Affiliation(s)
- Sabine Klaassen
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany; (J.K.); (A.S.-B.); (F.S.)
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, 13125 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Department of Paediatric Cardiology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- Correspondence: ; Tel.: +49-30-9406-3319; Fax: +49-30-9406-3358
| | - Jirko Kühnisch
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany; (J.K.); (A.S.-B.); (F.S.)
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, 13125 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Alina Schultze-Berndt
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany; (J.K.); (A.S.-B.); (F.S.)
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, 13125 Berlin, Germany
- Department of Paediatric Cardiology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Franziska Seidel
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany; (J.K.); (A.S.-B.); (F.S.)
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, 13125 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Department of Paediatric Cardiology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- Department of Congenital Heart Disease-Paediatric Cardiology, German Heart Institute Berlin, 13353 Berlin, Germany
| |
Collapse
|
17
|
Qu X, Harmelink C, Baldwin HS. Endocardial-Myocardial Interactions During Early Cardiac Differentiation and Trabeculation. Front Cardiovasc Med 2022; 9:857581. [PMID: 35600483 PMCID: PMC9116504 DOI: 10.3389/fcvm.2022.857581] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/24/2022] [Indexed: 01/27/2023] Open
Abstract
Throughout the continuum of heart formation, myocardial growth and differentiation occurs in concert with the development of a specialized population of endothelial cells lining the cardiac lumen, the endocardium. Once the endocardial cells are specified, they are in close juxtaposition to the cardiomyocytes, which facilitates communication between the two cell types that has been proven to be critical for both early cardiac development and later myocardial function. Endocardial cues orchestrate cardiomyocyte proliferation, survival, and organization. Additionally, the endocardium enables oxygenated blood to reach the cardiomyocytes. Cardiomyocytes, in turn, secrete factors that promote endocardial growth and function. As misregulation of this delicate and complex endocardial-myocardial interplay can result in congenital heart defects, further delineation of underlying genetic and molecular factors involved in cardiac paracrine signaling will be vital in the development of therapies to promote cardiac homeostasis and regeneration. Herein, we highlight the latest research that has advanced the elucidation of endocardial-myocardial interactions in early cardiac morphogenesis, including endocardial and myocardial crosstalk necessary for cellular differentiation and tissue remodeling during trabeculation, as well as signaling critical for endocardial growth during trabeculation.
Collapse
Affiliation(s)
- Xianghu Qu
- Department of Pediatrics (Cardiology), Vanderbilt University Medical Center, Nashville, TN, United States
| | - Cristina Harmelink
- Department of Pediatrics (Cardiology), Vanderbilt University Medical Center, Nashville, TN, United States
| | - H. Scott Baldwin
- Department of Pediatrics (Cardiology), Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Cell and Development Biology, Vanderbilt University, Nashville, TN, United States
- *Correspondence: H. Scott Baldwin
| |
Collapse
|
18
|
Gan P, Wang Z, Morales MG, Zhang Y, Bassel-Duby R, Liu N, Olson EN. RBPMS is an RNA-binding protein that mediates cardiomyocyte binucleation and cardiovascular development. Dev Cell 2022; 57:959-973.e7. [PMID: 35472321 PMCID: PMC9116735 DOI: 10.1016/j.devcel.2022.03.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/04/2022] [Accepted: 03/25/2022] [Indexed: 12/13/2022]
Abstract
Noncompaction cardiomyopathy is a common congenital cardiac disorder associated with abnormal ventricular cardiomyocyte trabeculation and impaired pump function. The genetic basis and underlying mechanisms of this disorder remain elusive. We show that the genetic deletion of RNA-binding protein with multiple splicing (Rbpms), an uncharacterized RNA-binding factor, causes perinatal lethality in mice due to congenital cardiovascular defects. The loss of Rbpms causes premature onset of cardiomyocyte binucleation and cell cycle arrest during development. Human iPSC-derived cardiomyocytes with RBPMS gene deletion have a similar blockade to cytokinesis. Sequencing analysis revealed that RBPMS plays a role in RNA splicing and influences RNAs involved in cytoskeletal signaling pathways. We found that RBPMS mediates the isoform switching of the heart-enriched LIM domain protein Pdlim5. The loss of Rbpms leads to an abnormal accumulation of Pdlim5-short isoforms, disrupting cardiomyocyte cytokinesis. Our findings connect premature cardiomyocyte binucleation to noncompaction cardiomyopathy and highlight the role of RBPMS in this process.
Collapse
Affiliation(s)
- Peiheng Gan
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhaoning Wang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Maria Gabriela Morales
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yu Zhang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ning Liu
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
19
|
Yang YY, Liu CT, Pai LF, Hu CF, Chen SJ, Hsu WF. Case Report: First Case of Non-restrictive Ventricular Septal Defect With Congestive Heart Failure in a Chinese Han Male Infant Carrying a Class II Chromosome 17p13.3 Microduplication. Front Pediatr 2022; 10:825298. [PMID: 35311053 PMCID: PMC8926061 DOI: 10.3389/fped.2022.825298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Chromosome 17p13.3 microduplication syndrome is considered a multisystem disorder that results in a wide variety of clinical manifestations including dysmorphic facial characteristics, brain structural malformations, developmental restriction, growth restriction, and neurocognitive disorders. The two major classes of chromosome 17p13.3 microduplication, which have different clinical presentations, are associated with specific genetic regions. Among the various known phenotypes, scattered cases with congenital heart disease (CHD) have been reported for both classes of chromosome 17p13.3 microduplication syndrome. Unfortunately, there is insufficient understanding of the correlation between chromosome anomaly induced alterations in gene expression and aberrant cardiac development, and thus early diagnosis of CHD among patients with chromosome 17p13.3 microduplication is difficult without routine prenatal cardiac assessment. One such congenital heart anomalies known to affect a substantial number of newborns worldwide is ventricular septal defect (VSD), which has been found in 17p13.3 microduplication carriers, and seems to sometimes undergo spontaneous closure. We report an unprecedented case of moderate sized perimembranous-outlet VSD and congestive heart failure (CHF) in a Chinese Han male infant with a class II chromosome 17p13.3 microduplication. Despite the fact that cytogenic testing and fetal echocardiography confirmed a 249-Kb chromosome duplication within 17p13.3 that encompassed the PAFAH1B1 gene and showed the presence of VSD during prenatal period, this patient still developed a range of symptoms including sustained prolonged feeding, dyspnea, diaphoresis and retarded growth. A physical examination indicated hepatomegaly and a grade III/VI pan-systolic murmur along the left upper sternal border. Laboratory testing showed a high serum pro-B-type natriuretic peptide (pro-BNP). Imaging studies revealed cardiomegaly and a persistent VSD with related pulmonary stenosis. Since the clinical findings were compatible with CHF, we provided mainline treatment with digoxin, captopril, and furosemide, as well as fluid restriction. Despite sustained poor weight gain, the feeding behavior and the respiratory conditions of the patient improved gradually. This case report and literature review suggest that patients carrying chromosome 17p13.3 microduplication who have VSD may have an increased risk of developing CHF as young infants and hence a comprehensive cardiac evaluation is warranted to allow the early diagnosis and management of any severe heart anomalies.
Collapse
Affiliation(s)
- Yung-Yu Yang
- Department of General Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Ting Liu
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Li-Fan Pai
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Fen Hu
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shyi-Jou Chen
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Wan-Fu Hsu
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
20
|
Zhang X, Wang Z, Xu Q, Chen Y, Liu W, Zhong T, Li H, Quan C, Zhang L, Cui CP. Splicing factor Srsf5 deletion disrupts alternative splicing and causes noncompaction of ventricular myocardium. iScience 2021; 24:103097. [PMID: 34622152 PMCID: PMC8482499 DOI: 10.1016/j.isci.2021.103097] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/17/2021] [Accepted: 09/06/2021] [Indexed: 11/21/2022] Open
Abstract
The serine/arginine-rich (SR) family of splicing factors plays important roles in mRNA splicing activation, repression, export, stabilization, and translation. SR-splicing factor 5 (SRSF5) is a glucose-inducible protein that promotes tumor cell growth. However, the functional role of SRSF5 in tissue development and disease remains unknown. Here, Srsf5 knockout (Srsf5−/−) mice were generated using CRISPR-Cas9. Mutant mice were perinatally lethal and exhibited cardiac dysfunction with noncompaction of the ventricular myocardium. The left ventricular internal diameter and volume were increased in Srsf5−/− mice during systole. Null mice had abnormal electrocardiogram patterns, indicative of a light atrioventricular block. Mechanistically, Srsf5 promoted the alternative splicing of Myom1 (myomesin-1), a protein that crosslinks myosin filaments to the sarcomeric M-line. The switch between embryonic and adult isoforms of Myom1 could not be completed in Srsf5-deficient heart. These findings indicate that Srsf5-regulated alternative splicing plays a critical role during heart development. Systemic loss of Srsf5 causes perinatal lethality in mice Srsf5 deficiency leads to cardiac dysfunction Alternative splicing of Myom1 in the heart around birth is regulated by Srsf5
Collapse
Affiliation(s)
- Xiaoli Zhang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, China.,State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 27 Taiping Road, Beijing 100850, China
| | - Ze Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 27 Taiping Road, Beijing 100850, China
| | - Qing Xu
- Core Facilities Centre, Capital Medical University, Beijing 100069, China
| | - Yuhan Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 27 Taiping Road, Beijing 100850, China
| | - Wen Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 27 Taiping Road, Beijing 100850, China
| | - Tong Zhong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 27 Taiping Road, Beijing 100850, China
| | - Hongchang Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 27 Taiping Road, Beijing 100850, China
| | - Chengshi Quan
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 27 Taiping Road, Beijing 100850, China
| | - Chun-Ping Cui
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 27 Taiping Road, Beijing 100850, China
| |
Collapse
|
21
|
Jorgensen R, Katta M, Wolfe J, Leach DF, Lavelle B, Chun J, Wilsbacher LD. Deletion of Sphingosine 1-Phosphate receptor 1 in cardiomyocytes during development leads to abnormal ventricular conduction and fibrosis. Physiol Rep 2021; 9:e15060. [PMID: 34618403 PMCID: PMC8496155 DOI: 10.14814/phy2.15060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 11/24/2022] Open
Abstract
Sphingosine 1-Phosphate receptor 1 (S1P1 , encoded by S1pr1) is a G protein-coupled receptor that signals in multiple cell types including endothelial cells and cardiomyocytes. Cardiomyocyte-specific deletion of S1pr1 during mouse development leads to ventricular noncompaction, with 44% of mutant mice surviving to adulthood. Adult survivors of embryonic cardiomyocyte S1pr1 deletion showed cardiac hypertrabeculation consistent with ventricular noncompaction. Surprisingly, systolic function in mutant mice was preserved through at least 1 year of age. Cardiac conduction was abnormal in cardiomyocyte S1pr1 mutant mice, with prolonged QRS intervals in mutants as compared with littermate control mice. Immunostaining of hearts from S1pr1 mutant embryos displayed a zone of intermediate Connexin 40 (Cx40) expression in the trabecular myocardium. However, we observed no significant differences in Cx40 and Connexin 43 immunostaining in hearts from adult survivors of embryonic cardiomyocyte S1pr1 deletion, which suggests normalized development of the ventricular conduction system in mutant mice. By contrast, the adult survivors of embryonic cardiomyocyte S1pr1 deletion showed increased cardiac fibrosis as compared with littermate controls. These results demonstrate that ventricular hypertrabeculation caused by embryonic deletion of cardiomyocyte S1pr1 correlates with cardiac fibrosis, which contributes to abnormal ventricular conduction. These results also reveal conduction abnormalities in the setting of hypertrabeculation with normal systolic function, which may be of clinical relevance in humans with ventricular hypertrabeculation.
Collapse
Affiliation(s)
- Ryan Jorgensen
- Feinberg Cardiovascular and Renal Research InstituteNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Meghna Katta
- Feinberg Cardiovascular and Renal Research InstituteNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Jayne Wolfe
- Feinberg Cardiovascular and Renal Research InstituteNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Desiree F. Leach
- Feinberg Cardiovascular and Renal Research InstituteNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Bianca Lavelle
- Feinberg Cardiovascular and Renal Research InstituteNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery InstituteLa JollaCaliforniaUSA
| | - Lisa D. Wilsbacher
- Feinberg Cardiovascular and Renal Research InstituteNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of PharmacologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| |
Collapse
|
22
|
Riekerk HCE, Coolen BF, J Strijkers G, van der Wal AC, Petersen SE, Sheppard MN, Oostra RJ, Christoffels VM, Jensen B. Higher spatial resolution improves the interpretation of the extent of ventricular trabeculation. J Anat 2021; 240:357-375. [PMID: 34569075 PMCID: PMC8742974 DOI: 10.1111/joa.13559] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/07/2021] [Accepted: 09/15/2021] [Indexed: 12/21/2022] Open
Abstract
The ventricular walls of the human heart comprise an outer compact layer and an inner trabecular layer. In the context of an increased pre-test probability, diagnosis left ventricular noncompaction cardiomyopathy is given when the left ventricle is excessively trabeculated in volume (trabecular vol >25% of total LV wall volume) or thickness (trabecular/compact (T/C) >2.3). Here, we investigated whether higher spatial resolution affects the detection of trabeculation and thus the assessment of normal and excessively trabeculated wall morphology. First, we screened left ventricles in 1112 post-natal autopsy hearts. We identified five excessively trabeculated hearts and this low prevalence of excessive trabeculation is in agreement with pathology reports but contrasts the prevalence of approximately 10% of the population found by in vivo non-invasive imaging. Using macroscopy, histology and low- and high-resolution MRI, the five excessively trabeculated hearts were compared with six normal hearts and seven abnormally trabeculated and excessive trabeculation-negative hearts. Some abnormally trabeculated hearts could be considered excessively trabeculated macroscopically because of a trabecular outflow or an excessive number of trabeculations, but they were excessive trabeculation-negative when assessed with MRI-based measurements (T/C <2.3 and vol <25%). The number of detected trabeculations and T/C ratio were positively correlated with higher spatial resolution. Using measurements on high resolution MRI and with histological validation, we could not replicate the correlation between trabeculations of the left and right ventricle that has been previously reported. In conclusion, higher spatial resolution may affect the sensitivity of diagnostic measurements and in addition could allow for novel measurements such as counting of trabeculations.
Collapse
Affiliation(s)
- Hanne C E Riekerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Bram F Coolen
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Gustav J Strijkers
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Allard C van der Wal
- Department of Pathology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Steffen E Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, UK.,Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Mary N Sheppard
- Department of Cardiovascular Pathology, Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Roelof-Jan Oostra
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Bjarke Jensen
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Una variante rara en HCN4 produce un fenotipo de hipertrabeculación/no compactación ventricular, dilatación auricular izquierda y bradicardia sinusal. Rev Esp Cardiol 2021. [DOI: 10.1016/j.recesp.2020.06.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
24
|
Hofbauer P, Jahnel SM, Papai N, Giesshammer M, Deyett A, Schmidt C, Penc M, Tavernini K, Grdseloff N, Meledeth C, Ginistrelli LC, Ctortecka C, Šalic Š, Novatchkova M, Mendjan S. Cardioids reveal self-organizing principles of human cardiogenesis. Cell 2021; 184:3299-3317.e22. [PMID: 34019794 DOI: 10.1016/j.cell.2021.04.034] [Citation(s) in RCA: 280] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 02/12/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022]
Abstract
Organoids capable of forming tissue-like structures have transformed our ability to model human development and disease. With the notable exception of the human heart, lineage-specific self-organizing organoids have been reported for all major organs. Here, we established self-organizing cardioids from human pluripotent stem cells that intrinsically specify, pattern, and morph into chamber-like structures containing a cavity. Cardioid complexity can be controlled by signaling that instructs the separation of cardiomyocyte and endothelial layers and by directing epicardial spreading, inward migration, and differentiation. We find that cavity morphogenesis is governed by a mesodermal WNT-BMP signaling axis and requires its target HAND1, a transcription factor linked to developmental heart chamber defects. Upon cryoinjury, cardioids initiated a cell-type-dependent accumulation of extracellular matrix, an early hallmark of both regeneration and heart disease. Thus, human cardioids represent a powerful platform to mechanistically dissect self-organization, congenital heart defects and serve as a foundation for future translational research.
Collapse
Affiliation(s)
- Pablo Hofbauer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Stefan M Jahnel
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Nora Papai
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Magdalena Giesshammer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Alison Deyett
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Clara Schmidt
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Mirjam Penc
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Katherina Tavernini
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Nastasja Grdseloff
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Christy Meledeth
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Lavinia Ceci Ginistrelli
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Claudia Ctortecka
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Šejla Šalic
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Maria Novatchkova
- Institute of Molecular Pathology (IMP), Vienna Biocenter 1, 1030 Vienna, Austria
| | - Sasha Mendjan
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria.
| |
Collapse
|
25
|
Khosravi F, Ahmadvand N, Bellusci S, Sauer H. The Multifunctional Contribution of FGF Signaling to Cardiac Development, Homeostasis, Disease and Repair. Front Cell Dev Biol 2021; 9:672935. [PMID: 34095143 PMCID: PMC8169986 DOI: 10.3389/fcell.2021.672935] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
The current focus on cardiovascular research reflects society’s concerns regarding the alarming incidence of cardiac-related diseases and mortality in the industrialized world and, notably, an urgent need to combat them by more efficient therapies. To pursue these therapeutic approaches, a comprehensive understanding of the mechanism of action for multifunctional fibroblast growth factor (FGF) signaling in the biology of the heart is a matter of high importance. The roles of FGFs in heart development range from outflow tract formation to the proliferation of cardiomyocytes and the formation of heart chambers. In the context of cardiac regeneration, FGFs 1, 2, 9, 16, 19, and 21 mediate adaptive responses including restoration of cardiac contracting rate after myocardial infarction and reduction of myocardial infarct size. However, cardiac complications in human diseases are correlated with pathogenic effects of FGF ligands and/or FGF signaling impairment. FGFs 2 and 23 are involved in maladaptive responses such as cardiac hypertrophic, fibrotic responses and heart failure. Among FGFs with known causative (FGFs 2, 21, and 23) or protective (FGFs 2, 15/19, 16, and 21) roles in cardiac diseases, FGFs 15/19, 21, and 23 display diagnostic potential. The effective role of FGFs on the induction of progenitor stem cells to cardiac cells during development has been employed to boost the limited capacity of postnatal cardiac repair. To renew or replenish damaged cardiomyocytes, FGFs 1, 2, 10, and 16 were tested in (induced-) pluripotent stem cell-based approaches and for stimulation of cell cycle re-entry in adult cardiomyocytes. This review will shed light on the wide range of beneficiary and detrimental actions mediated by FGF ligands and their receptors in the heart, which may open new therapeutic avenues for ameliorating cardiac complications.
Collapse
Affiliation(s)
- Farhad Khosravi
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Negah Ahmadvand
- Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Saverio Bellusci
- Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
26
|
Affiliation(s)
- Andrew D'Silva
- Department of Cardiology, Guy's and St Thomas' NHS Foundation Trust, London, UK .,School of Cardiovascular Medicine & Sciences, King's College London, London, UK
| | - Bjarke Jensen
- Department of Medical Biology, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Alonso-Fernández-Gatta M, Gallego-Delgado M, Caballero R, Villacorta E, Díaz-Peláez E, García-BerrocaL B, Crespo-García T, Plata-Izquierdo B, Marcos-Vadillo E, García-Cuenllas L, Barreiro-Pérez M, Isidoro-García M, Tamargo-Menéndez J, Delpón E, Sánchez PL. A rare HCN4 variant with combined sinus bradycardia, left atrial dilatation, and hypertrabeculation/left ventricular noncompaction phenotype. ACTA ACUST UNITED AC 2020; 74:781-789. [PMID: 33008772 DOI: 10.1016/j.rec.2020.06.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/24/2020] [Indexed: 01/20/2023]
Abstract
INTRODUCTION AND OBJECTIVES HCN4 variants have been reported to cause combined sick sinus syndrome (SSS) and left ventricular noncompaction (LVNC) cardiomyopathy. This relationship has been proven in few cases and no previous patients have associated left atrial dilatation (LAD). Our objective was to study a familial disorder characterized by SSS, LAD, and hypertrabeculation/LVNC and to identify the underlying genetic and electrophysiological characteristics. METHODS A family with SSS and LVNC underwent a clinical, genetic, and electrophysiological assessment. They were studied via electrocardiography, Holter recording, echocardiography, and exercise stress tests; cardiac magnetic resonance imaging was additionally performed in affected individuals. Genetic testing was undertaken with targeted next-generation sequencing, as well as a functional study of the candidate variant in Chinese hamster ovary cells. RESULTS Twelve members of the family had sinus bradycardia, associated with complete criteria of LVNC in 4 members and hypertrabeculation in 6 others, as well as LAD in 9 members. A HCN4 c.1123C>T;(p.R375C) variant was present in heterozygosis in all affected patients and absent in unaffected individuals. Electrophysiological analyses showed that the amplitude and densities of the HCN4 currents (IHCN4) generated by mutant p.R375C HCN4 channels were significantly lower than those generated by wild-type channels. CONCLUSIONS The combined phenotype of SSS, LAD, and LVNC is associated with the heritable HCN4 c.1123C>T;(p.R375C) variant. HCN4 variants should be included in the genetic diagnosis of LVNC cardiomyopathy and of patients with familial forms of SSS, as well as of individuals with sinus bradycardia and LAD.
Collapse
Affiliation(s)
- Marta Alonso-Fernández-Gatta
- Servicio de Cardiología, Complejo Asistencial Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - María Gallego-Delgado
- Servicio de Cardiología, Complejo Asistencial Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Centro de Referencia Nacional de Cardiopatías Familiares (CSUR), Salamanca, Spain
| | - Ricardo Caballero
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Eduardo Villacorta
- Servicio de Cardiología, Complejo Asistencial Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Centro de Referencia Nacional de Cardiopatías Familiares (CSUR), Salamanca, Spain.
| | - Elena Díaz-Peláez
- Servicio de Cardiología, Complejo Asistencial Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Centro de Referencia Nacional de Cardiopatías Familiares (CSUR), Salamanca, Spain
| | - Belén García-BerrocaL
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Centro de Referencia Nacional de Cardiopatías Familiares (CSUR), Salamanca, Spain; Unidad de Genética, Servicio de Bioquímica, Complejo Asistencial Universitario de Salamanca. Universidad de Salamanca, Salamanca, Spain
| | - Teresa Crespo-García
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Beatriz Plata-Izquierdo
- Centro de Referencia Nacional de Cardiopatías Familiares (CSUR), Salamanca, Spain; Servicio de Pediatría, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | - Elena Marcos-Vadillo
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Centro de Referencia Nacional de Cardiopatías Familiares (CSUR), Salamanca, Spain; Unidad de Genética, Servicio de Bioquímica, Complejo Asistencial Universitario de Salamanca. Universidad de Salamanca, Salamanca, Spain
| | - Luisa García-Cuenllas
- Centro de Referencia Nacional de Cardiopatías Familiares (CSUR), Salamanca, Spain; Servicio de Pediatría, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | - Manuel Barreiro-Pérez
- Servicio de Cardiología, Complejo Asistencial Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - María Isidoro-García
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Centro de Referencia Nacional de Cardiopatías Familiares (CSUR), Salamanca, Spain; Unidad de Genética, Servicio de Bioquímica, Complejo Asistencial Universitario de Salamanca. Universidad de Salamanca, Salamanca, Spain
| | - Juan Tamargo-Menéndez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Eva Delpón
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Pedro L Sánchez
- Servicio de Cardiología, Complejo Asistencial Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Centro de Referencia Nacional de Cardiopatías Familiares (CSUR), Salamanca, Spain
| |
Collapse
|
28
|
Mandrycky CJ, Williams NP, Batalov I, El-Nachef D, de Bakker BS, Davis J, Kim DH, DeForest CA, Zheng Y, Stevens KR, Sniadecki NJ. Engineering Heart Morphogenesis. Trends Biotechnol 2020; 38:835-845. [PMID: 32673587 DOI: 10.1016/j.tibtech.2020.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/22/2022]
Abstract
Recent advances in stem cell biology and tissue engineering have laid the groundwork for building complex tissues in a dish. We propose that these technologies are ready for a new challenge: recapitulating cardiac morphogenesis in vitro. In development, the heart transforms from a simple linear tube to a four-chambered organ through a complex process called looping. Here, we re-examine heart tube looping through the lens of an engineer and argue that the linear heart tube is an advantageous starting point for tissue engineering. We summarize the structures, signaling pathways, and stresses in the looping heart, and evaluate approaches that could be used to build a linear heart tube and guide it through the process of looping.
Collapse
Affiliation(s)
- Christian J Mandrycky
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Nisa P Williams
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Ivan Batalov
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Danny El-Nachef
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA
| | - Bernadette S de Bakker
- Clinical Anatomy and Embryology, Department of Medical Biology, AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jennifer Davis
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA
| | - Deok-Ho Kim
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Medicine/Cardiology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Cole A DeForest
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA; Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Ying Zheng
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Kelly R Stevens
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA
| | - Nathan J Sniadecki
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA; Department of Mechanical Engineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
29
|
Li C, Liu F, Liu S, Pan H, Du H, Huang J, Xie Y, Li Y, Zhao R, Wei Y. Elevated myocardial SORBS2 and the underlying implications in left ventricular noncompaction cardiomyopathy. EBioMedicine 2020; 53:102695. [PMID: 32143182 PMCID: PMC7058526 DOI: 10.1016/j.ebiom.2020.102695] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 01/19/2023] Open
Abstract
Background Left ventricular noncompaction cardiomyopathy (LVNC) is a hereditary heart disease characterized by an excessive trabecular meshwork of deep intertrabecular recesses within the ventricular myocardium. The guidelines for management of LVNC patients aim to improve quality of life by preventing cardiac heart failure. However, the mechanism underlying LVNC-associated heart failure remains poorly understood. Methods Using protein mass spectrometry analysis, we established that Sorbin And SH3 Domain Containing 2 (SORBS2) is up-regulated in LVNC hearts without changes to structure proteins. We conducted in vivo experiments wherein the heart tissues of wild-type mice were injected with an AAV9 vector to overexpress SORBS2, followed by analysis using echocardiography, T-tubule analysis and Ca2+ imaging to identify functional and morphological changes. In addition, we analyzed the function and structure of SORBS2 overexpressing human embryonic stem cell (hESC) derived cardiomyocytes (hESC-CM) via immunoblotting, immunohistochemistry, immunofluorescence, and confocal Ca2+ imaging. Findings LVNC myocardial tissues feature strongly elevated expression of SORBS2, microtubule densification and redistribution of Junctophilin 2 (JP2). SORBS2 interacts with β-tubulin, promoting its polymerization in 293T cells and hESC-derived CMs. In vivo, cardiac dysfunction, β-tubulin densification, JP2 translocation, T-tubule disorganization and Ca2+ handling dysfunction were observed in mice overexpressing SORBS2. Interpretation We identified a novel mechanism through which SORBS2 interacts with β-tubulin and promotes microtubule densification, eventually effecting JP2 distribution and T-tubule, potentially contributing to heart failure in LVNC disease. Fund This work was supported by a CAMS Initiative for Innovative Medicine grant (CAMS-I2M, 2016-I2M-1-015 to Y.J.Wei)
Collapse
Affiliation(s)
- Chunyan Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Fan Liu
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Shenghua Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Haizhou Pan
- Children's Heart Center, the Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haiwei Du
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Jian Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Yuanyuan Xie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Yanfen Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Ranxu Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Yingjie Wei
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China.
| |
Collapse
|
30
|
Tet inactivation disrupts YY1 binding and long-range chromatin interactions during embryonic heart development. Nat Commun 2019; 10:4297. [PMID: 31541101 PMCID: PMC6754421 DOI: 10.1038/s41467-019-12325-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 08/30/2019] [Indexed: 12/15/2022] Open
Abstract
Tet-mediated DNA demethylation plays an important role in shaping the epigenetic landscape and chromatin accessibility to control gene expression. While several studies demonstrated pivotal roles of Tet in regulating embryonic development, little is known about their functions in heart development. Here we analyze DNA methylation and hydroxymethylation dynamics during early cardiac development in both human and mice. We find that cardiac-specific deletion of Tet2 and Tet3 in mice (Tet2/3-DKO) leads to ventricular non-compaction cardiomyopathy (NCC) with embryonic lethality. Single-cell RNA-seq analyses reveal a reduction in cardiomyocyte numbers and transcriptional reprogramming in cardiac tissues upon Tet2/3 depletion. Impaired DNA demethylation and reduced chromatin accessibility in Tet2/3-DKO mice further compromised Ying-yang1 (YY1) binding to its genomic targets, and perturbed high-order chromatin organization at key genes involved in heart development. Our studies provide evidence of the physiological role of Tet in regulating DNA methylation dynamics and chromatin organization during early heart development. Tet-mediated DNA demethylation is intimately involved in reguatling embryonic development. Here the authors characterise DNA methylation and hydroxymethylation dynamics during early cardiac development in both human and mice and provide evidence that Tet-mediated DNA demethylation plays a role in regulating chromatin organization during early heart development.
Collapse
|
31
|
Yang Q, Wu F, Wang F, Cai K, Zhang Y, Sun Q, Zhao X, Gui Y, Li Q. Impact of DNA methyltransferase inhibitor 5-azacytidine on cardiac development of zebrafish in vivo and cardiomyocyte proliferation, apoptosis, and the homeostasis of gene expression in vitro. J Cell Biochem 2019; 120:17459-17471. [PMID: 31271227 DOI: 10.1002/jcb.29010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 12/15/2022]
Abstract
Cardiac development is a peculiar process involving coordinated cellular differentiation, migration, proliferation, and apoptosis. DNA methylation plays a key role in genomic stability, tissue-specific gene expression, cell proliferation, and apoptosis. Hypomethylation in the global genome has been reported in cardiovascular diseases. However, little is known about the impact and specific mechanism of global hypomethylation on cardiomyocytes. In the present study, we explored the impact of DNA methyltransferase inhibitors 5-azacytidine on cardiac development. In vivo experiment showed that hypomethylation of zebrafish embryos with 5-azacytidine exposure significantly reduced survival, induced malformations, and delayed general development process. Furthermore, zebrafish embryos injected with 5-azacytidine developed pericardial edema, ventricular volume reduction, looping deformity, and reduction in heart rate and ventricular shortening fraction. Cardiomyocytes treated with 5-azacytidine in vitro decreased proliferation and induced apoptosis in a concentration-dependent manner. Furthermore, 5-azacytidine treatment in cardiomyocytes resulted in 20 downregulated genes expression and two upregulated genes expression in 45 candidate genes, which indicated that DNA methylation functions as a bidirectional modulator in regulating gene expression. In conclusion, these results show the regulative effects of the epigenetic modifier 5-azacytidine in cardiac development of zebrafish embryos in vivo and cardiomyocyte proliferation and apoptosis and the homeostasis of gene expression in vitro, which offer a novel understanding of aberrant DNA methylation in the etiology of cardiovascular disease.
Collapse
Affiliation(s)
- Qian Yang
- Shanghai Key Laboratory of Birth Defect, Translational Medical Center for Development and Disease, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Fang Wu
- Shanghai Key Laboratory of Birth Defect, Translational Medical Center for Development and Disease, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Feng Wang
- Shanghai Key Laboratory of Birth Defect, Translational Medical Center for Development and Disease, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Ke Cai
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Yawen Zhang
- Shanghai Key Laboratory of Birth Defect, Translational Medical Center for Development and Disease, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Quanya Sun
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaolong Zhao
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yonghao Gui
- Shanghai Key Laboratory of Birth Defect, Translational Medical Center for Development and Disease, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Qiang Li
- Shanghai Key Laboratory of Birth Defect, Translational Medical Center for Development and Disease, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
32
|
Rasouli SJ, El-Brolosy M, Tsedeke AT, Bensimon-Brito A, Ghanbari P, Maischein HM, Kuenne C, Stainier DY. The flow responsive transcription factor Klf2 is required for myocardial wall integrity by modulating Fgf signaling. eLife 2018; 7:e38889. [PMID: 30592462 PMCID: PMC6329608 DOI: 10.7554/elife.38889] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 12/24/2018] [Indexed: 12/17/2022] Open
Abstract
Complex interplay between cardiac tissues is crucial for their integrity. The flow responsive transcription factor KLF2, which is expressed in the endocardium, is vital for cardiovascular development but its exact role remains to be defined. To this end, we mutated both klf2 paralogues in zebrafish, and while single mutants exhibit no obvious phenotype, double mutants display a novel phenotype of cardiomyocyte extrusion towards the abluminal side. This extrusion requires cardiac contractility and correlates with the mislocalization of N-cadherin from the lateral to the apical side of cardiomyocytes. Transgenic rescue data show that klf2 expression in endothelium, but not myocardium, prevents this cardiomyocyte extrusion phenotype. Transcriptome analysis of klf2 mutant hearts reveals that Fgf signaling is affected, and accordingly, we find that inhibition of Fgf signaling in wild-type animals can lead to abluminal cardiomyocyte extrusion. These studies provide new insights into how Klf2 regulates cardiovascular development and specifically myocardial wall integrity.
Collapse
Affiliation(s)
- Seyed Javad Rasouli
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Mohamed El-Brolosy
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Ayele Taddese Tsedeke
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Anabela Bensimon-Brito
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Parisa Ghanbari
- Department of Cardiac Development and RemodelingMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Hans-Martin Maischein
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Carsten Kuenne
- Bioinformatics Core UnitMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Didier Y Stainier
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| |
Collapse
|
33
|
Key Questions Relating to Left Ventricular Noncompaction Cardiomyopathy: Is the Emperor Still Wearing Any Clothes? Can J Cardiol 2017; 33:747-757. [DOI: 10.1016/j.cjca.2017.01.017] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 11/23/2022] Open
|
34
|
Arrieta A, Blackwood EA, Glembotski CC. ER Protein Quality Control and the Unfolded Protein Response in the Heart. Curr Top Microbiol Immunol 2017; 414:193-213. [PMID: 29026925 DOI: 10.1007/82_2017_54] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiac myocytes are the cells responsible for the robust ability of the heart to pump blood throughout the circulatory system. Cardiac myocytes grow in response to a variety of physiological and pathological conditions; this growth challenges endoplasmic reticulum-protein quality control (ER-PQC), a major feature of which includes the unfolded protein response (UPR). ER-PQC and the UPR in cardiac myocytes growing under physiological conditions, including normal development, exercise, and pregnancy, are sufficient to support hypertrophic growth of each cardiac myocyte. However, the ER-PQC and UPR are insufficient to respond to the challenge of cardiac myocyte growth under pathological conditions, including myocardial infarction and heart failure. In part, this insufficiency is due to a continual decline in the expression levels of important adaptive UPR components as a function of age and during myocardial pathology. This chapter will discuss the physiological and pathological conditions unique to the heart that involves ER-PQC, and whether the UPR is adaptive or maladaptive under these circumstances.
Collapse
Affiliation(s)
- A Arrieta
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, CA, 92182, USA
| | - E A Blackwood
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, CA, 92182, USA
| | - C C Glembotski
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, CA, 92182, USA.
| |
Collapse
|
35
|
Itoh N, Ohta H, Nakayama Y, Konishi M. Roles of FGF Signals in Heart Development, Health, and Disease. Front Cell Dev Biol 2016; 4:110. [PMID: 27803896 PMCID: PMC5067508 DOI: 10.3389/fcell.2016.00110] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/20/2016] [Indexed: 01/13/2023] Open
Abstract
The heart provides the body with oxygen and nutrients and assists in the removal of metabolic waste through the blood vessels of the circulatory system. It is the first organ to form during embryonic morphogenesis. FGFs with diverse functions in development, health, and disease are signaling proteins, mostly as paracrine growth factors or endocrine hormones. The human/mouse FGF family comprises 22 members. Findings obtained from mouse models and human diseases with FGF signaling disorders have indicated that several FGFs are involved in heart development, health, and disease. Paracrine FGFs including FGF8, FGF9, FGF10, and FGF16 act as paracrine signals in embryonic heart development. In addition, paracrine FGFs including FGF2, FGF9, FGF10, and FGF16 play roles as paracrine signals in postnatal heart pathophysiology. Although FGF15/19, FGF21, and FGF23 are typical endocrine FGFs, they mainly function as paracrine signals in heart development or pathophysiology. In heart diseases, serum FGF15/19 levels or FGF21 and FGF23 levels decrease or increase, respectively, indicating their possible roles in heart pathophysiology. FGF2 and FGF10 also stimulate the cardiac differentiation of cultured stem cells and cardiac reprogramming of cultured fibroblasts. These findings provide new insights into the roles of FGF signaling in the heart and potential therapeutic strategies for cardiac disorders.
Collapse
Affiliation(s)
- Nobuyuki Itoh
- Medical Innovation Center, Kyoto University Graduate School of Medicine Kyoto, Japan
| | - Hiroya Ohta
- Department of Microbial Chemistry, Kobe Pharmaceutical University Kobe, Japan
| | - Yoshiaki Nakayama
- Department of Microbial Chemistry, Kobe Pharmaceutical University Kobe, Japan
| | - Morichika Konishi
- Department of Microbial Chemistry, Kobe Pharmaceutical University Kobe, Japan
| |
Collapse
|
36
|
Sphingosine 1-phosphate receptor-1 in cardiomyocytes is required for normal cardiac development. Dev Biol 2016; 418:157-165. [PMID: 27333774 DOI: 10.1016/j.ydbio.2016.06.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 06/14/2016] [Accepted: 06/16/2016] [Indexed: 01/27/2023]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid that acts via G protein-coupled receptors. The S1P receptor S1P1, encoded by S1pr1, is expressed in developing heart but its roles there remain largely unexplored. Analysis of S1pr1 LacZ knockin embryos revealed β-galactosidase staining in cardiomyocytes in the septum and in the trabecular layer of hearts collected at 12.5 days post coitus (dpc) and weak staining in the inner aspect of the compact layer at 15.5 dpc and later. Nkx2-5-Cre- and Mlc2a-Cre-mediated conditional knockout of S1pr1 led to ventricular noncompaction and ventricular septal defects at 18.5 dpc and to perinatal lethality in the majority of mutants. Further analysis of Mlc2a-Cre conditional mutants revealed no gross phenotype at 12.5 dpc but absence of the normal increase in the number of cardiomyocytes and the thickness of the compact layer at 13.5 dpc and after. Consistent with relative lack of a compact layer, in situ hybridization at 13.5 dpc revealed expression of trabecular markers extending almost to the epicardium in mutants. Mutant hearts also showed decreased myofibril organization in the compact but not trabecular myocardium at 12.5 dpc. These results suggest that S1P signaling via S1P1 in cardiomyocytes plays a previously unknown and necessary role in heart development in mice.
Collapse
|