1
|
Liu Z, Liu Y, Yu Z, Tan C, Pek N, O'Donnell A, Wu A, Glass I, Winlaw DS, Guo M, Spence JR, Chen YW, Yutzey KE, Miao Y, Gu M. APOE-NOTCH axis governs elastogenesis during human cardiac valve remodeling. NATURE CARDIOVASCULAR RESEARCH 2024; 3:933-950. [PMID: 39196035 DOI: 10.1038/s44161-024-00510-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/19/2024] [Indexed: 08/29/2024]
Abstract
Valve remodeling is a process involving extracellular matrix organization and elongation of valve leaflets. Here, through single-cell RNA sequencing of human fetal valves, we identified an elastin-producing valve interstitial cell (VIC) subtype (apolipoprotein E (APOE)+, elastin-VICs) spatially located underneath valve endothelial cells (VECs) sensing unidirectional flow. APOE knockdown in fetal VICs resulted in profound elastogenesis defects. In valves with pulmonary stenosis (PS), we observed elastin fragmentation and decreased expression of APOE along with other genes regulating elastogenesis. Cell-cell interaction analysis revealed that jagged 1 (JAG1) from unidirectional VECs activates elastogenesis in elastin-VICs through NOTCH2. Similar observations were made in VICs cocultured with VECs under unidirectional flow. Notably, a drastic reduction of JAG1-NOTCH2 was also observed in PS valves. Lastly, we found that APOE controls JAG1-induced NOTCH activation and elastogenesis in VICs through the extracellular signal-regulated kinase pathway. Our study suggests important roles of both APOE and NOTCH in regulating elastogenesis during human valve remodeling.
Collapse
Affiliation(s)
- Ziyi Liu
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Yu Liu
- Cardiovascular Institute, Stanford School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Zhiyun Yu
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Cheng Tan
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Nicole Pek
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Anna O'Donnell
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Angeline Wu
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ian Glass
- Department of Pediatrics, Genetic Medicine, University of Washington, Seattle, WA, USA
| | - David S Winlaw
- Cardiothoracic Surgery, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Surgery, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Minzhe Guo
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Jason R Spence
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, USA
| | - Ya-Wen Chen
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute for Airway Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine E Yutzey
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yifei Miao
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA.
- Cardiovascular Institute, Stanford School of Medicine, Stanford, CA, USA.
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA.
| | - Mingxia Gu
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA.
- Cardiovascular Institute, Stanford School of Medicine, Stanford, CA, USA.
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
2
|
Zhang Q, Meng H, Wang X, Chen Y, Yan Z, Ruan J, Meng F. Low expression of Notch1 may be associated with acute myocardial infarction. Front Cardiovasc Med 2024; 11:1367675. [PMID: 38841263 PMCID: PMC11150703 DOI: 10.3389/fcvm.2024.1367675] [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: 02/11/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024] Open
Abstract
Background The transmembrane protein Notch1 is associated with cell growth, development, differentiation, proliferation, apoptosis, adhesion, and the epithelial mesenchymal transition. Proteomics, as a research method, uses a series of sequencing techniques to study the composition, expression levels, and modifications of proteins. Here, the association between Notch1 and acute myocardial infarction (AMI) was investigated using proteomics, to assess the possibility of using Notch1 as a biomarker for the disease. Methods Fifty-five eligible patients with AMI and 74 with chronic coronary syndrome (CCS) were enrolled, representing the experimental and control groups, respectively. The mRNA levels were assessed using RT-qPCR and proteins were measured using ELISA, and the results were compared and analyzed. Results Notch1 mRNA levels were 0.52 times higher in the peripheral blood mononuclear cells of the AMI group relative to the CCS group (p < 0.05) while Notch1 protein levels were 0.63 times higher in peripheral blood plasma in AMI patients (p < 0.05). Notch1 levels were not associated with older age, hypertension, smoking, high abdominal-blood glucose, high total cholesterol, and high LDL in AMI. Logistic regression indicated associations between AMI and reduced Notch1 expression, hypertension, smoking, and high fasting glucose. Conclusions Notch1 expression was reduced in the peripheral blood of patients with AMI relative to those with CCS. The low expression of Notch1 was found to be an independent risk factor for AMI and may thus be an indicator of the disease.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| | - Heyu Meng
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| | - Xue Wang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| | - Yanqiu Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| | - Zhaohan Yan
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| | - Jianjun Ruan
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| | - Fanbo Meng
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| |
Collapse
|
3
|
Wu B, Wu B, Benkaci S, Shi L, Lu P, Park T, Morrow BE, Wang Y, Zhou B. Crk and Crkl Are Required in the Endocardial Lineage for Heart Valve Development. J Am Heart Assoc 2023; 12:e029683. [PMID: 37702066 PMCID: PMC10547300 DOI: 10.1161/jaha.123.029683] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/02/2023] [Indexed: 09/14/2023]
Abstract
Background Endocardial cells are a major progenitor population that gives rise to heart valves through endocardial cushion formation by endocardial to mesenchymal transformation and the subsequent endocardial cushion remodeling. Genetic variants that affect these developmental processes can lead to congenital heart valve defects. Crk and Crkl are ubiquitously expressed genes encoding cytoplasmic adaptors essential for cell signaling. This study aims to explore the specific role of Crk and Crkl in the endocardial lineage during heart valve development. Methods and Results We deleted Crk and Crkl specifically in the endocardial lineage. The resultant heart valve morphology was evaluated by histological analysis, and the underlying cellular and molecular mechanisms were investigated by immunostaining and quantitative reverse transcription polymerase chain reaction. We found that the targeted deletion of Crk and Crkl impeded the remodeling of endocardial cushions at the atrioventricular canal into the atrioventricular valves. We showed that apoptosis was temporally increased in the remodeling atrioventricular endocardial cushions, and this developmentally upregulated apoptosis was repressed by deletion of Crk and Crkl. Loss of Crk and Crkl also resulted in altered extracellular matrix production and organization in the remodeling atrioventricular endocardial cushions. These morphogenic defects were associated with altered expression of genes in BMP (bone morphogenetic protein), connective tissue growth factor, and WNT signaling pathways, and reduced extracellular signal-regulated kinase signaling activities. Conclusions Our findings support that Crk and Crkl have shared functions in the endocardial lineage that critically regulate atrioventricular valve development; together, they likely coordinate the morphogenic signals involved in the remodeling of the atrioventricular endocardial cushions.
Collapse
Affiliation(s)
- Bingruo Wu
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
| | - Brian Wu
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
| | - Sonia Benkaci
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
| | - Lijie Shi
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
| | - Pengfei Lu
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
| | - Taeju Park
- Children’s Mercy Research Institute, Children’s Mercy Kansas City and Department of Pediatrics, University of Missouri‐Kansas City School of MedicineKansas CityMO
| | | | - Yidong Wang
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
- Cardiovascular Research Center, School of Basic Medical SciencesXi’an Jiaotong University Health Science CenterXi’anChina
| | - Bin Zhou
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
| |
Collapse
|
4
|
Liu H, Lu P, He S, Luo Y, Fang Y, Benkaci S, Wu B, Wang Y, Zhou B. β-Catenin regulates endocardial cushion growth by suppressing p21. Life Sci Alliance 2023; 6:e202302163. [PMID: 37385754 PMCID: PMC10310929 DOI: 10.26508/lsa.202302163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023] Open
Abstract
Endocardial cushion formation is essential for heart valve development and heart chamber separation. Abnormal endocardial cushion formation often causes congenital heart defects. β-Catenin is known to be essential for endocardial cushion formation; however, the underlying cellular and molecular mechanisms remain incompletely understood. Here, we show that endothelial-specific deletion of β-catenin in mice resulted in formation of hypoplastic endocardial cushions due to reduced cell proliferation and impaired cell migration. By using a β-catenin DM allele in which the transcriptional function of β-catenin is selectively disrupted, we further reveal that β-catenin regulated cell proliferation and migration through its transcriptional and non-transcriptional function, respectively. At the molecular level, loss of β-catenin resulted in increased expression of cell cycle inhibitor p21 in cushion endocardial and mesenchymal cells in vivo. In vitro rescue experiments with HUVECs and pig aortic valve interstitial cells confirmed that β-catenin promoted cell proliferation by suppressing p21. In addition, one savvy negative observation is that β-catenin was dispensable for endocardial-to-mesenchymal fate change. Taken together, our findings demonstrate that β-catenin is essential for cell proliferation and migration but dispensable for endocardial cells to gain mesenchymal fate during endocardial cushion formation. Mechanistically, β-catenin promotes cell proliferation by suppressing p21. These findings inform the potential role of β-catenin in the etiology of congenital heart defects.
Collapse
Affiliation(s)
- Huahua Liu
- Department of Cardiology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shan He
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Yuru Luo
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Yuan Fang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Sonia Benkaci
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yidong Wang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences; Department of Cardiology, First Affiliated Hospital; Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Bin Zhou
- Departments of Genetics, Pediatrics (Pediatric Genetic Medicine), and Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, The Einstein Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
5
|
O’Donnell A, Gonzalez BA, Mukherjee S, Wilson R, Alfieri CM, Swoboda CO, Millay DP, Zorn AM, Yutzey KE. Localized Prox1 Regulates Aortic Valve Endothelial Cell Diversity and Extracellular Matrix Stratification in Mice. Arterioscler Thromb Vasc Biol 2023; 43:1478-1493. [PMID: 37381982 PMCID: PMC10528305 DOI: 10.1161/atvbaha.123.319424] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Specialized valve endothelial cell (VEC) populations are localized oriented to blood flow in developing aortic and mitral valves, but their roles in valve development and disease are unknown. In the aortic valve (AoV), a population of VECs on the fibrosa side expresses the transcription factor Prox1 together with genes found in lymphatic ECs. In this study, we examine Prox1's role in regulating a lymphatic-like gene network and promoting VEC diversity required for the development of the stratified trilaminar extracellular matrix (ECM) of murine AoV leaflets. METHODS To determine whether disruption of Prox1 localization affects heart valve development, we generated mice (NFATc1enCre Prox1 gain-of-function) in which Prox1 is overexpressed on the ventricularis side of the AoV beginning in embryonic development. To identify potential targets of Prox1, we performed cleavage under targets and release using nuclease on wild-type and NFATc1enCre Prox1 gain-of-function AoVs with validation by colocalization in vivo using RNA in situ hybridization in NFATc1enCre Prox1 gain-of-function AoVs. Natural induction of Prox1 and target gene expression was evaluated in myxomatous AoVs in a mouse model of Marfan syndrome (Fbn1C1039G/+). RESULTS The overexpression of Prox1 is sufficient to cause enlargement of AoVs by postnatal day (P)0, as well as a decrease in ventricularis-specific gene expression and disorganized interstitial ECM layers at P7. We identified potential targets of Prox1 known to play roles in lymphatic ECs including Flt1, Efnb2, Egfl7, and Cx37. Ectopic Prox1 colocalized with induced Flt1, Efnb2, and Cx37 expression in NFATc1enCre Prox1 gain-of-function AoVs. Moreover, in Marfan syndrome myxomatous AoVs, endogenous Prox1, and its identified targets, were ectopically induced in ventricularis side VECs. CONCLUSIONS Our results support a role for Prox1 in localized lymphatic-like gene expression on the fibrosa side of the AoV. Furthermore, localized VEC specialization is required for development of the stratified trilaminar ECM critical for AoV function and is dysregulated in congenitally malformed valves.
Collapse
Affiliation(s)
- Anna O’Donnell
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Brittany A. Gonzalez
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Shreyasi Mukherjee
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Ruby Wilson
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Christina M. Alfieri
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Casey O. Swoboda
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Douglas P. Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Aaron M. Zorn
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Katherine E. Yutzey
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
6
|
Bishop D, Schwarz Q, Wiszniak S. Endothelial-derived angiocrine factors as instructors of embryonic development. Front Cell Dev Biol 2023; 11:1172114. [PMID: 37457293 PMCID: PMC10339107 DOI: 10.3389/fcell.2023.1172114] [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: 02/23/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Blood vessels are well-known to play roles in organ development and repair, primarily owing to their fundamental function in delivering oxygen and nutrients to tissues to promote their growth and homeostasis. Endothelial cells however are not merely passive conduits for carrying blood. There is now evidence that endothelial cells of the vasculature actively regulate tissue-specific development, morphogenesis and organ function, as well as playing roles in disease and cancer. Angiocrine factors are growth factors, cytokines, signaling molecules or other regulators produced directly from endothelial cells to instruct a diverse range of signaling outcomes in the cellular microenvironment, and are critical mediators of the vascular control of organ function. The roles of angiocrine signaling are only beginning to be uncovered in diverse fields such as homeostasis, regeneration, organogenesis, stem-cell maintenance, cell differentiation and tumour growth. While in some cases the specific angiocrine factor involved in these processes has been identified, in many cases the molecular identity of the angiocrine factor(s) remain to be discovered, even though the importance of angiocrine signaling has been implicated. In this review, we will specifically focus on roles for endothelial-derived angiocrine signaling in instructing tissue morphogenesis and organogenesis during embryonic and perinatal development.
Collapse
|
7
|
Hernández-García A, Pendleton KE, Kim S, Li Y, Kim BJ, Zaveri HP, Jordan VK, Berry AM, Ljungberg MC, Chen R, Lanz RB, Scott DA. SOX7 deficiency causes ventricular septal defects through its effects on endocardial-to-mesenchymal transition and the expression of Wnt4 and Bmp2. Hum Mol Genet 2023; 32:2152-2161. [PMID: 37000005 PMCID: PMC10281751 DOI: 10.1093/hmg/ddad050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/09/2023] [Accepted: 03/27/2023] [Indexed: 04/01/2023] Open
Abstract
SOX7 is a transcription factor-encoding gene located in a region on chromosome 8p23.1 that is recurrently deleted in individuals with ventricular septal defects (VSDs). We have previously shown that Sox7-/- embryos die of heart failure around E11.5. Here, we demonstrate that these embryos have hypocellular endocardial cushions with severely reduced numbers of mesenchymal cells. Ablation of Sox7 in the endocardium also resulted in hypocellular endocardial cushions, and we observed VSDs in rare E15.5 Sox7flox/-;Tie2-Cre and Sox7flox/flox;Tie2-Cre embryos that survived to E15.5. In atrioventricular explant studies, we showed that SOX7 deficiency leads to a severe reduction in endocardial-to-mesenchymal transition (EndMT). RNA-seq studies performed on E9.5 Sox7-/- heart tubes revealed severely reduced Wnt4 transcript levels. Wnt4 is expressed in the endocardium and promotes EndMT by acting in a paracrine manner to increase the expression of Bmp2 in the myocardium. Both WNT4 and BMP2 have been previously implicated in the development of VSDs in individuals with 46,XX sex reversal with dysgenesis of kidney, adrenals and lungs (SERKAL) syndrome and in individuals with short stature, facial dysmorphism and skeletal anomalies with or without cardiac anomalies 1 (SSFSC1) syndrome, respectively. We now show that Sox7 and Wnt4 interact genetically in the development of VSDs through their additive effects on endocardial cushion development with Sox7+/-;Wnt4+/- double heterozygous embryos having hypocellular endocardial cushions and perimembranous and muscular VSDs not seen in their Sox7+/- and Wnt4+/- littermates. These results provide additional evidence that SOX7, WNT4 and BMP2 function in the same pathway during mammalian septal development and that their deficiency can contribute to the development of VSDs in humans.
Collapse
Affiliation(s)
- Andrés Hernández-García
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Katherine E Pendleton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sangbae Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yumei Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bum J Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hitisha P Zaveri
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Valerie K Jordan
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Aliska M Berry
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - M Cecilia Ljungberg
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX 77030, USA
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rainer B Lanz
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daryl A Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
8
|
George RM, Firulli BA, Podicheti R, Rusch DB, Mannion BJ, Pennacchio LA, Osterwalder M, Firulli AB. Single cell evaluation of endocardial Hand2 gene regulatory networks reveals HAND2-dependent pathways that impact cardiac morphogenesis. Development 2023; 150:dev201341. [PMID: 36620995 PMCID: PMC10110492 DOI: 10.1242/dev.201341] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/26/2022] [Indexed: 01/10/2023]
Abstract
The transcription factor HAND2 plays essential roles during cardiogenesis. Hand2 endocardial deletion (H2CKO) results in tricuspid atresia or double inlet left ventricle with accompanying intraventricular septum defects, hypo-trabeculated ventricles and an increased density of coronary lumens. To understand the regulatory mechanisms of these phenotypes, single cell transcriptome analysis of mouse E11.5 H2CKO hearts was performed revealing a number of disrupted endocardial regulatory pathways. Using HAND2 DNA occupancy data, we identify several HAND2-dependent enhancers, including two endothelial enhancers for the shear-stress master regulator KLF2. A 1.8 kb enhancer located 50 kb upstream of the Klf2 TSS imparts specific endothelial/endocardial expression within the vasculature and endocardium. This enhancer is HAND2-dependent for ventricular endocardium expression but HAND2-independent for Klf2 vascular and valve expression. Deletion of this Klf2 enhancer results in reduced Klf2 expression within ventricular endocardium. These data reveal that HAND2 functions within endocardial gene regulatory networks including shear-stress response.
Collapse
Affiliation(s)
- Rajani M. George
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
| | - Beth A. Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
| | - Ram Podicheti
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA
| | - Douglas B. Rusch
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA
| | - Brandon J. Mannion
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA 94720, USA
| | - Len A. Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA 94720, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Marco Osterwalder
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Department for BioMedical Research (DBMR), University of Bern, Bern 3008, Switzerland
- Department of Cardiology, Bern University Hospital, Bern 3010, Switzerland
| | - Anthony B. Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
| |
Collapse
|
9
|
Siguero-Álvarez M, Salguero-Jiménez A, Grego-Bessa J, de la Barrera J, MacGrogan D, Prados B, Sánchez-Sáez F, Piñeiro-Sabarís R, Felipe-Medina N, Torroja C, Gómez MJ, Sabater-Molina M, Escribá R, Richaud-Patin I, Iglesias-García O, Sbroggio M, Callejas S, O'Regan DP, McGurk KA, Dopazo A, Giovinazzo G, Ibañez B, Monserrat L, Pérez-Pomares JM, Sánchez-Cabo F, Pendas AM, Raya A, Gimeno-Blanes JR, de la Pompa JL. A Human Hereditary Cardiomyopathy Shares a Genetic Substrate With Bicuspid Aortic Valve. Circulation 2023; 147:47-65. [PMID: 36325906 DOI: 10.1161/circulationaha.121.058767] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND The complex genetics underlying human cardiac disease is evidenced by its heterogenous manifestation, multigenic basis, and sporadic occurrence. These features have hampered disease modeling and mechanistic understanding. Here, we show that 2 structural cardiac diseases, left ventricular noncompaction (LVNC) and bicuspid aortic valve, can be caused by a set of inherited heterozygous gene mutations affecting the NOTCH ligand regulator MIB1 (MINDBOMB1) and cosegregating genes. METHODS We used CRISPR-Cas9 gene editing to generate mice harboring a nonsense or a missense MIB1 mutation that are both found in LVNC families. We also generated mice separately carrying these MIB1 mutations plus 5 additional cosegregating variants in the ASXL3, APCDD1, TMX3, CEP192, and BCL7A genes identified in these LVNC families by whole exome sequencing. Histological, developmental, and functional analyses of these mouse models were carried out by echocardiography and cardiac magnetic resonance imaging, together with gene expression profiling by RNA sequencing of both selected engineered mouse models and human induced pluripotent stem cell-derived cardiomyocytes. Potential biochemical interactions were assayed in vitro by coimmunoprecipitation and Western blot. RESULTS Mice homozygous for the MIB1 nonsense mutation did not survive, and the mutation caused LVNC only in heteroallelic combination with a conditional allele inactivated in the myocardium. The heterozygous MIB1 missense allele leads to bicuspid aortic valve in a NOTCH-sensitized genetic background. These data suggest that development of LVNC is influenced by genetic modifiers present in affected families, whereas valve defects are highly sensitive to NOTCH haploinsufficiency. Whole exome sequencing of LVNC families revealed single-nucleotide gene variants of ASXL3, APCDD1, TMX3, CEP192, and BCL7A cosegregating with the MIB1 mutations and LVNC. In experiments with mice harboring the orthologous variants on the corresponding Mib1 backgrounds, triple heterozygous Mib1 Apcdd1 Asxl3 mice showed LVNC, whereas quadruple heterozygous Mib1 Cep192 Tmx3;Bcl7a mice developed bicuspid aortic valve and other valve-associated defects. Biochemical analysis suggested interactions between CEP192, BCL7A, and NOTCH. Gene expression profiling of mutant mouse hearts and human induced pluripotent stem cell-derived cardiomyocytes revealed increased cardiomyocyte proliferation and defective morphological and metabolic maturation. CONCLUSIONS These findings reveal a shared genetic substrate underlying LVNC and bicuspid aortic valve in which MIB1-NOTCH variants plays a crucial role in heterozygous combination with cosegregating genetic modifiers.
Collapse
Affiliation(s)
- Marcos Siguero-Álvarez
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Center for Chromosome Stability and Institut for Cellulær og Molekylær Medicin, University of Copenhagen, Denmark (M.S.)
| | - Alejandro Salguero-Jiménez
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Joaquim Grego-Bessa
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Jorge de la Barrera
- Bioinformatics Unit (J.d.l.B., C.T., M.J.G., F.S.-C.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Donal MacGrogan
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Belén Prados
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Pluripotent Cell Technology Unit (B.P., G.G.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Fernando Sánchez-Sáez
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer Universidad de Salamanca, Spain (F.S.-S., N.F.-M., A.M.P.)
| | - Rebeca Piñeiro-Sabarís
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Natalia Felipe-Medina
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer Universidad de Salamanca, Spain (F.S.-S., N.F.-M., A.M.P.)
| | - Carlos Torroja
- Bioinformatics Unit (J.d.l.B., C.T., M.J.G., F.S.-C.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Manuel José Gómez
- Genomics Unit (S.C., A.D.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Laboratorio de Cardiogenética, Instituto Murciano de Investigación Biosanitaria, European Reference Networks and Unidad de Referencia-European Reference Networks Guard Heart de Cardiopatias Familiares, Hospital Universitario Virgen de la Arrixaca-Universidad de Murcia, El Palmar, Spain (M.S.-M., J.R.G.-B.)
| | - María Sabater-Molina
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Rubén Escribá
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research, Program for Clinical Translation of Regenerative Medicine in Catalonia, Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine and Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain (R.E., I.R.-P., O.I.-G., A.R.)
| | - Ivonne Richaud-Patin
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research, Program for Clinical Translation of Regenerative Medicine in Catalonia, Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine and Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain (R.E., I.R.-P., O.I.-G., A.R.)
| | - Olalla Iglesias-García
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research, Program for Clinical Translation of Regenerative Medicine in Catalonia, Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine and Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain (R.E., I.R.-P., O.I.-G., A.R.)
- Regenerative Medicine Program, Cima Universidad de Navarra, Navarra Institute for Health Research, Pamplona, Spain (O.I.-G.)
| | - Mauro Sbroggio
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Sergio Callejas
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Genomics Unit (S.C., A.D.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Declan P O'Regan
- Medical Research Council London Institute of Medical Sciences (D.P.O.' K.A.M.), Imperial College London, United Kingdom
| | - Kathryn A McGurk
- Medical Research Council London Institute of Medical Sciences (D.P.O.' K.A.M.), Imperial College London, United Kingdom
- National Heart and Lung Institute (K.A.M.), Imperial College London, United Kingdom
| | - Ana Dopazo
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Genomics Unit (S.C., A.D.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Giovanna Giovinazzo
- Pluripotent Cell Technology Unit (B.P., G.G.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Borja Ibañez
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Translational Laboratory (B.I.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Cardiology Department, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz Hospital, Madrid, Spain (B.I.)
| | - Lorenzo Monserrat
- Instituto de Investigación Biomédica de A Coruña and Departamento Científico, Health in Code S.L., A Coruña, Spain (L.M.)
| | - José María Pérez-Pomares
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Department of Animal Biology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga and Centro Andaluz de Nanomedicina y Biotecnología, Universidad de Málaga, Spain (J.M.P.-P.)
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit (J.d.l.B., C.T., M.J.G., F.S.-C.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Alberto M Pendas
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer Universidad de Salamanca, Spain (F.S.-S., N.F.-M., A.M.P.)
| | - Angel Raya
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research, Program for Clinical Translation of Regenerative Medicine in Catalonia, Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine and Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain (R.E., I.R.-P., O.I.-G., A.R.)
| | - Juan R Gimeno-Blanes
- Laboratorio de Cardiogenética, Instituto Murciano de Investigación Biosanitaria, European Reference Networks and Unidad de Referencia-European Reference Networks Guard Heart de Cardiopatias Familiares, Hospital Universitario Virgen de la Arrixaca-Universidad de Murcia, El Palmar, Spain (M.S.-M., J.R.G.-B.)
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| |
Collapse
|
10
|
Bao B, Hu H, Chen L, Lu S, Tang Q, Liang Z. SNP and DNA methylation analyses of a monozygotic twins discordant for complete endocardial cushion defect: a case report. Am J Transl Res 2022; 14:8271-8278. [PMID: 36505317 PMCID: PMC9730093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/24/2022] [Indexed: 12/15/2022]
Abstract
The exact cause of complete endocardial cushion defect (ECD) is still unknown. This report describes a unique pair of monozygotic twins (MZ twins) discordant for ECD. The chromosome karyotyping analysis revealed normal karyotype of 46, XY, 16qh+ and mat in both MZ twins. A genome-wide analysis of DNA using the Affymetrix SNP 6.0 revealed identical genotyping of single nucleotide polymorphisms (SNPs) and copy number variations (CNVs). An extensive methylation assay was carried out by NimbleGen 3 × 720 K CpG Island Plus RefSeq Promoter Arrays to analyze the potential epigenetic differences. The DNA methylation profiles of the affected twin seemed increased compared with that of the unaffected twin. However, further validation of Notch1 promoter hypermethylation and six top-ranked differentially methylated CpG sites by sodium bisulfate modification and methylation-specific PCR, failed to reveal consistent methylation differences between the twins. Other relevant factors, such as heritability and penetrance of the condition that place the MZ twins near to a threshold for ECD or variations in local epigenetic events in the twins' heart tissues, are probably responsible for the phenotypic discordance.
Collapse
Affiliation(s)
- Bihui Bao
- Department of Obstetrics and Gynecology, Qingbaijiang Women’s and Children’s Hospital (Maternal and Child Health Hospital), West China Second University Hospital, Sichuan UniversityChengdu 610300, Sichuan, China
| | - Hua Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital (Xinqiao Hospital), Army Medical UniversityChongqing 400037, China
| | - Limei Chen
- Department of Obstetrics and Gynecology, Qingbaijiang Women’s and Children’s Hospital (Maternal and Child Health Hospital), West China Second University Hospital, Sichuan UniversityChengdu 610300, Sichuan, China
| | - Shiyong Lu
- Department of Obstetrics and Gynecology, Qingbaijiang Women’s and Children’s Hospital (Maternal and Child Health Hospital), West China Second University Hospital, Sichuan UniversityChengdu 610300, Sichuan, China
| | - Qifeng Tang
- Department of Anesthesiology, Shanghai Hechuan-Rhine TCM HospitalShanghai 201103, China
| | - Zhiqing Liang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital (Southwest Hospital), Army Medical UniversityChongqing 400038, China
| |
Collapse
|
11
|
Lu P, Wang P, Wu B, Wang Y, Liu Y, Cheng W, Feng X, Yuan X, Atteya MM, Ferro H, Sugi Y, Rydquist G, Esmaily M, Butcher JT, Chang CP, Lenz J, Zheng D, Zhou B. A SOX17-PDGFB signaling axis regulates aortic root development. Nat Commun 2022; 13:4065. [PMID: 35831318 PMCID: PMC9279414 DOI: 10.1038/s41467-022-31815-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 06/30/2022] [Indexed: 11/08/2022] Open
Abstract
Developmental etiologies causing complex congenital aortic root abnormalities are unknown. Here we show that deletion of Sox17 in aortic root endothelium in mice causes underdeveloped aortic root leading to a bicuspid aortic valve due to the absence of non-coronary leaflet and mispositioned left coronary ostium. The respective defects are associated with reduced proliferation of non-coronary leaflet mesenchyme and aortic root smooth muscle derived from the second heart field cardiomyocytes. Mechanistically, SOX17 occupies a Pdgfb transcriptional enhancer to promote its transcription and Sox17 deletion inhibits the endothelial Pdgfb transcription and PDGFB growth signaling to the non-coronary leaflet mesenchyme. Restoration of PDGFB in aortic root endothelium rescues the non-coronary leaflet and left coronary ostium defects in Sox17 nulls. These data support a SOX17-PDGFB axis underlying aortic root development that is critical for aortic valve and coronary ostium patterning, thereby informing a potential shared disease mechanism for concurrent anomalous aortic valve and coronary arteries.
Collapse
Affiliation(s)
- Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ping Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yidong Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Cardiovascular Research Center, School of Basic Medical Sciences, Jiaotong University, Xi'an, Shanxi, China
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wei Cheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xuhui Feng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xinchun Yuan
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Miriam M Atteya
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Haleigh Ferro
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Yukiko Sugi
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Grant Rydquist
- School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Mahdi Esmaily
- School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | | | - Ching-Pin Chang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jack Lenz
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
- Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
- Departments of Pediatrics and Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
12
|
Pham DH, Dai CR, Lin B, Butcher JT. Local fluid shear stress operates a molecular switch to drive fetal semilunar valve extension. Dev Dyn 2022; 251:481-497. [PMID: 34535945 PMCID: PMC8891031 DOI: 10.1002/dvdy.419] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND While much is known about the genetic regulation of early valvular morphogenesis, mechanisms governing fetal valvular growth and remodeling remain unclear. Hemodynamic forces strongly influence morphogenesis, but it is unknown whether or how they interact with valvulogenic signaling programs. Side-specific activity of valvulogenic programs motivates the hypothesis that shear stress pattern-specific endocardial signaling controls the elongation of leaflets. RESULTS We determined that extension of the semilunar valve occurs via fibrosa sided endocardial proliferation. Low OSS was necessary and sufficient to induce canonical Wnt/β-catenin activation in fetal valve endothelium, which in turn drives BMP receptor/ligand expression, and pSmad1/5 activity essential for endocardial proliferation. In contrast, ventricularis endocardial cells expressed active Notch1 but minimal pSmad1/5. Endocardial monolayers exposed to LSS attenuate Wnt signaling in a Notch1 dependent manner. CONCLUSIONS Low OSS is transduced by endocardial cells into canonical Wnt signaling programs that regulate BMP signaling and endocardial proliferation. In contrast, high LSS induces Notch signaling in endocardial cells, inhibiting Wnt signaling and thereby restricting growth on the ventricular surface. Our results identify a novel mechanically regulated molecular switch, whereby fluid shear stress drives the growth of valve endothelium, orchestrating the extension of the valve in the direction of blood flow.
Collapse
Affiliation(s)
- Duc H. Pham
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Charles R. Dai
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Belle Lin
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jonathan T. Butcher
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA,Corresponding author:
| |
Collapse
|
13
|
Sen S, Hallee L, Lam CK. The Potential of Gamma Secretase as a Therapeutic Target for Cardiac Diseases. J Pers Med 2021; 11:jpm11121294. [PMID: 34945766 PMCID: PMC8703931 DOI: 10.3390/jpm11121294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
Heart diseases are some of the most common and pressing threats to human health worldwide. The American Heart Association and the National Institute of Health jointly work to annually update data on cardiac diseases. In 2018, 126.9 million Americans were reported as having some form of cardiac disorder, with an estimated direct and indirect total cost of USD 363.4 billion. This necessitates developing therapeutic interventions for heart diseases to improve human life expectancy and economic relief. In this review, we look into gamma-secretase as a potential therapeutic target for cardiac diseases. Gamma-secretase, an aspartyl protease enzyme, is responsible for the cleavage and activation of a number of substrates that are relevant to normal cardiac development and function as found in mutation studies. Some of these substrates are involved in downstream signaling processes and crosstalk with pathways relevant to heart diseases. Most of the substrates and signaling events we explored were found to be potentially beneficial to maintain cardiac function in diseased conditions. This review presents an updated overview of the current knowledge on gamma-secretase processing of cardiac-relevant substrates and seeks to understand if the modulation of gamma-secretase activity would be beneficial to combat cardiac diseases.
Collapse
Affiliation(s)
- Sujoita Sen
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Logan Hallee
- Department of Mathematical Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Chi Keung Lam
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Correspondence: ; Tel.: +1-302-831-3165
| |
Collapse
|
14
|
Abu Nahia K, Migdał M, Quinn TA, Poon KL, Łapiński M, Sulej A, Liu J, Mondal SS, Pawlak M, Bugajski Ł, Piwocka K, Brand T, Kohl P, Korzh V, Winata C. Genomic and physiological analyses of the zebrafish atrioventricular canal reveal molecular building blocks of the secondary pacemaker region. Cell Mol Life Sci 2021; 78:6669-6687. [PMID: 34557935 PMCID: PMC8558220 DOI: 10.1007/s00018-021-03939-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/06/2021] [Accepted: 09/10/2021] [Indexed: 01/06/2023]
Abstract
The atrioventricular canal (AVC) is the site where key structures responsible for functional division between heart regions are established, most importantly, the atrioventricular (AV) conduction system and cardiac valves. To elucidate the mechanism underlying AVC development and function, we utilized transgenic zebrafish line sqet31Et expressing EGFP in the AVC to isolate this cell population and profile its transcriptome at 48 and 72 hpf. The zebrafish AVC transcriptome exhibits hallmarks of mammalian AV node, including the expression of genes implicated in its development and those encoding connexins forming low conductance gap junctions. Transcriptome analysis uncovered protein-coding and noncoding transcripts enriched in AVC, which have not been previously associated with this structure, as well as dynamic expression of epithelial-to-mesenchymal transition markers and components of TGF-β, Notch, and Wnt signaling pathways likely reflecting ongoing AVC and valve development. Using transgenic line Tg(myl7:mermaid) encoding voltage-sensitive fluorescent protein, we show that abolishing the pacemaker-containing sinoatrial ring (SAR) through Isl1 loss of function resulted in spontaneous activation in the AVC region, suggesting that it possesses inherent automaticity although insufficient to replace the SAR. The SAR and AVC transcriptomes express partially overlapping species of ion channels and gap junction proteins, reflecting their distinct roles. Besides identifying conserved aspects between zebrafish and mammalian conduction systems, our results established molecular hallmarks of the developing AVC which underlies its role in structural and electrophysiological separation between heart chambers. This data constitutes a valuable resource for studying AVC development and function, and identification of novel candidate genes implicated in these processes.
Collapse
Affiliation(s)
- Karim Abu Nahia
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Maciej Migdał
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kar-Lai Poon
- Institute of Molecular and Cell Biology, 61 Biopolis Dr, Singapore , Singapore.,Developmental Dynamics, National Heart and Lung Institute, Imperial College London, London, UK
| | - Maciej Łapiński
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Agata Sulej
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, USA
| | - Shamba S Mondal
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Michał Pawlak
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | | | | | - Thomas Brand
- Developmental Dynamics, National Heart and Lung Institute, Imperial College London, London, UK
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Centre, Faculty of Medicine, and Faculty of Engineering, University of Freiburg, Freiburg im Breisgau, Germany
| | - Vladimir Korzh
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.
| | - Cecilia Winata
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.
| |
Collapse
|
15
|
Cheng L, Xie M, Qiao W, Song Y, Zhang Y, Geng Y, Xu W, Wang L, Wang Z, Huang K, Dong N, Sun Y. Generation and characterization of cardiac valve endothelial-like cells from human pluripotent stem cells. Commun Biol 2021; 4:1039. [PMID: 34489520 PMCID: PMC8421482 DOI: 10.1038/s42003-021-02571-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 08/18/2021] [Indexed: 12/31/2022] Open
Abstract
The cardiac valvular endothelial cells (VECs) are an ideal cell source that could be used for making the valve organoids. However, few studies have been focused on the derivation of this important cell type. Here we describe a two-step chemically defined xeno-free method for generating VEC-like cells from human pluripotent stem cells (hPSCs). HPSCs were specified to KDR+/ISL1+ multipotent cardiac progenitors (CPCs), followed by differentiation into valve endothelial-like cells (VELs) via an intermediate endocardial cushion cell (ECC) type. Mechanistically, administration of TGFb1 and BMP4 may specify VEC fate by activating the NOTCH/WNT signaling pathways and previously unidentified targets such as ATF3 and KLF family of transcription factors. When seeded onto the surface of the de-cellularized porcine aortic valve (DCV) matrix scaffolds, hPSC-derived VELs exhibit superior proliferative and clonogenic potential than the primary VECs and human aortic endothelial cells (HAEC). Our results show that hPSC-derived valvular cells could be efficiently generated from hPSCs, which might be used as seed cells for construction of valve organoids or next generation tissue engineered heart valves. Cheng et al. provide a detailed characterization of the differentiation of human pluripotent stem cells to valve endothelial cells and their function. Their results show that the valve endothelial-like cells express key markers for valve endothelial cells, exhibiting proliferative and clonogenic potential.
Collapse
Affiliation(s)
- LinXi Cheng
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - MingHui Xie
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - WeiHua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Song
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - YanYong Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - YingChao Geng
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - WeiLin Xu
- Wuhan Textile University, Wuhan, China
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Department of Cardiovascular Internal Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - NianGuo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - YuHua Sun
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China. .,University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
16
|
Kim H, Wang M, Paik DT. Endothelial-Myocardial Angiocrine Signaling in Heart Development. Front Cell Dev Biol 2021; 9:697130. [PMID: 34277641 PMCID: PMC8281241 DOI: 10.3389/fcell.2021.697130] [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: 04/18/2021] [Accepted: 06/10/2021] [Indexed: 12/23/2022] Open
Abstract
Vascular endothelial cells are a multifunctional cell type with organotypic specificity in their function and structure. In this review, we discuss various subpopulations of endothelial cells in the mammalian heart, which spatiotemporally regulate critical cellular and molecular processes of heart development via unique sets of angiocrine signaling pathways. In particular, elucidation of intercellular communication among the functional cell types in the developing heart has recently been accelerated by the use of single-cell sequencing. Specifically, we overview the heterogeneic nature of cardiac endothelial cells and their contribution to heart tube and chamber formation, myocardial trabeculation and compaction, and endocardial cushion and valve formation via angiocrine pathways.
Collapse
Affiliation(s)
- Hyeonyu Kim
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Mingqiang Wang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - David T Paik
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
17
|
Wang Y, Fang Y, Lu P, Wu B, Zhou B. NOTCH Signaling in Aortic Valve Development and Calcific Aortic Valve Disease. Front Cardiovasc Med 2021; 8:682298. [PMID: 34239905 PMCID: PMC8259786 DOI: 10.3389/fcvm.2021.682298] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/14/2021] [Indexed: 01/05/2023] Open
Abstract
NOTCH intercellular signaling mediates the communications between adjacent cells involved in multiple biological processes essential for tissue morphogenesis and homeostasis. The NOTCH1 mutations are the first identified human genetic variants that cause congenital bicuspid aortic valve (BAV) and calcific aortic valve disease (CAVD). Genetic variants affecting other genes in the NOTCH signaling pathway may also contribute to the development of BAV and the pathogenesis of CAVD. While CAVD occurs commonly in the elderly population with tri-leaflet aortic valve, patients with BAV have a high risk of developing CAVD at a young age. This observation indicates an important role of NOTCH signaling in the postnatal homeostasis of the aortic valve, in addition to its prenatal functions during aortic valve development. Over the last decade, animal studies, especially with the mouse models, have revealed detailed information in the developmental etiology of congenital aortic valve defects. In this review, we will discuss the molecular and cellular aspects of aortic valve development and examine the embryonic pathogenesis of BAV. We will focus our discussions on the NOTCH signaling during the endocardial-to-mesenchymal transformation (EMT) and the post-EMT remodeling of the aortic valve. We will further examine the involvement of the NOTCH mutations in the postnatal development of CAVD. We will emphasize the deleterious impact of the embryonic valve defects on the homeostatic mechanisms of the adult aortic valve for the purpose of identifying the potential therapeutic targets for disease intervention.
Collapse
Affiliation(s)
- Yidong Wang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yuan Fang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Bin Zhou
- Departments of Genetics, Pediatrics (Pediatric Genetic Medicine), and Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States
- The Einstein Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
18
|
Fontana F, Haack T, Reichenbach M, Knaus P, Puceat M, Abdelilah-Seyfried S. Antagonistic Activities of Vegfr3/Flt4 and Notch1b Fine-tune Mechanosensitive Signaling during Zebrafish Cardiac Valvulogenesis. Cell Rep 2021; 32:107883. [PMID: 32668254 DOI: 10.1016/j.celrep.2020.107883] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/10/2020] [Accepted: 06/17/2020] [Indexed: 12/22/2022] Open
Abstract
The formation of cardiac valves depends on mechanical forces exerted by blood flow. Endocardial cells lining the interior of the heart are sensitive to these stimuli and respond by rearranging into luminal cells subjected to shear stress and abluminal cells not exposed to it. The mechanisms by which endocardial cells sense these dynamic biomechanical stimuli and how they evoke different cellular responses are largely unknown. Here, we show that blood flow activates two parallel mechanosensitive pathways, one mediated by Notch and the other by Klf2a. Both pathways negatively regulate the angiogenesis receptor Vegfr3/Flt4, which becomes restricted to abluminal endocardial cells. Its loss disrupts valve morphogenesis and results in the occurrence of Notch signaling within abluminal endocardial cells. Our work explains how antagonistic activities by Vegfr3/Flt4 on the abluminal side and by Notch on the luminal side shape cardiac valve leaflets by triggering unique differences in the fates of endocardial cells.
Collapse
Affiliation(s)
- Federica Fontana
- Institute of Biochemistry and Biology, Potsdam University, D-14476 Potsdam, Germany
| | - Timm Haack
- Institute of Molecular Biology, Hannover Medical School, D-30625 Hannover, Germany
| | - Maria Reichenbach
- Institute of Biochemistry, Freie Universität Berlin, D-14195 Berlin, Germany
| | - Petra Knaus
- Institute of Biochemistry, Freie Universität Berlin, D-14195 Berlin, Germany
| | - Michel Puceat
- INSERM U-1251, MMG, Aix-Marseille University, 13885 Marseille, France
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, D-14476 Potsdam, Germany; Institute of Molecular Biology, Hannover Medical School, D-30625 Hannover, Germany.
| |
Collapse
|
19
|
Hong N, Zhang E, Xie H, Jin L, Zhang Q, Lu Y, Chen AF, Yu Y, Zhou B, Chen S, Yu Y, Sun K. The transcription factor Sox7 modulates endocardiac cushion formation contributed to atrioventricular septal defect through Wnt4/Bmp2 signaling. Cell Death Dis 2021; 12:393. [PMID: 33846290 PMCID: PMC8041771 DOI: 10.1038/s41419-021-03658-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 03/22/2021] [Indexed: 02/02/2023]
Abstract
Cardiac septum malformations account for the largest proportion in congenital heart defects. The transcription factor Sox7 has critical functions in the vascular development and angiogenesis. It is unclear whether Sox7 also contributes to cardiac septation development. We identified a de novo 8p23.1 deletion with Sox7 haploinsufficiency in an atrioventricular septal defect (AVSD) patient using whole exome sequencing in 100 AVSD patients. Then, multiple Sox7 conditional loss-of-function mice models were generated to explore the role of Sox7 in atrioventricular cushion development. Sox7 deficiency mice embryos exhibited partial AVSD and impaired endothelial to mesenchymal transition (EndMT). Transcriptome analysis revealed BMP signaling pathway was significantly downregulated in Sox7 deficiency atrioventricular cushions. Mechanistically, Sox7 deficiency reduced the expressions of Bmp2 in atrioventricular canal myocardium and Wnt4 in endocardium, and Sox7 binds to Wnt4 and Bmp2 directly. Furthermore, WNT4 or BMP2 protein could partially rescue the impaired EndMT process caused by Sox7 deficiency, and inhibition of BMP2 by Noggin could attenuate the effect of WNT4 protein. In summary, our findings identify Sox7 as a novel AVSD pathogenic candidate gene, and it can regulate the EndMT involved in atrioventricular cushion morphogenesis through Wnt4-Bmp2 signaling. This study contributes new strategies to the diagnosis and treatment of congenital heart defects.
Collapse
Affiliation(s)
- Nanchao Hong
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China ,grid.8547.e0000 0001 0125 2443Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Erge Zhang
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Huilin Xie
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China ,grid.8547.e0000 0001 0125 2443Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Lihui Jin
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Qi Zhang
- grid.16821.3c0000 0004 0368 8293Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 200092 Shanghai, China
| | - Yanan Lu
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Alex F. Chen
- grid.16821.3c0000 0004 0368 8293Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 200092 Shanghai, China
| | - Yongguo Yu
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Bin Zhou
- grid.9227.e0000000119573309Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China
| | - Sun Chen
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Yu Yu
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China ,grid.16821.3c0000 0004 0368 8293Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 200092 Shanghai, China
| | - Kun Sun
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| |
Collapse
|
20
|
Ford SM, Pedersen CJ, Ford MR, Kim JW, Karunamuni GH, McPheeters MT, Jawaid S, Jenkins MW, Rollins AM, Watanabe M. Folic acid prevents functional and structural heart defects induced by prenatal ethanol exposure. Am J Physiol Heart Circ Physiol 2021. [DOI: 10.1152/ajpheart.00817.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
State-of-the-art biophotonic tools captured blood flow and endocardial cushion volumes in tiny beating quail embryo hearts, an accessible model for studying four-chambered heart development. Both hemodynamic flow and endocardial cushion volumes were altered with ethanol exposure but normalized when folic acid was introduced with ethanol. Folic acid supplementation preserved hemodynamic function that is intimately involved in sculpting the heart from the earliest stages of heart development.
Collapse
Affiliation(s)
- Stephanie M. Ford
- Division of Neonatology, Department of Pediatrics, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Cameron J. Pedersen
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Matthew R. Ford
- Department of Ophthalmology, Cole Eye Institute, Cleveland Clinic, Cleveland Ohio
| | - Jun W. Kim
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Ganga H. Karunamuni
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Matthew T. McPheeters
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Safdar Jawaid
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Michael W. Jenkins
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Andrew M. Rollins
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Michiko Watanabe
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
21
|
Wang Y, Lu P, Jiang L, Wu B, Zhou B. Control of sinus venous valve and sinoatrial node development by endocardial NOTCH1. Cardiovasc Res 2021; 116:1473-1486. [PMID: 31591643 DOI: 10.1093/cvr/cvz249] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 08/06/2019] [Accepted: 10/01/2019] [Indexed: 12/22/2022] Open
Abstract
AIMS Sinus venous valve (SVV) and sinoatrial node (SAN) develop together at the sinoatrial junction during embryogenesis. SVV ensures unidirectional cardiac input and SAN generates sinus rhythmic contraction, respectively; both functions are essential for embryonic survival. We aim to reveal the potential role of endocardial NOTCH signalling in SVV and SAN formation. METHODS AND RESULTS We specifically deleted Notch1 in the endocardium using an Nfatc1Cre line. This deletion resulted in underdeveloped SVV and SAN, associated with reduced expression of T-box transcription factors, Tbx5 andTbx18, which are essential for the formation of SVV and SAN. The deletion also led to decreased expression of Wnt2 in myocardium of SVV and SAN. WNT2 treatment was able to rescue the growth defect of SVV and SAN resulted from the Notch1 deletion in whole embryo cultures. Furthermore, the Notch1 deletion reduced the expression of Nrg1 in the SVV myocardium and supplement of NRG1 restored the growth of SVV in cultured Notch1 knockout embryos. CONCLUSION Our findings support that endocardial NOTCH1 controls the development of SVV and SAN by coordinating myocardial WNT and NRG1 signalling functions.
Collapse
Affiliation(s)
- Yidong Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shanxi 710061, China.,Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Liping Jiang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Department of Ultrasound, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Bin Zhou
- Department of Genetics, Paediatrics, and Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Institute for Aging Research, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, USA.,Department of Cardiology of First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| |
Collapse
|
22
|
Li H, Chang C, Li X, Zhang R. The roles and activation of endocardial Notch signaling in heart regeneration. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:3. [PMID: 33521843 PMCID: PMC7847831 DOI: 10.1186/s13619-020-00060-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022]
Abstract
As a highly conserved signaling pathway in metazoans, the Notch pathway plays important roles in embryonic development and tissue regeneration. Recently, cardiac injury and regeneration have become an increasingly popular topic for biomedical research, and Notch signaling has been shown to exert crucial functions during heart regeneration as well. In this review, we briefly summarize the molecular functions of the endocardial Notch pathway in several cardiac injury and stress models. Although there is an increase in appreciating the importance of endocardial Notch signaling in heart regeneration, the mechanism of its activation is not fully understood. This review highlights recent findings on the activation of the endocardial Notch pathway by hemodynamic blood flow change in larval zebrafish ventricle after partial ablation, a process involving primary cilia, mechanosensitive ion channel Trpv4 and mechanosensitive transcription factor Klf2.
Collapse
Affiliation(s)
- Huicong Li
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Cheng Chang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xueyu Li
- School of Life Sciences, Fudan University, Shanghai, China.
| | - Ruilin Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
23
|
Sharma V, Goessling LS, Brar AK, Joshi CS, Mysorekar IU, Eghtesady P. Coxsackievirus B3 Infection Early in Pregnancy Induces Congenital Heart Defects Through Suppression of Fetal Cardiomyocyte Proliferation. J Am Heart Assoc 2021; 10:e017995. [PMID: 33440998 PMCID: PMC7955305 DOI: 10.1161/jaha.120.017995] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022]
Abstract
Background Coxsackievirus B (CVB) is the most common cause of viral myocarditis. It targets cardiomyocytes through coxsackie and adenovirus receptor, which is highly expressed in the fetal heart. We hypothesized CVB3 can precipitate congenital heart defects when fetal infection occurs during critical window of gestation. Methods and Results We infected C57Bl/6 pregnant mice with CVB3 during time points in early gestation (embryonic day [E] 5, E7, E9, and E11). We used different viral titers to examine possible dose-response relationship and assessed viral loads in various fetal organs. Provided viral exposure occurred between E7 and E9, we observed characteristic features of ventricular septal defect (33.6%), abnormal myocardial architecture resembling noncompaction (23.5%), and double-outlet right ventricle (4.4%) among 209 viable fetuses examined. We observed a direct relationship between viral titers and severity of congenital heart defects, with apparent predominance among female fetuses. Infected dams remained healthy; we did not observe any maternal heart or placental injury suggestive of direct viral effects on developing heart as likely cause of congenital heart defects. We examined signaling pathways in CVB3-exposed hearts using RNA sequencing, Kyoto Encyclopedia of Genes and Genomes enrichment analysis, and immunohistochemistry. Signaling proteins of the Hippo, tight junction, transforming growth factor-β1, and extracellular matrix proteins were the most highly enriched in CVB3-infected fetuses with ventricular septal defects. Moreover, cardiomyocyte proliferation was 50% lower in fetuses with ventricular septal defects compared with uninfected controls. Conclusions We conclude prenatal CVB3 infection induces congenital heart defects. Alterations in myocardial proliferate capacity and consequent changes in cardiac architecture and trabeculation appear to account for most of observed phenotypes.
Collapse
Affiliation(s)
- Vipul Sharma
- Division of Pediatric Cardiothoracic SurgeryDepartment of SurgeryWashington University School of MedicineSt. LouisMO
| | - Lisa S. Goessling
- Division of Pediatric Cardiothoracic SurgeryDepartment of SurgeryWashington University School of MedicineSt. LouisMO
| | - Anoop K. Brar
- Division of Pediatric Cardiothoracic SurgeryDepartment of SurgeryWashington University School of MedicineSt. LouisMO
| | - Chetanchandra S. Joshi
- Department of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMO
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMO
| | - Indira U. Mysorekar
- Department of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMO
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMO
| | - Pirooz Eghtesady
- Division of Pediatric Cardiothoracic SurgeryDepartment of SurgeryWashington University School of MedicineSt. LouisMO
| |
Collapse
|
24
|
Miao Y, Tian L, Martin M, Paige SL, Galdos FX, Li J, Klein A, Zhang H, Ma N, Wei Y, Stewart M, Lee S, Moonen JR, Zhang B, Grossfeld P, Mital S, Chitayat D, Wu JC, Rabinovitch M, Nelson TJ, Nie S, Wu SM, Gu M. Intrinsic Endocardial Defects Contribute to Hypoplastic Left Heart Syndrome. Cell Stem Cell 2020; 27:574-589.e8. [PMID: 32810435 PMCID: PMC7541479 DOI: 10.1016/j.stem.2020.07.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 05/21/2020] [Accepted: 07/15/2020] [Indexed: 01/03/2023]
Abstract
Hypoplastic left heart syndrome (HLHS) is a complex congenital heart disease characterized by abnormalities in the left ventricle, associated valves, and ascending aorta. Studies have shown intrinsic myocardial defects but do not sufficiently explain developmental defects in the endocardial-derived cardiac valve, septum, and vasculature. Here, we identify a developmentally impaired endocardial population in HLHS through single-cell RNA profiling of hiPSC-derived endocardium and human fetal heart tissue with an underdeveloped left ventricle. Intrinsic endocardial defects contribute to abnormal endothelial-to-mesenchymal transition, NOTCH signaling, and extracellular matrix organization, key factors in valve formation. Endocardial abnormalities cause reduced cardiomyocyte proliferation and maturation by disrupting fibronectin-integrin signaling, consistent with recently described de novo HLHS mutations associated with abnormal endocardial gene and fibronectin regulation. Together, these results reveal a critical role for endocardium in HLHS etiology and provide a rationale for considering endocardial function in regenerative strategies.
Collapse
Affiliation(s)
- Yifei Miao
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lei Tian
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Marcy Martin
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Sharon L Paige
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Francisco X Galdos
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Jibiao Li
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alyssa Klein
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Hao Zhang
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Ning Ma
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Yuning Wei
- Center for Personal Dynamic Regulomes, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Maria Stewart
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Soah Lee
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Jan-Renier Moonen
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Bing Zhang
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Xin Hua Hospital, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Paul Grossfeld
- Department of Pediatrics, UCSD School of Medicine, La Jolla, CA 92093, USA
| | - Seema Mital
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - David Chitayat
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada; The Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Marlene Rabinovitch
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Timothy J Nelson
- Division of General Internal Medicine, Division of Pediatric Cardiology, and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Shuyi Nie
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Sean M Wu
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Mingxia Gu
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
25
|
New Concepts in the Development and Malformation of the Arterial Valves. J Cardiovasc Dev Dis 2020; 7:jcdd7040038. [PMID: 32987700 PMCID: PMC7712390 DOI: 10.3390/jcdd7040038] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022] Open
Abstract
Although in many ways the arterial and atrioventricular valves are similar, both being derived for the most part from endocardial cushions, we now know that the arterial valves and their surrounding structures are uniquely dependent on progenitors from both the second heart field (SHF) and neural crest cells (NCC). Here, we will review aspects of arterial valve development, highlighting how our appreciation of NCC and the discovery of the SHF have altered our developmental models. We will highlight areas of research that have been particularly instructive for understanding how the leaflets form and remodel, as well as those with limited or conflicting results. With this background, we will explore how this developmental knowledge can help us to understand human valve malformations, particularly those of the bicuspid aortic valve (BAV). Controversies and the current state of valve genomics will be indicated.
Collapse
|
26
|
Andrés-Delgado L, Galardi-Castilla M, Münch J, Peralta M, Ernst A, González-Rosa JM, Tessadori F, Santamaría L, Bakkers J, Vermot J, de la Pompa JL, Mercader N. Notch and Bmp signaling pathways act coordinately during the formation of the proepicardium. Dev Dyn 2020; 249:1455-1469. [PMID: 33103836 PMCID: PMC7754311 DOI: 10.1002/dvdy.229] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The epicardium is the outer mesothelial layer of the heart. It encloses the myocardium and plays key roles in heart development and regeneration. It derives from the proepicardium (PE), cell clusters that appear in the dorsal pericardium (DP) close to the atrioventricular canal and the venous pole of the heart, and are released into the pericardial cavity. PE cells are advected around the beating heart until they attach to the myocardium. Bmp and Notch signaling influence PE formation, but it is unclear how both signaling pathways interact during this process in the zebrafish. RESULTS Here, we show that the developing PE is influenced by Notch signaling derived from the endothelium. Overexpression of the intracellular receptor of notch in the endothelium enhances bmp expression, increases the number of pSmad1/5 positive cells in the DP and PE, and enhances PE formation. On the contrary, pharmacological inhibition of Notch1 impairs PE formation. bmp2b overexpression can rescue loss of PE formation in the presence of a Notch1 inhibitor, but Notch gain-of-function could not recover PE formation in the absence of Bmp signaling. CONCLUSIONS Endothelial Notch signaling activates bmp expression in the heart tube, which in turn induces PE cluster formation from the DP layer.
Collapse
Affiliation(s)
- Laura Andrés-Delgado
- Development of the Epicardium and its Role During Regeneration Laboratory, National Center of Cardiovascular Research Carlos III, Madrid, Spain.,Department of Anatomy, Histology, and Neuroscience, School of Medicine, Autonoma University of Madrid, Madrid, Spain
| | - María Galardi-Castilla
- Development of the Epicardium and its Role During Regeneration Laboratory, National Center of Cardiovascular Research Carlos III, Madrid, Spain
| | - Juliane Münch
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, National Center of Cardiovascular Research Carlos III, Madrid, Spain.,Ciber CV, Madrid, Spain.,Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
| | - Marina Peralta
- Development of the Epicardium and its Role During Regeneration Laboratory, National Center of Cardiovascular Research Carlos III, Madrid, Spain.,Institute of Genetics and Molecular and Cellular Biology (IGBMC), Illkirch, France.,Australian Regenerative Institute, Monash University, Clayton, Victoria, Australia
| | | | - Juan Manuel González-Rosa
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Luis Santamaría
- Department of Anatomy, Histology, and Neuroscience, School of Medicine, Autonoma University of Madrid, Madrid, Spain
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and UMC Utrecht, Utrecht, The Netherlands.,Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands
| | - Julien Vermot
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Illkirch, France.,Department of Bioengineering, Imperial College London, London, UK
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, National Center of Cardiovascular Research Carlos III, Madrid, Spain.,Ciber CV, Madrid, Spain
| | - Nadia Mercader
- Development of the Epicardium and its Role During Regeneration Laboratory, National Center of Cardiovascular Research Carlos III, Madrid, Spain.,Institute of Anatomy, University of Bern, Bern, Switzerland
| |
Collapse
|
27
|
Biomechanical Cues Direct Valvulogenesis. J Cardiovasc Dev Dis 2020; 7:jcdd7020018. [PMID: 32438610 PMCID: PMC7345189 DOI: 10.3390/jcdd7020018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/27/2020] [Accepted: 05/12/2020] [Indexed: 12/30/2022] Open
Abstract
The vertebrate embryonic heart initially forms with two chambers, a ventricle and an atrium, separated by the atrioventricular junction. Localized genetic and biomechanical information guides the development of valves, which function to ensure unidirectional blood flow. If the valve development process goes awry, pathology associated with congenital valve defects can ensue. Congenital valve defects (CVD) are estimated to affect 1–2% of the population and can often require a lifetime of treatment. Despite significant clinical interest, molecular genetic mechanisms that direct valve development remain incompletely elucidated. Cells in the developing valve must contend with a dynamic hemodynamic environment. A growing body of research supports the idea that cells in the valve are highly sensitive to biomechanical forces, which cue changes in gene expression required for normal development or for maintenance of the adult valve. This review will focus on mechanotransductive pathways involved in valve development across model species. We highlight current knowledge regarding how cells sense physical forces associated with blood flow and pressure in the forming heart, and summarize how these changes are transduced into genetic and developmental responses. Lastly, we provide perspectives on how altered biomechanical cues may lead to CVD pathogenesis.
Collapse
|
28
|
Lan Y, Pan H, Li C, Banks KM, Sam J, Ding B, Elemento O, Goll MG, Evans T. TETs Regulate Proepicardial Cell Migration through Extracellular Matrix Organization during Zebrafish Cardiogenesis. Cell Rep 2020; 26:720-732.e4. [PMID: 30650362 PMCID: PMC6366638 DOI: 10.1016/j.celrep.2018.12.076] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 10/30/2018] [Accepted: 12/18/2018] [Indexed: 12/22/2022] Open
Abstract
Ten-eleven translocation (Tet) enzymes (Tet1/2/3) mediate 5-methylcytosine (5mC) hydroxylation, which can facilitate DNA demethylation and thereby impact gene expression. Studied mostly for how mutant isoforms impact cancer, the normal roles for Tet enzymes during organogenesis are largely unknown. By analyzing compound mutant zebrafish, we discovered a requirement for Tet2/3 activity in the embryonic heart for recruitment of epicardial progenitors, associated with development of the atrial-ventricular canal (AVC). Through a combination of methylation, hydroxymethylation, and transcript profiling, the genes encoding the activin A subunit Inhbaa (in endocardium) and Sox9b (in myocardium) were implicated as demethylation targets of Tet2/3 and critical for organization of AVC-localized extracellular matrix (ECM), facilitating migration of epicardial progenitors onto the developing heart tube. This study elucidates essential DNA demethylation modifications that govern gene expression changes during cardiac development with striking temporal and lineage specificities, highlighting complex interactions in multiple cell populations during development of the vertebrate heart. Lan et al. show that zebrafish larvae mutant for tet2 and tet3 fail to demethylate genes encoding Inhbaa (in endocardium) and Sox9b (in myocardium), leading to defects in ECM needed to form valves and to recruit epicardial progenitors onto the heart tube.
Collapse
Affiliation(s)
- Yahui Lan
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Heng Pan
- Department of Physiology and Biophysics, Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Cheng Li
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Program in Biochemistry and Structural Biology, Cell and Developmental Biology, and Molecular Biology, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Kelly M Banks
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jessica Sam
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Bo Ding
- Bonacept, LLC, 7699 Palmilla Drive, Apt. 3312, San Diego, CA 92122, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mary G Goll
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
29
|
Kong J, Wang W. A Systemic Review on the Regulatory Roles of miR-34a in Gastrointestinal Cancer. Onco Targets Ther 2020; 13:2855-2872. [PMID: 32308419 PMCID: PMC7138617 DOI: 10.2147/ott.s234549] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/22/2019] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of endogenous non-coding single-stranded small-molecule RNAs that regulate gene expression by repressing target messenger RNA (mRNA) translation or degrading mRNA. miR-34a is one of the most important miRNAs participating in various physiological and pathological processes. miR-34a is abnormally expressed in a variety of tumors. The roles of miR-34a in gastrointestinal cancer (GIC) draw lots of attention. Numerous studies have demonstrated that dysregulated miR-34a is closely related to the proliferation, differentiation, migration, and invasion of tumor cells, as well as the diagnosis, prognosis, treatment, and chemo-resistance of tumors. Thus, we systematically reviewed the abnormal expression and regulatory roles of miR-34a in GICs including esophageal cancer (EC), gastric cancer (GC), colorectal cancer (CRC), hepatocellular carcinoma (HCC), pancreatic cancer (PC), and gallbladder cancer (GBC). It may provide a profile of versatile roles of miR-34a in GICs.
Collapse
Affiliation(s)
- Jiehong Kong
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Weipeng Wang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| |
Collapse
|
30
|
Flinn MA, Link BA, O'Meara CC. Upstream regulation of the Hippo-Yap pathway in cardiomyocyte regeneration. Semin Cell Dev Biol 2019; 100:11-19. [PMID: 31606277 DOI: 10.1016/j.semcdb.2019.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/27/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022]
Abstract
The response of the adult mammalian heart to injury such as myocardial infarction has long been described as primarily fibrotic scarring and adverse remodeling with little to no regeneration of cardiomyocytes. Emerging studies have challenged this paradigm by demonstrating that, indeed, adult mammalian cardiomyocytes are capable of completing cytokinesis albeit at levels vastly insufficient to compensate for the loss of functional cardiomyocytes following ischemic injury. Thus, there is great interest in identifying mechanisms to guide adult cardiomyocyte cell cycle re-entry and facilitate endogenous heart regeneration. The Hippo signaling pathway is a core kinase cascade that functions to suppress the transcriptional co-activators Yap and Taz by phosphorylation and therefore cytoplasmic retention or phospho-degradation. This pathway has recently sparked interest in the field of cardiac regeneration as inhibition of Hippo kinase signaling or overdriving the transcriptional co-activator, Yap, significantly promotes proliferation of terminally differentiated adult mammalian cardiomyocytes and can restore function in failing mouse hearts. Thus, the Hippo pathway is an attractive therapeutic target for promoting cardiomyocyte renewal and cardiac regeneration. Although the core kinases and transcriptional activators of the Hippo pathway have been studied extensively over the last twenty years, the regulatory inputs of this pathway, particularly in vertebrates, are poorly understood. Recent studies have elucidated several upstream regulatory inputs to the Hippo pathway in adult mammalian cardiomyocytes that influence cell proliferation and heart regeneration. Considering upstream inputs to the Hippo pathway are thought to be context and cell type specific, targeting these various components could serve as a therapeutic approach for refining Hippo-Yap signaling in the heart. Here, we provide an overview of the emerging regulatory inputs to the Hippo pathway as they relate to mammalian cardiomyocytes and heart regeneration.
Collapse
Affiliation(s)
- Michael A Flinn
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian A Link
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Caitlin C O'Meara
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Genomics Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
31
|
Abstract
Cardiogenesis is a complex developmental process involving multiple overlapping stages of cell fate specification, proliferation, differentiation, and morphogenesis. Precise spatiotemporal coordination between the different cardiogenic processes is ensured by intercellular signalling crosstalk and tissue-tissue interactions. Notch is an intercellular signalling pathway crucial for cell fate decisions during multicellular organismal development and is aptly positioned to coordinate the complex signalling crosstalk required for progressive cell lineage restriction during cardiogenesis. In this Review, we describe the role of Notch signalling and the crosstalk with other signalling pathways during the differentiation and patterning of the different cardiac tissues and in cardiac valve and ventricular chamber development. We examine how perturbation of Notch signalling activity is linked to congenital heart diseases affecting the neonate and adult, and discuss studies that shed light on the role of Notch signalling in heart regeneration and repair after injury.
Collapse
|
32
|
Epigenetics and Mechanobiology in Heart Development and Congenital Heart Disease. Diseases 2019; 7:diseases7030052. [PMID: 31480510 PMCID: PMC6787645 DOI: 10.3390/diseases7030052] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/30/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
: Congenital heart disease (CHD) is the most common birth defect worldwide and the number one killer of live-born infants in the United States. Heart development occurs early in embryogenesis and involves complex interactions between multiple cell populations, limiting the understanding and consequent treatment of CHD. Furthermore, genome sequencing has largely failed to predict or yield therapeutics for CHD. In addition to the underlying genome, epigenetics and mechanobiology both drive heart development. A growing body of evidence implicates the aberrant regulation of these two extra-genomic systems in the pathogenesis of CHD. In this review, we describe the stages of human heart development and the heart defects known to manifest at each stage. Next, we discuss the distinct and overlapping roles of epigenetics and mechanobiology in normal development and in the pathogenesis of CHD. Finally, we highlight recent advances in the identification of novel epigenetic biomarkers and environmental risk factors that may be useful for improved diagnosis and further elucidation of CHD etiology.
Collapse
|
33
|
Zhang L, Hao C, Wu Y, Zhu Y, Ren Y, Tong Z. Microcalcification and BMP-2 in breast cancer: correlation with clinicopathological features and outcomes. Onco Targets Ther 2019; 12:2023-2033. [PMID: 30936719 PMCID: PMC6421899 DOI: 10.2147/ott.s187835] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Microcalcification is a very important diagnostic information in breast cancer. The purpose of this study was to determine the relationship of clinicopathological features and prognosis of breast cancer with microcalcification and to detect biomarkers related to the possible mechanisms of microcalcifications. Patients and methods All 529 subjects with microcalcifications were selected from patients who had been examined using breast mammography. The control group did not have detectable microcalcifications, and was matched in a ratio of 1:3. The clinicopathological factors, progression-free survival (PFS), and overall survival were evaluated by SPSS. Results There was a significant difference in tumor size between the two groups, with larger tumors in the calcification group than the control group, and the proportion of patients in the calcification group with tumors of >5 cm was 20.4% vs 17.2% in the control group (P=0.041). The proportion of patients with lymph node metastasis in the calcification group was higher than that of the control group (35% vs 27.9%, P=0.027). The recurrence rate in ductal carcinoma in situ (DCIS) and invasive ductal carcinoma (IDC) patients with microcalcification was higher than that in the control group (P=0.035 and 0.044). BMP-2 expression was higher in breast cancer tissues, especially in breast cancer tissues with microcalcifications. The recurrence rate in the BMP-2(+) group was higher than that in the BMP-2(-) group both in DCIS and IDC (P=0.044 and 0.049). Microcalcifications and the positive expression of BMP-2 were independent factors affecting the PFS of the breast cancer patients. Conclusion Through the analysis of this study, it was found that the prognosis of the patients with microcalcification was relatively poor. BMP-2 was highly expressed in the breast cancer with microcalcification and was associated with poor prognosis.
Collapse
Affiliation(s)
- Li Zhang
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China,
| | - Chunfang Hao
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China,
| | - Yansheng Wu
- Department of Maxillofacial and Otorhinolaryngology Head and Neck Surgery, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
| | - Yuying Zhu
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China,
| | - Yulin Ren
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China,
| | - Zhongsheng Tong
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China,
| |
Collapse
|
34
|
Gunawan F, Gentile A, Fukuda R, Tsedeke AT, Jiménez-Amilburu V, Ramadass R, Iida A, Sehara-Fujisawa A, Stainier DYR. Focal adhesions are essential to drive zebrafish heart valve morphogenesis. J Cell Biol 2019; 218:1039-1054. [PMID: 30635353 PMCID: PMC6400548 DOI: 10.1083/jcb.201807175] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/07/2018] [Accepted: 12/13/2018] [Indexed: 12/17/2022] Open
Abstract
Gunawan et al. analyze at single-cell resolution collective endocardial cell migration into the extracellular matrix and the cellular rearrangements forming leaflets during zebrafish heart valve formation. They show that focal adhesion activity driven by Integrin α5β1 and Talin1 are essential to drive cardiac valve morphogenesis in zebrafish. Elucidating the morphogenetic events that shape vertebrate heart valves, complex structures that prevent retrograde blood flow, is critical to understanding valvular development and aberrations. Here, we used the zebrafish atrioventricular (AV) valve to investigate these events in real time and at single-cell resolution. We report the initial events of collective migration of AV endocardial cells (ECs) into the extracellular matrix (ECM), and their subsequent rearrangements to form the leaflets. We functionally characterize integrin-based focal adhesions (FAs), critical mediators of cell–ECM interactions, during valve morphogenesis. Using transgenes to block FA signaling specifically in AV ECs as well as loss-of-function approaches, we show that FA signaling mediated by Integrin α5β1 and Talin1 promotes AV EC migration and overall shaping of the valve leaflets. Altogether, our investigation reveals the critical processes driving cardiac valve morphogenesis in vivo and establishes the zebrafish AV valve as a vertebrate model to study FA-regulated tissue morphogenesis.
Collapse
Affiliation(s)
- Felix Gunawan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Alessandra Gentile
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ryuichi Fukuda
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ayele Taddese Tsedeke
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Vanesa Jiménez-Amilburu
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Radhan Ramadass
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Atsuo Iida
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | | | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
35
|
Myocardial Notch1-Rbpj deletion does not affect NOTCH signaling, heart development or function. PLoS One 2018; 13:e0203100. [PMID: 30596653 PMCID: PMC6312338 DOI: 10.1371/journal.pone.0203100] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/11/2018] [Indexed: 01/09/2023] Open
Abstract
During vertebrate cardiac development NOTCH signaling activity in the endocardium is essential for the crosstalk between endocardium and myocardium that initiates ventricular trabeculation and valve primordium formation. This crosstalk leads later to the maturation and compaction of the ventricular chambers and the morphogenesis of the cardiac valves, and its alteration may lead to disease. Although endocardial NOTCH signaling has been shown to be crucial for heart development, its physiological role in the myocardium has not been clearly established. Here we have used mouse genetics to evaluate the role of NOTCH in myocardial development. We have inactivated the unique and ubiquitous NOTCH effector RBPJ in early cardiomyocytes progenitors, and examined its consequences in cardiac development and function. Our results show that mice with Tnnt2-Cre-mediated myocardial-specific deletion of Rbpj develop to term, with homozygous mutant animals showing normal expression of cardiac development markers, and normal adult heart function. Similar observations have been obtained after Notch1 deletion with Tnnt2-Cre. We have also deleted Rbpj in both myocardial and endocardial progenitor cells, using the Nkx2.5-Cre driver, resulting in ventricular septal defect (VSD), double outlet right ventricle (DORV), and bicuspid aortic valve (BAV), due to NOTCH signaling abrogation in the endocardium of cardiac valves. Our data demonstrate that NOTCH-RBPJ inactivation in the myocardium does not affect heart development or adult cardiac function.
Collapse
|
36
|
Bhakta M, Padanad MS, Harris JP, Lubczyk C, Amatruda JF, Munshi NV. pouC Regulates Expression of bmp4 During Atrioventricular Canal Formation in Zebrafish. Dev Dyn 2018; 248:173-188. [PMID: 30444277 DOI: 10.1002/dvdy.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/04/2018] [Accepted: 10/24/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Many human gene mutations have been linked to congenital heart disease (CHD), yet CHD remains a major health issue worldwide due in part to an incomplete understanding of the molecular basis for cardiac malformation. RESULTS Here we identify the orthologous mouse Pou6f1 and zebrafish pouC as POU homeodomain transcription factors enriched in the developing heart. We find that pouC is a multi-functional transcriptional regulator containing separable activation, repression, protein-protein interaction, and DNA binding domains. Using zebrafish heart development as a model system, we demonstrate that pouC knockdown impairs cardiac morphogenesis and affects cardiovascular function. We also find that levels of pouC expression must be fine-tuned to enable proper heart formation. At the cellular level, we demonstrate that pouC knockdown disrupts atrioventricular canal (AVC) cardiomyocyte maintenance, although chamber myocyte specification remains intact. Mechanistically, we show that pouC binds a bmp4 intronic regulatory element to mediate transcriptional activation. CONCLUSIONS Taken together, our study establishes pouC as a novel transcriptional input into the regulatory hierarchy that drives AVC morphogenesis in zebrafish. We anticipate that these findings will inform future efforts to explore functional conservation in mammals and potential association with atrioventricular septal defects in humans. Developmental Dynamics 248:173-188, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Minoti Bhakta
- Department of Internal Medicine - Cardiology, UT Southwestern Medical Center, Dallas, Texas
| | - Mahesh S Padanad
- Department of Internal Medicine - Cardiology, UT Southwestern Medical Center, Dallas, Texas
| | - John P Harris
- Department of Internal Medicine - Cardiology, UT Southwestern Medical Center, Dallas, Texas
| | - Christina Lubczyk
- Department of Internal Medicine - Cardiology, UT Southwestern Medical Center, Dallas, Texas
| | - James F Amatruda
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas.,Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas
| | - Nikhil V Munshi
- Department of Internal Medicine - Cardiology, UT Southwestern Medical Center, Dallas, Texas.,Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas.,McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas.,Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
37
|
Wang Y, Lu P, Wu B, Morrow BE, Zhou B. NOTCH maintains developmental cardiac gene network through WNT5A. J Mol Cell Cardiol 2018; 125:98-105. [PMID: 30347193 DOI: 10.1016/j.yjmcc.2018.10.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/19/2018] [Accepted: 10/15/2018] [Indexed: 11/25/2022]
Abstract
NOTCH and WNT signaling pathways play critical roles in cardiac chamber formation. Here we explored the potential interactions between the two pathways in this developmental process by using genetically modified mouse models and whole embryo culture systems. By deletion of Notch1 to inactivate NOTCH1 signaling in the endocardium in vivo and ex vivo rescue experiments, we showed that myocardial WNT5A mediated endocardial NOTCH1 signaling to maintain the gene regulatory network essential for cardiac chamber formation. Furthermore, genetic deletion of β-catenin in the myocardium and inhibition of the WNT/Ca2+ signaling by FK506 resulted in a similar disruption of the gene regulatory network as inactivation of endocardial NOTCH1 signaling. Together, these findings identify WNT5A as a key myocardial factor that mediates the endocardial NOTCH signaling to maintain the gene regulatory network essential for cardiac chamber formation through WNT/β-catenin and WNT/Ca2+ signaling pathways.
Collapse
Affiliation(s)
- Yidong Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China; Department of Genetics, Albert Einstein College of Medicine, New York 10461, USA.
| | - Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, New York 10461, USA
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, New York 10461, USA
| | - Bernice E Morrow
- Departments of Genetics, Obstetrics & Gynecology, and Pediatrics, Albert Einstein College of Medicine, Wilf Cardiovascular Research Institute, New York 10461, USA
| | - Bin Zhou
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Albert Einstein College of Medicine, Institute for Aging Research, Wilf Cardiovascular Research Institute, New York 10461, USA; Department of Cardiology of First Affiliated Hospital, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
38
|
Wang Y, Lu P, Wu B, Riascos-Bernal DF, Sibinga NES, Valenta T, Basler K, Zhou B. Myocardial β-Catenin-BMP2 signaling promotes mesenchymal cell proliferation during endocardial cushion formation. J Mol Cell Cardiol 2018; 123:150-158. [PMID: 30201295 PMCID: PMC10662972 DOI: 10.1016/j.yjmcc.2018.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/02/2018] [Accepted: 09/01/2018] [Indexed: 01/09/2023]
Abstract
Abnormal endocardial cushion formation is a major cause of congenital heart valve disease, which is a common birth defect with significant morbidity and mortality. Although β-catenin and BMP2 are two well-known regulators of endocardial cushion formation, their interaction in this process is largely unknown. Here, we report that deletion of β-catenin in myocardium results in formation of hypoplastic endocardial cushions accompanying a decrease of mesenchymal cell proliferation. Loss of β-catenin reduced Bmp2 expression in myocardium and SMAD signaling in cushion mesenchyme. Exogenous BMP2 recombinant proteins fully rescued the proliferation defect of mesenchymal cells in cultured heart explants from myocardial β-catenin knockout embryos. Using a canonical WNT signaling reporter mouse line, we showed that cushion myocardium exhibited high WNT/β-catenin activities during endocardial cushion growth. Selective disruption of the signaling function of β-catenin resulted in a cushion growth defect similar to that caused by the complete loss of β-catenin. Together, these observations demonstrate that myocardial β-catenin signaling function promotes mesenchymal cell proliferation and endocardial cushion expansion through inducing BMP signaling.
Collapse
Affiliation(s)
- Yidong Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China; Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States.
| | - Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States
| | - Dario F Riascos-Bernal
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, United States
| | - Nicholas E S Sibinga
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, United States
| | - Tomas Valenta
- Institute of Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
| | - Konrad Basler
- Institute of Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States; Department of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York 10461, United States; Department of Cardiology, First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
39
|
Huang S, Park J, Qiu C, Chung KW, Li SY, Sirin Y, Han SH, Taylor V, Zimber-Strobl U, Susztak K. Jagged1/Notch2 controls kidney fibrosis via Tfam-mediated metabolic reprogramming. PLoS Biol 2018; 16:e2005233. [PMID: 30226866 PMCID: PMC6161902 DOI: 10.1371/journal.pbio.2005233] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 09/28/2018] [Accepted: 09/03/2018] [Indexed: 12/14/2022] Open
Abstract
While Notch signaling has been proposed to play a key role in fibrosis, the direct molecular pathways targeted by Notch signaling and the precise ligand and receptor pair that are responsible for kidney disease remain poorly defined. In this study, we found that JAG1 and NOTCH2 showed the strongest correlation with the degree of interstitial fibrosis in a genome-wide expression analysis of a large cohort of human kidney samples. Transcript analysis of mouse kidney disease models, including folic-acid (FA)-induced nephropathy, unilateral ureteral obstruction (UUO), or apolipoprotein L1 (APOL1)-associated kidney disease, indicated that Jag1 and Notch2 levels were higher in all analyzed kidney fibrosis models. Mice with tubule-specific deletion of Jag1 or Notch2 (Kspcre/Jag1flox/flox and Kspcre/Notch2flox/flox) had no kidney-specific alterations at baseline but showed protection from FA-induced kidney fibrosis. Tubule-specific genetic deletion of Notch1 and global knockout of Notch3 had no effect on fibrosis. In vitro chromatin immunoprecipitation experiments and genome-wide expression studies identified the mitochondrial transcription factor A (Tfam) as a direct Notch target. Re-expression of Tfam in tubule cells prevented Notch-induced metabolic and profibrotic reprogramming. Tubule-specific deletion of Tfam resulted in fibrosis. In summary, Jag1 and Notch2 play a key role in kidney fibrosis development by regulating Tfam expression and metabolic reprogramming.
Collapse
Affiliation(s)
- Shizheng Huang
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jihwan Park
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Chengxiang Qiu
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ki Wung Chung
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Szu-yuan Li
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Yasemin Sirin
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Seung Hyeok Han
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Ursula Zimber-Strobl
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environment and Health, Munich, Germany
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
40
|
Cai C, Sang C, Du J, Jia H, Tu J, Wan Q, Bao B, Xie S, Huang Y, Li A, Li J, Yang K, Wang S, Lu Q. Knockout of tnni1b in zebrafish causes defects in atrioventricular valve development via the inhibition of the myocardial wnt signaling pathway. FASEB J 2018; 33:696-710. [PMID: 30044923 DOI: 10.1096/fj.201800481rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The proper development of atrioventricular (AV) valves is critical for heart morphogenesis and for the formation of the cardiac conduction system. Defects in AV valve development are the most common type of congenital heart defect. Cardiac troponin I ( ctnni), a structural and regulatory protein involved in cardiac muscle contraction, is a subunit of the troponin complex, but the functions and molecular mechanisms of ctnni during early heart development remain unclear. We created a knockout zebrafish model in which troponin I type 1b ( tnni1b) ( Tnni-HC, heart and craniofacial) was deleted using the clustered regularly interspaced short palindromic repeat/clustered regularly interspaced short palindromic repeat-associated protein system. In the homozygous mutant, the embryos showed severe pericardial edema, malformation of the heart tube, reduction of heart rate without contraction and with almost no blood flow, heart cavity congestion, and lack of an endocardial ring or valve leaflet, resulting in 88.8 ± 6.0% lethality at 7 d postfertilization. Deletion of tnni1b caused the abnormal expression of several markers involved in AV valve development, including bmp4, cspg2, has2, notch1b, spp1, and Alcam. Myocardial re-expression of tnni1b in mutants partially rescued the pericardial edema phenotype and AV canal (AVC) developmental defects. We further showed that tnni1b knockout in zebrafish and ctnni knockdown in rat h9c2 myocardial cells inhibited cardiac wnt signaling and that myocardial reactivation of wnt signaling partially rescued the abnormal expression of AVC markers caused by the tnni1b deletion. Taken together, our data suggest that tnni1b plays a vital role in zebrafish AV valve development by regulating the myocardial wnt signaling pathway.-Cai, C., Sang, C., Du, J., Jia, H., Tu, J., Wan, Q., Bao, B., Xie, S., Huang, Y., Li, A., Li, J., Yang, K., Wang, S., Lu, Q. Knockout of tnni1b in zebrafish causes defects in atrioventricular valve development via the inhibition of myocardial wnt signaling pathway.
Collapse
Affiliation(s)
- Chen Cai
- Key Laboratory of Molecular Biophysics, Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Caijun Sang
- Key Laboratory of Molecular Biophysics, Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Du
- School Hospital, Huazhong University of Science and Technology, Wuhan, China; and
| | - Haibo Jia
- Key Laboratory of Molecular Biophysics, Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Jiayi Tu
- Key Laboratory of Molecular Biophysics, Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Wan
- Key Laboratory of Molecular Biophysics, Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Binghao Bao
- Key Laboratory of Molecular Biophysics, Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Shanglun Xie
- Key Laboratory of Molecular Biophysics, Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Huang
- Key Laboratory of Molecular Biophysics, Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Ao Li
- Key Laboratory of Molecular Biophysics, Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Jiayu Li
- Key Laboratory of Molecular Biophysics, Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Yang
- Exercise Immunology Center, Wuhan Sports University, Wuhan, China
| | - Song Wang
- Exercise Immunology Center, Wuhan Sports University, Wuhan, China
| | - Qunwei Lu
- Key Laboratory of Molecular Biophysics, Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
41
|
Segert J, Schneider I, Berger IM, Rottbauer W, Just S. Mediator complex subunit Med12 regulates cardiac jelly development and AV valve formation in zebrafish. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 138:20-31. [PMID: 30036562 DOI: 10.1016/j.pbiomolbio.2018.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/30/2018] [Accepted: 07/17/2018] [Indexed: 11/25/2022]
Abstract
The molecular mechanism essential for the formation of heart valves involves complex interactions of signaling molecules and transcription factors. The Mediator Complex (MC) functions as multi-subunit machinery to orchestrate gene transcription, especially for tissue-specific fine-tuning of transcriptional processes during development, also in the heart. Here, we analyzed the role of the MC subunit Med12 during atrioventricular canal (AVC) development and endocardial cushion formation, using the Med12-deficient zebrafish mutant trapped (tpd). Whereas primary heart formation was only slightly affected in tpd, we identified defects in AVC development and cardiac jelly formation. We found that although misexpression of bmp4 and versican in tpd hearts can be restored by overexpression of a modified version of the Sox9b transcription factor (harboring VP16 transactivation domain) that functions independent of its co-activator Med12, endocardial cushion development in tpd was not reconstituted. Interestingly, expression of tbx2b and its target hyaluronan synthase 2 (has2) - the synthase of hyaluronan (HA) in the heart - was absent in both uninjected and Sox9b-VP16 overexpressing tpd hearts. HA is a major ECM component of the cardiac jelly and required for endocardial cushion formation. Furthermore, we found secreted phosphoprotein 1 (spp1), an endocardial marker of activated AV endocardial cells, completely absent in tpd hearts, suggesting that crucial steps of the transformation of AV endocardial cells into endocardial cushions is blocked. We demonstrate that Med12 controls cardiac jelly formation Sox9-independently by regulating tbx2b and has2 expression and therefore the production of the glycosaminoglycan HA at the AVC to guarantee proper endocardial cushion development.
Collapse
Affiliation(s)
- Julia Segert
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Isabelle Schneider
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Ina M Berger
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | | | - Steffen Just
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany.
| |
Collapse
|
42
|
Eley L, Alqahtani AM, MacGrogan D, Richardson RV, Murphy L, Salguero-Jimenez A, Sintes Rodriguez San Pedro M, Tiurma S, McCutcheon L, Gilmore A, de La Pompa JL, Chaudhry B, Henderson DJ. A novel source of arterial valve cells linked to bicuspid aortic valve without raphe in mice. eLife 2018; 7:34110. [PMID: 29956664 PMCID: PMC6025960 DOI: 10.7554/elife.34110] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 06/12/2018] [Indexed: 12/20/2022] Open
Abstract
Abnormalities of the arterial valve leaflets, predominantly bicuspid aortic valve, are the commonest congenital malformations. Although many studies have investigated the development of the arterial valves, it has been assumed that, as with the atrioventricular valves, endocardial to mesenchymal transition (EndMT) is the predominant mechanism. We show that arterial is distinctly different from atrioventricular valve formation. Whilst the four septal valve leaflets are dominated by NCC and EndMT-derived cells, the intercalated leaflets differentiate directly from Tnnt2-Cre+/Isl1+ progenitors in the outflow wall, via a Notch-Jag dependent mechanism. Further, when this novel group of progenitors are disrupted, development of the intercalated leaflets is disrupted, resulting in leaflet dysplasia and bicuspid valves without raphe, most commonly affecting the aortic valve. This study thus overturns the dogma that heart valves are formed principally by EndMT, identifies a new source of valve interstitial cells, and provides a novel mechanism for causation of bicuspid aortic valves without raphe.
Collapse
Affiliation(s)
- Lorriane Eley
- Institute of Genetic Medicine, Cardiovascular Research Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ahlam Ms Alqahtani
- Institute of Genetic Medicine, Cardiovascular Research Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Donal MacGrogan
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Rachel V Richardson
- Institute of Genetic Medicine, Cardiovascular Research Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lindsay Murphy
- Institute of Genetic Medicine, Cardiovascular Research Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Alejandro Salguero-Jimenez
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Shindi Tiurma
- Institute of Genetic Medicine, Cardiovascular Research Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lauren McCutcheon
- Institute of Genetic Medicine, Cardiovascular Research Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Adam Gilmore
- Institute of Genetic Medicine, Cardiovascular Research Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - José Luis de La Pompa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Bill Chaudhry
- Institute of Genetic Medicine, Cardiovascular Research Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Deborah J Henderson
- Institute of Genetic Medicine, Cardiovascular Research Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
43
|
Wang Y, Wu B, Farrar E, Lui W, Lu P, Zhang D, Alfieri CM, Mao K, Chu M, Yang D, Xu D, Rauchman M, Taylor V, Conway SJ, Yutzey KE, Butcher JT, Zhou B. Notch-Tnf signalling is required for development and homeostasis of arterial valves. Eur Heart J 2018; 38:675-686. [PMID: 26491108 DOI: 10.1093/eurheartj/ehv520] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 09/15/2015] [Indexed: 01/04/2023] Open
Abstract
Aims Congenital anomalies of arterial valves are common birth defects, leading to valvar stenosis. With no pharmaceutical treatment that can prevent the disease progression, prosthetic replacement is the only choice of treatment, incurring considerable morbidity and mortality. Animal models presenting localized anomalies and stenosis of congenital arterial valves similar to that of humans are critically needed research tools to uncover developmental molecular mechanisms underlying this devastating human condition. Methods and results We generated and characterized mouse models with conditionally altered Notch signalling in endothelial or interstitial cells of developing valves. Mice with inactivation of Notch1 signalling in valvar endothelial cells (VEC) developed congenital anomalies of arterial valves including bicuspid aortic valves and valvar stenosis. Notch1 signalling in VEC was required for repressing proliferation and activating apoptosis of valvar interstitial cells (VIC) after endocardial-to-mesenchymal transformation (EMT). We showed that Notch signalling regulated Tnfα expression in vivo, and Tnf signalling was necessary for apoptosis of VIC and post-EMT development of arterial valves. Furthermore, activation or inhibition of Notch signalling in cultured pig aortic VEC-promoted or suppressed apoptosis of VIC, respectively. Conclusion We have now met the need of critical animal models and shown that Notch-Tnf signalling balances proliferation and apoptosis for post-EMT development of arterial valves. Our results suggest that mutations in its components may lead to congenital anomaly of aortic valves and valvar stenosis in humans.
Collapse
Affiliation(s)
- Yidong Wang
- Department of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Price 420, 1301 Morris Park Avenue, Bronx, NY 14061, USA
| | - Bingruo Wu
- Department of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Price 420, 1301 Morris Park Avenue, Bronx, NY 14061, USA
| | - Emily Farrar
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Wendy Lui
- Department of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Price 420, 1301 Morris Park Avenue, Bronx, NY 14061, USA
| | - Pengfei Lu
- Department of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Price 420, 1301 Morris Park Avenue, Bronx, NY 14061, USA
| | - Donghong Zhang
- Department of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Price 420, 1301 Morris Park Avenue, Bronx, NY 14061, USA
| | - Christina M Alfieri
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Medical Centre, Cincinnati, OH, USA
| | - Kai Mao
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ming Chu
- Department of Medicine and Geriatrics (Cardiology), First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Di Yang
- Department of Medicine and Geriatrics (Cardiology), First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Di Xu
- Department of Medicine and Geriatrics (Cardiology), First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Michael Rauchman
- Department of Internal Medicine, Saint Louis University, St. Louis, MO, USA
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Simon J Conway
- Department of Paediatrics, Indiana University, Indianapolis, IN, USA
| | - Katherine E Yutzey
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Medical Centre, Cincinnati, OH, USA
| | - Jonathan T Butcher
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Bin Zhou
- Department of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Price 420, 1301 Morris Park Avenue, Bronx, NY 14061, USA.,Department of Medicine and Geriatrics (Cardiology), First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
44
|
Peng C, Shen J, Lin X, Su KJ, Greenbaum J, Zhu W, Lou HL, Liu F, Zeng CP, Deng WF, Deng HW. Genetic sharing with coronary artery disease identifies potential novel loci for bone mineral density. Bone 2017; 103:70-77. [PMID: 28651948 PMCID: PMC5796548 DOI: 10.1016/j.bone.2017.06.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 12/30/2022]
Abstract
Bone mineral density (BMD) is a complex trait with high missing heritability. Numerous evidences have shown that BMD variation has a relationship with coronary artery disease (CAD). This relationship may come from a common genetic basis called pleiotropy. By leveraging the pleiotropy with CAD, we may be able to improve the detection power of genetic variants associated with BMD. Using a recently developed conditional false discovery rate (cFDR) method, we jointly analyzed summary statistics from two large independent genome wide association studies (GWAS) of lumbar spine (LS) BMD and CAD. Strong pleiotropic enrichment and 7 pleiotropic SNPs were found for the two traits. We identified 41 SNPs for LS BMD (cFDR<0.05), of which 20 were replications of previous GWASs and 21 were potential novel SNPs that were not reported before. Four genes encompassed by 9 cFDR-significant SNPs were partially validated in the gene expression assay. Further functional enrichment analysis showed that genes corresponding to the cFDR-significant LS BMD SNPs were enriched in GO terms and KEGG pathways that played crucial roles in bone metabolism (adjP<0.05). In protein-protein interaction analysis, strong interactions were found between the proteins produced by the corresponding genes. Our study demonstrated the reliability and high-efficiency of the cFDR method on the detection of trait-associated genetic variants, the present findings shed novel insights into the genetic variability of BMD as well as the shared genetic basis underlying osteoporosis and CAD.
Collapse
Affiliation(s)
- Cheng Peng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China; Department of Geriatrics, National Key Clinical Specialty, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, China
| | - Jie Shen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Xu Lin
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Kuan-Jui Su
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, Tulane University, New Orleans, LA, USA
| | - Jonathan Greenbaum
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, Tulane University, New Orleans, LA, USA
| | - Wei Zhu
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, Tulane University, New Orleans, LA, USA
| | - Hui-Ling Lou
- Department of Geriatrics, National Key Clinical Specialty, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, China
| | - Feng Liu
- Department of Geriatrics, National Key Clinical Specialty, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, China
| | - Chun-Ping Zeng
- Department of Endocrinology and Metabolism, Affiliated Nanhai Hospital of Southern Medical University, Guangzhou, China
| | | | - Hong-Wen Deng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China; Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
45
|
Mašek J, Andersson ER. The developmental biology of genetic Notch disorders. Development 2017; 144:1743-1763. [PMID: 28512196 DOI: 10.1242/dev.148007] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Notch signaling regulates a vast array of crucial developmental processes. It is therefore not surprising that mutations in genes encoding Notch receptors or ligands lead to a variety of congenital disorders in humans. For example, loss of function of Notch results in Adams-Oliver syndrome, Alagille syndrome, spondylocostal dysostosis and congenital heart disorders, while Notch gain of function results in Hajdu-Cheney syndrome, serpentine fibula polycystic kidney syndrome, infantile myofibromatosis and lateral meningocele syndrome. Furthermore, structure-abrogating mutations in NOTCH3 result in CADASIL. Here, we discuss these human congenital disorders in the context of known roles for Notch signaling during development. Drawing on recent analyses by the exome aggregation consortium (EXAC) and on recent studies of Notch signaling in model organisms, we further highlight additional Notch receptors or ligands that are likely to be involved in human genetic diseases.
Collapse
Affiliation(s)
- Jan Mašek
- Karolinska Institutet, Huddinge 14183, Sweden
| | | |
Collapse
|
46
|
Wang Y, Wu B, Lu P, Zhang D, Wu B, Varshney S, Del Monte-Nieto G, Zhuang Z, Charafeddine R, Kramer AH, Sibinga NE, Frangogiannis NG, Kitsis RN, Adams RH, Alitalo K, Sharp DJ, Harvey RP, Stanley P, Zhou B. Uncontrolled angiogenic precursor expansion causes coronary artery anomalies in mice lacking Pofut1. Nat Commun 2017; 8:578. [PMID: 28924218 PMCID: PMC5603578 DOI: 10.1038/s41467-017-00654-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/18/2017] [Indexed: 12/13/2022] Open
Abstract
Coronary artery anomalies may cause life-threatening cardiac complications; however, developmental mechanisms underpinning coronary artery formation remain ill-defined. Here we identify an angiogenic cell population for coronary artery formation in mice. Regulated by a DLL4/NOTCH1/VEGFA/VEGFR2 signaling axis, these angiogenic cells generate mature coronary arteries. The NOTCH modulator POFUT1 critically regulates this signaling axis. POFUT1 inactivation disrupts signaling events and results in excessive angiogenic cell proliferation and plexus formation, leading to anomalous coronary arteries, myocardial infarction and heart failure. Simultaneous VEGFR2 inactivation fully rescues these defects. These findings show that dysregulated angiogenic precursors link coronary anomalies to ischemic heart disease.Though coronary arteries are crucial for heart function, the mechanisms guiding their formation are largely unknown. Here, Wang et al. identify a unique, endocardially-derived angiogenic precursor cell population for coronary artery formation in mice and show that a DLL4/NOTCH1/VEGFA/VEGFR2 signaling axis is key for coronary artery development.
Collapse
Affiliation(s)
- Yidong Wang
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Bingruo Wu
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Pengfei Lu
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Donghong Zhang
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Brian Wu
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Shweta Varshney
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Gonzalo Del Monte-Nieto
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia.,St. Vincent's Clinical School, University of New South Wales, Kensington, NSW, 2052, Australia
| | - Zhenwu Zhuang
- Department of Medicine, Yale University, New Haven, Connecticut, 06510, USA
| | - Rabab Charafeddine
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Adam H Kramer
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Nicolas E Sibinga
- Departments of Medicine, Developmental and Molecular Biology, Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Nikolaos G Frangogiannis
- Departments of Medicine, Microbiology and Immunology, Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology, Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Röntgenstraße 20, and Faculty of Medicine, University of Münster, 48149, Münster, Germany
| | - Kari Alitalo
- Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu 8, FI-00290, Helsinki, Finland
| | - David J Sharp
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia.,St. Vincent's Clinical School, University of New South Wales, Kensington, NSW, 2052, Australia.,School of Biotechnology and Biomolecular Science, University of New South Wales, Kensington, NSW, 2052, Australia
| | - Pamela Stanley
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Bin Zhou
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA. .,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
47
|
BMP2 expression in the endocardial lineage is required for AV endocardial cushion maturation and remodeling. Dev Biol 2017; 430:113-128. [PMID: 28790014 DOI: 10.1016/j.ydbio.2017.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 07/16/2017] [Accepted: 08/04/2017] [Indexed: 12/13/2022]
Abstract
Distal outgrowth, maturation and remodeling of the endocardial cushion mesenchyme in the atrioventricular (AV) canal are the essential morphogenetic events during four-chambered heart formation. Mesenchymalized AV endocardial cushions give rise to the AV valves and the membranous ventricular septum (VS). Failure of these processes results in several human congenital heart defects. Despite this clinical relevance, the mechanisms governing how mesenchymalized AV endocardial cushions mature and remodel into the membranous VS and AV valves have only begun to be elucidated. The role of BMP signaling in the myocardial and secondary heart forming lineage has been well studied; however, little is known about the role of BMP2 expression in the endocardial lineage. To fill this knowledge gap, we generated Bmp2 endocardial lineage-specific conditional knockouts (referred to as Bmp2 cKOEndo) by crossing conditionally-targeted Bmp2flox/flox mice with a Cre-driver line, Nfatc1Cre, wherein Cre-mediated recombination was restricted to the endocardial cells and their mesenchymal progeny. Bmp2 cKOEndo mouse embryos did not exhibit failure or delay in the initial AV endocardial cushion formation at embryonic day (ED) 9.5-11.5; however, significant reductions in AV cushion size were detected in Bmp2 cKOEndo mouse embryos when compared to control embryos at ED13.5 and ED16.5. Moreover, deletion of Bmp2 from the endocardial lineage consistently resulted in membranous ventricular septal defects (VSDs), and mitral valve deficiencies, as evidenced by the absence of stratification of mitral valves at birth. Muscular VSDs were not found in Bmp2 cKOEndo mouse hearts. To understand the underlying morphogenetic mechanisms leading to a decrease in cushion size, cell proliferation and cell death were examined for AV endocardial cushions. Phospho-histone H3 analyses for cell proliferation and TUNEL assays for apoptotic cell death did not reveal significant differences between control and Bmp2 cKOEndo in AV endocardial cushions. However, mRNA expression of the extracellular matrix components, versican, Has2, collagen 9a1, and periostin was significantly reduced in Bmp2 cKOEndo AV cushions. Expression of transcription factors implicated in the cardiac valvulogenesis, Snail2, Twist1 and Sox9, was also significantly reduced in Bmp2 cKOEndo AV cushions. These data provide evidence that BMP2 expression in the endocardial lineage is essential for the distal outgrowth, maturation and remodeling of AV endocardial cushions into the normal membranous VS and the stratified AV valves.
Collapse
|
48
|
Zhang X, Ai F, Li X, Tian L, Wang X, Shen S, Liu F. MicroRNA-34a suppresses colorectal cancer metastasis by regulating Notch signaling. Oncol Lett 2017; 14:2325-2333. [PMID: 28781671 PMCID: PMC5530141 DOI: 10.3892/ol.2017.6444] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 04/04/2017] [Indexed: 12/19/2022] Open
Abstract
Dysregulation of microRNA (miRNA/miR) expression is causally associated with cancer initiation and progression. However, the precise mechanisms by which dysregulated miRNAs induce colorectal tumorigenesis remain unknown. In the present study, downregulation of miR-34a was identified in colorectal cancer cell lines and clinical specimens. Clinical studies revealed that miR-34a expression was negatively associated with distant metastasis, and positively associated with differentiation and survival of human colorectal cancer specimens. In vitro miRNA functional assays demonstrated that miR-34a bound to the putative 3'-untranslated regions of Notch1 and Jagged1 in SW480 cells, and thereby attenuated the migration and invasion of the colon cancer cells. It was additionally identified that miR-34a downregulated the expression of vimentin and fibronectin via Notch1 and Jagged1. Overall, these data indicate that miR-34a serves a key role in suppressing colorectal cancer metastasis by targeting and regulating Notch signaling.
Collapse
Affiliation(s)
- Xuemei Zhang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan 410008, P.R. China.,Cancer Research Institute, Central South University, Changsha, Hunan 410013, P.R. China
| | - Feiyan Ai
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan 410008, P.R. China
| | - Xiayu Li
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan 410008, P.R. China
| | - Li Tian
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan 410008, P.R. China
| | - Xiaoyan Wang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan 410008, P.R. China
| | - Shourong Shen
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan 410008, P.R. China
| | - Fen Liu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
49
|
Wu B, Wang Y, Xiao F, Butcher JT, Yutzey KE, Zhou B. Developmental Mechanisms of Aortic Valve Malformation and Disease. Annu Rev Physiol 2017; 79:21-41. [DOI: 10.1146/annurev-physiol-022516-034001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Bingruo Wu
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York 10461;
| | - Yidong Wang
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York 10461;
| | - Feng Xiao
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York 10461;
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029 China
| | - Jonathan T. Butcher
- Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853;
| | - Katherine E. Yutzey
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio 45229;
| | - Bin Zhou
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York 10461;
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029 China
| |
Collapse
|
50
|
Albanese I, Yu B, Al-Kindi H, Barratt B, Ott L, Al-Refai M, de Varennes B, Shum-Tim D, Cerruti M, Gourgas O, Rhéaume E, Tardif JC, Schwertani A. Role of Noncanonical Wnt Signaling Pathway in Human Aortic Valve Calcification. Arterioscler Thromb Vasc Biol 2016; 37:543-552. [PMID: 27932350 DOI: 10.1161/atvbaha.116.308394] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 11/28/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The mechanisms underlying the pathogenesis of aortic valve calcification remain unclear. With accumulating evidence demonstrating that valve calcification recapitulates bone development, the crucial roles of noncanonical Wnt ligands WNT5a, WNT5b, and WNT11 in osteogenesis make them critical targets in the study of aortic valve calcification. APPROACH AND RESULTS Using immunohistochemistry, real-time qPCR, Western blotting, and tissue culture, we examined the tissue distribution of WNT5a, WNT5b, and WNT11 in noncalcified and calcified aortic valves and their effects on human aortic valve interstitial cells (HAVICs). Only focal strong immunostaining for WNT5a was seen in and around areas of calcification. Abundant immunostaining for WNT5b and WNT11 was seen in inflammatory cells, fibrosis, and activated myofibroblasts in areas of calcified foci. There was significant correlation between WNT5b and WNT11 overall staining and presence of calcification, lipid score, fibrosis, and microvessels (P<0.05). Real-time qPCR and Western blotting revealed abundant expression of both Wnts in stenotic aortic valves, particularly in bicuspid valves. Incubation of HAVICs from noncalcified valves with the 3 noncanonical Wnts significantly increased cell apoptosis and calcification (P<0.05). Treatment of HAVICs with the mitogen-activated protein kinase-38β and GSK3β inhibitors significantly reduced their mineralization (P<0.01). Raman spectroscopy identified the inorganic phosphate deposits as hydroxyapatite and showed a significant increase in hydroxyapatite deposition in HAVICs in response to WNT5a and WNT11 (P<0.05). Similar crystallinity was seen in the deposits found in HAVICs treated with Wnts and in calcified human aortic valves. CONCLUSIONS These findings suggest a potential role for noncanonical Wnt signaling in the pathogenesis of aortic valve calcification.
Collapse
Affiliation(s)
- Isabella Albanese
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Bin Yu
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Hamood Al-Kindi
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Bianca Barratt
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Leah Ott
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Mohammad Al-Refai
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Benoit de Varennes
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Dominique Shum-Tim
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Marta Cerruti
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Ophélie Gourgas
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Eric Rhéaume
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Jean-Claude Tardif
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Adel Schwertani
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.).
| |
Collapse
|