1
|
Thakkar C, Alikunju S, Venkatasubramanium A, Yang Z, Sayed N, Abdellatif M, Sayed D. Constitutive expression of cardiomyocyte Klf9 precipitates metabolic dysfunction and spontaneous heart failure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633464. [PMID: 39896585 PMCID: PMC11785073 DOI: 10.1101/2025.01.16.633464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Metabolic adaptations and flexibility during development and disease play an essential in cardiomyocyte function and survival. We recently reported Glucocorticoid receptor (GR)-Krüppel-like factor 9 (Klf9) axis in mediating metabolic adaptations in cardiomyocytes stimulated with Dexamethasone. Klf9 expression decreases in hypertrophic and failing hearts, suggesting its importance in cardiac homeostasis and its potential contribution to dysfunction under pressure overload. Genome wide Klf9 occupancy in adult hearts revealed 2,242 genes directly associated with Klf9, with enrichment in metabolic pathways, autophagy, ubiquitin-mediated proteolysis, and cellular senescence. We generated and characterized a conditional cardiac specific Klf9 knock-In (Klf9KI) mice, which developed progressive cardiac hypertrophy, cardiac dysfunction and cardiac failure by 8wks of age, with mortality by 12-14wks. RNA-seq analysis at 1wk, 4wks, and 8wks showed stage-specific transcriptional changes. At 1 week, 64.81% of differentially expressed genes were downregulated, aligning with Klf9's predicted role as a transcriptional repressor. At 4wks and 8wks, more genes were upregulated, suggesting more of secondary targets in response to cardiac phenotype. KEGG pathway analysis showed dysregulation in lipid, carbohydrate and glutathione metabolism, transcriptional regulation, apoptosis, and innate immunity. Untargeted Metabolomics at 4wks identified significant alterations in tissue metabolite levels, particularly in pathways involving fatty acid metabolism, amino acids, and glucose, correlating with transcriptome data. Mitochondrial function assays revealed progressive dysregulation. At 2 weeks, complex I activity was significantly reduced, while complex II and IV activities were partially preserved. By 4 weeks, all measured respiratory complexes showed significant declines, consistent with decline in mitochondrial function. These mitochondrial deficits preceded overt cardiac dysfunction and likely contributed to the development of hypertrophy and failure. In conclusion, constitutive Klf9 overexpression disrupts transcriptional and metabolic homeostasis, driving progressive hypertrophy, cardiac dysfunction, and failure.
Collapse
|
2
|
Harbaum L, Hennigs JK, Pott J, Ostermann J, Sinning CR, Sau A, Sieliwonczyk E, Ng FS, Rhodes CJ, Tello K, Klose H, Gräf S, Wilkins MR. Sex-specific Genetic Determinants of Right Ventricular Structure and Function. Am J Respir Crit Care Med 2024; 211:113-123. [PMID: 39374572 PMCID: PMC11755371 DOI: 10.1164/rccm.202404-0721oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 10/07/2024] [Indexed: 10/09/2024] Open
Abstract
RATIONALE While sex differences in right heart phenotypes have been observed, the molecular drivers remain unknown. OBJECTIVES To provide biological insights into sex differences in the structure and function of the right ventricle (RV) using common genetic variation. METHODS RV phenotypes were obtained from cardiac magnetic resonance imaging in 18,156 women and 16,171 men from the UK Biobank. Observational analyses and sex-stratified genome-wide association studies were performed. Candidate female-specific loci were evaluated against invasively measured cardiac performance in 479 female patients with idiopathic or heritable pulmonary arterial hypertension (PAH), recruited to the UK NIHR BioResource Rare Diseases study. MEASUREMENTS AND MAIN RESULTS Sex was associated with differences in RV volumes and ejection fraction in models adjusting for left heart counterparts, blood pressure, lung function and sex hormone levels. Six genome-wide significant loci (13%) revealed heterogeneity of allelic effects between women and men, and significant sex-by-genotype interaction. These included two sex-specific candidate loci present in women only: a locus for RV ejection fraction in BMPR1A and a locus for RV end-systolic volume near DMRT2. Epigenetic data in RV tissue indicate that variation at the BMPR1A locus likely alters transcriptional regulation. In female patients with PAH, a variant located in the promoter of BMPR1A was significantly associated with cardiac index (effect size 0.16 l/min/m2), despite similar RV afterload. CONCLUSIONS BMPR1A has emerged as a biologically plausible candidate gene for female-specific genetic determination of RV function, showing associations with cardiac performance under chronically increased afterload in female patients with PAH.
Collapse
Affiliation(s)
- Lars Harbaum
- Imperial College London, London, United Kingdom of Great Britain and Northern Ireland
- Hamburg, Germany;
| | - Jan K Hennigs
- University Medical Center Hamburg-Eppendorf, Department of Medicine II, Hamburg, Germany
- Stanford University, Wall Center for Pulmonary Vascular Disease, Stanford, California, United States
| | - Julian Pott
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonna Ostermann
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph R Sinning
- University Heart Center Hamburg, Department of General and Interventional Cardiology, Hamburg, 20246 , Germany
| | - Arunashis Sau
- Imperial College London, London, United Kingdom of Great Britain and Northern Ireland
| | - Ewa Sieliwonczyk
- Imperial College London, London, United Kingdom of Great Britain and Northern Ireland
| | - Fu Siong Ng
- Imperial College London, London, United Kingdom of Great Britain and Northern Ireland
| | - Christopher J Rhodes
- Imperial College London, National Heart & Lung Institute, London, United Kingdom of Great Britain and Northern Ireland
| | - Khodr Tello
- University Hospital Giessen und Marburg GmbH, Pulmonary Hypertension Division, Medical Clinic II, Giessen, Germany
| | - Hans Klose
- University of Hamburg-Eppendorf, Pneumology, Hamburg, Germany
| | - Stefan Gräf
- University of Cambridge, Medicine, Cambridge, Cambridgeshire, United Kingdom of Great Britain and Northern Ireland
| | - Martin R Wilkins
- Imperial College London, London, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
3
|
Danielpour D. Advances and Challenges in Targeting TGF-β Isoforms for Therapeutic Intervention of Cancer: A Mechanism-Based Perspective. Pharmaceuticals (Basel) 2024; 17:533. [PMID: 38675493 PMCID: PMC11054419 DOI: 10.3390/ph17040533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The TGF-β family is a group of 25 kDa secretory cytokines, in mammals consisting of three dimeric isoforms (TGF-βs 1, 2, and 3), each encoded on a separate gene with unique regulatory elements. Each isoform plays unique, diverse, and pivotal roles in cell growth, survival, immune response, and differentiation. However, many researchers in the TGF-β field often mistakenly assume a uniform functionality among all three isoforms. Although TGF-βs are essential for normal development and many cellular and physiological processes, their dysregulated expression contributes significantly to various diseases. Notably, they drive conditions like fibrosis and tumor metastasis/progression. To counter these pathologies, extensive efforts have been directed towards targeting TGF-βs, resulting in the development of a range of TGF-β inhibitors. Despite some clinical success, these agents have yet to reach their full potential in the treatment of cancers. A significant challenge rests in effectively targeting TGF-βs' pathological functions while preserving their physiological roles. Many existing approaches collectively target all three isoforms, failing to target just the specific deregulated ones. Additionally, most strategies tackle the entire TGF-β signaling pathway instead of focusing on disease-specific components or preferentially targeting tumors. This review gives a unique historical overview of the TGF-β field often missed in other reviews and provides a current landscape of TGF-β research, emphasizing isoform-specific functions and disease implications. The review then delves into ongoing therapeutic strategies in cancer, stressing the need for more tools that target specific isoforms and disease-related pathway components, advocating mechanism-based and refined approaches to enhance the effectiveness of TGF-β-targeted cancer therapies.
Collapse
Affiliation(s)
- David Danielpour
- Case Comprehensive Cancer Center Research Laboratories, The Division of General Medical Sciences-Oncology, Case Western Reserve University, Cleveland, OH 44106, USA; ; Tel.: +1-216-368-5670; Fax: +1-216-368-8919
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
- Institute of Urology, University Hospitals, Cleveland, OH 44106, USA
| |
Collapse
|
4
|
Schmidt KE, Höving AL, Kiani Zahrani S, Trevlopoulou K, Kaltschmidt B, Knabbe C, Kaltschmidt C. Serum-Induced Proliferation of Human Cardiac Stem Cells Is Modulated via TGFβRI/II and SMAD2/3. Int J Mol Sci 2024; 25:959. [PMID: 38256034 PMCID: PMC10815425 DOI: 10.3390/ijms25020959] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The ageing phenotype is strongly driven by the exhaustion of adult stem cells (ASCs) and the accumulation of senescent cells. Cardiovascular diseases (CVDs) and heart failure (HF) are strongly linked to the ageing phenotype and are the leading cause of death. As the human heart is considered as an organ with low regenerative capacity, treatments targeting the rejuvenation of human cardiac stem cells (hCSCs) are of great interest. In this study, the beneficial effects of human blood serum on proliferation and senescence of hCSCs have been investigated at the molecular level. We show the induction of a proliferation-related gene expression response by human blood serum at the mRNA level. The concurrent differential expression of the TGFβ target and inhibitor genes indicates the participation of TGFβ signalling in this context. Surprisingly, the application of TGFβ1 as well as the inhibition of TGFβ type I and type II receptor (TGFβRI/II) signalling strongly increased the proliferation of hCSCs. Likewise, both human blood serum and TGFβ1 reduced the senescence in hCSCs. The protective effect of serum on senescence in hCSCs was enhanced by simultaneous TGFβRI/II inhibition. These results strongly indicate a dual role of TGFβ signalling in terms of the serum-mediated effects on hCSCs. Further analysis via RNA sequencing (RNA-Seq) revealed the participation of Ras-inactivating genes wherefore a prevention of hyperproliferation upon serum-treatment in hCSCs via TGFβ signalling and Ras-induced senescence is suggested. These insights may improve treatments of heart failure in the future.
Collapse
Affiliation(s)
- Kazuko E. Schmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (S.K.Z.); (K.T.); (B.K.); (C.K.)
- Institute for Laboratory and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
- Medical Faculty OWL, University of Bielefeld, 33615 Bielefeld, Germany
| | - Anna L. Höving
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (S.K.Z.); (K.T.); (B.K.); (C.K.)
- Institute for Laboratory and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
- Medical Faculty OWL, University of Bielefeld, 33615 Bielefeld, Germany
| | - Sina Kiani Zahrani
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (S.K.Z.); (K.T.); (B.K.); (C.K.)
| | - Katerina Trevlopoulou
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (S.K.Z.); (K.T.); (B.K.); (C.K.)
| | - Barbara Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (S.K.Z.); (K.T.); (B.K.); (C.K.)
- AG Molecular Neurobiology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany
| | - Cornelius Knabbe
- Institute for Laboratory and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
- Medical Faculty OWL, University of Bielefeld, 33615 Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (S.K.Z.); (K.T.); (B.K.); (C.K.)
| |
Collapse
|
5
|
Prapa M, Ho SY. Human Genetics of Semilunar Valve and Aortic Arch Anomalies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:761-775. [PMID: 38884747 DOI: 10.1007/978-3-031-44087-8_45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Lesions of the semilunar valve and the aortic arch can occur either in isolation or as part of well-described clinical syndromes. The polygenic cause of calcific aortic valve disease will be discussed including the key role of NOTCH1 mutations. In addition, the complex trait of bicuspid aortic valve disease will be outlined, both in sporadic/familial cases and in the context of associated syndromes, such as Alagille, Williams, and Kabuki syndromes. Aortic arch abnormalities particularly coarctation of the aorta and interrupted aortic arch, including their association with syndromes such as Turner and 22q11 deletion, respectively, are also discussed. Finally, the genetic basis of congenital pulmonary valve stenosis is summarized, with particular note to Ras-/mitogen-activated protein kinase (Ras/MAPK) pathway syndromes and other less common associations, such as Holt-Oram syndrome.
Collapse
Affiliation(s)
- Matina Prapa
- Department of Clinical Genetics, St George's University Hospitals NHS Foundation Trust, London, UK.
| | - Siew Yen Ho
- Cardiac Morphology, Royal Brompton & Harefield Hospitals, London, UK
| |
Collapse
|
6
|
Li Y, Johnson JP, Yang Y, Yu D, Kubo H, Berretta RM, Wang T, Zhang X, Foster M, Yu J, Tilley DG, Houser SR, Chen X. Effects of maternal hypothyroidism on postnatal cardiomyocyte proliferation and cardiac disease responses of the progeny. Am J Physiol Heart Circ Physiol 2023; 325:H702-H719. [PMID: 37539452 PMCID: PMC10659327 DOI: 10.1152/ajpheart.00320.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/05/2023]
Abstract
Maternal hypothyroidism (MH) could adversely affect the cardiac disease responses of the progeny. This study tested the hypothesis that MH reduces early postnatal cardiomyocyte (CM) proliferation so that the adult heart of MH progeny has a smaller number of larger cardiac myocytes, which imparts adverse cardiac disease responses following injury. Thyroidectomy (TX) was used to establish MH. The progeny from mice that underwent sham or TX surgery were termed Ctrl (control) or MH (maternal hypothyroidism) progeny, respectively. MH progeny had similar heart weight (HW) to body weight (BW) ratios and larger CM size consistent with fewer CMs at postnatal day 60 (P60) compared with Ctrl (control) progeny. MH progeny had lower numbers of EdU+, Ki67+, and phosphorylated histone H3 (PH3)+ CMs, which suggests they had a decreased CM proliferation in the postnatal timeframe. RNA-seq data showed that genes related to DNA replication were downregulated in P5 MH hearts, including bone morphogenetic protein 10 (Bmp10). Both in vivo and in vitro studies showed Bmp10 treatment increased CM proliferation. After transverse aortic constriction (TAC), the MH progeny had more severe cardiac pathological remodeling compared with the Ctrl progeny. Thyroid hormone (T4) treatment for MH mothers preserved their progeny's postnatal CM proliferation capacity and prevented excessive pathological remodeling after TAC. Our results suggest that CM proliferation during early postnatal development was significantly reduced in MH progeny, resulting in fewer CMs with hypertrophy in adulthood. These changes were associated with more severe cardiac disease responses after pressure overload.NEW & NOTEWORTHY Our study shows that compared with Ctrl (control) progeny, the adult progeny of mothers who have MH (MH progeny) had fewer CMs. This reduction of CM numbers was associated with decreased postnatal CM proliferation. Gene expression studies showed a reduced expression of Bmp10 in MH progeny. Bmp10 has been linked to myocyte proliferation. In vivo and in vitro studies showed that Bmp10 treatment of MH progeny and their myocytes could increase CM proliferation. Differences in CM number and size in adult hearts of MH progeny were linked to more severe cardiac structural and functional remodeling after pressure overload. T4 (synthetic thyroxine) treatment of MH mothers during their pregnancy, prevented the reduction in CM number in their progeny and the adverse response to disease stress.
Collapse
Affiliation(s)
- Yijia Li
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Jaslyn P Johnson
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Yijun Yang
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Daohai Yu
- Department of Biomedical Education and Data Science, Center for Biostatistics and Epidemiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Hajime Kubo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Remus M Berretta
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Tao Wang
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Xiaoying Zhang
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, Pennsylvania, United States
| | - Michael Foster
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Jun Yu
- Department of Cardiovascular Sciences, Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, Pennsylvania, United States
| | - Douglas G Tilley
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, Pennsylvania, United States
| | - Steven R Houser
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Xiongwen Chen
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
7
|
Ye D, Liu Y, Pan H, Feng Y, Lu X, Gan L, Wan J, Ye J. Insights into bone morphogenetic proteins in cardiovascular diseases. Front Pharmacol 2023; 14:1125642. [PMID: 36909186 PMCID: PMC9996008 DOI: 10.3389/fphar.2023.1125642] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are secretory proteins belonging to the transforming growth factor-β (TGF-β) superfamily. These proteins play important roles in embryogenesis, bone morphogenesis, blood vessel remodeling and the development of various organs. In recent years, as research has progressed, BMPs have been found to be closely related to cardiovascular diseases, especially atherosclerosis, vascular calcification, cardiac remodeling, pulmonary arterial hypertension (PAH) and hereditary hemorrhagic telangiectasia (HHT). In this review, we summarized the potential roles and related mechanisms of the BMP family in the cardiovascular system and focused on atherosclerosis and PAH.
Collapse
Affiliation(s)
- Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yinghui Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Heng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiyi Lu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liren Gan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
8
|
Yang Y, Yang H, Lian X, Yang S, Shen H, Wu S, Wang X, Lyu G. Circulating microRNA: Myocardium-derived prenatal biomarker of ventricular septal defects. Front Genet 2022; 13:899034. [PMID: 36035156 PMCID: PMC9403759 DOI: 10.3389/fgene.2022.899034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Recently, circulating microRNAs (miRNAs) from maternal blood and amniotic fluid have been used as biomarkers for ventricular septal defect (VSD) diagnosis. However, whether circulating miRNAs are associated with fetal myocardium remains unknown.Methods: Dimethadione (DMO) induced a VSD rat model. The miRNA expression profiles of the myocardium, amniotic fluid and maternal serum were analyzed. Differentially expressed microRNAs (DE-microRNAs) were verified by qRT–PCR. The target gene of miR-1-3p was confirmed by dual luciferase reporter assays. Expression of amniotic fluid-derived DE-microRNAs was verified in clinical samples.Results: MiRNAs were differentially expressed in VSD fetal rats and might be involved in cardiomyocyte differentiation and apoptosis. MiR-1-3p, miR-1b and miR-293-5p were downregulated in the myocardium and upregulated in amniotic fluid/maternal serum. The expression of amniotic fluid-derived DE-microRNAs (miR-1-3p, miR-206 and miR-184) was verified in clinical samples. Dual luciferase reporter assays confirmed that miR-1-3p directly targeted SLC8A1/NCX1.Conclusion: MiR-1-3p, miR-1b and miR-293-5p are downregulated in VSD myocardium and upregulated in circulation and may be released into circulation by cardiomyocytes. MiR-1-3p targets SLC8A1/NCX1 and participates in myocardial apoptosis. MiR-1-3p upregulation in circulation is a direct and powerful indicator of fetal VSD and is expected to serve as a prenatal VSD diagnostic marker.
Collapse
Affiliation(s)
- Yiru Yang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Hainan Yang
- Department of Ultrasound, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xihua Lian
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Shuping Yang
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Haolin Shen
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Shufen Wu
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Xiali Wang
- Collaborative Innovation Center for Maternal and Infant Health Service Application Technology, Quanzhou Medical College, Quanzhou, Fujian, China
| | - Guorong Lyu
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Collaborative Innovation Center for Maternal and Infant Health Service Application Technology, Quanzhou Medical College, Quanzhou, Fujian, China
- *Correspondence: Guorong Lyu,
| |
Collapse
|
9
|
Mitochondrial calcium uniporter stabilization preserves energetic homeostasis during Complex I impairment. Nat Commun 2022; 13:2769. [PMID: 35589699 PMCID: PMC9120069 DOI: 10.1038/s41467-022-30236-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
Calcium entering mitochondria potently stimulates ATP synthesis. Increases in calcium preserve energy synthesis in cardiomyopathies caused by mitochondrial dysfunction, and occur due to enhanced activity of the mitochondrial calcium uniporter channel. The signaling mechanism that mediates this compensatory increase remains unknown. Here, we find that increases in the uniporter are due to impairment in Complex I of the electron transport chain. In normal physiology, Complex I promotes uniporter degradation via an interaction with the uniporter pore-forming subunit, a process we term Complex I-induced protein turnover. When Complex I dysfunction ensues, contact with the uniporter is inhibited, preventing degradation, and leading to a build-up in functional channels. Preventing uniporter activity leads to early demise in Complex I-deficient animals. Conversely, enhancing uniporter stability rescues survival and function in Complex I deficiency. Taken together, our data identify a fundamental pathway producing compensatory increases in calcium influx during Complex I impairment.
Collapse
|
10
|
Chloe Li KY, Cook AC, Lovering RC. GOing Forward With the Cardiac Conduction System Using Gene Ontology. Front Genet 2022; 13:802393. [PMID: 35309148 PMCID: PMC8924464 DOI: 10.3389/fgene.2022.802393] [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: 10/26/2021] [Accepted: 02/09/2022] [Indexed: 02/03/2023] Open
Abstract
The cardiac conduction system (CCS) comprises critical components responsible for the initiation, propagation, and coordination of the action potential. Aberrant CCS development can cause conduction abnormalities, including sick sinus syndrome, accessory pathways, and atrioventricular and bundle branch blocks. Gene Ontology (GO; http://geneontology.org/) is an invaluable global bioinformatics resource which provides structured, computable knowledge describing the functions of gene products. Many gene products are known to be involved in CCS development; however, this information is not comprehensively captured by GO. To address the needs of the heart development research community, this study aimed to describe the specific roles of proteins reported in the literature to be involved with CCS development and/or function. 14 proteins were prioritized for GO annotation which led to the curation of 15 peer-reviewed primary experimental articles using carefully selected GO terms. 152 descriptive GO annotations, including those describing sinoatrial node and atrioventricular node development were created and submitted to the GO Consortium database. A functional enrichment analysis of 35 key CCS development proteins confirmed that this work has improved the in-silico interpretation of this CCS dataset. This work may improve future investigations of the CCS with application of high-throughput methods such as genome-wide association studies analysis, proteomics, and transcriptomics.
Collapse
Affiliation(s)
- Kan Yan Chloe Li
- Department of Preclinical and Fundamental Science, Institute of Cardiovascular Science, Functional Gene Annotation, University College London, London, United Kingdom,Department of Children’s Cardiovascular Disease, Centre for Morphology and Structural Heart Disease, Institute of Cardiovascular Science, University College London, London, United Kingdom,*Correspondence: Kan Yan Chloe Li,
| | - Andrew C Cook
- Department of Children’s Cardiovascular Disease, Centre for Morphology and Structural Heart Disease, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Ruth C Lovering
- Department of Preclinical and Fundamental Science, Institute of Cardiovascular Science, Functional Gene Annotation, University College London, London, United Kingdom
| |
Collapse
|
11
|
Bi Y, Yang Z, Jin M, Zhai K, Wang J, Mao Y, Liu Y, Ding M, Wang H, Wang F, Cai H, Ji G. ERp44 is required for endocardial cushion development by regulating VEGFA secretion in myocardium. Cell Prolif 2022; 55:e13179. [PMID: 35088919 PMCID: PMC8891561 DOI: 10.1111/cpr.13179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/22/2021] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES Endocardial cushions are precursors of the valve septum complex that separates the four heart chambers. Several genes have been implicated in the development of endocardial cushions. Specifically, ERp44 has been found to play a role in the early secretory pathway, but its function in heart development has not been well studied. MATERIALS AND METHODS In this study, we established conditional and tissue-specific knockout mouse models. The morphology, survival rate, the development of heart and endocardial cushion were under evaluation. The relationship between ERp44 and VEGFA was investigated by transcriptome, qPCR, WB, immunofluorescence and immunohistochemistry. RESULTS ERp44 knockout (KO) mice were smaller in size, and most mice died during early postnatal life. KO hearts exhibited the typical phenotypes of congenital heart diseases, such as abnormal heart shapes and severe septal and valvular defects. Similar phenotypes were found in cTNT-Cre+/- ; ERp44fl / fl mice, which indicated that myocardial ERp44 principally controls endocardial cushion formation. Further studies demonstrated that the deletion of ERp44 significantly decreased the proliferation of cushion cells and impaired the endocardial-mesenchymal transition (EndMT), which was followed by endocardial cushion dysplasia. Finally, we found that ERp44 was directly bound to VEGFA and controlled its release, further regulating EndMT. CONCLUSION We demonstrated that ERp44 plays a specific role in heart development. ERp44 contributes to the development of the endocardial cushion by affecting VEGFA-mediated EndMT.
Collapse
Affiliation(s)
- Youkun Bi
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Zhiguang Yang
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Meng Jin
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Kui Zhai
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jun Wang
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yang Mao
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yang Liu
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Mingqin Ding
- National Institute of Biological SciencesBeijingChina
| | - Huiwen Wang
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Fengchao Wang
- National Institute of Biological SciencesBeijingChina
| | - Hong Cai
- Department of DermatologyAir Force Medical CenterPLABeijingChina
| | - Guangju Ji
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
| |
Collapse
|
12
|
Atli EI, Atli E, Yalcintepe S, Demir S, Kalkan R, Akurut C, Ozen Y, Gurkan H. Investigation of Genetic Alterations in Congenital Heart Diseases in Prenatal Period. Glob Med Genet 2021; 9:29-33. [PMID: 35169781 PMCID: PMC8837410 DOI: 10.1055/s-0041-1736566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/09/2021] [Indexed: 11/09/2022] Open
Abstract
The prenatal diagnosis of congenital heart disease (CHD) is important because of mortality risk. The onset of CHD varies, and depending on the malformation type, the risk of aneuploidy is changed. To identify possible genetic alterations in CHD, G-banding, chromosomal microarray or if needed DNA mutation analysis and direct sequence analysis should be planned. In present study, to identify genetic alterations, cell culture, karyotype analysis, and single nucleotide polymorphism, array analyses were conducted on a total 950 samples. Interventional prenatal genetic examination was performed on 23 (2, 4%, 23/950) fetal CHD cases. Chromosomal abnormalities were detected in 5 out of 23 cases (21, 7%). Detected chromosomal abnormalities were 10q23.2 deletion, trisomy 18, 8p22.3-p23.2 deletion, 8q21.3-q24.3 duplication, 11q24.2q24.5 (9 Mb) deletion, and 8p22p12 (16.8 Mb) deletion. Our study highlights the importance of genetic testing in CHD.
Collapse
Affiliation(s)
- Emine Ikbal Atli
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Engin Atli
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Sinem Yalcintepe
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Selma Demir
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Rasime Kalkan
- Department of Medical Genetics, Faculty of Medicine, Near East University, Nicosia, Cyprus
| | - Cisem Akurut
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Yasemin Ozen
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Hakan Gurkan
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
13
|
Generation and characterization of a Myh6-driven Cre knockin mouse line. Transgenic Res 2021; 30:821-835. [PMID: 34542814 PMCID: PMC8580938 DOI: 10.1007/s11248-021-00285-4] [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: 08/02/2021] [Accepted: 09/10/2021] [Indexed: 10/25/2022]
Abstract
Gene deletion by the Cre-Loxp system has facilitated functional studies of many critical genes in mice, offering important insights and allowing deeper understanding on the mechanisms underlying organ development and diseases, such as heart development and diseases. In this study, we generated a Myh6-Cre knockin mouse model by inserting the IRES-Cre-wpre-polyA cassette between the translational stop codon and the 3' untranslated region of the endogenous Myh6 gene. By crossing knockin mice with the Rosa26 reporter lines, we found that Myh6-Cre targeted cardiomyocytes at the embryonic and postnatal stages. In addition, we were able to inactivate the desmosome gene Desmoplakin (Dsp) by breeding Myh6-Cre mice with a conditional Dspflox knockout mouse line, which resulted in embryonic lethality during the mid-term pregnancy. These results suggest that the new Myh6-Cre mouse line can serve as a robust tool to dissect the distinct roles of genes involved in heart development and function.
Collapse
|
14
|
Ding Q, Qi Y, Tsang SY. Mitochondrial Biogenesis, Mitochondrial Dynamics, and Mitophagy in the Maturation of Cardiomyocytes. Cells 2021; 10:cells10092463. [PMID: 34572112 PMCID: PMC8466139 DOI: 10.3390/cells10092463] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 01/01/2023] Open
Abstract
Pluripotent stem cells (PSCs) can undergo unlimited self-renewal and can differentiate into all the cell types present in our body, including cardiomyocytes. Therefore, PSCs can be an excellent source of cardiomyocytes for future regenerative medicine and medical research studies. However, cardiomyocytes obtained from PSC differentiation culture are regarded as immature structurally, electrophysiologically, metabolically, and functionally. Mitochondria are organelles responsible for various cellular functions such as energy metabolism, different catabolic and anabolic processes, calcium fluxes, and various signaling pathways. Cells can respond to cellular needs to increase the mitochondrial mass by mitochondrial biogenesis. On the other hand, cells can also degrade mitochondria through mitophagy. Mitochondria are also dynamic organelles that undergo continuous fusion and fission events. In this review, we aim to summarize previous findings on the changes of mitochondrial biogenesis, mitophagy, and mitochondrial dynamics during the maturation of cardiomyocytes. In addition, we intend to summarize whether changes in these processes would affect the maturation of cardiomyocytes. Lastly, we aim to discuss unanswered questions in the field and to provide insights for the possible strategies of enhancing the maturation of PSC-derived cardiomyocytes.
Collapse
Affiliation(s)
- Qianqian Ding
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China;
| | - Yanxiang Qi
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China;
| | - Suk-Ying Tsang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China;
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China;
- Key Laboratory for Regenerative Medicine, Ministry of Education, The Chinese University of Hong Kong, Hong Kong, China
- The Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Hong Kong, China
- Correspondence: ; Tel.: +852-39431020
| |
Collapse
|
15
|
Zheng M, Erhardt S, Ai D, Wang J. Bmp Signaling Regulates Hand1 in a Dose-Dependent Manner during Heart Development. Int J Mol Sci 2021; 22:ijms22189835. [PMID: 34576009 PMCID: PMC8465227 DOI: 10.3390/ijms22189835] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 01/22/2023] Open
Abstract
The bone morphogenetic protein (Bmp) signaling pathway and the basic helix–loop–helix (bHLH) transcription factor Hand1 are known key regulators of cardiac development. In this study, we investigated the Bmp signaling regulation of Hand1 during cardiac outflow tract (OFT) development. In Bmp2 and Bmp4loss-of-function embryos with varying levels of Bmp in the heart, Hand1 is sensitively decreased in response to the dose of Bmp expression. In contrast, Hand1 in the heart is dramatically increased in Bmp4 gain-of-function embryos. We further identified and characterized the Bmp/Smad regulatory elements in Hand1. Combined transfection assays and chromatin immunoprecipitation (ChIP) experiments indicated that Hand1 is directly activated and bound by Smads. In addition, we found that upon the treatment of Bmp2 and Bmp4, P19 cells induced Hand1 expression and favored cardiac differentiation. Together, our data indicated that the Bmp signaling pathway directly regulates Hand1 expression in a dose-dependent manner during heart development.
Collapse
Affiliation(s)
- Mingjie Zheng
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.Z.); (S.E.)
| | - Shannon Erhardt
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.Z.); (S.E.)
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Di Ai
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA;
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.Z.); (S.E.)
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
16
|
Abstract
Congenital heart disease is the most frequent birth defect and the leading cause of death for the fetus and in the first year of life. The wide phenotypic diversity of congenital heart defects requires expert diagnosis and sophisticated repair surgery. Although these defects have been described since the seventeenth century, it was only in 2005 that a consensus international nomenclature was adopted, followed by an international classification in 2017 to help provide better management of patients. Advances in genetic engineering, imaging, and omics analyses have uncovered mechanisms of heart formation and malformation in animal models, but approximately 80% of congenital heart defects have an unknown genetic origin. Here, we summarize current knowledge of congenital structural heart defects, intertwining clinical and fundamental research perspectives, with the aim to foster interdisciplinary collaborations at the cutting edge of each field. We also discuss remaining challenges in better understanding congenital heart defects and providing benefits to patients.
Collapse
Affiliation(s)
- Lucile Houyel
- Unité de Cardiologie Pédiatrique et Congénitale and Centre de Référence des Malformations Cardiaques Congénitales Complexes (M3C), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France.,Université de Paris, 75015 Paris, France
| | - Sigolène M Meilhac
- Université de Paris, 75015 Paris, France.,Imagine-Institut Pasteur Unit of Heart Morphogenesis, INSERM UMR 1163, 75015 Paris, France;
| |
Collapse
|
17
|
Yonebayashi S, Tajiri K, Hara M, Saito H, Suzuki N, Sakai S, Kimura T, Sato A, Sekimoto A, Fujita S, Okamoto R, Schwartz RJ, Yoshida T, Imanaka-Yoshida K. Generation of Transgenic Mice that Conditionally Overexpress Tenascin-C. Front Immunol 2021; 12:620541. [PMID: 33763067 PMCID: PMC7982461 DOI: 10.3389/fimmu.2021.620541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/10/2021] [Indexed: 02/05/2023] Open
Abstract
Tenascin-C (TNC) is an extracellular matrix glycoprotein that is expressed during embryogenesis. It is not expressed in normal adults, but is up-regulated under pathological conditions. Although TNC knockout mice do not show a distinct phenotype, analyses of disease models using TNC knockout mice combined with in vitro experiments revealed the diverse functions of TNC. Since high TNC levels often predict a poor prognosis in various clinical settings, we developed a transgenic mouse that overexpresses TNC through Cre recombinase-mediated activation. Genomic walking showed that the transgene was integrated into and truncated the Atp8a2 gene. While homozygous transgenic mice showed a severe neurological phenotype, heterozygous mice were viable, fertile, and did not exhibit any distinct abnormalities. Breeding hemizygous mice with Nkx2.5 promoter-Cre or α-myosin heavy chain promoter Cre mice induced the heart-specific overexpression of TNC in embryos and adults. TNC-overexpressing mouse hearts did not have distinct histological or functional abnormalities. However, the expression of proinflammatory cytokines/chemokines was significantly up-regulated and mortality rates during the acute stage after myocardial infarction were significantly higher than those of the controls. Our novel transgenic mouse may be applied to investigations on the role of TNC overexpression in vivo in various tissue/organ pathologies using different Cre donors.
Collapse
Affiliation(s)
- Saori Yonebayashi
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kazuko Tajiri
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Mari Hara
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, Tsu, Japan.,Research Center for Matrix Biology, Mie University, Tsu, Japan
| | - Hiromitsu Saito
- Department of Animal Genomics, Functional Genomics Institute, Mie University Life Science Research Center, Tsu, Japan
| | - Noboru Suzuki
- Department of Animal Genomics, Functional Genomics Institute, Mie University Life Science Research Center, Tsu, Japan
| | - Satoshi Sakai
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Taizo Kimura
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Akira Sato
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Akiyo Sekimoto
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Satoshi Fujita
- Department of Cardiology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Ryuji Okamoto
- Department of Cardiology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Robert J Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Toshimichi Yoshida
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, Tsu, Japan.,Research Center for Matrix Biology, Mie University, Tsu, Japan
| | - Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, Tsu, Japan.,Research Center for Matrix Biology, Mie University, Tsu, Japan
| |
Collapse
|
18
|
Bhattacharya A, Al-Sammarraie N, Gebere MG, Johnson J, Eberth JF, Azhar M. Myocardial TGFβ2 Is Required for Atrioventricular Cushion Remodeling and Myocardial Development. J Cardiovasc Dev Dis 2021; 8:jcdd8030026. [PMID: 33801433 PMCID: PMC7999251 DOI: 10.3390/jcdd8030026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/18/2021] [Accepted: 02/26/2021] [Indexed: 12/05/2022] Open
Abstract
Among the three transforming growth factor beta (TGFβ) ligands, TGFβ2 is essential for heart development and is produced by multiple cell types, including myocardium. Heterozygous mutations in TGFB2 in patients of connective tissue disorders result in congenital heart defects and adult valve malformations, including mitral valve prolapse (MVP) with or without regurgitation. Tgfb2 germline knockout fetuses exhibit multiple cardiac defects but the role of myocardial-TGFβ2 in heart development is yet to be elucidated. Here, myocardial Tgfb2 conditional knockout (CKO) embryos were generated by crossing Tgfb2flox mice with Tgfb2+/−; cTntCre mice. Tgfb2flox/− embryos were normal, viable. Cell fate mapping was done using dual-fluorescent mT/mG+/− mice. Cre-mediated Tgfb2 deletion was assessed by genomic PCR. RNAscope in situ hybridization was used to detect the loss of myocardial Tgfb2 expression. Histological, morphometric, immunohistochemical, and in situ hybridization analyses of CKOs and littermate controls at different stages of heart development (E12.5–E18.5) were used to determine the role of myocardium-derived TGFβ2 in atrioventricular (AV) cushion remodeling and myocardial development. CKOs exhibit a thin ventricular myocardium, AV cushion remodeling defects and developed incomplete AV septation defects. The loss of myocardial Tgfb2 resulted in impaired cushion maturation and dysregulated cell death. Phosphorylated SMAD2, a surrogate for TGFβ signaling, was “paradoxically” increased in both AV cushion mesenchyme and ventricular myocardium in the CKOs. Our results indicate that TGFβ2 produced by cardiomyocytes acting as cells autonomously on myocardium and via paracrine signaling on AV cushions are required for heart development.
Collapse
Affiliation(s)
- Aniket Bhattacharya
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA; (A.B.); (N.A.-S.); (M.G.G.); (J.J.); (J.F.E.)
| | - Nadia Al-Sammarraie
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA; (A.B.); (N.A.-S.); (M.G.G.); (J.J.); (J.F.E.)
| | - Mengistu G. Gebere
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA; (A.B.); (N.A.-S.); (M.G.G.); (J.J.); (J.F.E.)
| | - John Johnson
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA; (A.B.); (N.A.-S.); (M.G.G.); (J.J.); (J.F.E.)
| | - John F. Eberth
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA; (A.B.); (N.A.-S.); (M.G.G.); (J.J.); (J.F.E.)
| | - Mohamad Azhar
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA; (A.B.); (N.A.-S.); (M.G.G.); (J.J.); (J.F.E.)
- William Jennings Bryan Dorn VA Medical Center, Dorn Research Institute, Columbia, SC 29209, USA
- Correspondence: ; Tel.: +1-803-216-3831
| |
Collapse
|
19
|
Cui N, Sakurai T, Kamiyoshi A, Ichikawa-Shindo Y, Kawate H, Tanaka M, Tanaka M, Wei Y, Kakihara S, Zhao Y, Aruga K, Kawagishi H, Nakada T, Yamada M, Shindo T. Adrenomedullin-RAMP2 and -RAMP3 Systems Regulate Cardiac Homeostasis during Cardiovascular Stress. Endocrinology 2021; 162:6129198. [PMID: 33545715 DOI: 10.1210/endocr/bqab001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Indexed: 12/26/2022]
Abstract
Adrenomedullin (AM) is a peptide hormone with multiple physiological functions, which are regulated by its receptor activity-modifying proteins, RAMP2 and RAMP3. We previously reported that AM or RAMP2 knockout (KO) (AM-/-, RAMP2-/-) is embryonically lethal in mice, whereas RAMP3-/- mice are apparently normal. AM, RAMP2, and RAMP3 are all highly expressed in the heart; however, their functions there are not fully understood. Here, we analyzed the pathophysiological functions of the AM-RAMP2 and AM-RAMP3 systems in hearts subjected to cardiovascular stress. Cardiomyocyte-specific RAMP2-/- (C-RAMP2-/-) and RAMP3-/- showed no apparent heart failure at base line. After 1 week of transverse aortic constriction (TAC), however, C-RAMP2-/- exhibited significant cardiac hypertrophy, decreased ejection fraction, and increased fibrosis compared with wild-type mice. Both dP/dtmax and dP/dtmin were significantly reduced in C-RAMP2-/-, indicating reduced ventricular contractility and relaxation. Exposing C-RAMP2-/- cardiomyocytes to isoproterenol enhanced their hypertrophy and oxidative stress compared with wild-type cells. C-RAMP2-/- cardiomyocytes also contained fewer viable mitochondria and showed reduced mitochondrial membrane potential and respiratory capacity. RAMP3-/- also showed reduced systolic function and enhanced fibrosis after TAC, but those only became apparent after 4 weeks. A reduction in cardiac lymphatic vessels was the characteristic feature in RAMP3-/-. These observations indicate the AM-RAMP2 system is necessary for early adaptation to cardiovascular stress through regulation of cardiac mitochondria. AM-RAMP3 is necessary for later adaptation through regulation of lymphatic vessels. The AM-RAMP2 and AM-RAMP3 systems thus play separate critical roles in the maintenance of cardiovascular homeostasis against cardiovascular stress.
Collapse
Affiliation(s)
- Nanqi Cui
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takayuki Sakurai
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
- Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| | - Akiko Kamiyoshi
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
- Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| | - Yuka Ichikawa-Shindo
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hisaka Kawate
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Megumu Tanaka
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Masaaki Tanaka
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yangxuan Wei
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shinji Kakihara
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yunlu Zhao
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kohsuke Aruga
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hiroyuki Kawagishi
- Department of Molecular Pharmacology, Shinshu University School of Medicine, Matsumoto, Japan
- Department of Biotechnology, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| | - Tsutomu Nakada
- Department of Instrumental Analysis, Research Center for Supports to Advanced Science, Shinshu University, Matsumoto, Japan
| | - Mitsuhiko Yamada
- Department of Molecular Pharmacology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takayuki Shindo
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
- Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| |
Collapse
|
20
|
Deepe R, Fitzgerald E, Wolters R, Drummond J, Guzman KD, van den Hoff MJ, Wessels A. The Mesenchymal Cap of the Atrial Septum and Atrial and Atrioventricular Septation. J Cardiovasc Dev Dis 2020; 7:jcdd7040050. [PMID: 33158164 PMCID: PMC7712865 DOI: 10.3390/jcdd7040050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/28/2020] [Accepted: 11/02/2020] [Indexed: 12/26/2022] Open
Abstract
In this publication, dedicated to Professor Robert H. Anderson and his contributions to the field of cardiac development, anatomy, and congenital heart disease, we will review some of our earlier collaborative studies. The focus of this paper is on our work on the development of the atrioventricular mesenchymal complex, studies in which Professor Anderson has played a significant role. We will revisit a number of events relevant to atrial and atrioventricular septation and present new data on the development of the mesenchymal cap of the atrial septum, a component of the atrioventricular mesenchymal complex which, thus far, has received only moderate attention.
Collapse
Affiliation(s)
- Ray Deepe
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (R.D.); (E.F.); (R.W.); (J.D.); (K.D.G.)
| | - Emily Fitzgerald
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (R.D.); (E.F.); (R.W.); (J.D.); (K.D.G.)
| | - Renélyn Wolters
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (R.D.); (E.F.); (R.W.); (J.D.); (K.D.G.)
| | - Jenna Drummond
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (R.D.); (E.F.); (R.W.); (J.D.); (K.D.G.)
| | - Karen De Guzman
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (R.D.); (E.F.); (R.W.); (J.D.); (K.D.G.)
| | - Maurice J.B. van den Hoff
- Amsterdam UMC, Academic Medical Center, Department of Medical Biology, Meibergdreef 15, 1105AZ Amsterdam, The Netherlands;
| | - Andy Wessels
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (R.D.); (E.F.); (R.W.); (J.D.); (K.D.G.)
- Correspondence: ; Tel.: +1-843-792-8183
| |
Collapse
|
21
|
Fang X, Cai Z, Wang H, Han D, Cheng Q, Zhang P, Gao F, Yu Y, Song Z, Wu Q, An P, Huang S, Pan J, Chen HZ, Chen J, Linkermann A, Min J, Wang F. Loss of Cardiac Ferritin H Facilitates Cardiomyopathy via Slc7a11-Mediated Ferroptosis. Circ Res 2020; 127:486-501. [PMID: 32349646 DOI: 10.1161/circresaha.120.316509] [Citation(s) in RCA: 508] [Impact Index Per Article: 101.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
RATIONALE Maintaining iron homeostasis is essential for proper cardiac function. Both iron deficiency and iron overload are associated with cardiomyopathy and heart failure via complex mechanisms. Although ferritin plays a central role in iron metabolism by storing excess cellular iron, the molecular function of ferritin in cardiomyocytes remains unknown. OBJECTIVE To characterize the functional role of Fth (ferritin H) in mediating cardiac iron homeostasis and heart disease. METHODS AND RESULTS Mice expressing a conditional Fth knockout allele were crossed with 2 distinct Cre recombinase-expressing mouse lines, resulting in offspring that lack Fth expression specifically in myocytes (MCK-Cre) or cardiomyocytes (Myh6-Cre). Mice lacking Fth in cardiomyocytes had decreased cardiac iron levels and increased oxidative stress, resulting in mild cardiac injury upon aging. However, feeding these mice a high-iron diet caused severe cardiac injury and hypertrophic cardiomyopathy, with molecular features typical of ferroptosis, including reduced glutathione (GSH) levels and increased lipid peroxidation. Ferrostatin-1, a specific inhibitor of ferroptosis, rescued this phenotype, supporting the notion that ferroptosis plays a pathophysiological role in the heart. Finally, we found that Fth-deficient cardiomyocytes have reduced expression of the ferroptosis regulator Slc7a11, and overexpressing Slc7a11 selectively in cardiomyocytes increased GSH levels and prevented cardiac ferroptosis. CONCLUSIONS Our findings provide compelling evidence that ferritin plays a major role in protecting against cardiac ferroptosis and subsequent heart failure, thereby providing a possible new therapeutic target for patients at risk of developing cardiomyopathy.
Collapse
Affiliation(s)
- Xuexian Fang
- From the First Affiliated Hospital, School of Public Health (X.F., Z.C., D.H., Q.C., P.Z., Y.Y., Z.S., Q.W., S.H., J.P., J.M., F.W.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University (X.F., P.A., F.W.).,Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, China (X.F., H.W., F.W.)
| | - Zhaoxian Cai
- From the First Affiliated Hospital, School of Public Health (X.F., Z.C., D.H., Q.C., P.Z., Y.Y., Z.S., Q.W., S.H., J.P., J.M., F.W.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Wang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, China (X.F., H.W., F.W.)
| | - Dan Han
- From the First Affiliated Hospital, School of Public Health (X.F., Z.C., D.H., Q.C., P.Z., Y.Y., Z.S., Q.W., S.H., J.P., J.M., F.W.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Cheng
- From the First Affiliated Hospital, School of Public Health (X.F., Z.C., D.H., Q.C., P.Z., Y.Y., Z.S., Q.W., S.H., J.P., J.M., F.W.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Pan Zhang
- From the First Affiliated Hospital, School of Public Health (X.F., Z.C., D.H., Q.C., P.Z., Y.Y., Z.S., Q.W., S.H., J.P., J.M., F.W.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Gao
- The Second Affiliated Hospital (F.G., J.C.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yingying Yu
- From the First Affiliated Hospital, School of Public Health (X.F., Z.C., D.H., Q.C., P.Z., Y.Y., Z.S., Q.W., S.H., J.P., J.M., F.W.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zijun Song
- From the First Affiliated Hospital, School of Public Health (X.F., Z.C., D.H., Q.C., P.Z., Y.Y., Z.S., Q.W., S.H., J.P., J.M., F.W.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Wu
- From the First Affiliated Hospital, School of Public Health (X.F., Z.C., D.H., Q.C., P.Z., Y.Y., Z.S., Q.W., S.H., J.P., J.M., F.W.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng An
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University (X.F., P.A., F.W.)
| | - Sicong Huang
- From the First Affiliated Hospital, School of Public Health (X.F., Z.C., D.H., Q.C., P.Z., Y.Y., Z.S., Q.W., S.H., J.P., J.M., F.W.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianwei Pan
- From the First Affiliated Hospital, School of Public Health (X.F., Z.C., D.H., Q.C., P.Z., Y.Y., Z.S., Q.W., S.H., J.P., J.M., F.W.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Hou-Zao Chen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (H.-Z.C.)
| | - Jinghai Chen
- The Second Affiliated Hospital (F.G., J.C.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Andreas Linkermann
- Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany (A.L.)
| | - Junxia Min
- From the First Affiliated Hospital, School of Public Health (X.F., Z.C., D.H., Q.C., P.Z., Y.Y., Z.S., Q.W., S.H., J.P., J.M., F.W.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- From the First Affiliated Hospital, School of Public Health (X.F., Z.C., D.H., Q.C., P.Z., Y.Y., Z.S., Q.W., S.H., J.P., J.M., F.W.), Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University (X.F., P.A., F.W.).,Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, China (X.F., H.W., F.W.)
| |
Collapse
|
22
|
Dronkers E, Wauters MMM, Goumans MJ, Smits AM. Epicardial TGFβ and BMP Signaling in Cardiac Regeneration: What Lesson Can We Learn from the Developing Heart? Biomolecules 2020; 10:biom10030404. [PMID: 32150964 PMCID: PMC7175296 DOI: 10.3390/biom10030404] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/31/2022] Open
Abstract
The epicardium, the outer layer of the heart, has been of interest in cardiac research due to its vital role in the developing and diseased heart. During development, epicardial cells are active and supply cells and paracrine cues to the myocardium. In the injured adult heart, the epicardium is re-activated and recapitulates embryonic behavior that is essential for a proper repair response. Two indispensable processes for epicardial contribution to heart tissue formation are epithelial to mesenchymal transition (EMT), and tissue invasion. One of the key groups of cytokines regulating both EMT and invasion is the transforming growth factor β (TGFβ) family, including TGFβ and Bone Morphogenetic Protein (BMP). Abundant research has been performed to understand the role of TGFβ family signaling in the developing epicardium. However, less is known about signaling in the adult epicardium. This review provides an overview of the current knowledge on the role of TGFβ in epicardial behavior both in the development and in the repair of the heart. We aim to describe the presence of involved ligands and receptors to establish if and when signaling can occur. Finally, we discuss potential targets to improve the epicardial contribution to cardiac repair as a starting point for future investigation.
Collapse
|
23
|
Dituri F, Cossu C, Mancarella S, Giannelli G. The Interactivity between TGFβ and BMP Signaling in Organogenesis, Fibrosis, and Cancer. Cells 2019; 8:E1130. [PMID: 31547567 PMCID: PMC6829314 DOI: 10.3390/cells8101130] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022] Open
Abstract
The Transforming Growth Factor beta (TGFβ) and Bone Morphogenic Protein (BMP) pathways intersect at multiple signaling hubs and cooperatively or counteractively participate to bring about cellular processes which are critical not only for tissue morphogenesis and organogenesis during development, but also for adult tissue homeostasis. The proper functioning of the TGFβ/BMP pathway depends on its communication with other signaling pathways and any deregulation leads to developmental defects or diseases, including fibrosis and cancer. In this review we explore the cellular and physio-pathological contexts in which the synergism or antagonism between the TGFβ and BMP pathways are crucial determinants for the normal developmental processes, as well as the progression of fibrosis and malignancies.
Collapse
Affiliation(s)
- Francesco Dituri
- National Institute of Gastroenterology "S. De Bellis", Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Carla Cossu
- National Institute of Gastroenterology "S. De Bellis", Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Serena Mancarella
- National Institute of Gastroenterology "S. De Bellis", Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Gianluigi Giannelli
- National Institute of Gastroenterology "S. De Bellis", Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| |
Collapse
|
24
|
A familial congenital heart disease with a possible multigenic origin involving a mutation in BMPR1A. Sci Rep 2019; 9:2959. [PMID: 30814609 PMCID: PMC6393482 DOI: 10.1038/s41598-019-39648-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 01/28/2019] [Indexed: 12/12/2022] Open
Abstract
The genetics of many congenital heart diseases (CHDs) can only unsatisfactorily be explained by known chromosomal or Mendelian syndromes. Here, we present sequencing data of a family with a potentially multigenic origin of CHD. Twelve of nineteen family members carry a familial mutation [NM_004329.2:c.1328 G > A (p.R443H)] which encodes a predicted deleterious variant of BMPR1A. This mutation co-segregates with a linkage region on chromosome 1 that associates with the emergence of severe CHDs including Ebstein's anomaly, atrioventricular septal defect, and others. We show that the continuous overexpression of the zebrafish homologous mutation bmpr1aap.R438H within endocardium causes a reduced AV valve area, a downregulation of Wnt/ß-catenin signalling at the AV canal, and growth of additional tissue mass in adult zebrafish hearts. This finding opens the possibility of testing genetic interactions between BMPR1A and other candidate genes within linkage region 1 which may provide a first step towards unravelling more complex genetic patterns in cardiovascular disease aetiology.
Collapse
|
25
|
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: 1.3] [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
|
26
|
Gomez-Puerto MC, Iyengar PV, García de Vinuesa A, Ten Dijke P, Sanchez-Duffhues G. Bone morphogenetic protein receptor signal transduction in human disease. J Pathol 2018; 247:9-20. [PMID: 30246251 PMCID: PMC6587955 DOI: 10.1002/path.5170] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/03/2018] [Accepted: 09/13/2018] [Indexed: 12/23/2022]
Abstract
Bone morphogenetic proteins (BMPs) are secreted cytokines that were initially discovered on the basis of their ability to induce bone. Several decades of research have now established that these proteins function in a large variety of physiopathological processes. There are about 15 BMP family members, which signal via three transmembrane type II receptors and four transmembrane type I receptors. Mechanistically, BMP binding leads to phosphorylation of the type I receptor by the type II receptor. This activated heteromeric complex triggers intracellular signaling that is initiated by phosphorylation of receptor‐regulated SMAD1, 5, and 8 (also termed R‐SMADs). Activated R‐SMADs form heteromeric complexes with SMAD4, which engage in specific transcriptional responses. There is convergence along the signaling pathway and, besides the canonical SMAD pathway, BMP‐receptor activation can also induce non‐SMAD signaling. Each step in the pathway is fine‐tuned by positive and negative regulation and crosstalk with other signaling pathways. For example, ligand bioavailability for the receptor can be regulated by ligand‐binding proteins that sequester the ligand from interacting with receptors. Accessory co‐receptors, also known as BMP type III receptors, lack intrinsic enzymatic activity but enhance BMP signaling by presenting ligands to receptors. In this review, we discuss the role of BMP receptor signaling and how corruption of this pathway contributes to cardiovascular and musculoskeletal diseases and cancer. We describe pharmacological tools to interrogate the function of BMP receptor signaling in specific biological processes and focus on how these agents can be used as drugs to inhibit or activate the function of the receptor, thereby normalizing dysregulated BMP signaling. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Maria Catalina Gomez-Puerto
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Prasanna Vasudevan Iyengar
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Amaya García de Vinuesa
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Gonzalo Sanchez-Duffhues
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
27
|
Yao R, Yu T, Xu Y, Li G, Yin L, Zhou Y, Wang J, Yan Z. Concurrent somatic KRAS mutation and germline 10q22.3-q23.2 deletion in a patient with juvenile myelomonocytic leukemia, developmental delay, and multiple malformations: a case report. BMC Med Genomics 2018; 11:60. [PMID: 30012129 PMCID: PMC6048798 DOI: 10.1186/s12920-018-0377-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/03/2018] [Indexed: 11/10/2022] Open
Abstract
Background The proto-oncogene KRAS performs an essential function in normal tissue signaling, and the mutation of KRAS gene is a key step in the development of many cancers. Somatic KRAS mutations are often detected in patients with solid and non-solid tumors, whereas germline KRAS mutations are implicated in patients with the Noonan syndrome, cardio-facio-cutaneous (CFC) syndrome and Costello syndrome. The deletion of chromosome 10q22.3-q23.2 is a rare cytogenetic abnormality, which often leads to distinct facial appearance and delays in speech and global development. Case presentation Herein, we report the case of a 4-year-old boy diagnosed with juvenile myelomonocytic leukemia. The boy also had syndromic features, such as speech and motor developmental delay, multiple congenital malformations, including distinct facial features, club feet, and cryptorchidism. Using whole-exome sequencing, we identified a pathogenic mutation in KRAS [c.34G > A, p.Gly12Ser] isolated from peripheral blood DNA. Sanger sequencing confirmed the wild-type sequence in the parents and patient’s salivary cell DNA indicating its somatic state. A 7311-kb deletion in 10q22.3-q23.2 was also revealed by chromosomal microarray analysis, which was later proved as a germline de novo variant. Conclusion Juvenile myelomonocytic leukemia in the patient was attributed to a somatic KRAS mutation, whereas the syndromic features of the patient were considered a consequence of germline chromosome 10q22.3-q23.2 deletion. Genetic testing for patients with complicated phenotypes can be valuable in detecting multiple pathogenic variants.
Collapse
Affiliation(s)
- Ruen Yao
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China.,Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Tingting Yu
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China. .,Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, People's Republic of China.
| | - Yufei Xu
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China.,Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Guoqiang Li
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China.,Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Lei Yin
- Department of Internal Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, People's Republic of China.,Rare Diseases Outpatient Clinic, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Yunfang Zhou
- Rare Diseases Outpatient Clinic, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Jian Wang
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China.,Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Zhilong Yan
- Department of Pediatric Surgery, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
28
|
Abstract
Ventricular myocardial development is a well-orchestrated process involving different cardiac structures, multiple signal pathways, and myriad proteins. Dysregulation of this important developmental event can result in cardiomyopathies, such as left ventricle non-compaction, which affect the pediatric population and the adults. Human and mouse studies have shed light upon the etiology of some cardiomyopathy cases and highlighted the contribution of both genetic and environmental factors. However, the regulation of ventricular myocardial development remains incompletely understood. Zinc is an essential trace metal with structural, enzymatic, and signaling function. Perturbation of zinc homeostasis has resulted in developmental and physiological defects including cardiomyopathy. In this review, we summarize several mechanisms by which zinc and zinc transporters can impact the regulation of ventricular myocardial development. Based on our review, we propose that zinc deficiency and mutations of zinc transporters may underlie some cardiomyopathy cases especially those involving ventricular myocardial development defects.
Collapse
Affiliation(s)
- Wen Lin
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Deqiang Li
- Division of Cardiac Surgery, School of Medicine, University of Maryland, 800 West Baltimore ST, Rm 314, Baltimore, MD, 21201, USA.
| |
Collapse
|
29
|
Zhang M, Zhang Y, Xu E, Mohibi S, de Anda DM, Jiang Y, Zhang J, Chen X. Rbm24, a target of p53, is necessary for proper expression of p53 and heart development. Cell Death Differ 2018; 25:1118-1130. [PMID: 29358667 PMCID: PMC5988652 DOI: 10.1038/s41418-017-0029-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/29/2017] [Accepted: 10/19/2017] [Indexed: 01/08/2023] Open
Abstract
Activation of p53-dependent apoptosis is critical for tumor suppression but aberrant activation of p53 also leads to developmental defects and heart failure. Here, we found that Rbm24 RNA-binding protein, a target of p53, regulates p53 mRNA translation. Mechanistically, we found that through binding to p53 mRNA and interaction with translation initiation factor eIF4E, Rbm24 prevents eIF4E from binding to p53 mRNA and inhibits the assembly of translation initiation complex. Importantly, we showed that mice deficient in Rbm24 die in utero due to the endocardial cushion defect in the heart at least in part due to aberrant activation of p53-dependent apoptosis. We also showed that the heart developmental defect in Rbm24-null mice can be partially rescued by p53 deficiency through decreased apoptosis in the heart. Together, we postulate that the p53-Rbm24 loop is critical for the heart development and may be explored for mitigating congenital heart diseases and heart failure.
Collapse
Affiliation(s)
- Min Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yanhong Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Enshun Xu
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Shakur Mohibi
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Danielle Michelle de Anda
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Yuqian Jiang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA.
| |
Collapse
|
30
|
Ahmed A, Wang T, Delgado-Olguin P. Ezh2 is not required for cardiac regeneration in neonatal mice. PLoS One 2018; 13:e0192238. [PMID: 29466371 PMCID: PMC5821314 DOI: 10.1371/journal.pone.0192238] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 01/18/2018] [Indexed: 12/22/2022] Open
Abstract
The neonatal mouse heart has the remarkable capacity to regenerate lost myocardium within the first week of life. Neonatal cardiomyocytes re-express fetal genes that control cell proliferation after injury to promote regeneration. The loss of regenerative capacity of the heart one week after birth coincides with repression of a fetal transcriptional program coordinated by epigenetic regulators. The histone methyltransferase enhancer of zeste homolog 2 (Ezh2) is a repressor of fetal cardiac transcriptional programs and suppresses cardiomyocyte cell proliferation, suggesting a potential function in heart regeneration. However, it was recently demonstrated that Ezh2 is dispensable for heart regeneration in the neonatal heart. Here, we provide evidence supporting this finding and demonstrate that Ezh2 deficiency does not affect regeneration of the neonatal heart. We inactivated Ezh2 in differentiating embryonic cardiomyocytes, which led to depletion of histone H3 trimethylated at lysine 27 (H3K27me3). Ezh2 deficiency in cardiomyocytes did not affect clearance of the fibrotic scar in myocardial infarction (MI) and apical resection models of cardiac injury at post-natal day 1 (P1). Similarly, cardiomyocyte-specific loss of Ezh2 did not affect fibrotic scar size after MI or apical resection at P7, suggesting that it does not extend the regenerative time window. Our results demonstrate that Ezh2 is not required for innate neonatal cardiac regeneration.
Collapse
Affiliation(s)
- Abdalla Ahmed
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Tao Wang
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Human Biology Program, University of Toronto, Toronto, Ontario, Canada
| | - Paul Delgado-Olguin
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Heart & Stroke Richard Lewar Centre of Excellence, Toronto, ON, Canada
- * E-mail:
| |
Collapse
|
31
|
Sommakia S, Houlihan PR, Deane SS, Simcox JA, Torres NS, Jeong MY, Winge DR, Villanueva CJ, Chaudhuri D. Mitochondrial cardiomyopathies feature increased uptake and diminished efflux of mitochondrial calcium. J Mol Cell Cardiol 2017; 113:22-32. [PMID: 28962857 PMCID: PMC5652072 DOI: 10.1016/j.yjmcc.2017.09.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 09/07/2017] [Accepted: 09/25/2017] [Indexed: 12/26/2022]
Abstract
Calcium (Ca2+) influx into the mitochondrial matrix stimulates ATP synthesis. Here, we investigate whether mitochondrial Ca2+ transport pathways are altered in the setting of deficient mitochondrial energy synthesis, as increased matrix Ca2+ may provide a stimulatory boost. We focused on mitochondrial cardiomyopathies, which feature such dysfunction of oxidative phosphorylation. We study a mouse model where the main transcription factor for mitochondrial DNA (transcription factor A, mitochondrial, Tfam) has been disrupted selectively in cardiomyocytes. By the second postnatal week (10-15day old mice), these mice have developed a dilated cardiomyopathy associated with impaired oxidative phosphorylation. We find evidence of increased mitochondrial Ca2+ during this period using imaging, electrophysiology, and biochemistry. The mitochondrial Ca2+ uniporter, the main portal for Ca2+ entry, displays enhanced activity, whereas the mitochondrial sodium-calcium (Na+-Ca2+) exchanger, the main portal for Ca2+ efflux, is inhibited. These changes in activity reflect changes in protein expression of the corresponding transporter subunits. While decreased transcription of Nclx, the gene encoding the Na+-Ca2+ exchanger, explains diminished Na+-Ca2+ exchange, the mechanism for enhanced uniporter expression appears to be post-transcriptional. Notably, such changes allow cardiac mitochondria from Tfam knockout animals to be far more sensitive to Ca2+-induced increases in respiration. In the absence of Ca2+, oxygen consumption declines to less than half of control values in these animals, but rebounds to control levels when incubated with Ca2+. Thus, we demonstrate a phenotype of enhanced mitochondrial Ca2+ in a mitochondrial cardiomyopathy model, and show that such Ca2+ accumulation is capable of rescuing deficits in energy synthesis capacity in vitro.
Collapse
Affiliation(s)
- Salah Sommakia
- Nora Eccles Harrison Cardiovascular Research and Training Institute, Cardiology Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Patrick R Houlihan
- Department of Cardiology, Boston Children's Hospital, Boston, MA, United States
| | - Sadiki S Deane
- Nora Eccles Harrison Cardiovascular Research and Training Institute, Cardiology Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Judith A Simcox
- Department of Biochemistry, University of Utah, Salt Lake City, UT, United States
| | - Natalia S Torres
- Nora Eccles Harrison Cardiovascular Research and Training Institute, Cardiology Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Mi-Young Jeong
- Department of Biochemistry, University of Utah, Salt Lake City, UT, United States; Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Dennis R Winge
- Department of Biochemistry, University of Utah, Salt Lake City, UT, United States; Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Claudio J Villanueva
- Department of Biochemistry, University of Utah, Salt Lake City, UT, United States
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute, Cardiology Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
32
|
Vijayan V, Gupta S, Gupta S. Bone morphogenetic protein-5, a key molecule that mediates differentiation in MC3T3E1 osteoblast cell line. Biofactors 2017; 43:558-566. [PMID: 28497879 DOI: 10.1002/biof.1360] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/01/2017] [Accepted: 03/19/2017] [Indexed: 01/17/2023]
Abstract
Bone morphogenetic protein-5 (BMP-5) is a member of the TGF receptor-β family with osteoinductive property. However, its physiological role in osteoblast differentiation is not defined. This study highlights the importance of BMP-5 in MC3T3E1 osteoblast differentiation. Pre-osteoblasts exposed to osteogenic media (ascorbic acid, 50 µg/ml and β-glycerophosphate, 10 mM) showed high protein expression of BMP-5 in cell lysates and cell culture supernatants, which peaked during early time-points of differentiation and declined with onset of mineralization. Attenuation of endogenous BMP-5 protein expression by RNA interference downregulated the expression of type I collagen (COLIA1), an early osteoblast differentiation marker but not osteocalcin, a late osteoblast differentiation marker. Further experiments to analyze the cell signaling components revealed that BMP-5 modulates COLIA1 expression via p38-Runx2 axis involving Runx2 (Ser19) phosphorylation. These effects were also observed when recombinant BMP-5 was added to pre-osteoblast cultures reinforcing the fact that BMP-5 is a modulator of COLIA1 expression. We conclude that BMP-5 has stage-specific role to play during MC3T3E1 osteoblast differentiation in part by autocrine p38/Runx2/COLIA1 signaling. © 2017 BioFactors, 43(4):558-566, 2017.
Collapse
Affiliation(s)
- Viji Vijayan
- Molecular Sciences Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110 067, India
| | - Sakshi Gupta
- Molecular Sciences Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110 067, India
| | - Sarika Gupta
- Molecular Sciences Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110 067, India
| |
Collapse
|
33
|
Shi W, Wang H, Zheng X, Jiang X, Xu Z, Shen H, Li M. HNF-4alpha Negatively Regulates Hepcidin Expression Through BMPR1A in HepG2 Cells. Biol Trace Elem Res 2017; 176:294-304. [PMID: 27660075 DOI: 10.1007/s12011-016-0846-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/06/2016] [Indexed: 12/19/2022]
Abstract
Hepcidin synthesis is reported to be inadequate according to the body iron store in patients with non-alcoholic fatty liver disease (NAFLD) undergoing hepatic iron overload (HIO). However, the underlying mechanisms remain unclear. We hypothesize that hepatocyte nuclear factor-4α (HNF-4α) may negatively regulate hepcidin expression and contribute to hepcidin deficiency in NAFLD patients. The effect of HNF-4α on hepcidin expression was observed by transfecting specific HNF-4α small interfering RNA (siRNA) or plasmids into HepG2 cells. Both direct and indirect mechanisms involved in the regulation of HNF-4α on hepcidin were detected by real-time PCR, Western blotting, chromatin immunoprecipitation (chIP), and reporter genes. It was found that HNF-4α suppressed hepcidin messenger RNA (mRNA) and protein expressions in HepG2 cells, and this suppressive effect was independent of the potential HNF-4α response elements. Phosphorylation of SMAD1 but not STAT3 was inactivated by HNF-4α, and the SMAD4 response element was found essential to HNF-4α-induced hepcidin reduction. Neither inhibitory SMADs, SMAD6, and SMAD7 nor BMPR ligands, BMP2, BMP4, BMP6, and BMP7 were regulated by HNF-4α in HepG2 cells. BMPR1A, but not BMPR1B, BMPR2, ActR2A, ActR2B, or HJV, was decreased by HNF-4α, and HNF4α-knockdown-induced stimulation of hepcidin could be entirely blocked when BMPR1A was interfered with at the same time. In conclusion, the present study suggests that HNF-4α has a suppressive effect on hepcidin expression by inactivating the BMP pathway, specifically via BMPR1A, in HepG2 cells.
Collapse
Affiliation(s)
- Wencai Shi
- Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, No. 800 Xiangyin Road, Shanghai, 200433, China
- Department of Clinical Nutrition, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Heyang Wang
- Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, No. 800 Xiangyin Road, Shanghai, 200433, China
| | - Xuan Zheng
- Department of Clinical Nutrition, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xin Jiang
- Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, No. 800 Xiangyin Road, Shanghai, 200433, China
| | - Zheng Xu
- Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, No. 800 Xiangyin Road, Shanghai, 200433, China
| | - Hui Shen
- Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, No. 800 Xiangyin Road, Shanghai, 200433, China
| | - Min Li
- Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, No. 800 Xiangyin Road, Shanghai, 200433, China.
| |
Collapse
|
34
|
Bolar N, Verstraeten A, Van Laer L, Loeys B. Molecular Insights into Bicuspid Aortic Valve Development and the associated aortopathy. AIMS MOLECULAR SCIENCE 2017. [DOI: 10.3934/molsci.2017.4.478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
35
|
Yu Z, Mouillesseaux KP, Kushner EJ, Bautch VL. Tumor-Derived Factors and Reduced p53 Promote Endothelial Cell Centrosome Over-Duplication. PLoS One 2016; 11:e0168334. [PMID: 27977771 PMCID: PMC5158050 DOI: 10.1371/journal.pone.0168334] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/30/2016] [Indexed: 01/28/2023] Open
Abstract
Approximately 30% of tumor endothelial cells have over-duplicated (>2) centrosomes, which may contribute to abnormal vessel function and drug resistance. Elevated levels of vascular endothelial growth factor A induce excess centrosomes in endothelial cells, but how other features of the tumor environment affect centrosome over-duplication is not known. To test this, we treated endothelial cells with tumor-derived factors, hypoxia, or reduced p53, and assessed centrosome numbers. We found that hypoxia and elevated levels of bone morphogenetic protein 2, 6 and 7 induced excess centrosomes in endothelial cells through BMPR1A and likely via SMAD signaling. In contrast, inflammatory mediators IL-8 and lipopolysaccharide did not induce excess centrosomes. Finally, down-regulation in endothelial cells of p53, a critical regulator of DNA damage and proliferation, caused centrosome over-duplication. Our findings suggest that some tumor-derived factors and genetic changes in endothelial cells contribute to excess centrosomes in tumor endothelial cells.
Collapse
Affiliation(s)
- Zhixian Yu
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Kevin P. Mouillesseaux
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Erich J. Kushner
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Victoria L. Bautch
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
36
|
Kong GWS, Cao Y, Huang J, Cheng KY, Pursley AN, Rosenfeld JA, Edwards JG, Chan YM, Cheung SW, Leung TY, Choy KW. Prenatal detection of 10q22q23 duplications: dilemmas in phenotype prediction. Prenat Diagn 2016; 36:1211-1216. [PMID: 27859473 DOI: 10.1002/pd.4959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/31/2016] [Accepted: 10/31/2016] [Indexed: 11/08/2022]
Abstract
OBJECTIVES The phenotype for 10q22q23 duplication is diverse, ranging from intellectual disability and dysmorphism to normal development. Interpreting the clinical significance of the duplication identified in this region is difficult, especially in the prenatal setting. This study aimed to characterize the prenatal findings associated with this submicroscopic imbalance and discuss the dilemmas in predicting the phenotype of 10q22q23 duplications. METHODS This is a retrospective study of three cases of 10q22q23 duplications diagnosed prenatally by chromosomal microarray analysis. Detailed pregnancy outcome and pediatric follow-up were documented. RESULTS The genotypic and phenotypic features of the reported cases were discussed. 10q22q23 duplications are associated with an unpredictable and variable phenotypic outcome. Despite there was no phenotype found to be shared by 50% of the duplication cases, congenital heart defects, hypotelorism, and developmental delays including speech and motor delay seem to be more common. CONCLUSIONS The phenotype of 10q22q23 duplication is highly variable prenatally and postnatally. Identification of additional affected individuals with similar duplications is needed to provide further insights into the pathogenesis of this microduplication. © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Grace Wing Shan Kong
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong
| | - Ye Cao
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Jin Huang
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong
| | - Kwun Yue Cheng
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong
| | - Amber Nolen Pursley
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | - Janice G Edwards
- Genetic Counseling Program, University of South Carolina, Columbia, SC, USA
| | - Yiu Man Chan
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong
| | - Sau Wai Cheung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Tak Yeung Leung
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.,Hong Kong Branches of Chinese National Engineering Research Centers-Center for Assisted Reproductive Technology and Reproductive Genetics, The Chinese University of Hong Kong, Hong Kong
| | - Kwong Wai Choy
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.,Hong Kong Branches of Chinese National Engineering Research Centers-Center for Assisted Reproductive Technology and Reproductive Genetics, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
37
|
Beets K, Staring MW, Criem N, Maas E, Schellinx N, de Sousa Lopes SMC, Umans L, Zwijsen A. BMP-SMAD signalling output is highly regionalized in cardiovascular and lymphatic endothelial networks. BMC DEVELOPMENTAL BIOLOGY 2016; 16:34. [PMID: 27724845 PMCID: PMC5057272 DOI: 10.1186/s12861-016-0133-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 09/12/2016] [Indexed: 11/13/2022]
Abstract
BACKGROUND Bone morphogenetic protein (BMP) signalling has emerged as a fundamental pathway in endothelial cell biology and deregulation of this pathway is implicated in several vascular disorders. BMP signalling output in endothelial cells is highly context- and dose-dependent. Phosphorylation of the BMP intracellular effectors, SMAD1/5/9, is routinely used to monitor BMP signalling activity. To better understand the in vivo context-dependency of BMP-SMAD signalling, we investigated differences in BMP-SMAD transcriptional activity in different vascular beds during mouse embryonic and postnatal stages. For this, we used the BRE::gfp BMP signalling reporter mouse in which the BMP response element (BRE) from the ID1-promotor, a SMAD1/5/9 target gene, drives the expression of GFP. RESULTS A mosaic pattern of GFP was present in various angiogenic sprouting plexuses and in endocardium of cardiac cushions and trabeculae in the heart. High calibre veins seemed to be more BRE::gfp transcriptionally active than arteries, and ubiquitous activity was present in embryonic lymphatic vasculature. Postnatal lymphatic vessels showed however only discrete micro-domains of transcriptional activity. Dynamic shifts in transcriptional activity were also observed in the endocardium of the developing heart, with a general decrease in activity over time. Surprisingly, proliferative endothelial cells were almost never GFP-positive. Patches of transcriptional activity seemed to correlate with vasculature undergoing hemodynamic alterations. CONCLUSION The BRE::gfp mouse allows to investigate selective context-dependent aspects of BMP-SMAD signalling. Our data reveals the highly dynamic nature of BMP-SMAD mediated transcriptional regulation in time and space throughout the vascular tree, supporting that BMP-SMAD signalling can be a source of phenotypic diversity in some, but not all, healthy endothelium. This knowledge can provide insight in vascular bed or organ-specific diseases and phenotypic heterogeneity within an endothelial cell population.
Collapse
Affiliation(s)
- Karen Beets
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Michael W. Staring
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Nathan Criem
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Elke Maas
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Niels Schellinx
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | | | - Lieve Umans
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - An Zwijsen
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|
38
|
Morrell NW, Bloch DB, ten Dijke P, Goumans MJTH, Hata A, Smith J, Yu PB, Bloch KD. Targeting BMP signalling in cardiovascular disease and anaemia. Nat Rev Cardiol 2016; 13:106-20. [PMID: 26461965 PMCID: PMC4886232 DOI: 10.1038/nrcardio.2015.156] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone morphogenetic proteins (BMPs) and their receptors, known to be essential regulators of embryonic patterning and organogenesis, are also critical for the regulation of cardiovascular structure and function. In addition to their contributions to syndromic disorders including heart and vascular development, BMP signalling is increasingly recognized for its influence on endocrine-like functions in postnatal cardiovascular and metabolic homeostasis. In this Review, we discuss several critical and novel aspects of BMP signalling in cardiovascular health and disease, which highlight the cell-specific and context-specific nature of BMP signalling. Based on advancing knowledge of the physiological roles and regulation of BMP signalling, we indicate opportunities for therapeutic intervention in a range of cardiovascular conditions including atherosclerosis and pulmonary arterial hypertension, as well as for anaemia of inflammation. Depending on the context and the repertoire of ligands and receptors involved in specific disease processes, the selective inhibition or enhancement of signalling via particular BMP ligands (such as in atherosclerosis and pulmonary arterial hypertension, respectively) might be beneficial. The development of selective small molecule antagonists of BMP receptors, and the identification of ligands selective for BMP receptor complexes expressed in the vasculature provide the most immediate opportunities for new therapies.
Collapse
Affiliation(s)
- Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Donald B Bloch
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Peter ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medicine Centre, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Marie-Jose T H Goumans
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medicine Centre, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Akiko Hata
- Cardiovascular Research Institute, University of California, 500 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Jim Smith
- MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Paul B Yu
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Kenneth D Bloch
- Anaesthesia Centre for Critical Care Research, Department of Anaesthesia, Critical Care and Pain Medicine, 55 Fruit Street, Boston, MA 02114, USA
| |
Collapse
|
39
|
Wu Y, Zhou X, Huang X, Xia Q, Chen Z, Zhang X, Yang D, Geng YJ. Pax8 plays a pivotal role in regulation of cardiomyocyte growth and senescence. J Cell Mol Med 2016; 20:644-54. [PMID: 26781745 PMCID: PMC5125375 DOI: 10.1111/jcmm.12779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 12/05/2015] [Indexed: 12/18/2022] Open
Abstract
Congenital heart disease (CHD) is a worldwide health problem, particularly in young populations. In spite of the advancement and progress in medical research and technology, the underlying causative factors and mechanisms of CHD still remain unclear. Bone morphogenetic protein receptor IA (ALK3) mediates the development of ventricular septal defect (VSD). We have recently found that paired box gene 8 (Pax8) may be the downstream molecule of ALK3. Paired box gene 8 plays an essential role in VSD, and apoptosis and proliferation imbalance leads to septal dysplasia. Recent studies have also disclosed that cellular senescence also participates in embryonic development. Whether programmed senescence exists in cardiac organogenesis has not ever been reported. We hypothesized that together with various biological processes, such as apoptosis, enhanced cellular senescence may occur actively in the development of Pax8 null mice murine hearts. In H9C2 myogenic cells, Pax8 overexpression can rescue caspase‐dependent apoptosis induced by ALK3 silencing. Senescent cells and senescence‐associated mediators in Pax8 knockout hearts increased compared with the wild‐type ones in an age‐dependent manner. These results suggest that Pax8 maybe the downstream molecule of ALK3, it mediates the murine heart development perhaps via cellular senescence, which may serve as a mechanism that compensates for the cell loss via apoptosis in heart development.
Collapse
Affiliation(s)
- Yihao Wu
- Division of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xi Zhou
- Division of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoyan Huang
- Division of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Quan Xia
- Division of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Zhe Chen
- Division of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Xingwei Zhang
- Division of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Deye Yang
- Division of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Division of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Yong-jian Geng
- The University of Texas School of Medicine at Houston, Houston, TX, USA
| |
Collapse
|
40
|
Ayuzawa N, Nagase M, Ueda K, Nishimoto M, Kawarazaki W, Marumo T, Aiba A, Sakurai T, Shindo T, Fujita T. Rac1-Mediated Activation of Mineralocorticoid Receptor in Pressure Overload–Induced Cardiac Injury. Hypertension 2016; 67:99-106. [DOI: 10.1161/hypertensionaha.115.06054] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/09/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Nobuhiro Ayuzawa
- From the Division of Clinical Epigenetics, Research Center for Advanced Science and Technology (N.A., K.U., M. Nishimoto, W.K., T.M., T.F.), and Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine (A.A.), The University of Tokyo, Tokyo, Japan; Department of Anatomy and Life Structure, Faculty of Medicine, Juntendo University, Tokyo, Japan (M. Nagase); CREST, Japan Science and Technology Agency, Tokyo, Japan (T.M., T.F.); and Department of
| | - Miki Nagase
- From the Division of Clinical Epigenetics, Research Center for Advanced Science and Technology (N.A., K.U., M. Nishimoto, W.K., T.M., T.F.), and Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine (A.A.), The University of Tokyo, Tokyo, Japan; Department of Anatomy and Life Structure, Faculty of Medicine, Juntendo University, Tokyo, Japan (M. Nagase); CREST, Japan Science and Technology Agency, Tokyo, Japan (T.M., T.F.); and Department of
| | - Kohei Ueda
- From the Division of Clinical Epigenetics, Research Center for Advanced Science and Technology (N.A., K.U., M. Nishimoto, W.K., T.M., T.F.), and Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine (A.A.), The University of Tokyo, Tokyo, Japan; Department of Anatomy and Life Structure, Faculty of Medicine, Juntendo University, Tokyo, Japan (M. Nagase); CREST, Japan Science and Technology Agency, Tokyo, Japan (T.M., T.F.); and Department of
| | - Mitsuhiro Nishimoto
- From the Division of Clinical Epigenetics, Research Center for Advanced Science and Technology (N.A., K.U., M. Nishimoto, W.K., T.M., T.F.), and Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine (A.A.), The University of Tokyo, Tokyo, Japan; Department of Anatomy and Life Structure, Faculty of Medicine, Juntendo University, Tokyo, Japan (M. Nagase); CREST, Japan Science and Technology Agency, Tokyo, Japan (T.M., T.F.); and Department of
| | - Wakako Kawarazaki
- From the Division of Clinical Epigenetics, Research Center for Advanced Science and Technology (N.A., K.U., M. Nishimoto, W.K., T.M., T.F.), and Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine (A.A.), The University of Tokyo, Tokyo, Japan; Department of Anatomy and Life Structure, Faculty of Medicine, Juntendo University, Tokyo, Japan (M. Nagase); CREST, Japan Science and Technology Agency, Tokyo, Japan (T.M., T.F.); and Department of
| | - Takeshi Marumo
- From the Division of Clinical Epigenetics, Research Center for Advanced Science and Technology (N.A., K.U., M. Nishimoto, W.K., T.M., T.F.), and Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine (A.A.), The University of Tokyo, Tokyo, Japan; Department of Anatomy and Life Structure, Faculty of Medicine, Juntendo University, Tokyo, Japan (M. Nagase); CREST, Japan Science and Technology Agency, Tokyo, Japan (T.M., T.F.); and Department of
| | - Atsu Aiba
- From the Division of Clinical Epigenetics, Research Center for Advanced Science and Technology (N.A., K.U., M. Nishimoto, W.K., T.M., T.F.), and Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine (A.A.), The University of Tokyo, Tokyo, Japan; Department of Anatomy and Life Structure, Faculty of Medicine, Juntendo University, Tokyo, Japan (M. Nagase); CREST, Japan Science and Technology Agency, Tokyo, Japan (T.M., T.F.); and Department of
| | - Takayuki Sakurai
- From the Division of Clinical Epigenetics, Research Center for Advanced Science and Technology (N.A., K.U., M. Nishimoto, W.K., T.M., T.F.), and Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine (A.A.), The University of Tokyo, Tokyo, Japan; Department of Anatomy and Life Structure, Faculty of Medicine, Juntendo University, Tokyo, Japan (M. Nagase); CREST, Japan Science and Technology Agency, Tokyo, Japan (T.M., T.F.); and Department of
| | - Takayuki Shindo
- From the Division of Clinical Epigenetics, Research Center for Advanced Science and Technology (N.A., K.U., M. Nishimoto, W.K., T.M., T.F.), and Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine (A.A.), The University of Tokyo, Tokyo, Japan; Department of Anatomy and Life Structure, Faculty of Medicine, Juntendo University, Tokyo, Japan (M. Nagase); CREST, Japan Science and Technology Agency, Tokyo, Japan (T.M., T.F.); and Department of
| | - Toshiro Fujita
- From the Division of Clinical Epigenetics, Research Center for Advanced Science and Technology (N.A., K.U., M. Nishimoto, W.K., T.M., T.F.), and Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine (A.A.), The University of Tokyo, Tokyo, Japan; Department of Anatomy and Life Structure, Faculty of Medicine, Juntendo University, Tokyo, Japan (M. Nagase); CREST, Japan Science and Technology Agency, Tokyo, Japan (T.M., T.F.); and Department of
| |
Collapse
|
41
|
Briggs LE, Burns TA, Lockhart MM, Phelps AL, Van den Hoff MJB, Wessels A. Wnt/β-catenin and sonic hedgehog pathways interact in the regulation of the development of the dorsal mesenchymal protrusion. Dev Dyn 2015; 245:103-13. [PMID: 26297872 DOI: 10.1002/dvdy.24339] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 07/29/2015] [Accepted: 08/18/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The dorsal mesenchymal protrusion (DMP) is a second heart field (SHF) derived tissue involved in cardiac septation. Molecular mechanisms controlling SHF/DMP development include the Bone Morphogenetic Protein and Wnt/β-catenin signaling pathways. Reduced expression of components in these pathways leads to inhibition of proliferation of the SHF/DMP precursor population and failure of the DMP to develop. While the Sonic Hedgehog (Shh) pathway has also been demonstrated to be critically important for SHF/DMP development and atrioventricular septation, its role in the regulation of SHF proliferation is contentious. RESULTS Tissue-specific deletion of the Shh receptor Smoothened from the SHF resulted in compromised DMP formation and atrioventricular septal defects (AVSDs). Immunohistochemical analysis at critical stages of DMP development showed significant proliferation defect as well as reduction in levels of the Wnt/β-catenin pathway-intermediates β-catenin, Lef1, and Axin2. To determine whether the defects seen in the conditional Smoothened knock-out mouse could be attributed to reduced Wnt/β-catenin signaling, LiCl, a pharmacological activator of this Wnt/β-catenin pathway, was administered. This resulted in restoration of proliferation and partial rescue of the AVSD phenotype. CONCLUSIONS The data presented suggest that the Wnt/β-catenin pathway interact with the Shh pathway in the regulation of SHF/DMP-precursor proliferation and, hence, the development of the DMP.
Collapse
Affiliation(s)
- Laura E Briggs
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Tara A Burns
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Marie M Lockhart
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Aimee L Phelps
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Maurice J B Van den Hoff
- Heart Failure Research Center, Department of Anatomy, Embryology and Physiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Andy Wessels
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
42
|
Fang X, Wang H, An P, Min J, Wang F. Cardiomyocyte-specific deletion of ferroportin using MCK-Cre has no apparent effect on cardiac iron homeostasis. Int J Cardiol 2015; 201:90-2. [DOI: 10.1016/j.ijcard.2015.07.089] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/29/2015] [Indexed: 01/15/2023]
|
43
|
Martin PS, Kloesel B, Norris RA, Lindsay M, Milan D, Body SC. Embryonic Development of the Bicuspid Aortic Valve. J Cardiovasc Dev Dis 2015; 2:248-272. [PMID: 28529942 PMCID: PMC5438177 DOI: 10.3390/jcdd2040248] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bicuspid aortic valve (BAV) is the most common congenital valvular heart defect with an overall frequency of 0.5%–1.2%. BAVs result from abnormal aortic cusp formation during valvulogenesis, whereby adjacent cusps fuse into a single large cusp resulting in two, instead of the normal three, aortic cusps. Individuals with BAV are at increased risk for ascending aortic disease, aortic stenosis and coarctation of the aorta. The frequent occurrence of BAV and its anatomically discrete but frequent co-existing diseases leads us to suspect a common cellular origin. Although autosomal-dominant transmission of BAV has been observed in a few pedigrees, notably involving the gene NOTCH1, no single-gene model clearly explains BAV inheritance, implying a complex genetic model involving interacting genes. Several sequencing studies in patients with BAV have identified rare and uncommon mutations in genes of cardiac embryogenesis. But the extensive cell-cell signaling and multiple cellular origins involved in cardiac embryogenesis preclude simplistic explanations of this disease. In this review, we examine the series of events from cellular and transcriptional embryogenesis of the heart, to development of the aortic valve.
Collapse
Affiliation(s)
- Peter S. Martin
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St., Th724, Boston, MA 02115, USA; E-Mails: (P.S.M.); (B.K.)
| | - Benjamin Kloesel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St., Th724, Boston, MA 02115, USA; E-Mails: (P.S.M.); (B.K.)
| | - Russell A. Norris
- Department of Regenerative Medicine and Cell Biology, Children’s Research Institute, Medical University of South Carolina, 173 Ashley St, Charleston, SC 29403, USA; E-Mail:
| | - Mark Lindsay
- Cardiovascular Research Center, Richard B. Simches Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; E-Mails: (M.L.); (D.M.)
| | - David Milan
- Cardiovascular Research Center, Richard B. Simches Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; E-Mails: (M.L.); (D.M.)
| | - Simon C. Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St., Th724, Boston, MA 02115, USA; E-Mails: (P.S.M.); (B.K.)
- Author to whom correspondence should be addressed: E-Mail: ; Tel.: +1-617-732-7330; Fax: +1-617-730-2813
| |
Collapse
|
44
|
Tang M, Yang YF, Xie L, Chen JL, Zhang WZ, Wang J, Zhao TL, Yang JF, Tan ZP. Duplication of 10q22.3-q23.3 encompassingBMPR1AandNGR3associated with congenital heart disease, microcephaly, and mild intellectual disability. Am J Med Genet A 2015; 167A:3174-9. [PMID: 26383923 DOI: 10.1002/ajmg.a.37347] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 08/10/2015] [Indexed: 12/30/2022]
Affiliation(s)
- Mi Tang
- Department of Cardiothoracic Surgery; The Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Yi-Feng Yang
- Department of Cardiothoracic Surgery; The Second Xiangya Hospital; Central South University; Changsha Hunan China
- Clinical Center for Gene Diagnosis and Therapy of State Key Laboratory of Medical Genetics; The Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Li Xie
- Department of Cardiothoracic Surgery; The Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Jin-Lan Chen
- Department of Cardiothoracic Surgery; The Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Wei-Zhi Zhang
- Department of Cardiothoracic Surgery; The Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Jian Wang
- Department of Cardiothoracic Surgery; The Second Xiangya Hospital; Central South University; Changsha Hunan China
- Clinical Center for Gene Diagnosis and Therapy of State Key Laboratory of Medical Genetics; The Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Tian-Li Zhao
- Department of Cardiothoracic Surgery; The Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Jin-Fu Yang
- Department of Cardiothoracic Surgery; The Second Xiangya Hospital; Central South University; Changsha Hunan China
- Clinical Center for Gene Diagnosis and Therapy of State Key Laboratory of Medical Genetics; The Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Zhi-Ping Tan
- Department of Cardiothoracic Surgery; The Second Xiangya Hospital; Central South University; Changsha Hunan China
- Clinical Center for Gene Diagnosis and Therapy of State Key Laboratory of Medical Genetics; The Second Xiangya Hospital; Central South University; Changsha Hunan China
| |
Collapse
|
45
|
Bonet F, Dueñas Á, López-Sánchez C, García-Martínez V, Aránega AE, Franco D. MiR-23b and miR-199a impair epithelial-to-mesenchymal transition during atrioventricular endocardial cushion formation. Dev Dyn 2015. [PMID: 26198058 DOI: 10.1002/dvdy.24309] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Valve development is a multistep process involving the activation of the cardiac endothelium, epithelial-mesenchymal transition (EMT) and the progressive alignment and differentiation of distinct mesenchymal cell types. Several pathways such as Notch/delta, Tgf-beta and/or Vegf signaling have been implicated in crucial steps of valvulogenesis. We have previously demonstrated discrete changes in microRNAs expression during cardiogenesis, which are predicted to target Bmp- and Tgf-beta signaling. We now analyzed the expression profile of 20 candidate microRNAs in atrial, ventricular, and atrioventricular canal regions at four different developmental stages. RESULTS qRT-PCR analyses of microRNAs demonstrated a highly dynamic and distinct expression profiles within the atrial, ventricular, and atrioventricular canal regions of the developing chick heart. miR-23b, miR-199a, and miR-15a displayed increased expression during early AVC development whereas others such as miR-130a and miR-200a display decreased expression levels. Functional analyses of miR-23b, miR-199a, and miR-15a overexpression led to in vitro EMT blockage. Molecular analyses demonstrate that distinct EMT signaling pathways are impaired after microRNA expression, including a large subset of EMT-related genes that are predicted to be targeted by these microRNAs. CONCLUSIONS Our data demonstrate that miR-23b and miR-199a over-expression can impair atrioventricular EMT.
Collapse
Affiliation(s)
- Fernando Bonet
- Cardiovascular Research Group, Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | - Ángel Dueñas
- Cardiovascular Research Group, Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | - Carmen López-Sánchez
- Department of Anatomy and Embryology, Faculty of Medicine, University of Extremadura, Badajoz, Spain
| | - Virginio García-Martínez
- Department of Anatomy and Embryology, Faculty of Medicine, University of Extremadura, Badajoz, Spain
| | - Amelia E Aránega
- Cardiovascular Research Group, Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | - Diego Franco
- Cardiovascular Research Group, Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| |
Collapse
|
46
|
Chen SY, Han B, Zhu YT, Mahabole M, Huang J, Beebe DC, Tseng SCG. HC-HA/PTX3 Purified From Amniotic Membrane Promotes BMP Signaling in Limbal Niche Cells to Maintain Quiescence of Limbal Epithelial Progenitor/Stem Cells. Stem Cells 2015; 33:3341-55. [PMID: 26148958 DOI: 10.1002/stem.2091] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/08/2015] [Accepted: 05/23/2015] [Indexed: 12/13/2022]
Abstract
To explore how limbal niche cells (LNCs) may control quiescence, self-renewal, and corneal epithelial lineage commitment/differentiation of limbal epithelial progenitor/stem cells (LEPCs), we have established an in vitro sphere assay by reunion between the two cell types in three-dimensional Matrigel. The resultant sphere exhibits inhibition of corneal epithelial lineage commitment/differentiation and marked clonal growth of LEPCs, of which the latter is correlated with activation of canonical Wnt signaling. Herein, we have created a similar reunion assay in immobilized heavy chain-hyaluronic acid/pentraxin 3 (HC-HA/PTX3), which is purified from amniotic membrane (AM) and consists of a complex formed by hyaluronic covalently linked to heavy chain 1 of inter-α-inhibitor and noncovalently linked to pentraxin 3. The resultant spheres exhibited similar suppression of corneal epithelial lineage commitment/differentiation but upregulation of quiescence markers including nuclear translocation of Bmi-1, and negligible clonal growth of LEPCs. This outcome was correlated with the suppression of canonical Wnt but activation of noncanonical (Planar cell polarity) Wnt signaling as well as BMP signaling in both LEPCs and LNCs. The activation of BMP signaling in LNCs was pivotal because nuclear translocation of pSmad1/5/8 was prohibited in hLEPCs when reunioned with mLNCs of conditionally deleted Bmpr1a;Acvr1(DCKO) mice. Furthermore, ablation of BMP signaling in LEPCs led to upregulation of cell cycle genes, downregulation of Bmi-1, nuclear exclusion of phosphorylated Bmi-1, and marked promotion of the clonal growth of LEPCs. Hence, HC-HA/PTX3 uniquely upregulates BMP signaling in LNCs which leads to BMP signaling in LEPCs to achieve quiescence, helping explain how AM transplantation is clinically useful to be used as a matrix for ex vivo expansion of LEPCs and to treat corneal blindness caused by limbal stem cells deficiency.
Collapse
Affiliation(s)
- Szu-Yu Chen
- R&D Department, TissueTech, Inc., Miami, Florida, USA
| | - Bo Han
- Ocular Surface Research & Education Foundation, Miami, Florida, USA.,Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Ying-Ting Zhu
- R&D Department, TissueTech, Inc., Miami, Florida, USA
| | | | - Jie Huang
- Department of Ophthalmology and Visual Sciences, Washington University, St. Louis, Missouri, USA
| | - David C Beebe
- Department of Ophthalmology and Visual Sciences, Washington University, St. Louis, Missouri, USA
| | - Scheffer C G Tseng
- R&D Department, TissueTech, Inc., Miami, Florida, USA.,Ocular Surface Research & Education Foundation, Miami, Florida, USA
| |
Collapse
|
47
|
hnRNP U protein is required for normal pre-mRNA splicing and postnatal heart development and function. Proc Natl Acad Sci U S A 2015; 112:E3020-9. [PMID: 26039991 DOI: 10.1073/pnas.1508461112] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We report that mice lacking the heterogeneous nuclear ribonucleoprotein U (hnRNP U) in the heart develop lethal dilated cardiomyopathy and display numerous defects in cardiac pre-mRNA splicing. Mutant hearts have disorganized cardiomyocytes, impaired contractility, and abnormal excitation-contraction coupling activities. RNA-seq analyses of Hnrnpu mutant hearts revealed extensive defects in alternative splicing of pre-mRNAs encoding proteins known to be critical for normal heart development and function, including Titin and calcium/calmodulin-dependent protein kinase II delta (Camk2d). Loss of hnRNP U expression in cardiomyocytes also leads to aberrant splicing of the pre-mRNA encoding the excitation-contraction coupling component Junctin. We found that the protein product of an alternatively spliced Junctin isoform is N-glycosylated at a specific asparagine site that is required for interactions with specific protein partners. Our findings provide conclusive evidence for the essential role of hnRNP U in heart development and function and in the regulation of alternative splicing.
Collapse
|
48
|
Paolini P, Pick D, Lapira J, Sannino G, Pasqualini L, Ludka C, Sprague LJ, Zhang X, Bartolotta EA, Vazquez-Hidalgo E, Barba DT, Bazan C, Hardiman G. Developmental and extracellular matrix-remodeling processes in rosiglitazone-exposed neonatal rat cardiomyocytes. Pharmacogenomics 2015; 15:759-74. [PMID: 24897284 DOI: 10.2217/pgs.14.39] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE The objective of this study was to investigate the effects of rosiglitazone (Avandia(®)) on gene expression in neonatal rat ventricular myocytes. MATERIALS & METHODS Myocytes were exposed to rosiglitazone ex vivo. The two factors examined in the experiment were drug exposure (rosiglitazone and dimethyl sulfoxide vs dimethyl sulfoxide), and length of exposure to drug (½ h, 1 h, 2 h, 4 h, 6 h, 8 h, 12 h, 18 h, 24 h, 36 h and 48 h). RESULTS Transcripts that were consistently expressed in response to the drug were identified. Cardiovascular system development, extracellular matrix and immune response are represented prominently among the significantly modified gene ontology terms. CONCLUSION Hmgcs2, Angptl4, Cpt1a, Cyp1b1, Ech1 and Nqo1 mRNAs were strongly upregulated in cells exposed to rosiglitazone. Enrichment of transcripts involved in cardiac muscle cell differentiation and the extracellular matrix provides a panel of biomarkers for further analysis in the context of adverse cardiac outcomes in humans. Original submitted 15 November 2013; Revision submitted 14 February 2014.
Collapse
Affiliation(s)
- Paul Paolini
- Department of Biology, San Diego State University, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kageyama Y, Hoshijima M, Seo K, Bedja D, Sysa-Shah P, Andrabi SA, Chen W, Höke A, Dawson VL, Dawson TM, Gabrielson K, Kass DA, Iijima M, Sesaki H. Parkin-independent mitophagy requires Drp1 and maintains the integrity of mammalian heart and brain. EMBO J 2014; 33:2798-813. [PMID: 25349190 PMCID: PMC4282557 DOI: 10.15252/embj.201488658] [Citation(s) in RCA: 352] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 08/18/2014] [Accepted: 09/19/2014] [Indexed: 12/22/2022] Open
Abstract
Mitochondrial dynamics and mitophagy have been linked to cardiovascular and neurodegenerative diseases. Here, we demonstrate that the mitochondrial division dynamin Drp1 and the Parkinson's disease-associated E3 ubiquitin ligase parkin synergistically maintain the integrity of mitochondrial structure and function in mouse heart and brain. Mice lacking cardiac Drp1 exhibited lethal heart defects. In Drp1KO cardiomyocytes, mitochondria increased their connectivity, accumulated ubiquitinated proteins, and decreased their respiration. In contrast to the current views of the role of parkin in ubiquitination of mitochondrial proteins, mitochondrial ubiquitination was independent of parkin in Drp1KO hearts, and simultaneous loss of Drp1 and parkin worsened cardiac defects. Drp1 and parkin also play synergistic roles in neuronal mitochondrial homeostasis and survival. Mitochondrial degradation was further decreased by combination of Drp1 and parkin deficiency, compared with their single loss. Thus, the physiological importance of parkin in mitochondrial homeostasis is revealed in the absence of mitochondrial division in mammals.
Collapse
Affiliation(s)
- Yusuke Kageyama
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Masahiko Hoshijima
- Center for Research in Biological Systems and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kinya Seo
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Djahida Bedja
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Polina Sysa-Shah
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shaida A Andrabi
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering Johns Hopkins University School of Medicine, Baltimore, MD, USA Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
| | - Weiran Chen
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ahmet Höke
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering Johns Hopkins University School of Medicine, Baltimore, MD, USA Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering Johns Hopkins University School of Medicine, Baltimore, MD, USA Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
| | - Kathleen Gabrielson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Miho Iijima
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
50
|
Lockhart MM, Boukens BJD, Phelps AL, Brown CLM, Toomer KA, Burns TA, Mukherjee RD, Norris RA, Trusk TC, van den Hoff MJB, Wessels A. Alk3 mediated Bmp signaling controls the contribution of epicardially derived cells to the tissues of the atrioventricular junction. Dev Biol 2014; 396:8-18. [PMID: 25300579 DOI: 10.1016/j.ydbio.2014.09.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 08/30/2014] [Accepted: 09/26/2014] [Indexed: 11/29/2022]
Abstract
Recent studies using mouse models for cell fate tracing of epicardial derived cells (EPDCs) have demonstrated that at the atrioventricular (AV) junction EPDCs contribute to the mesenchyme of the AV sulcus, the annulus fibrosus, and the parietal leaflets of the AV valves. There is little insight, however, into the mechanisms that govern the contribution of EPDCs to these tissues. While it has been demonstrated that bone morphogenetic protein (Bmp) signaling is required for AV cushion formation, its role in regulating EPDC contribution to the AV junction remains unexplored. To determine the role of Bmp signaling in the contribution of EPDCs to the AV junction, the Bmp receptor activin-like kinase 3 (Alk3; or Bmpr1a) was conditionally deleted in the epicardium and EPDCs using the mWt1/IRES/GFP-Cre (Wt1(Cre)) mouse. Embryonic Wt1(Cre);Alk3(fl/fl) specimens showed a significantly smaller AV sulcus and a severely underdeveloped annulus fibrosus. Electrophysiological analysis of adult Wt1(Cre);Alk3(fl/fl) mice showed, unexpectedly, no ventricular pre-excitation. Cell fate tracing revealed a significant decrease in the number of EPDCs within the parietal leaflets of the AV valves. Postnatal Wt1(Cre);Alk3(fl/fl) specimens showed myxomatous changes in the leaflets of the mitral valve. Together these observations indicate that Alk3 mediated Bmp signaling is important in the cascade of events that regulate the contribution of EPDCs to the AV sulcus, annulus fibrosus, and the parietal leaflets of the AV valves. Furthermore, this study shows that EPDCs do not only play a critical role in early developmental events at the AV junction, but that they also are important in the normal maturation of the AV valves.
Collapse
Affiliation(s)
- Marie M Lockhart
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | | | - Aimee L Phelps
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Christina-Lin M Brown
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Katelynn A Toomer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Tara A Burns
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Rupak D Mukherjee
- Division of Cardiothoracic Surgery, Department of Surgery and Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Russell A Norris
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Thomas C Trusk
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Maurice J B van den Hoff
- Heart Failure Research Center, Department of Anatomy, Embryology and Physiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Andy Wessels
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|