1
|
Zhan R, Zhou F, Liu C, Chen C, Li M, Huang D, Zheng N, Lin T, Zuo Z, He C, Chen X. Resveratrol ameliorates cyprodinil-induced zebrafish cardiac developmental defects as an aryl hydrocarbon receptor antagonist. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:44789-44799. [PMID: 38954331 DOI: 10.1007/s11356-024-34024-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
Cyprodinil, a globally utilized broad-spectrum pyrimidine amine fungicide, has been observed to elicit cardiac abnormality. Resveratrol (RSV), a naturally occurring polyphenolic compound, showcases remarkable defensive properties in nurturing cardiac development. To investigate whether RSV could protect against cyprodinil-induced cardiac defects, we exposed zebrafish embryos to cyprodinil (500 μg/L) in the presence or absence of RSV (1 μM). Our results showed that RSV significantly mitigated the decrease of survival rate and embryo movement and the hatching delay induced by cyprodinil. In addition, RSV also improved cyprodinil-induced zebrafish cardiac developmental toxicity, including pericardial edema and cardiac function impairment. In mechanism, RSV attenuated the cyprodinil-induced changes in mRNA expression involved in cardiac development, such as myh6, myl7, tbx5, and gata4, and calcium ion channels, such as ncx1h, slc8a4a, and atp2a2b. We further showed that RSV might inhibit the activity of aryl hydrocarbon receptor (AhR) signaling pathways induced by cyprodinil. In summary, our findings establish that the protective effects of RSV against the cardiac developmental toxicity are induced by cyprodinil due to its remarkable ability to inhibit AhR activity. Our findings not only shed light on a new avenue for regulating and ensuring the safe utilization of cyprodinil but also presents a novel concept to promote its responsible use.
Collapse
Affiliation(s)
- Ruyu Zhan
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Fushan Zhou
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Chaoyang Liu
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Chuanchang Chen
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Mingmei Li
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Dongqin Huang
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Naying Zheng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Tingting Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Xintan Chen
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China.
| |
Collapse
|
2
|
Pitchika GK, Naik BK, Ramana GVV, Nirupama R, Ranjani TS, Venkaiah K, Reddy MH, Sainath SB, Pradeepkiran JA. Transcriptomic profile in carbendazim-induced developmental defects in zebrafish (Danio rerio) embryos/larvae. Comp Biochem Physiol C Toxicol Pharmacol 2024; 280:109907. [PMID: 38522711 DOI: 10.1016/j.cbpc.2024.109907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/26/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Carbendazim is a widely used fungicide to protect agricultural and horticultural crops against a wide array of fungal species. Published reports have shown that the wide usage of carbendazim resulted in reprotoxicity, carcinogenicity, immunotoxicity, and developmental toxicity in mammalian models. However, studies related to the developmental toxicity of carbendazim in aquatic organisms are not clear. To address this gap, an attempt was made by exposing zebrafish embryos to carbendazim (800 μg/L) and assessing the phenotypic and transcriptomic profile at different developmental stages [24 hour post fertilization (hpf), 48 hpf, 72 hpf and 96 hpf). At 48 hpf, phenotypic abnormalities such as delay in hatching rate, deformed spinal axial curvature, and pericardial edema were observed in zebrafish larvae over its respective controls. At 72 hpf, exposure of zebrafish embryos exposed to carbendazim resulted in scoliosis; however, unexposed larvae did not exhibit signs of scoliosis. Interestingly, the transcriptomic analysis revealed a total of 1253 DEGs were observed at selected time points, while unique genes at 24 hpf, 48 hpf, 72 hpf and 96 hpf was found to be 76.54 %, 61.14 %, 92.98 %, and 68.28 %, respectively. Functional profiling of downregulated genes revealed altered transcriptomic markers associated with phototransduction (24 hpf and 72 hpf), immune system (48 hpf), and SNARE interactions in the vesicular pathway (96 hpf). Whereas functional profiling of upregulated genes revealed altered transcriptomic markers associated with riboflavin metabolism (24 hpf), basal transcription factors (48 hpf), insulin signaling pathway (72 hpf), and primary bile acid biosynthesis (96 hpf). Taken together, carbendazim-induced developmental toxicity could be ascribed to pleiotropic responses at the molecular level, which in turn might reflect phenotypic abnormalities.
Collapse
Affiliation(s)
- Gopi Krishna Pitchika
- Department of Zoology, Vikrama Simhapuri University College, Kavali 524201, A.P., India.
| | - B Krishna Naik
- Department of Zoology, Vikrama Simhapuri University College, Kavali 524201, A.P., India
| | - G V V Ramana
- Department of Zoology, Vikrama Simhapuri University College, Kavali 524201, A.P., India
| | - R Nirupama
- Department of Zoology, Vikrama Simhapuri University College, Kavali 524201, A.P., India
| | - T Sri Ranjani
- Department of Zoology, D.K. Govt. College for Women (A), Dargamitta, Nellore 524003, A.P., India
| | - K Venkaiah
- Department of Biotechnology, Vikrama Simhapuri University, Nellore 524324, A.P., India
| | - M Hanuma Reddy
- Department of Marine Biology, Vikrama Simhapuri University, Nellore 524324, A.P., India
| | - S B Sainath
- Department of Biotechnology, Vikrama Simhapuri University, Nellore 524324, A.P., India.
| | | |
Collapse
|
3
|
Jumppanen M, Kinnunen SM, Zore M, Välimäki MJ, Talman V, Gennäs GBA, Ruskoaho HJ, Yli-Kauhaluoma J. Affinity chromatography reveals direct binding of the GATA4-NKX2-5 interaction inhibitor (3i-1000) with GATA4. Sci Rep 2024; 14:8938. [PMID: 38637629 PMCID: PMC11026519 DOI: 10.1038/s41598-024-59418-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
Heart failure is a serious medical condition with a poor prognosis. Current treatments can only help manage the symptoms and slow the progression of heart failure. However, there is currently no cure to prevent and reverse cardiac remodeling. Transcription factors are in a central role in various cellular processes, and in the heart, GATA4 and NKX2-5 transcription factors mediate hypertrophic responses and remodeling. We have identified compounds that modulate the synergistic interaction of GATA4 and NKX2-5 and shown that the most promising compound (1, 3i-1000) is cardioprotective in vitro and in vivo. However, direct evidence of its binding site and mechanism of action has not been available. Due to the disordered nature of transcription factors, classical target engagement approaches cannot be utilized. Here, we synthesized a small-molecule ligand-binding pulldown probe of compound 1 to utilize affinity chromatography alongside CETSA, AlphaScreen, and molecular modeling to study ligand binding. These results provide the first evidence of direct physical binding of compound 1 selectively to GATA4. While developing drugs that target transcription factors presents challenges, advances in technologies and knowledge of intrinsically disordered proteins enable the identification of small molecules that can selectively target transcription factors.
Collapse
Affiliation(s)
- Mikael Jumppanen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Sini M Kinnunen
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Matej Zore
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Mika J Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Virpi Talman
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Gustav Boije Af Gennäs
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Heikki J Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland.
| |
Collapse
|
4
|
Liu Y, Jin X, Ye Y, Xu Z, Du Z, Hong H, Yu H, Lin H, Huang X, Sun H. Emerging disinfection byproducts 3-bromine carbazole induces cardiac developmental toxicity via aryl hydrocarbon receptor activation in zebrafish larvae. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 346:123609. [PMID: 38395134 DOI: 10.1016/j.envpol.2024.123609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024]
Abstract
3-bromine carbazole (3-BCZ) represents a group of emerging aromatic disinfection byproducts (DBP) detected in drinking water; however, limited information is available regarding its potential cardiotoxicity. To assess its impacts, zebrafish embryos were exposed to 0, 0.06, 0.14, 0.29, 0.58, 1.44 or 2.88 mg/L of 3-BCZ for 120 h post fertilization (hpf). Our results revealed that ≥1.44 mg/L 3-BCZ exposure induced a higher incidence of heart malformation and an elevated pericardial area in zebrafish larvae; it also decreased the number of cardiac muscle cells and thins the walls of the ventricle and atrium while increasing cardiac output and impeding cardiac looping. Furthermore, 3-BCZ exposure also exhibited significant effects on the transcriptional levels of genes related to both cardiac development (nkx2.5, vmhc, gata4, tbx5, tbx2b, bmp4, bmp10, and bmp2b) and cardiac function (cacna1ab, cacna1da, atp2a1l, atp1b2b, atp1a3b, and tnnc1a). Notably, N-acetyl-L-cysteine, a reactive oxygen species scavenger, may alleviate the failure of cardiac looping induced by 3-BCZ but not the associated cardiac dysfunction or malformation; conversely, the aryl hydrocarbon receptor agonist CH131229 can completely eliminate the cardiotoxicity caused by 3-BCZ. This study provides new evidence for potential risks associated with ingesting 3-BCZ as well as revealing underlying mechanisms responsible for its cardiotoxic effects on zebrafish embryos.
Collapse
Affiliation(s)
- Yingying Liu
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Xudong Jin
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Yanan Ye
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Zeqiong Xu
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Zhongkun Du
- College of Resources and Environment, Shandong Agricultural University, Key Laboratory of Agricultural Environment in Universities of Shandong, 61 Daizong Road, Taian, 271018, PR China
| | - Huachang Hong
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Haiying Yu
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Hongjun Lin
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Xianfeng Huang
- National and Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, PR China
| | - Hongjie Sun
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China.
| |
Collapse
|
5
|
Pohjolainen L, Kinnunen SM, Auno S, Kiriazis A, Pohjavaara S, Kari-Koskinen J, Zore M, Jumppanen M, Yli-Kauhaluoma J, Talman V, Ruskoaho H, Välimäki MJ. Switching of hypertrophic signalling towards enhanced cardiomyocyte identity and maturity by a GATA4-targeted compound. Stem Cell Res Ther 2024; 15:5. [PMID: 38167208 PMCID: PMC10763434 DOI: 10.1186/s13287-023-03623-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The prevalence of heart failure is constantly increasing, and the prognosis of patients remains poor. New treatment strategies to preserve cardiac function and limit cardiac hypertrophy are therefore urgently needed. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are increasingly used as an experimental platform for cardiac in vitro studies. However, in contrast to adult cardiomyocytes, hiPSC-CMs display immature morphology, contractility, gene expression and metabolism and hence express a naive phenotype that resembles more of a foetal cardiomyocyte. METHODS A library of 14 novel compounds was synthesized in-house and screened for GATA4-NKX2-5 reporter activity and cellular toxicity. The most potent compound, 3i-1262, along with previously reported GATA4-acting compounds, were selected to investigate their effects on hypertrophy induced by endothelin-1 or mechanical stretch. Morphological changes and protein expression were characterized using immunofluorescence staining and high-content analysis. Changes in gene expression were studied using qPCR and RNA sequencing. RESULTS The prototype compound 3i-1262 inhibited GATA4-NKX2-5 synergy in a luciferase reporter assay. Additionally, the isoxazole compound 3i-1262 inhibited the hypertrophy biomarker B-type natriuretic peptide (BNP) by reducing BNP promoter activity and proBNP expression in neonatal rat ventricular myocytes and hiPSC-CMs, respectively. Treatment with 3i-1262 increased metabolic activity and cardiac troponin T expression in hiPSC-CMs without affecting GATA4 protein levels. RNA sequencing analysis revealed that 3i-1262 induces gene expression related to metabolic activity and cell cycle exit, indicating a change in the identity and maturity status of hiPSC-CMs. The biological processes that were enriched in upregulated genes in response to 3i-1262 were downregulated in response to mechanical stretch, and conversely, the downregulated processes in response to 3i-1262 were upregulated in response to mechanical stretch. CONCLUSIONS There is currently a lack of systematic understanding of the molecular modulation and control of hiPSC-CM maturation. In this study, we demonstrated that the GATA4-interfering compound 3i-1262 reorganizes the cardiac transcription factor network and converts hypertrophic signalling towards enhanced cardiomyocyte identity and maturity. This conceptually unique approach provides a novel structural scaffold for further development as a modality to promote cardiomyocyte specification and maturity.
Collapse
Affiliation(s)
- Lotta Pohjolainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Sini M Kinnunen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Samuli Auno
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Alexandros Kiriazis
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Saana Pohjavaara
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Julia Kari-Koskinen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Matej Zore
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Mikael Jumppanen
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jari Yli-Kauhaluoma
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Heikki Ruskoaho
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Mika J Välimäki
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland.
| |
Collapse
|
6
|
Cao C, Li L, Zhang Q, Li H, Wang Z, Wang A, Liu J. Nkx2.5: a crucial regulator of cardiac development, regeneration and diseases. Front Cardiovasc Med 2023; 10:1270951. [PMID: 38124890 PMCID: PMC10732152 DOI: 10.3389/fcvm.2023.1270951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Cardiomyocytes fail to regenerate after birth and respond to mitotic signals through cellular hypertrophy rather than cellular proliferation. Necrotic cardiomyocytes in the infarcted ventricular tissue are eventually replaced by fibroblasts, generating scar tissue. Cardiomyocyte loss causes localized systolic dysfunction. Therefore, achieving the regeneration of cardiomyocytes is of great significance for cardiac function and development. Heart development is a complex biological process. An integral cardiac developmental network plays a decisive role in the regeneration of cardiomyocytes. During this process, genetic epigenetic factors, transcription factors, signaling pathways and small RNAs are involved in regulating the developmental process of the heart. Cardiomyocyte-specific genes largely promote myocardial regeneration, among which the Nkx2.5 transcription factor is one of the earliest markers of cardiac progenitor cells, and the loss or overexpression of Nkx2.5 affects cardiac development and is a promising candidate factor. Nkx2.5 affects the development and function of the heart through its multiple functional domains. However, until now, the specific mechanism of Nkx2.5 in cardiac development and regeneration is not been fully understood. Therefore, this article will review the molecular structure, function and interaction regulation of Nkx2.5 to provide a new direction for cardiac development and the treatment of heart regeneration.
Collapse
Affiliation(s)
- Ce Cao
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lei Li
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Qian Zhang
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Haoran Li
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ziyan Wang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Aoao Wang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Jianxun Liu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
7
|
Fear VS, Forbes CA, Shaw NC, Farley KO, Mantegna JL, Htun JP, Syn G, Viola H, Cserne Szappanos H, Hool L, Ward M, Baynam G, Lassmann T. Gene editing and cardiac disease modelling for the interpretation of genetic variants of uncertain significance in congenital heart disease. Stem Cell Res Ther 2023; 14:345. [PMID: 38049901 PMCID: PMC10696868 DOI: 10.1186/s13287-023-03592-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Genomic sequencing in congenital heart disease (CHD) patients often discovers novel genetic variants, which are classified as variants of uncertain significance (VUS). Functional analysis of each VUS is required in specialised laboratories, to determine whether the VUS is disease causative or not, leading to lengthy diagnostic delays. We investigated stem cell cardiac disease modelling and transcriptomics for the purpose of genetic variant classification using a GATA4 (p.Arg283Cys) VUS in a patient with CHD. METHODS We performed high efficiency CRISPR gene editing with homology directed repair in induced pluripotent stem cells (iPSCs), followed by rapid clonal selection with amplicon sequencing. Genetic variant and healthy matched control cells were compared using cardiomyocyte disease modelling and transcriptomics. RESULTS Genetic variant and healthy cardiomyocytes similarly expressed Troponin T (cTNNT), and GATA4. Transcriptomics analysis of cardiomyocyte differentiation identified changes consistent with the patient's clinical human phenotype ontology terms. Further, transcriptomics revealed changes in calcium signalling, and cardiomyocyte adrenergic signalling in the variant cells. Functional testing demonstrated, altered action potentials in GATA4 genetic variant cardiomyocytes were consistent with patient cardiac abnormalities. CONCLUSIONS This work provides in vivo functional studies supportive of a damaging effect on the gene or gene product. Furthermore, we demonstrate the utility of iPSCs, CRISPR gene editing and cardiac disease modelling for genetic variant interpretation. The method can readily be applied to other genetic variants in GATA4 or other genes in cardiac disease, providing a centralised assessment pathway for patient genetic variant interpretation.
Collapse
Affiliation(s)
- Vanessa S Fear
- Translational Genetics, Precision Health, Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, 6009, Australia.
- Centre for Child Health Research, University of Western Australia, Crawley, Australia.
- Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, WA, 6009, Australia.
| | - Catherine A Forbes
- Translational Genetics, Precision Health, Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, 6009, Australia
| | - Nicole C Shaw
- Translational Genetics, Precision Health, Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, 6009, Australia
| | - Kathryn O Farley
- Translational Genetics, Precision Health, Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, 6009, Australia
- Computational Biology, Precision Health, Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, 6009, Australia
| | - Jessica L Mantegna
- Translational Genetics, Precision Health, Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, 6009, Australia
| | - Jasmin P Htun
- Translational Genetics, Precision Health, Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, 6009, Australia
| | - Genevieve Syn
- Computational Biology, Precision Health, Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, 6009, Australia
| | - Helena Viola
- University of Western Australia, Crawley, Australia
| | | | - Livia Hool
- University of Western Australia, Crawley, Australia
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Michelle Ward
- Undiagnosed Diseases Program, Genetic Services of WA, Subiaco, Australia
| | - Gareth Baynam
- Western Australian Register of Developmental Anomalies, King Edward Memorial Hospital, Subiaco, WA, 6008, Australia
- Undiagnosed Diseases Program, Genetic Services of WA, Subiaco, Australia
| | - Timo Lassmann
- Translational Genetics, Precision Health, Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, 6009, Australia
- Computational Biology, Precision Health, Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, 6009, Australia
- Centre for Child Health Research, University of Western Australia, Crawley, Australia
| |
Collapse
|
8
|
Wang Z, Wang X, Lan X, Zhu H, Qu L, Pan C. Polymorphism within the GATA binding protein 4 gene is significantly associated with goat litter size. Anim Biotechnol 2023; 34:4291-4300. [PMID: 36421983 DOI: 10.1080/10495398.2022.2147533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
GATA binding protein 4 (GATA4) is a typical transcription binding factor, and its main functions include regulating the proliferation, differentiation and apoptosis of ovarian granulosa cells, promoting spermatogenesis and sex differentiation, implying that this gene have possibly roles in animal reproduction. This study aims to detect five potential insertion/deletions (indels) of the GATA4 gene in 606 healthy unrelated Shaanbei white cashmere (SBWC) goats and analyze its association with the litter size. The electrophoresis and DNA sequencing identified two polymorphic indels (e.g., P4-Del-8bp and P5-Ins-9bp indel). Then T-test analysis showed that P4-Del-8bp was significantly correlated with litter size (p = 0.022) because of two different genotypes detected, e.g., insertion-deletion (ID) and deletion-deletion (DD), and the average litter size of individuals with DD genotype goats was higher than that of others. However, there was no correlation between P5-Ins-9bp and lambing of goats. Chi-square (X2) test found that the distribution of and P4-Del-8bp genotypes (X2 = 6.475, p = 0.011) was significantly different between single and multiple-lamb groups, while P5-Ins-9bp (X2 = 0.030, p = 0.862) was not. Therefore, these findings revealed that P4-Del-8bp polymorphism of goat GATA4 gene was a potential molecular marker significantly associated with litter size, which can be used for the marker-assisted selection (MAS) breeding to improve goat industry.
Collapse
Affiliation(s)
- Zhiying Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xinyu Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xianyong Lan
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Haijing Zhu
- Life Science Research Center, Shaanxi Provincial Engineering and Technology Research Center of Cashmere Goats, Yulin University, Yulin, China
| | - Lei Qu
- Life Science Research Center, Shaanxi Provincial Engineering and Technology Research Center of Cashmere Goats, Yulin University, Yulin, China
| | - Chuanying Pan
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
9
|
Neidviecky E, Deng H. Determination of Complex Formation between Drosophila Nrf2 and GATA4 Factors at Selective Chromatin Loci Demonstrates Transcription Coactivation. Cells 2023; 12:938. [PMID: 36980279 PMCID: PMC10047698 DOI: 10.3390/cells12060938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/03/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Nrf2 is the dominant cellular stress response factor that protects cells through transcriptional responses to xenobiotic and oxidative stimuli. Nrf2 malfunction is highly correlated with many human diseases, but the underlying molecular mechanisms remain to be fully uncovered. GATA4 is a conserved GATA family transcription factor that is essential for cardiac and dorsal epidermal development. Here, we describe a novel interaction between Drosophila Nrf2 and GATA4 proteins, i.e., cap'n'collar C (CncC) and Pannier (Pnr), respectively. Using the bimolecular fluorescence complementation (BiFC) assay-a unique imaging tool for probing protein complexes in living cells-we detected CncC-Pnr complexes in the nuclei of Drosophila embryonic and salivary gland cells. Visualization of CncC-Pnr BiFC signals on the polytene chromosome revealed that CncC and Pnr tend to form complexes in euchromatic regions, with a preference for loci that are not highly occupied by CncC or Pnr alone. Most genes within these loci are activated by the CncC-Pnr BiFC, but not by individually expressed CncC or Pnr fusion proteins, indicating a novel mechanism whereby CncC and Pnr interact at specific genomic loci and coactivate genes at these loci. Finally, CncC-induced early lethality can be rescued by Pnr depletion, suggesting that CncC and Pnr function in the same genetic pathway during the early development of Drosophila. Taken together, these results elucidate a novel crosstalk between the Nrf2 xenobiotic/oxidative response factor and GATA factors in the transcriptional regulation of development. This study also demonstrates that the polytene chromosome BiFC assay is a valuable tool for mapping genes that are targeted by specific transcription factor complexes.
Collapse
Affiliation(s)
| | - Huai Deng
- Department of Biology, University of Minnesota Duluth, 1035 Kirby Drive, Duluth, MN 55812, USA
| |
Collapse
|
10
|
Shafique S, Ali SR, Rajput SN, Salim A, Khan I. Cardiac Transcription Regulators Differentiate Human Umbilical Cord Mesenchymal Stem Cells into Cardiac Cells. Altern Lab Anim 2023; 51:12-29. [PMID: 36484201 DOI: 10.1177/02611929221143774] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Stem cell-based therapy presents an attractive alternative to conventional therapies for degenerative diseases. Numerous studies have investigated the capability of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) to contribute to the regeneration of cardiomyocytes, and the results have encouraged further basic and clinical studies on the MSC-based treatment of cardiomyopathies. This study aimed to determine the potential of cardiomyogenic transcription factors in differentiating hUC-MSCs into cardiac-like cells in vitro. MSCs were isolated from umbilical cord tissue and were transduced with the transcription factor genes, GATA-4 and Nkx 2.5, via infection with lentiviruses, to promote differentiation into the cardiomyogenic lineage. Gene and protein expression were analysed with qPCR and immunocytochemical staining. After transduction, differentiated cardiac-like cells showed significant expression of cardiac genes and proteins, namely GATA-4, Nkx-2.5, cardiac troponin I (cTnI) and myosin heavy chain (MHC). The cardiomyogenic-induced group significantly overexpressed cardiac-specific genes (GATA-4, Nkx-2.5, cTnI, MHC, α-actinin and Wnt2). Expression of the calcium channel gene was also significantly increased, while the sodium channel gene was downregulated in the transduced hUC-MSCs, as compared to non-transduced cells. The results suggest that GATA-4 and Nkx-2.5 interact synergistically in the activation of downstream cardiac transcription factors, demonstrating the functional convergence of hUC-MSC differentiation into cardiac-like cells. These findings could potentially be utilised in the efficient production of cardiac-like cells from stem cells; these cardiac-like cells could then be used in various applications, such as for in vivo implantation in infarcted myocardium, and for drug screening in toxicity testing.
Collapse
Affiliation(s)
- Shumaila Shafique
- 208246Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Syeda Roohina Ali
- 208246Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Shafiqa Naeem Rajput
- 208246Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Asmat Salim
- 208246Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Irfan Khan
- 208246Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
11
|
Yi T, Hao X, Sun H, Zhang Y, Han J, Gu X, Sun L, Liu X, Zhao Y, Guo Y, Zhou X, He Y. Genetic aetiology distribution of 398 foetuses with congenital heart disease in the prenatal setting. ESC Heart Fail 2022; 10:917-930. [PMID: 36478645 PMCID: PMC10053168 DOI: 10.1002/ehf2.14209] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/24/2022] [Accepted: 10/02/2022] [Indexed: 12/12/2022] Open
Abstract
AIMS Copy number variant-sequencing (CNV-seq) and exome sequencing (ES) have been used as powerful tools in understanding the role of genetic variants in congenital heart diseases (CHDs). A few previous large cohort studies have utilized CNV-seq and ES to investigate prenatally diagnosed CHD. Here, we sought to determine the value of CNV-seq and ES for genetic evaluation of foetal CHDs. METHODS AND RESULTS We recruited 398 pregnant women diagnosed with CHDs between 8 January 2017 and 30 November 2020. CNV-seq and ES were performed on foetal and parent samples. CHD cases were classified following the guidelines of the International Paediatric and Congenital Cardiac Code and the Tenth and Eleventh Revisions of the International Classification of Diseases. Data on aneuploids (AUP), pathogenic CNVs (pCNVs), and single nucleotide variants (SNVs) were collected and compared, following appropriate procedures. We identified genetic abnormalities in 129 (32.41%) foetuses. These abnormalities included AUP (10.80%), pCNVs (13.32%), and SNVs (8.04%). ES analysis yielded higher SNVs in cases without AUP or pCNVs. Non-isolated CHDs were associated with higher genetic abnormalities than isolated CHDs, mainly due to AUP differences between the two groups. The prevalence of genetic defects was the highest in foetuses with atrioventricular septal defects (AVSD), left ventricular outflow tract obstruction (LVOTO), and conotruncal defects (CTD). AVSD and anomalies of atrioventricular junctions and valves were associated with AUP abnormalities. CTD, anomalies of extrapericardial arterial trunks, and anomalies of the ventricular outflow tracts were the most common CHD categories diagnosed using CNVs. The most common CHDs associated with single ventricle (SV) abnormalities were heterotaxy (Hex) (14.89%), LVOTO (14.58%), and ventricular septal defect (VSD) (26.67%, 4/15). Although the ES yields were higher than CNV-seq for VSD (44.4%, 4/9), LVOTO (20%, 7/35), Hex (14.89%, 7/47), and CTD (9.1%, 11/121), its diagnostic yield did not increase for SV (6.7%, 1/15), AVSD (3.8%, 1/26), or right ventricular obstruction defects (2.6%, 1/38). The most common mutations were observed in KMT2D, CHD7, and NOTCH1. CONCLUSIONS To our knowledge, this is the largest cohort study to investigate the incidence of SNVs using ES in foetal CHD. CNV-seq and ES identified genetic abnormalities in nearly 1/3 of foetal CHD cases. Thus, CNV-seq and ES can provide clinically relevant information for pregnancy management.
Collapse
Affiliation(s)
- Tong Yi
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Xiaoyan Hao
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Hairui Sun
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Ye Zhang
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Jiancheng Han
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Xiaoyan Gu
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Lin Sun
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Xiaowei Liu
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Ying Zhao
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Yong Guo
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Xiaoxue Zhou
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Yihua He
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| |
Collapse
|
12
|
Razzaq SS, Khan I, Naeem N, Salim A, Begum S, Haneef K. Overexpression of GATA binding protein 4 and myocyte enhancer factor 2C induces differentiation of mesenchymal stem cells into cardiac-like cells. World J Stem Cells 2022; 14:700-713. [PMID: 36188117 PMCID: PMC9516467 DOI: 10.4252/wjsc.v14.i9.700] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/20/2022] [Accepted: 08/30/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Heart diseases are the primary cause of death all over the world. Following myocardial infarction, billions of cells die, resulting in a huge loss of cardiac function. Stem cell-based therapies have appeared as a new area to support heart regeneration. The transcription factors GATA binding protein 4 (GATA-4) and myocyte enhancer factor 2C (MEF2C) are considered prominent factors in the development of the cardiovascular system.
AIM To explore the potential of GATA-4 and MEF2C for the cardiac differentiation of human umbilical cord mesenchymal stem cells (hUC-MSCs).
METHODS hUC-MSCs were characterized morphologically and immunologically by the presence of specific markers of MSCs via immunocytochemistry and flow cytometry, and by their potential to differentiate into osteocytes and adipocytes. hUC-MSCs were transfected with GATA-4, MEF2C, and their combination to direct the differentiation. Cardiac differentiation was confirmed by semiquantitative real-time polymerase chain reaction and immunocytochemistry.
RESULTS hUC-MSCs expressed specific cell surface markers CD105, CD90, CD44, and vimentin but lack the expression of CD45. The transcription factors GATA-4 and MEF2C, and their combination induced differentiation in hUC-MSCs with significant expression of cardiac genes i.e., GATA-4, MEF2C, NK2 homeobox 5 (NKX2.5), MHC, and connexin-43, and cardiac proteins GATA-4, NKX2.5, cardiac troponin T, and connexin-43.
CONCLUSION Transfection with GATA-4, MEF2C, and their combination effectively induces cardiac differentiation in hUC-MSCs. These genetically modified MSCs could be a promising treatment option for heart diseases in the future.
Collapse
Affiliation(s)
- Syeda Saima Razzaq
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Nadia Naeem
- Dow Research Institute of Biotechnology & Biomedical Sciences (DRIBBS), Dow University of Health Sciences (DUHS), Ojha Campus, Karachi 75200, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Sumreen Begum
- Stem Cells Research Laboratory (SCRL), Sindh Institute of Urology and Transplantation (SIUT), Karachi 74200, Pakistan
| | - Kanwal Haneef
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
13
|
Pustovit KB, Samoilova DV, Abramochkin DV, Filatova TS, Kuzmin VS. α1-adrenergic receptors accompanied by GATA4 expression are related to proarrhythmic conduction and automaticity in rat interatrial septum. J Physiol Biochem 2022; 78:793-805. [PMID: 35802254 DOI: 10.1007/s13105-022-00902-8] [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: 06/02/2021] [Accepted: 05/19/2022] [Indexed: 11/25/2022]
Abstract
The development of interatrial septum (IAS) is a complicated process, which continues during postnatal life. The hypertrophic signals in developing heart are mediated among others by α-adrenergic pathways. These facts suggest the presence of specific electrophysiological features in developing IAS. This study was aimed to investigate the electrical activity in the tissue preparations of IAS from rat heart in normal conditions and under stimulation of adrenoreceptors. Intracellular recording of electrical activity revealed less negative level of resting membrane potential in IAS if compared to myocardium of left atrium. In normal conditions, non-paced IAS preparations were quiescent, but noradrenaline (10-5 M) and phenylephrine (10-5 M) induced spontaneous action potentials, which could be abolished by α1-blocker prazosin (10-5 M), but not β1-blocker atenolol (10-5 M). Optical mapping showed drastic phenylephrine-induced slowing of conduction in adult rat IAS. The α1-dependent ectopic automaticity of IAS myocardium might be explained by immunohistochemical data indicating the presence of transcription factor GATA4 and abundant α1A-adrenoreceptors in myocytes from adult rat IAS. An elevated sensitivity to adrenergic stimulation due to involvement of α1-adrenergic pathways may underlie increased proarrhythmic potential of adult IAS at least in rats.
Collapse
Affiliation(s)
- Ksenia B Pustovit
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia
| | - Daria V Samoilova
- N. N. Blokhin National Medical Research Centre of Oncology, Kashirskoye sh., 24, Moscow, Russia
| | - Denis V Abramochkin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia.
| | - Tatiana S Filatova
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia.,Laboratory of Cardiac Electrophysiology, National Medical Research Center for Cardiology, 3rd Cherepkovskaya, 15a, Moscow, Russia.,Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova str., 1, Moscow, Russia
| | - Vladislav S Kuzmin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia
| |
Collapse
|
14
|
Välimäki MJ, Leigh RS, Kinnunen SM, March AR, de Sande AH, Kinnunen M, Varjosalo M, Heinäniemi M, Kaynak BL, Ruskoaho H. GATA-targeted compounds modulate cardiac subtype cell differentiation in dual reporter stem cell line. Stem Cell Res Ther 2021; 12:190. [PMID: 33736688 PMCID: PMC7977156 DOI: 10.1186/s13287-021-02259-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
Background Pharmacological modulation of cell fate decisions and developmental gene regulatory networks holds promise for the treatment of heart failure. Compounds that target tissue-specific transcription factors could overcome non-specific effects of small molecules and lead to the regeneration of heart muscle following myocardial infarction. Due to cellular heterogeneity in the heart, the activation of gene programs representing specific atrial and ventricular cardiomyocyte subtypes would be highly desirable. Chemical compounds that modulate atrial and ventricular cell fate could be used to improve subtype-specific differentiation of endogenous or exogenously delivered progenitor cells in order to promote cardiac regeneration. Methods Transcription factor GATA4-targeted compounds that have previously shown in vivo efficacy in cardiac injury models were tested for stage-specific activation of atrial and ventricular reporter genes in differentiating pluripotent stem cells using a dual reporter assay. Chemically induced gene expression changes were characterized by qRT-PCR, global run-on sequencing (GRO-seq) and immunoblotting, and the network of cooperative proteins of GATA4 and NKX2-5 were further explored by the examination of the GATA4 and NKX2-5 interactome by BioID. Reporter gene assays were conducted to examine combinatorial effects of GATA-targeted compounds and bromodomain and extraterminal domain (BET) inhibition on chamber-specific gene expression. Results GATA4-targeted compounds 3i-1000 and 3i-1103 were identified as differential modulators of atrial and ventricular gene expression. More detailed structure-function analysis revealed a distinct subclass of GATA4/NKX2-5 inhibitory compounds with an acetyl lysine-like domain that contributed to ventricular cells (%Myl2-eGFP+). Additionally, BioID analysis indicated broad interaction between GATA4 and BET family of proteins, such as BRD4. This indicated the involvement of epigenetic modulators in the regulation of GATA-dependent transcription. In this line, reporter gene assays with combinatorial treatment of 3i-1000 and the BET bromodomain inhibitor (+)-JQ1 demonstrated the cooperative role of GATA4 and BRD4 in the modulation of chamber-specific cardiac gene expression. Conclusions Collectively, these results indicate the potential for therapeutic alteration of cell fate decisions and pathological gene regulatory networks by GATA4-targeted compounds modulating chamber-specific transcriptional programs in multipotent cardiac progenitor cells and cardiomyocytes. The compound scaffolds described within this study could be used to develop regenerative strategies for myocardial regeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02259-z.
Collapse
Affiliation(s)
- Mika J Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Robert S Leigh
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Sini M Kinnunen
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Alexander R March
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Ana Hernández de Sande
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Matias Kinnunen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Merja Heinäniemi
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Bogac L Kaynak
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.
| | - Heikki Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.
| |
Collapse
|
15
|
Tang C, Shen C, Zhu K, Zhou Y, Chuang YJ, He C, Zuo Z. Exposure to the AhR agonist cyprodinil impacts the cardiac development and function of zebrafish larvae. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 201:110808. [PMID: 32516676 DOI: 10.1016/j.ecoenv.2020.110808] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/19/2020] [Accepted: 05/25/2020] [Indexed: 06/11/2023]
Abstract
Cyprodinil is a broad-spectrum pyrimidine amine fungicide that has been reportedly used worldwide. However, toxicity studies of cyprodinil on aquatic organisms, specifically zebrafish (Danio rerio), are lacking. In our present study, we predicted cyprodinil binding to the aryl hydrocarbon receptor (AhR) by using molecular docking simulation. Then, we used recombinant HepG2 cells and Tg(cyp1a1-12DRE:egfp) transgenic zebrafish to further assess the AhR agonistic activity of cyprodinil. Besides, the significant upregulation of cyp1a1 further verified that statement. Moreover, we found that zebrafish exposure to cyprodinil induced developmental toxicity in the larvae, particularly during cardiac development. The expression levels of cardiac development-related genes, namely tbx5, nkx2.5, gata4, and tnnt2, were markedly altered, which might cause the adverse effects of cyprodinil on cardiac function and development. In summary, we found that cyprodinil, as an AhR agonist, induced development toxicity in zebrafish larvae, especially on cardiac. Data here can assess the potential effects on organisms in the aquatic environment and promote the regulation and safe use of cyprodinil.
Collapse
Affiliation(s)
- Chen Tang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chao Shen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Kongyang Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yixi Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yung-Jen Chuang
- Department of Medical Science, Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China.
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China; State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, Fujian, 361005, China.
| |
Collapse
|
16
|
Floriani MA, Glaeser AB, Dorfman LE, Agnes G, Rosa RFM, Zen PRG. GATA 4 Deletions Associated with Congenital Heart Diseases in South Brazil. J Pediatr Genet 2020; 10:92-97. [PMID: 33996178 DOI: 10.1055/s-0040-1714691] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/10/2020] [Indexed: 01/10/2023]
Abstract
The normal development of the heart comprises a highly regulated machinery of genetic events, involving transcriptional factors. Congenital heart disease (CHD), have been associated with chromosomal abnormalities and copy number variants (CNVs). Our goal was to investigate through the multiplex ligation-dependent probe amplification (MLPA) technique, the presence of CNVs in reference genes for normal cardiac development in patients with CHD. GATA4 , NKX2-5 , TBX5 , BMP4 , and CRELD1 genes and 22q11.2 chromosome region were analyzed in 207 children with CHD admitted for the first time in a cardiac intensive care unit from a pediatric hospital. CNVs were detected in seven patients (3.4%): four had a 22q11.2 deletion (22q11DS) (1.9%), two had a GATA4 deletion (1%) and one had a 22q11.2 duplication (0.5%). No patients with CNVs in the NKX2-5 , TBX5 , BMP4 , and CRELD1 genes were identified. GATA4 deletions appear to be present in a significant number of CHD patients, especially those with septal defects, persistent left superior vena cava, pulmonary artery abnormalities, and extracardiac findings. GATA4 screening seems to be more effective when directed to these CHDs. The investigation of CNVs in GATA4 and 22q11 chromosome region in patients with CHD is important to anticipating the diagnosis, and to contributing to family planning.
Collapse
Affiliation(s)
- Maiara A Floriani
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Andressa B Glaeser
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Luiza E Dorfman
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Grasiela Agnes
- Molecular Biology Laboratory, Universidade Federal de Ciências da Saúde de Porto Alegre, Rio Grande do Sul, Brazil
| | - Rafael F M Rosa
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.,Department of Internal Medicine, Clinical Genetics, Universidade Federal de Ciências da Saúde de Porto Alegre and Irmandade Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Paulo R G Zen
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.,Department of Internal Medicine, Clinical Genetics, Universidade Federal de Ciências da Saúde de Porto Alegre and Irmandade Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
17
|
Abstract
Various strategies have been applied to replace the loss of cardiomyocytes in order to restore reduced cardiac function and prevent the progression of heart disease. Intensive research efforts in the field of cellular reprogramming and cell transplantation may eventually lead to efficient in vivo applications for the treatment of cardiac injuries, representing a novel treatment strategy for regenerative medicine. Modulation of cardiac transcription factor (TF) networks by chemical entities represents another viable option for therapeutic interventions. Comprehensive screening projects have revealed a number of molecular entities acting on molecular pathways highly critical for cellular lineage commitment and differentiation, including compounds targeting Wnt- and transforming growth factor beta (TGFβ)-signaling. Furthermore, previous studies have demonstrated that GATA4 and NKX2-5 are essential TFs in gene regulation of cardiac development and hypertrophy. For example, both of these TFs are required to fully activate mechanical stretch-responsive genes such as atrial natriuretic peptide and brain natriuretic peptide (BNP). We have previously reported that the compound 3i-1000 efficiently inhibited the synergy of the GATA4-NKX2-5 interaction. Cellular effects of 3i-1000 have been further characterized in a number of confirmatory in vitro bioassays, including rat cardiac myocytes and animal models of ischemic injury and angiotensin II-induced pressure overload, suggesting the potential for small molecule-induced cardioprotection.
Collapse
Affiliation(s)
- Mika J. Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiHelsinki, Finland
| | - Heikki J. Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiHelsinki, Finland
| |
Collapse
|
18
|
Jumppanen M, Kinnunen SM, Välimäki MJ, Talman V, Auno S, Bruun T, Boije Af Gennäs G, Xhaard H, Aumüller IB, Ruskoaho H, Yli-Kauhaluoma J. Synthesis, Identification, and Structure-Activity Relationship Analysis of GATA4 and NKX2-5 Protein-Protein Interaction Modulators. J Med Chem 2019; 62:8284-8310. [PMID: 31431011 PMCID: PMC7076710 DOI: 10.1021/acs.jmedchem.9b01086] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
Transcription factors GATA4 and NKX2-5
directly interact and synergistically
activate several cardiac genes and stretch-induced cardiomyocyte hypertrophy.
Previously, we identified phenylisoxazole carboxamide 1 as a hit compound, which inhibited the GATA4–NKX2-5 transcriptional
synergy. Here, the chemical space around the molecular structure of 1 was explored by synthesizing and characterizing 220 derivatives
and structurally related compounds. In addition to the synergistic
transcriptional activation, selected compounds were evaluated for
their effects on transcriptional activities of GATA4 and NKX2-5 individually
as well as potential cytotoxicity. The structure–activity relationship
(SAR) analysis revealed that the aromatic isoxazole substituent in
the southern part regulates the inhibition of GATA4–NKX2-5
transcriptional synergy. Moreover, inhibition of GATA4 transcriptional
activity correlated with the reduced cell viability. In summary, comprehensive
SAR analysis accompanied by data analysis successfully identified
potent and selective inhibitors of GATA4–NKX2-5 transcriptional
synergy and revealed structural features important for it.
Collapse
Affiliation(s)
| | | | | | - Virpi Talman
- Imperial College London, Imperial Centre for Translational and Experimental Medicine , National Heart and Lung Institute , Du Cane Road , London W12 0NN , United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Liang X, Chu G, Wang L, Lai G, Zhao Y. Role of Nkx2.5 in H 2O 2-induced Nsd1 suppression. Cell Stress Chaperones 2019; 24:697-707. [PMID: 31104268 PMCID: PMC6629736 DOI: 10.1007/s12192-019-00995-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 03/18/2019] [Accepted: 04/04/2019] [Indexed: 11/24/2022] Open
Abstract
Nuclear receptor-binding SET domain-containing protein 1 (Nsd1) acts as a histone lysine methyltransferase, and its role in oxidative stress-related abnormal embryonic heart development remains poorly understood. In the present study, H2O2 decreased the expression of Nsd1 and NK2 transcription factor related locus 5 (Nkx2.5). We further focused on Nkx2.5 modulating the transcription of Nsd1 in response to H2O2. Luciferase activity analysis indicated that a regulatory region from - 646 to - 282 is essential for the basal transcriptional activity, in which, an a Nkx2.5-binding element (NKE) was identified at - 412/- 406 of the Nsd1 promoter by electrophoresis mobility shift assay and a chromatin immunoprecipitation assay. H2O2 obviously reduced the p646-luc promoter activity, and the depletion of Nkx2.5 expression weakened H2O2 inhibition on the p646-luc promoter. The overexpression of Nkx2.5 increase Nsd1 p646-luc promoter activity, but did not affected p646-luc-mut. Furthermore, overexpression and depletion of Nkx2.5 led to the increase and decrease of Nsd1 protein and mRNA levels. These data indicated that H2O2-induced Nsd1 suppression resulted from the decrease of Nkx2.5 expression through the NKE element.
Collapse
Affiliation(s)
- Xiaoyan Liang
- Department of Clinical Genetics, China Medical University, No.36 San Hao Street, Shenyang, 110004, Liaoning, China
- Central Laboratory, Binzhou People's Hospital, Binzhou, 256600, Shandong, China
| | - Guoming Chu
- Department of Clinical Genetics, China Medical University, No.36 San Hao Street, Shenyang, 110004, Liaoning, China
| | - Leitong Wang
- Department of Clinical Genetics, China Medical University, No.36 San Hao Street, Shenyang, 110004, Liaoning, China
| | - Guangrui Lai
- Department of Clinical Genetics, China Medical University, No.36 San Hao Street, Shenyang, 110004, Liaoning, China
| | - Yanyan Zhao
- Department of Clinical Genetics, China Medical University, No.36 San Hao Street, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
20
|
Zhou W, Jiang D, Tian J, Liu L, Lu T, Huang X, Sun H. Acetylation of H3K4, H3K9, and H3K27 mediated by p300 regulates the expression of GATA4 in cardiocytes. Genes Dis 2018; 6:318-325. [PMID: 32042871 PMCID: PMC6997570 DOI: 10.1016/j.gendis.2018.10.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/08/2018] [Indexed: 12/23/2022] Open
Abstract
GATA4 is a particularly important cardiogenic transcription factor and serves as a potent driver of cardiogenesis. Recent progress in the field has made it clear that histone acetylation can influence gene expression through changing the structure of chromatin. Our previous research had revealed that hypo-acetylation could repress gata4 expression in cardiocytes, however the underlying mechanism by which this occurred was still unclear. To reveal the mechanism of histone acetylation involved in the regulation of gata4 transcription, we concentrated on P300, one of the important histone acetyltransferase associated with cardiogenesis. We found that P300 participated in gata4 expression through regulating histone acetylation in embryonic mouse hearts. RNAi-mediated downregulation of P300 modulated the global acetylation of H3 and the acetylation of H3K4, H3K9, and H3K27 in gata4 and Tbx5 promoters. Interestingly, there was an obvious inhibition of gata4 transcription, whereas Tbx5 was not influenced. Furthermore, SGC-CBP30, the selective inhibitor of the bromodomain in CBP/P300, downregulated gata4 transcription by repressing the acetylation of H3K4, H3K9, and H3K27 in the gata4 promoters. Taken together, our results identified that acetylation of H3K4, H3K9, and H3K27 mediated by P300 plays an important role in regulation of gata4 expression in cardiogenesis.
Collapse
Affiliation(s)
- Wei Zhou
- Heart Centre, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Dagui Jiang
- Heart Centre, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Jie Tian
- Heart Centre, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Lingjuan Liu
- Heart Centre, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Tiewei Lu
- Heart Centre, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Xupei Huang
- Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Huichao Sun
- Heart Centre, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China.,Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| |
Collapse
|
21
|
Dixit R, Narasimhan C, Balekundri VI, Agrawal D, Kumar A, Mohapatra B. Functionally significant, novel GATA4
variants are frequently associated with Tetralogy of Fallot. Hum Mutat 2018; 39:1957-1972. [DOI: 10.1002/humu.23620] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 08/13/2018] [Accepted: 08/20/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Ritu Dixit
- Cytogenetics Laboratory; Department of Zoology; Banaras Hindu University; Varanasi Uttar Pradesh India
| | - Chitra Narasimhan
- Department of Pediatric Cardiology; Sri Jayadeva Institute of Cardiovascular Sciences and Research; Bengaluru Karnataka India
| | - Vijyalakshmi I. Balekundri
- Super Speciality Hospital; Prime Minister Swasth Suraksha Yojana (PMSSY); Bengaluru Medical College and Research Institute; Bengaluru Karnataka India
| | - Damyanti Agrawal
- Department of Cardio-vascular and Thoracic Surgery; Institute of Medical Science; Banaras Hindu University; Varanasi Uttar Pradesh India
| | - Ashok Kumar
- Department of Pediatrics; Institute of Medical Sciences; Banaras Hindu University; Varanasi Uttar Pradesh India
| | - Bhagyalaxmi Mohapatra
- Cytogenetics Laboratory; Department of Zoology; Banaras Hindu University; Varanasi Uttar Pradesh India
| |
Collapse
|
22
|
Stem cells are the most sensitive screening tool to identify toxicity of GATA4-targeted novel small-molecule compounds. Arch Toxicol 2018; 92:2897-2911. [PMID: 29987409 PMCID: PMC6132687 DOI: 10.1007/s00204-018-2257-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 07/04/2018] [Indexed: 01/10/2023]
Abstract
Safety assessment of drug candidates in numerous in vitro and experimental animal models is expensive, time consuming and animal intensive. More thorough toxicity profiling already in the early drug discovery projects using human cell models, which more closely resemble the physiological cell types, would help to decrease drug development costs. In this study we aimed to compare different cardiac and stem cell models for in vitro toxicity testing and to elucidate structure–toxicity relationships of novel compounds targeting the cardiac transcription factor GATA4. By screening the effects of eight compounds at concentrations ranging from 10 nM up to 30 µM on the viability of eight different cell types, we identified significant cell type- and structure-dependent toxicity profiles. We further characterized two compounds in more detail using high-content analysis. The results highlight the importance of cell type selection for toxicity screening and indicate that stem cells represent the most sensitive screening model, which can detect toxicity that may otherwise remain unnoticed. Furthermore, our structure–toxicity analysis reveals a characteristic dihedral angle in the GATA4-targeted compounds that causes stem cell toxicity and thus helps to direct further drug development efforts towards non-toxic derivatives.
Collapse
|
23
|
Thomford NE, Dzobo K, Yao NA, Chimusa E, Evans J, Okai E, Kruszka P, Muenke M, Awandare G, Wonkam A, Dandara C. Genomics and Epigenomics of Congenital Heart Defects: Expert Review and Lessons Learned in Africa. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 22:301-321. [PMID: 29762087 PMCID: PMC6016577 DOI: 10.1089/omi.2018.0033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Congenital heart defects (CHD) are structural malformations found at birth with a prevalence of 1%. The clinical trajectory of CHD is highly variable and thus in need of robust diagnostics and therapeutics. Major surgical interventions are often required for most CHDs. In Africa, despite advances in life sciences infrastructure and improving education of medical scholars, the limited clinical data suggest that CHD detection and correction are still not at par with the rest of the world. But the toll and genetics of CHDs in Africa has seldom been systematically investigated. We present an expert review on CHD with lessons learned on Africa. We found variable CHD phenotype prevalence in Africa across countries and populations. There are important gaps and paucity in genomic studies of CHD in African populations. Among the available genomic studies, the key findings in Africa were variants in GATA4 (P193H), MTHFR 677TT, and MTHFR 1298CC that were associated with atrial septal defect, ventricular septal defect (VSD), Tetralogy of Fallot (TOF), and patent ductus arteriosus phenotypes and 22q.11 deletion, which is associated with TOF. There were no data on epigenomic association of CHD in Africa, however, other studies have shown an altered expression of miR-421 and miR-1233-3p to be associated with TOF and hypermethylation of CpG islands in the promoter of SCO2 gene also been associated with TOF and VSD in children with non-syndromic CHD. These findings signal the urgent need to develop and implement genetic and genomic research on CHD to identify the hereditary and genome-environment interactions contributing to CHD. These projected studies would also offer comparisons on CHD pathophysiology between African and other populations worldwide. Genomic research on CHD in Africa should be developed in parallel with next generation technology policy research and responsible innovation frameworks that examine the social and political factors that shape the emergence and societal embedding of new technologies.
Collapse
Affiliation(s)
- Nicholas Ekow Thomford
- 1 Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute for Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
- 2 School of Medical Sciences, University of Cape Coast , Cape Coast, Ghana
| | - Kevin Dzobo
- 3 ICGEB, Cape Town Component, University of Cape Town , Cape Town, South Africa
- 4 Division of Medical Biochemistry, IIDMM, Department of IBM, Faculty of Health Sciences, University of Cape Town , Cape Town, South Africa
| | - Nana Akyaa Yao
- 5 National Cardiothoracic Centre, Korle Bu Teaching Hospital , Accra, Ghana
- 6 University of Ghana Medical School, University of Ghana , Accra, Ghana
| | - Emile Chimusa
- 1 Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute for Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Jonathan Evans
- 1 Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute for Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Emmanuel Okai
- 2 School of Medical Sciences, University of Cape Coast , Cape Coast, Ghana
- 7 Cape Coast Teaching Hospital , Cape Coast, Ghana
| | - Paul Kruszka
- 8 National Human Genome Research Institute, Medical Genetics Branch, National Institutes of Health , Bethesda, Maryland, USA
| | - Maximilian Muenke
- 8 National Human Genome Research Institute, Medical Genetics Branch, National Institutes of Health , Bethesda, Maryland, USA
| | - Gordon Awandare
- 9 Department of Biochemistry, WACCBIP, University of Ghana , Legon, Accra, Ghana
| | - Ambroise Wonkam
- 1 Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute for Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Collet Dandara
- 1 Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute for Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| |
Collapse
|
24
|
Kinnunen SM, Tölli M, Välimäki MJ, Gao E, Szabo Z, Rysä J, Ferreira MPA, Ohukainen P, Serpi R, Correia A, Mäkilä E, Salonen J, Hirvonen J, Santos HA, Ruskoaho H. Cardiac Actions of a Small Molecule Inhibitor Targeting GATA4-NKX2-5 Interaction. Sci Rep 2018; 8:4611. [PMID: 29545582 PMCID: PMC5854571 DOI: 10.1038/s41598-018-22830-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/23/2018] [Indexed: 02/07/2023] Open
Abstract
Transcription factors are fundamental regulators of gene transcription, and many diseases, such as heart diseases, are associated with deregulation of transcriptional networks. In the adult heart, zinc-finger transcription factor GATA4 is a critical regulator of cardiac repair and remodelling. Previous studies also suggest that NKX2-5 plays function role as a cofactor of GATA4. We have recently reported the identification of small molecules that either inhibit or enhance the GATA4–NKX2-5 transcriptional synergy. Here, we examined the cardiac actions of a potent inhibitor (3i-1000) of GATA4–NKX2-5 interaction in experimental models of myocardial ischemic injury and pressure overload. In mice after myocardial infarction, 3i-1000 significantly improved left ventricular ejection fraction and fractional shortening, and attenuated myocardial structural changes. The compound also improved cardiac function in an experimental model of angiotensin II -mediated hypertension in rats. Furthermore, the up-regulation of cardiac gene expression induced by myocardial infarction and ischemia reduced with treatment of 3i-1000 or when micro- and nanoparticles loaded with 3i-1000 were injected intramyocardially or intravenously, respectively. The compound inhibited stretch- and phenylephrine-induced hypertrophic response in neonatal rat cardiomyocytes. These results indicate significant potential for small molecules targeting GATA4–NKX2-5 interaction to promote myocardial repair after myocardial infarction and other cardiac injuries.
Collapse
Affiliation(s)
- Sini M Kinnunen
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland.,Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Marja Tölli
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Mika J Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland.,Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Erhe Gao
- Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, United States of America
| | - Zoltan Szabo
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Jaana Rysä
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mónica P A Ferreira
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Pauli Ohukainen
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| | - Raisa Serpi
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Alexandra Correia
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Ermei Mäkilä
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, Turku, Finland
| | - Jarno Salonen
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, Turku, Finland
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Heikki Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland. .,Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
25
|
Wu Y, Zuo Z, Chen M, Zhou Y, Yang Q, Zhuang S, Wang C. The developmental effects of low-level procymidone towards zebrafish embryos and involved mechanism. CHEMOSPHERE 2018; 193:928-935. [PMID: 29874768 DOI: 10.1016/j.chemosphere.2017.11.105] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 06/08/2023]
Abstract
Procymidone (PCM), a dicarboximide fungicide, is widely used in agriculture to control plant diseases. In the present study, zebrafish embryos were exposed to PCM at 0, 10, 100 and 1000 ng/L for 72 h, the development and cardiac functioning of larvae were observed and determined. The results showed that hatching rate was significantly decreased in the 1000 ng/L treatment, and pericardial edema rate and spine curvature rate were significantly increased in the 100 and 1000 ng/L groups. The PCM-treated larvae exhibited an increased heart rate as well as arrhythmia, and shortened low jaw. The transcription levels of cardiac development-related genes tbx5, nkx2.5, tnnt2, gata4, myh6, myl7, cdh2, ryr2 were altered, which might be responsible for the cardiac developmental and functioning defects in the larvae. The deformation in bone development might be related with the impaired transcription levels of ihh, shh, bmp2b, bmp4, gh, igf1, sox9, gli2. The activities of Na+/K+-ATPase and Ca2+-ATPase were significantly inhibited by 100 ng/L and 1000 ng/L PCM exposure, which might be a cause for the occurrence of pericardial edema and skeletal deformation. The results of this study will be helpful in evaluating the potential threat of PCM to fish population in the aquatic environment.
Collapse
Affiliation(s)
- Yuqiong Wu
- Wuyi University, College of Tea and Food Science, Wuyishan, Fujian 354300, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Meng Chen
- Key Laboratory of the Coastal and Wetland Ecosystems (Xiamen University), Ministry of Education, China
| | - Yixi Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Qihong Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Shanshan Zhuang
- Key Laboratory of the Coastal and Wetland Ecosystems (Xiamen University), Ministry of Education, China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China.
| |
Collapse
|
26
|
Välimäki MJ, Tölli MA, Kinnunen SM, Aro J, Serpi R, Pohjolainen L, Talman V, Poso A, Ruskoaho HJ. Discovery of Small Molecules Targeting the Synergy of Cardiac Transcription Factors GATA4 and NKX2-5. J Med Chem 2017; 60:7781-7798. [PMID: 28858485 DOI: 10.1021/acs.jmedchem.7b00816] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Transcription factors are pivotal regulators of gene transcription, and many diseases are associated with the deregulation of transcriptional networks. In the heart, the transcription factors GATA4 and NKX2-5 are required for cardiogenesis. GATA4 and NKX2-5 interact physically, and the activation of GATA4, in cooperation with NKX2-5, is essential for stretch-induced cardiomyocyte hypertrophy. Here, we report the identification of four small molecule families that either inhibit or enhance the GATA4-NKX2-5 transcriptional synergy. A fragment-based screening, reporter gene assay, and pharmacophore search were utilized for the small molecule screening, identification, and optimization. The compounds modulated the hypertrophic agonist-induced cardiac gene expression. The most potent hit compound, N-[4-(diethylamino)phenyl]-5-methyl-3-phenylisoxazole-4-carboxamide (3, IC50 = 3 μM), exhibited no activity on the protein kinases involved in the regulation of GATA4 phosphorylation. The identified and chemically and biologically characterized active compound, and its derivatives may provide a novel class of small molecules for modulating heart regeneration.
Collapse
Affiliation(s)
- Mika J Välimäki
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki , Helsinki FI-00014, Finland.,Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu , Oulu FI-90014, Finland
| | - Marja A Tölli
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu , Oulu FI-90014, Finland
| | - Sini M Kinnunen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki , Helsinki FI-00014, Finland.,Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu , Oulu FI-90014, Finland
| | - Jani Aro
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu , Oulu FI-90014, Finland
| | - Raisa Serpi
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu , Oulu FI-90014, Finland
| | - Lotta Pohjolainen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki , Helsinki FI-00014, Finland
| | - Virpi Talman
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki , Helsinki FI-00014, Finland
| | - Antti Poso
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland , Kuopio FI-70211, Finland
| | - Heikki J Ruskoaho
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki , Helsinki FI-00014, Finland.,Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu , Oulu FI-90014, Finland
| |
Collapse
|
27
|
Tong YF. Mutations of NKX2.5 and GATA4 genes in the development of congenital heart disease. Gene 2016; 588:86-94. [DOI: 10.1016/j.gene.2016.04.061] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/27/2016] [Accepted: 04/29/2016] [Indexed: 12/17/2022]
|