1
|
Feng H, Yang S, Zhang L, Zhu J, Li J, Yang Z. A new Prdm1-Cre line is suitable for studying the second heart field development. Dev Biol 2024; 514:78-86. [PMID: 38880275 DOI: 10.1016/j.ydbio.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
The second heart field (SHF) plays a pivotal role in heart development, particularly in outflow tract (OFT) morphogenesis and septation, as well as in the expansion of the right ventricle (RV). Two mouse Cre lines, the Mef2c-AHF-Cre (Mef2c-Cre) and Isl1-Cre, have been widely used to study the SHF development. However, Cre activity is triggered not only in the SHF but also in the RV in the Mef2c-Cre mice, and in the Isl1-Cre mice, Cre activation is not SHF-specific. Therefore, a more suitable SHF-Cre line is desirable for better understanding SHF development. Here, we generated and characterized the Prdm1-Cre knock-in mice. In comparison with Mef2c-Cre mice, the Cre activity is similar in the pharyngeal and splanchnic mesoderm, and in the OFT of the Prdm1-Cre mice. Nonetheless, it was noticed that Cre expression is largely reduced in the RV of Prdm1-Cre mice compared to the Mef2c-Cre mice. Furthermore, we deleted Hand2, Nkx2-5, Pdk1 and Tbx20 using both Mef2c-Cre and Prdm1-Cre mice to study OFT morphogenesis and septation, making a comparison between these two Cre lines. New insights were obtained in understanding SHF development including differentiation into cardiomyocytes in the OFT using Prdm1-Cre mice. In conclusion, we found that Prdm1-Cre mouse line is a more appropriate tool to monitor SHF development, while the Mef2c-Cre mice are excellent in studying the role and function of the SHF in OFT morphogenesis and septation.
Collapse
Affiliation(s)
- Haiyue Feng
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Suming Yang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Lijun Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Jingai Zhu
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Jinsong Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
2
|
Zhang C, Ge L, Xie H, Liu X, Xun C, Chen Y, Chen H, Lu M, Chen P. Retinoic acid induced specific changes in the phosphoproteome of C17.2 neural stem cells. J Cell Mol Med 2024; 28:e18205. [PMID: 38506089 PMCID: PMC10951872 DOI: 10.1111/jcmm.18205] [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: 09/30/2023] [Revised: 01/16/2024] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
Retinoic acid (RA), a vitamin A derivative, is an effective cell differentiating factor which plays critical roles in neuronal differentiation induction and the production of neurotransmitters in neurons. However, the specific changes in phosphorylation levels and downstream signalling pathways associated with RA remain unclear. This study employed qualitative and quantitative phosphoproteomics approaches based on mass spectrometry to investigate the phosphorylation changes induced by RA in C17.2 neural stem cells (NSCs). Dimethyl labelling, in conjunction with TiO2 phosphopeptide enrichment, was utilized to profile the phosphoproteome of self-renewing and RA-induced differentiated cells in C17.2 NSCs. The results of our study revealed that, qualitatively, 230 and 14 phosphoproteins were exclusively identified in the self-renewal and RA-induced groups respectively. Quantitatively, we successfully identified and quantified 177 unique phosphoproteins, among which 70 exhibited differential phosphorylation levels. Analysis of conserved phosphorylation motifs demonstrated enrichment of motifs corresponding to cyclin-dependent kinase and MAPK in the RA-induced group. Additionally, through a comprehensive literature and database survey, we found that the differentially expressed proteins were associated with the Wnt/β-catenin and Hippo signalling pathways. This work sheds light on the changes in phosphorylation levels induced by RA in C17.2 NSCs, thereby expanding our understanding of the molecular mechanisms underlying RA-induced neuronal differentiation.
Collapse
Affiliation(s)
- Cheng Zhang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life SciencesHunan Normal UniversityChangshaPR China
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life ScienceSouth China Normal UniversityGuangzhouPR China
| | - Lite Ge
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life SciencesHunan Normal UniversityChangshaPR China
- Hunan Provincial Key Laboratory of Neurorestoratology, the Second Affiliated HospitalHunan Normal UniversityChangshaPR China
- Department of Neurology, Second Xiangya HospitalCentral South UniversityChangshaPR China
| | - Huali Xie
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life SciencesHunan Normal UniversityChangshaPR China
| | - Xiaoqian Liu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life SciencesHunan Normal UniversityChangshaPR China
| | - Chengfeng Xun
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life SciencesHunan Normal UniversityChangshaPR China
| | - Yan Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life SciencesHunan Normal UniversityChangshaPR China
| | - Haiyan Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life SciencesHunan Normal UniversityChangshaPR China
| | - Ming Lu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life SciencesHunan Normal UniversityChangshaPR China
- Department of Neurology, Second Xiangya HospitalCentral South UniversityChangshaPR China
| | - Ping Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life SciencesHunan Normal UniversityChangshaPR China
| |
Collapse
|
3
|
Luo H, Yang Z, Li J, Jin H, Jiang M, Shan C. Deletion of PDK 1 Caused Cardiac Malmorphogenesis and Heart Defects Due to Profound Protein Phosphorylation Changes Mediated by SHP 2. J Cardiovasc Transl Res 2023; 16:1220-1231. [PMID: 36988860 DOI: 10.1007/s12265-023-10380-y] [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: 02/17/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023]
Abstract
Phosphoinositide-dependent protein kinase-1 (PDK1), a master kinase and involved in multiple signaling transduction, participates in regulating embryonic cardiac development and postnatal cardiac remodeling. Germline PDK1 knockout mice displayed no heart development; in this article, we deleted PDK1 in heart tissue with different cre to characterize the temporospatial features and find the relevance with congenital heart disease(CHD), furthermore to investigate the underlying mechanism. Knocking out PDK1 with Nkx2.5-cre, the heart showed prominent pulmonic stenosis. Ablated PDK1 with Mef2cSHF-cre, the second heart field (SHF) exhibited severe hypoplasia. And deleted PDK1 with αMHC-cre, the mice displayed dilated heart disease, protein analysis indicated PI3K and ERK were activated; meanwhile, PDK1-AKT-GSK3, and S6K-S6 were disrupted; phosphorylation level of Akt473, S6k421/424, and Gsk3α21 enhanced; however, Akt308, S6k389, and Gsk3β9 decreased. In mechanism investigation, we found SHP2 membrane localization and phosphorylation level of SHP2542 elevated, which suggested SHP2 likely mediated the disruption.
Collapse
Affiliation(s)
- Hongmei Luo
- Guangdong Medical University, Guangdong Dongguan, 523808, China.
- Model Animal Research Center, Nanjing University, Jiangsu Nanjing, 210028, China.
| | - Zhongzhou Yang
- Model Animal Research Center, Nanjing University, Jiangsu Nanjing, 210028, China
| | - Jie Li
- Model Animal Research Center, Nanjing University, Jiangsu Nanjing, 210028, China
| | - Hengwei Jin
- Model Animal Research Center, Nanjing University, Jiangsu Nanjing, 210028, China
| | - Mingyang Jiang
- Model Animal Research Center, Nanjing University, Jiangsu Nanjing, 210028, China
| | - Congjia Shan
- Model Animal Research Center, Nanjing University, Jiangsu Nanjing, 210028, China
| |
Collapse
|
4
|
Bragança J, Pinto R, Silva B, Marques N, Leitão HS, Fernandes MT. Charting the Path: Navigating Embryonic Development to Potentially Safeguard against Congenital Heart Defects. J Pers Med 2023; 13:1263. [PMID: 37623513 PMCID: PMC10455635 DOI: 10.3390/jpm13081263] [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: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Congenital heart diseases (CHDs) are structural or functional defects present at birth due to improper heart development. Current therapeutic approaches to treating severe CHDs are primarily palliative surgical interventions during the peri- or prenatal stages, when the heart has fully developed from faulty embryogenesis. However, earlier interventions during embryonic development have the potential for better outcomes, as demonstrated by fetal cardiac interventions performed in utero, which have shown improved neonatal and prenatal survival rates, as well as reduced lifelong morbidity. Extensive research on heart development has identified key steps, cellular players, and the intricate network of signaling pathways and transcription factors governing cardiogenesis. Additionally, some reports have indicated that certain adverse genetic and environmental conditions leading to heart malformations and embryonic death may be amendable through the activation of alternative mechanisms. This review first highlights key molecular and cellular processes involved in heart development. Subsequently, it explores the potential for future therapeutic strategies, targeting early embryonic stages, to prevent CHDs, through the delivery of biomolecules or exosomes to compensate for faulty cardiogenic mechanisms. Implementing such non-surgical interventions during early gestation may offer a prophylactic approach toward reducing the occurrence and severity of CHDs.
Collapse
Affiliation(s)
- José Bragança
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Rute Pinto
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Bárbara Silva
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- PhD Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve, 8005-139 Faro, Portugal
| | - Nuno Marques
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- School of Health, University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
5
|
Balatskyi VV, Sowka A, Dobrzyn P, Piven OO. WNT/β-catenin pathway is a key regulator of cardiac function and energetic metabolism. Acta Physiol (Oxf) 2023; 237:e13912. [PMID: 36599355 DOI: 10.1111/apha.13912] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/24/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
The WNT/β-catenin pathway is a master regulator of cardiac development and growth, and its activity is low in healthy adult hearts. However, even this low activity is essential for maintaining normal heart function. Acute activation of the WNT/β-catenin signaling cascade is considered to be cardioprotective after infarction through the upregulation of prosurvival genes and reprogramming of metabolism. Chronically high WNT/β-catenin pathway activity causes profibrotic and hypertrophic effects in the adult heart. New data suggest more complex functions of β-catenin in metabolic maturation of the perinatal heart, establishing an adult pattern of glucose and fatty acid utilization. Additionally, low basal activity of the WNT/β-catenin cascade maintains oxidative metabolism in the adult heart, and this pathway is reactivated by physiological or pathological stimuli to meet the higher energy needs of the heart. This review summarizes the current state of knowledge of the organization of canonical WNT signaling and its function in cardiogenesis, heart maturation, adult heart function, and remodeling. We also discuss the role of the WNT/β-catenin pathway in cardiac glucose, lipid metabolism, and mitochondrial physiology.
Collapse
Affiliation(s)
- Volodymyr V Balatskyi
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Adrian Sowka
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Pawel Dobrzyn
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Oksana O Piven
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Department of Human Genetics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
6
|
Gu Y, Zhou Y, Ju S, Liu X, Zhang Z, Guo J, Gao J, Zang J, Sun H, Chen Q, Wang J, Xu J, Xu Y, Chen Y, Guo Y, Dai J, Ma H, Wang C, Jin G, Li C, Xia Y, Shen H, Yang Y, Guo X, Hu Z. Multi-omics profiling visualizes dynamics of cardiac development and functions. Cell Rep 2022; 41:111891. [PMID: 36577384 DOI: 10.1016/j.celrep.2022.111891] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 09/14/2022] [Accepted: 12/05/2022] [Indexed: 12/29/2022] Open
Abstract
Cardiogenesis is a tightly regulated dynamic process through a continuum of differentiation and proliferation events. Key factors and pathways governing this process remain incompletely understood. Here, we investigate mice hearts from embryonic day 10.5 to postnatal week 8 and dissect developmental changes in phosphoproteome-, proteome-, metabolome-, and transcriptome-encompassing cardiogenesis and cardiac maturation. We identify mitogen-activated protein kinases as core kinases involved in transcriptional regulation by mediating the phosphorylation of chromatin remodeling proteins during early cardiogenesis. We construct the reciprocal regulatory network of transcription factors (TFs) and identify a series of TFs controlling early cardiogenesis involved in cycling-dependent proliferation. After birth, we identify cardiac resident macrophages with high arachidonic acid metabolism activities likely involved in the clearance of injured apoptotic cardiomyocytes. Together, our comprehensive multi-omics data offer a panoramic view of cardiac development and maturation that provides a resource for further in-depth functional exploration.
Collapse
Affiliation(s)
- Yayun Gu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Yan Zhou
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Sihan Ju
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaofei Liu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Zicheng Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Jia Guo
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Jimiao Gao
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Jie Zang
- School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Hao Sun
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Qi Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Jinghan Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Jiani Xu
- School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Yiqun Xu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Yingjia Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Yueshuai Guo
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Juncheng Dai
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Hongxia Ma
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Cheng Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Guangfu Jin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Chaojun Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Hongbing Shen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Yang Yang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211100, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211100, China.
| |
Collapse
|
7
|
Zhao K, Yang Z. The second heart field: the first 20 years. Mamm Genome 2022:10.1007/s00335-022-09975-8. [PMID: 36550326 DOI: 10.1007/s00335-022-09975-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
In 2001, three independent groups reported the identification of a novel cluster of progenitor cells that contribute to heart development in mouse and chicken embryos. This population of progenitor cells was designated as the second heart field (SHF), and a new research direction in heart development was launched. Twenty years have since passed and a comprehensive understanding of the SHF has been achieved. This review provides retrospective insights in to the contribution, the signaling regulatory networks and the epithelial properties of the SHF. It also includes the spatiotemporal characteristics of SHF development and interactions between the SHF and other types of cells during heart development. Although considerable efforts will be required to investigate the cellular heterogeneity of the SHF, together with its intricate regulatory networks and undefined mechanisms, it is expected that the burgeoning new technology of single-cell sequencing and precise lineage tracing will advance the comprehension of SHF function and its molecular signals. The advances in SHF research will translate to clinical applications and to the treatment of congenital heart diseases, especially conotruncal defects, as well as to regenerative medicine.
Collapse
Affiliation(s)
- Ke Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China.
| |
Collapse
|
8
|
Kim YJ. Activity-induced synaptic structural modifications by Akt. Biochem Biophys Res Commun 2022; 621:94-100. [PMID: 35820284 DOI: 10.1016/j.bbrc.2022.06.093] [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: 05/31/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022]
Abstract
The activity-dependent regulation of synaptic structures plays a key role in synaptic development and plasticity; however, the signaling mechanisms involved remain largely unknown. The serine/threonine protein kinase Akt, a downstream effector of phosphoinositide 3-kinase (PI3K), plays a pivotal role in a wide range of physiological functions. We focused on the importance of Akt in rapid synaptic structural changes after stimulation at the Drosophila neuromuscular junction, a well-studied model synapse. Compared with wild-type larvae, akt mutants showed significantly reduced muscle size and an increased number of boutons per area, suggesting that Akt is required for proper pre- and postsynaptic growth. In addition, the level of cysteine string protein (CSP) was significantly increased, and its distribution was different in akt mutants. After high K+ single stimulation, the CSP level of akt mutant NMJs increased dramatically compared with that of wild-type NMJs. Interestingly, ghost boutons without postsynaptic specialization were found in akt mutant NMJs, and the number of these boutons was significantly increased by patterned stimulation. In contrast, the postsynaptic change in the subsynaptic reticulum (SSR) in the akt mutant occurred independent of stimulation. These results suggest that Akt functions in both pre- and postsynaptic growth and differentiation, and in particular, presynaptic action occurs in an activity-dependent manner.
Collapse
Affiliation(s)
- Yoon-Jung Kim
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, South Korea.
| |
Collapse
|
9
|
Wang W, Lu G, Liu H, Xiong Z, Leung H, Cao R, Pang AL, Su X, Law PWN, Zhao Z, Chen Z, Chan W. Pten Regulates Cardiomyocyte Differentiation by Modulating Non-CG Methylation via Dnmt3. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100849. [PMID: 34247447 PMCID: PMC8425920 DOI: 10.1002/advs.202100849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/04/2021] [Indexed: 06/13/2023]
Abstract
The regulation of cardiomyocyte differentiation is a fundamental aspect of cardiac development and regenerative medicine. PTEN plays important roles during embryonic development. However, its role in cardiomyocyte differentiation remains unknown. In this study, a low-cost protocol for cardiomyocyte differentiation from mouse embryonic stem cells (ESCs) is presented and it is shown that Pten deletion potently suppresses cardiomyocyte differentiation. Transcriptome analysis shows that the expression of a series of cardiomyocyte marker genes is downregulated in Pten-/- cardiomyocytes. Pten ablation induces Dnmt3b expression via the AKT/FoxO3a pathway and regulates the expression of a series of imprinted genes, including Igf2. Double knockout of Dnmt3l and Dnmt3b rescues the deficiency of cardiomyocyte differentiation of Pten-/- ESCs. The DNA methylomes from wild-type and Pten-/- embryoid bodies and cardiomyocytes are analyzed by whole-genome bisulfite sequencing. Pten deletion significantly promotes the non-CG (CHG and CHH) methylation levels of genomic DNA during cardiomyocyte differentiation, and the non-CG methylation levels of cardiomyocyte genes and Igf2 are increased in Pten-/- cardiomyocytes. Igf2 or Igf1r deletion also suppresses cardiomyocyte differentiation through the MAPK/ERK signaling pathway, and IGF2 supplementation partially rescues the cardiomyocyte differentiation. Finally, Pten conditional knockout mice are generated and the role of PTEN in cardiomyocyte differentiation is verified in vivo.
Collapse
Affiliation(s)
- Wuming Wang
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Gang Lu
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Hong‐Bin Liu
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Zhiqiang Xiong
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
- SDIVF R&D Centre12W, Hong Kong Science ParkShatinHong KongChina
| | - Ho‐Duen Leung
- SDIVF R&D Centre12W, Hong Kong Science ParkShatinHong KongChina
| | - Ruican Cao
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Alan Lap‐Yin Pang
- R&D DivisionTGD Life Company Limited15W, Hong Kong Science ParkShatinHong KongChina
| | - Xianwei Su
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
- SDIVF R&D Centre12W, Hong Kong Science ParkShatinHong KongChina
| | - Patrick Wai Nok Law
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
| | - Zhiju Zhao
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
| | - Zi‐Jiang Chen
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Wai‐Yee Chan
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| |
Collapse
|
10
|
Khosravi F, Ahmadvand N, Bellusci S, Sauer H. The Multifunctional Contribution of FGF Signaling to Cardiac Development, Homeostasis, Disease and Repair. Front Cell Dev Biol 2021; 9:672935. [PMID: 34095143 PMCID: PMC8169986 DOI: 10.3389/fcell.2021.672935] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
The current focus on cardiovascular research reflects society’s concerns regarding the alarming incidence of cardiac-related diseases and mortality in the industrialized world and, notably, an urgent need to combat them by more efficient therapies. To pursue these therapeutic approaches, a comprehensive understanding of the mechanism of action for multifunctional fibroblast growth factor (FGF) signaling in the biology of the heart is a matter of high importance. The roles of FGFs in heart development range from outflow tract formation to the proliferation of cardiomyocytes and the formation of heart chambers. In the context of cardiac regeneration, FGFs 1, 2, 9, 16, 19, and 21 mediate adaptive responses including restoration of cardiac contracting rate after myocardial infarction and reduction of myocardial infarct size. However, cardiac complications in human diseases are correlated with pathogenic effects of FGF ligands and/or FGF signaling impairment. FGFs 2 and 23 are involved in maladaptive responses such as cardiac hypertrophic, fibrotic responses and heart failure. Among FGFs with known causative (FGFs 2, 21, and 23) or protective (FGFs 2, 15/19, 16, and 21) roles in cardiac diseases, FGFs 15/19, 21, and 23 display diagnostic potential. The effective role of FGFs on the induction of progenitor stem cells to cardiac cells during development has been employed to boost the limited capacity of postnatal cardiac repair. To renew or replenish damaged cardiomyocytes, FGFs 1, 2, 10, and 16 were tested in (induced-) pluripotent stem cell-based approaches and for stimulation of cell cycle re-entry in adult cardiomyocytes. This review will shed light on the wide range of beneficiary and detrimental actions mediated by FGF ligands and their receptors in the heart, which may open new therapeutic avenues for ameliorating cardiac complications.
Collapse
Affiliation(s)
- Farhad Khosravi
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Negah Ahmadvand
- Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Saverio Bellusci
- Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
11
|
Abstract
Dynamic changes in microRNAs in oocyte and cumulus cells before and after maturation may explain the spatiotemporal post-transcriptional gene regulation within bovine follicular cells during the oocyte maturation process. miR-20a has been previously shown to regulate proliferation and differentiation as well as progesterone levels in cultured bovine granulosa cells. In the present study, we aimed to demonstrate the function of miR-20a during the bovine oocyte maturation process. Maturation of cumulus-oocyte complexes (COCs) was performed at 39°C in an humidified atmosphere with 5% CO2 in air. The expression of miR-20a was investigated in the cumulus cells and oocytes at 22 h post culture. The functional role of miR-20a was examined by modulating the expression of miR-20a in COCs during in vitro maturation (IVM). We found that the miR-20a expression was increased in cumulus cells but decreased in oocytes after IVM. Overexpression of miR-20a increased the oocyte maturation rate. Even though not statistically significant, miR-20a overexpression during IVM increased progesterone levels in the spent medium. This was further supported by the expression of STAR and CYP11A1 genes in cumulus cells. The phenotypes observed due to overexpression of miR-20a were validated by BMP15 supplementation during IVM and subsequent transfection of BMP15-treated COCs using miR-20a mimic or BMPR2 siRNA. We found that miR-20a mimic or BMPR2 siRNA transfection rescued BMP15-reduced oocyte maturation and progesterone levels. We concluded that miR-20a regulates oocyte maturation by increasing cumulus cell progesterone synthesis by simultaneous suppression of BMPR2 expression.
Collapse
|
12
|
Guo R, Li J, Chen C, Xiao M, Liao M, Hu Y, Liu Y, Li D, Zou J, Sun D, Torre V, Zhang Q, Chai R, Tang M. Biomimetic 3D bacterial cellulose-graphene foam hybrid scaffold regulates neural stem cell proliferation and differentiation. Colloids Surf B Biointerfaces 2021; 200:111590. [PMID: 33529926 DOI: 10.1016/j.colsurfb.2021.111590] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/29/2020] [Accepted: 01/22/2021] [Indexed: 01/09/2023]
Abstract
Neural stem cell (NSC)-based therapy is a promising candidate for treating neurodegenerative diseases and the preclinical researches call an urgent need for regulating the growth and differentiation of such cells. The recognition that three-dimensional culture has the potential to be a biologically significant system has stimulated an extraordinary impetus for scientific researches in tissue engineering and regenerative medicine. Here, A novel scaffold for culturing NSCs, three-dimensional bacterial cellulose-graphene foam (3D-BC/G), which was prepared via in situ bacterial cellulose interfacial polymerization on the skeleton surface of porous graphene foam has been reported. 3D-BC/G not only supports NSC growth and adhesion, but also maintains NSC stemness and enhances their proliferative capacity. Further phenotypic analysis indicated that 3D-BC/G induces NSCs to selectively differentiate into neurons, forming a neural network in a short amount of time. The scaffold has good biocompatibility with primary cortical neurons enhancing the neuronal network activities. To explore the underlying mechanisms, RNA-Seq analysis to identify genes and signaling pathways was performed and it suggests that 3D-BC/G offers a more promising three-dimensional conductive substrate for NSC research and neural tissue engineering, and the repertoire of gene expression serves as a basis for further studies to better understand NSC biology.
Collapse
Affiliation(s)
- Rongrong Guo
- School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China; Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Jian Li
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China
| | - Chuntao Chen
- Institute of Chemicobiology and Functional Materials, Key Laboratory for Soft Chemistry and Functional Materials of Ministry Education, School of Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, 210094, Jiangsu Province, China
| | - Miao Xiao
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, China; International School for Advanced Studies (SISSA), via Bonomea 265, Trieste, 34136, Italy
| | - Menghui Liao
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yangnan Hu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yun Liu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Dan Li
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China; Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China
| | - Jun Zou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, China
| | - Dongping Sun
- Institute of Chemicobiology and Functional Materials, Key Laboratory for Soft Chemistry and Functional Materials of Ministry Education, School of Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, 210094, Jiangsu Province, China
| | - Vincent Torre
- International School for Advanced Studies (SISSA), via Bonomea 265, Trieste, 34136, Italy
| | - Qi Zhang
- School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China; Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China; Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China.
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China; Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
13
|
Calore M, Lorenzon A, Vitiello L, Poloni G, Khan MAF, Beffagna G, Dazzo E, Sacchetto C, Polishchuk R, Sabatelli P, Doliana R, Carnevale D, Lembo G, Bonaldo P, De Windt L, Braghetta P, Rampazzo A. A novel murine model for arrhythmogenic cardiomyopathy points to a pathogenic role of Wnt signalling and miRNA dysregulation. Cardiovasc Res 2020; 115:739-751. [PMID: 30304392 DOI: 10.1093/cvr/cvy253] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 09/06/2018] [Accepted: 10/09/2018] [Indexed: 12/19/2022] Open
Abstract
AIMS Arrhythmogenic cardiomyopathy (AC) is one of the most common inherited cardiomyopathies, characterized by progressive fibro-fatty replacement in the myocardium. Clinically, AC manifests itself with ventricular arrhythmias, syncope, and sudden death and shows wide inter- and intra-familial variability. Among the causative genes identified so far, those encoding for the desmosomal proteins plakophilin-2 (PKP2), desmoplakin (DSP), and desmoglein-2 (DSG2) are the most commonly mutated. So far, little is known about the molecular mechanism(s) behind such a varied spectrum of phenotypes, although it has been shown that the causative mutations not only lead to structural abnormalities but also affect the miRNA profiling of cardiac tissue. Here, we aimed at studying the pathogenic effects of a nonsense mutation of the desmoglein-2 gene, both at the structural level and in terms of miRNA expression pattern. METHODS AND RESULTS We generated transgenic mice with cardiomyocyte-specific overexpression of a FLAG-tagged human desmoglein-2 harbouring the Q558* nonsense mutation found in an AC patient. The hearts of these mice showed signs of fibrosis, decrease in desmosomal size and number, and reduction of the Wnt/β-catenin signalling. Genome-wide RNA-Seq performed in Tg-hQ hearts and non-transgenic hearts revealed that 24 miRNAs were dysregulated in transgenic animals. Further bioinformatic analyses for selected miRNAs suggested that miR-217-5p, miR-499-5p, and miR-708-5p might be involved in the pathogenesis of the disease. CONCLUSION Down-regulation of the canonical Wnt/β-catenin signalling might be considered a common key event in the AC pathogenesis. We identified the miRNA signature in AC hearts, with miR-708-5p and miR-217-5p being the most up-regulated and miR-499-5p the most down-regulated miRNAs. All of them were predicted to be involved in the regulation of the Wnt/β-catenin pathway and might reveal the potential pathophysiology mechanisms of AC, as well as be useful as therapeutic targets for the disease.
Collapse
Affiliation(s)
- Martina Calore
- Department of Biology, University of Padua, Via Ugo Bassi 58/B, Padua, Italy.,Department of Cardiology, Faculty of Health, Medicine and Life Sciences, Maastricht University, MD Maastricht, The Netherlands
| | - Alessandra Lorenzon
- Department of Biology, University of Padua, Via Ugo Bassi 58/B, Padua, Italy
| | - Libero Vitiello
- Department of Biology, University of Padua, Via Ugo Bassi 58/B, Padua, Italy.,Italian Inter-University Institute of Myology, Padua, Italy
| | - Giulia Poloni
- Department of Biology, University of Padua, Via Ugo Bassi 58/B, Padua, Italy
| | - Mohsin A F Khan
- Department of Experimental Cardiology, Biostatistics and Bioinformatics, Academic Medical Center, Amsterdam, The Netherlands
| | - Giorgia Beffagna
- Department of Biology, University of Padua, Via Ugo Bassi 58/B, Padua, Italy
| | - Emanuela Dazzo
- Department of Biology, University of Padua, Via Ugo Bassi 58/B, Padua, Italy
| | - Claudia Sacchetto
- Department of Biology, University of Padua, Via Ugo Bassi 58/B, Padua, Italy
| | - Roman Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Patrizia Sabatelli
- National Research Council of Italy, Institute of Molecular Genetics, Bologna, Italy
| | - Roberto Doliana
- Department of Translational Research, CRO-IRCCS National Cancer Institute, Aviano, Italy
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Pozzilli, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Pozzilli, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padua, Via Ugo Bassi 58/B, Padua, Italy
| | - Leon De Windt
- Department of Cardiology, Faculty of Health, Medicine and Life Sciences, Maastricht University, MD Maastricht, The Netherlands
| | - Paola Braghetta
- Department of Molecular Medicine, University of Padua, Via Ugo Bassi 58/B, Padua, Italy
| | - Alessandra Rampazzo
- Department of Biology, University of Padua, Via Ugo Bassi 58/B, Padua, Italy
| |
Collapse
|
14
|
Vahdat S, Pahlavan S, Mahmoudi E, Barekat M, Ansari H, Bakhshandeh B, Aghdami N, Baharvand H. Expansion of Human Pluripotent Stem Cell-derived Early Cardiovascular Progenitor Cells by a Cocktail of Signaling Factors. Sci Rep 2019; 9:16006. [PMID: 31690816 PMCID: PMC6831601 DOI: 10.1038/s41598-019-52516-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 10/16/2019] [Indexed: 11/15/2022] Open
Abstract
Cardiovascular progenitor cells (CPCs) derived from human pluripotent stem cells (hPSCs) are proposed to be invaluable cell sources for experimental and clinical studies. This wide range of applications necessitates large-scale production of CPCs in an in vitro culture system, which enables both expansion and maintenance of these cells. In this study, we aimed to develop a defined and efficient culture medium that uses signaling factors for large-scale expansion of early CPCs, called cardiogenic mesodermal cells (CMCs), which were derived from hPSCs. Chemical screening resulted in a medium that contained a reproducible combination of three factors (A83-01, bFGF, and CHIR99021) that generated 1014 CMCs after 10 passages without the propensity for tumorigenicity. Expanded CMCs retained their gene expression pattern, chromosomal stability, and differentiation tendency through several passages and showed both the safety and possible cardio-protective potentials when transplanted into the infarcted rat myocardium. These CMCs were efficiently cryopreserved for an extended period of time. This culture medium could be used for both adherent and suspension culture conditions, for which the latter is required for large-scale CMC production. Taken together, hPSC-derived CMCs exhibited self-renewal capacity in our simple, reproducible, and defined medium. These cells might ultimately be potential, promising cell sources for cardiovascular studies.
Collapse
Affiliation(s)
- Sadaf Vahdat
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Elena Mahmoudi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Barekat
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hassan Ansari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Nasser Aghdami
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
15
|
Conway SJ, McConnell R, Simmons O, Snider PL. Armadillo-like helical domain containing-4 is dynamically expressed in both the first and second heart fields. Gene Expr Patterns 2019; 34:119077. [PMID: 31655130 DOI: 10.1016/j.gep.2019.119077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/18/2019] [Accepted: 10/19/2019] [Indexed: 12/19/2022]
Abstract
Armadillo repeat and Armadillo-like helical domain containing proteins form a large family with diverse and fundamental functions in many eukaryotes. Herein we investigated the spatiotemporal expression pattern of Armadillo-like helical domain containing 4 (or Armh4) as an uncharacterized protein coding mouse gene, within the mouse embryo during the initial stages of heart morphogenesis. We found Armh4 is initially expressed in both first heart field as well as the second heart field progenitors and subsequently within predominantly their cardiomyocyte derivatives. Armh4 expression is initially cardiac-restricted in the developing embryo and is expressed in second heart field subpharyngeal mesoderm prior to cardiomyocyte differentiation, but Armh4 diminishes as the embryonic heart matures into the fetal heart. Armh4 is subsequently expressed in craniofacial structures and neural crest-derived dorsal root and trigeminal ganglia. Whereas lithium chloride-induced stimulation of Wnt/β-catenin signaling elevated Armh4 expression in both second heart field subpharyngeal mesodermal progenitors and outflow tract, right ventricle and atrial cardiomyocytes, neither a systemic loss of Islet-1 nor an absence of cardiac neural crest cells had any effect upon Armh4 expression. These results confirm that Wnt/β-catenin-responsive Armh4 is a useful specific biomarker of the FHF and SHF cardiomyocyte derivatives only.
Collapse
Affiliation(s)
- Simon J Conway
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Reagan McConnell
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, Northern Ireland, UK
| | - Olga Simmons
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Paige L Snider
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
16
|
Xie J, Lin J, Wei M, Teng Y, He Q, Yang G, Yang X. Sustained Akt signaling in articular chondrocytes causes osteoarthritis via oxidative stress-induced senescence in mice. Bone Res 2019; 7:23. [PMID: 31646013 PMCID: PMC6804644 DOI: 10.1038/s41413-019-0062-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/25/2019] [Accepted: 06/13/2019] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is an age-related disorder that is strongly associated with chondrocyte senescence. The causal link between disruptive PTEN/Akt signaling and chondrocyte senescence and the underlying mechanism are unclear. In this study, we found activated Akt signaling in human OA cartilage as well as in a mouse OA model with surgical destabilization of the medial meniscus. Genetic mouse models mimicking sustained Akt signaling in articular chondrocytes via PTEN deficiency driven by either Col2a1-Cre or Col2a1-CreERT2 developed OA, whereas restriction of Akt signaling reversed the OA phenotypes in PTEN-deficient mice. Mechanistically, prolonged activation of Akt signaling caused an accumulation of reactive oxygen species and triggered chondrocyte senescence as well as a senescence-associated secretory phenotype, whereas chronic administration of the antioxidant N-acetylcysteine suppressed chondrocyte senescence and mitigated OA progression in PTEN-deficient mice. Therefore, inhibition of Akt signaling by PTEN is required for the maintenance of articular cartilage. Disrupted Akt signaling in articular chondrocytes triggers oxidative stress-induced chondrocyte senescence and causes OA.
Collapse
Affiliation(s)
- Jing Xie
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Jingting Lin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Min Wei
- 2Department of Orthopaedics, Chinese PLA General Hospital, Beijing, 100853 China
| | - Yan Teng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Qi He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Guan Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206 China
| |
Collapse
|
17
|
Affiliation(s)
- Francisco X Galdos
- From the Cardiovascular Institute (F.X.G., S.M.W.), Stanford University School of Medicine, CA.,Institute for Stem Cell Biology and Regenerative Medicine (F.X.G.), Stanford University School of Medicine, CA
| | - Sean M Wu
- From the Cardiovascular Institute (F.X.G., S.M.W.), Stanford University School of Medicine, CA.,Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA
| |
Collapse
|
18
|
Jin H, Wang H, Li J, Yu S, Xu M, Qiu Z, Xia M, Zhu J, Feng Q, Xie J, Xu B, Yang Z. Differential contribution of the two waves of cardiac progenitors and their derivatives to aorta and pulmonary artery. Dev Biol 2019; 450:82-89. [DOI: 10.1016/j.ydbio.2019.03.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/07/2019] [Accepted: 03/27/2019] [Indexed: 10/27/2022]
|
19
|
Vahdat S, Bakhshandeh B. Prediction of putative small molecules for manipulation of enriched signalling pathways in hESC-derived early cardiovascular progenitors by bioinformatics analysis. IET Syst Biol 2019; 13:77-83. [PMID: 33444476 DOI: 10.1049/iet-syb.2018.5037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 08/25/2018] [Accepted: 09/21/2018] [Indexed: 11/20/2022] Open
Abstract
Human pluripotent stem cell-derived cardiovascular progenitor cells (CPCs) are considered as powerful tools for cardiac regenerative medicine and developmental study. Mesoderm posterior1+ (MESP1+ ) cells are identified as the earliest CPCs from which almost all cardiac cell types are generated. Molecular insights to the transcriptional regulatory factors of early CPCs are required to control cell fate decisions. Herein, the microarray data set of human embryonic stem cells (hESCs)-derived MESP1+ cells was analysed and differentially expressed genes (DEGs) were identified in comparison to undifferentiated hESCs and MESP1-negative cells. Then, gene ontology and pathway enrichment analysis of DEGs were carried out with the subsequent prediction of putative regulatory small molecules for modulation of CPC fate. Some key signalling cascades of cardiogenesis including Hippo, Wnt, transforming growth factor-β, and PI3K/Akt were highlighted in MESP1+ cells. The transcriptional regulatory network of MESP1+ cells were visualised through interaction networks of DEGs. Additionally, 35 promising chemicals were predicted based on correlations with gene expression signature of MESP1+ cells for effective in vitro CPC manipulation. Studying the transcriptional profile of MESP1+ cells resulted into the identification of important signalling pathways and chemicals which could be introduced as powerful tools to manage proliferation and differentiation of hESC-derived CPCs more efficiently.
Collapse
Affiliation(s)
- Sadaf Vahdat
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
20
|
Xiang Q, Yang B, Li L, Qiu B, Qiu C, Gao X, Zhou H(J, Min W. Critical role of Lin28-TNFR2 signalling in cardiac stem cell activation and differentiation. J Cell Mol Med 2019; 23:0. [PMID: 30734494 PMCID: PMC6433861 DOI: 10.1111/jcmm.14202] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/04/2019] [Accepted: 01/15/2019] [Indexed: 12/28/2022] Open
Abstract
Tumour necrotic factor receptor-2 (TNFR2) has been to be cardiac-protective and is expressed in cardiac progenitor cells. Our goal is to define the mechanism for TNFR2-mediated cardiac stem cell activation and differentiation. By employing a protocol of in vitro cardiac stem cell (CSC) differentiation from human inducible pluripotent stem cell (hiPSC), we show that expression of TNFR2 precedes expression of CSC markers followed by expression of mature cardiomyocyte proteins. Activation of TNFR2 by a specific agonist promotes whereas inhibition of TNFR2 by neutralizing antibody diminishes hiPSC-based CSC differentiation. Interestingly, pluripotent cell factor RNA-binding protein Lin28 enhances TNFR2 protein expression in early CSC activation by directly binding to a conserved Lin28-motif within the 3'UTR of Tnfr2 mRNA. Furthermore, inhibition of Lin28 blunts TNFR2 expression and TNFR2-dependent CSC activation and differentiation. Our study demonstrates a critical role of Lin28-TNFR2 axis in CSC activation and survival, providing a novel strategy to enhance stem cell-based therapy for the ischaemic heart diseases.
Collapse
Affiliation(s)
- Qiuling Xiang
- Yale Stem CenterInterdepartmental Program in Vascular Biology and TherapeuticsDepartment of PathologyYale University School of MedicineNew HavenConnecticut
- Translational Medicine Center, the First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdongChina
- Center for Stem Cell Biology and Tissue EngineeringKey Laboratory for Stem Cells and Tissue EngineeringMinistry of Education, Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Bicheng Yang
- Yale Stem CenterInterdepartmental Program in Vascular Biology and TherapeuticsDepartment of PathologyYale University School of MedicineNew HavenConnecticut
| | - Li Li
- Yale Stem CenterInterdepartmental Program in Vascular Biology and TherapeuticsDepartment of PathologyYale University School of MedicineNew HavenConnecticut
- Translational Medicine Center, the First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Bin Qiu
- Yale Stem CenterInterdepartmental Program in Vascular Biology and TherapeuticsDepartment of PathologyYale University School of MedicineNew HavenConnecticut
| | - Caihong Qiu
- Yale Stem CenterInterdepartmental Program in Vascular Biology and TherapeuticsDepartment of PathologyYale University School of MedicineNew HavenConnecticut
| | - Xiao‐Bing Gao
- Department of Comparative Medicine and Obstetrics, Gynecology, and Reproductive SciencesYale University School of MedicineNew HavenConnecticut
| | - Huanjiao (Jenny) Zhou
- Yale Stem CenterInterdepartmental Program in Vascular Biology and TherapeuticsDepartment of PathologyYale University School of MedicineNew HavenConnecticut
| | - Wang Min
- Yale Stem CenterInterdepartmental Program in Vascular Biology and TherapeuticsDepartment of PathologyYale University School of MedicineNew HavenConnecticut
| |
Collapse
|
21
|
Xia M, Luo W, Jin H, Yang Z. HAND2-mediated epithelial maintenance and integrity in cardiac outflow tract morphogenesis. Development 2019; 146:dev.177477. [PMID: 31201155 DOI: 10.1242/dev.177477] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/03/2019] [Indexed: 01/06/2023]
Abstract
During embryogenesis, epithelial organization is the prerequisite for organogenesis, in particular, for establishing the tubular structure. Recent studies provided hints about epithelial formation in early heart development, which has not been systemically explored. Here, we revealed a gradient of HAND2 protein in the cardiac progenitors in the anterior dorsal pericardial wall (aDPW) and adjacent transition zone (TZ) in the outflow tract (OFT). Deletion of Hand2 caused cell arrest and accumulation in the TZ leading to defective morphogenesis. While apicobasal cell polarity was unaffected, the key epithelial elements of adherens junction and cell-matrix adhesion were disrupted in the TZ of Hand2 mutant mice, indicating poorly formed epithelium. RNA-seq analysis revealed altered regulation of the contractile fiber and actin cytoskeleton, which affected cardiomyocyte differentiation. Furthermore, we have identified Stars as being transcriptionally controlled by HAND2. STARS facilitates actin polymerization that is essential for anchoring the adhesive molecules to create cell adhesion. Thus, we have uncovered a new function of HAND2 in mediating epithelial maintenance and integrity in OFT morphogenesis. Meanwhile, this study provides insights to understanding cardiac progenitor contribution to OFT development.
Collapse
Affiliation(s)
- Meng Xia
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Wen Luo
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Hengwei Jin
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| |
Collapse
|
22
|
Hubert F, Payan SM, Rochais F. FGF10 Signaling in Heart Development, Homeostasis, Disease and Repair. Front Genet 2018; 9:599. [PMID: 30546382 PMCID: PMC6279889 DOI: 10.3389/fgene.2018.00599] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/15/2018] [Indexed: 12/17/2022] Open
Abstract
Essential muscular organ that provides the whole body with oxygen and nutrients, the heart is the first organ to function during embryonic development. Cardiovascular diseases, including acquired and congenital heart defects, are the leading cause of mortality in industrialized countries. Fibroblast Growth Factors (FGFs) are involved in a variety of cellular responses including proliferation, differentiation, and migration. Among the 22 human/mouse FGFs, the secreted FGF10 ligand through the binding of its specific receptors (FGFR1b and FGFR2b) and subsequent activation of downstream signaling is known to play essential role in cardiac development, homeostasis and disease. FGF10 is one of the major marker of the early cardiac progenitor cells and a crucial regulator of differentiated cardiomyocyte proliferation in the developing embryo. Increasing evidence support the hypothesis that a detailed understanding of developmental processes is essential to identify targets for cardiac repair and regeneration. Indeed the activation of resident cardiomyocyte proliferation together with the injection of cardiac progenitors represent the most promising therapeutical strategies for cardiac regenerative medicine. The recent findings showing that FGF10 promotes adult cardiomyocyte cell cycle reentry and directs stem cell differentiation and cell reprogramming toward the cardiogenic lineage provide new insights into therapeutical strategies for cardiac regeneration and repair.
Collapse
Affiliation(s)
- Fabien Hubert
- Aix-Marseille Univ, INSERM, MMG, U1251, Marseille, France
| | - Sandy M Payan
- Aix-Marseille Univ, INSERM, MMG, U1251, Marseille, France
| | | |
Collapse
|
23
|
Giancotti V, Bergamin N, Cataldi P, Rizzi C. Epigenetic Contribution of High-Mobility Group A Proteins to Stem Cell Properties. Int J Cell Biol 2018; 2018:3698078. [PMID: 29853899 PMCID: PMC5941823 DOI: 10.1155/2018/3698078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 03/01/2018] [Accepted: 03/18/2018] [Indexed: 02/07/2023] Open
Abstract
High-mobility group A (HMGA) proteins have been examined to understand their participation as structural epigenetic chromatin factors that confer stem-like properties to embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and cancer stem cells (CSCs). The function of HMGA was evaluated in conjunction with that of other epigenetic factors such as histones and microRNAs (miRs), taking into consideration the posttranscriptional modifications (PTMs) of histones (acetylation and methylation) and DNA methylation. HMGA proteins were coordinated or associated with histone and DNA modification and the expression of the factors related to pluripotency. CSCs showed remarkable differences compared with ESCs and iPSCs.
Collapse
Affiliation(s)
- Vincenzo Giancotti
- Department of Life Science, University of Trieste, Trieste, Italy
- Trieste Proteine Ricerche, Palmanova, Udine, Italy
| | - Natascha Bergamin
- Division of Pathology, Azienda Ospedaliero-Universitaria, Udine, Italy
| | - Palmina Cataldi
- Division of Pathology, Azienda Ospedaliero-Universitaria, Udine, Italy
| | - Claudio Rizzi
- Division of Pathology, Azienda Ospedaliero-Universitaria, Udine, Italy
| |
Collapse
|
24
|
Hu J, Shi Y, Xia M, Liu Z, Zhang R, Luo H, Zhang T, Yang Z, Yuan B. WDR1-regulated actin dynamics is required for outflow tract and right ventricle development. Dev Biol 2018; 438:124-137. [PMID: 29654745 DOI: 10.1016/j.ydbio.2018.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 04/05/2018] [Accepted: 04/08/2018] [Indexed: 10/17/2022]
Abstract
Outflow tract (OFT) anomalies account for about 30% of human congenital heart defects detected at birth. The second heart field (SHF) progenitors contribute to OFT and right ventricle (RV) development, but the process largely remains unknown. WDR1 (WD-repeat domain 1) is a major co-factor of actin depolymerizing factor (ADF)/cofilin that actively disassembles ADF/cofilin-bound actin filaments. Its function in embryonic heart development has been unknown. Using Wdr1 floxed mice and Nkx2.5-Cre, we deleted Wdr1 in embryonic heart (Wdr1F/F;Nkx2.5-Cre) and found that these mice exhibited embryonic lethality, and hypoplasia of OFT and RV. To investigate the role of WDR1 in OFT and RV development, we generated SHF progenitors-specific Wdr1 deletion mice (shfKO). shfKO mice began to die at embryonic day 11.5 (E11.5), and displayed decreased size of the proximal OFT and RV at E10.5. In shfKO embryos, neither the number of SHF cells deployment to OFT nor cell proliferation and the cell number were changed, whereas the cellular organization and myofibrillar assembly of cardiomyocytes were severely disrupted. In the proximal OFT and RV of both shfKO and Wdr1F/F;Nkx2.5-Cre embryos, cardiomyocytes were dissociated from the outer compact myocardial layer and loosely and disorderly arranged into multilayered myocardium. Our results demonstrate that WDR1 is indispensable for normal OFT and RV development, and suggest that WDR1-mediated actin dynamics functions in controlling the size of OFT and RV, which might through regulating the spatial arrangement of cardiomyocytes.
Collapse
Affiliation(s)
- Jisheng Hu
- Biomedical Research Institute, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| | - Yingchao Shi
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Meng Xia
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Zhongying Liu
- Biomedical Research Institute, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| | - Ruirui Zhang
- Biomedical Research Institute, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| | - Hongmei Luo
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Tongcun Zhang
- Biomedical Research Institute, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| | - Zhongzhou Yang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China.
| | - Baiyin Yuan
- Biomedical Research Institute, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China.
| |
Collapse
|
25
|
Goumans MJ, Zwijsen A, Ten Dijke P, Bailly S. Bone Morphogenetic Proteins in Vascular Homeostasis and Disease. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a031989. [PMID: 28348038 DOI: 10.1101/cshperspect.a031989] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is well established that control of vascular morphogenesis and homeostasis is regulated by vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), Delta-like 4 (Dll4), angiopoietin, and ephrin signaling. It has become clear that signaling by bone morphogenetic proteins (BMPs), which have a long history of studies in bone and early heart development, are also essential for regulating vascular function. Indeed, mutations that cause deregulated BMP signaling are linked to two human vascular diseases, hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension. These observations are corroborated by data obtained with vascular cells in cell culture and in mouse models. BMPs are required for normal endothelial cell differentiation and for venous/arterial and lymphatic specification. In adult life, BMP signaling orchestrates neo-angiogenesis as well as vascular inflammation, remodeling, and calcification responses to shear and oxidative stress. This review emphasizes the pivotal role of BMPs in the vascular system, based on studies of mouse models and human vascular disorders.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - An Zwijsen
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium.,KU Leuven Department of Human Genetics, 3000 Leuven, Belgium
| | - Peter Ten Dijke
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Mécale (INSERM), U1036, 38000 Grenoble, France.,Laboratoire Biologie du Cancer et de l'Infection, Commissariat à l'Énergie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France.,University of Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
26
|
Higgins DF, Ewart LM, Masterson E, Tennant S, Grebnev G, Prunotto M, Pomposiello S, Conde-Knape K, Martin FM, Godson C. BMP7-induced-Pten inhibits Akt and prevents renal fibrosis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3095-3104. [PMID: 28923783 DOI: 10.1016/j.bbadis.2017.09.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 09/12/2017] [Accepted: 09/14/2017] [Indexed: 12/18/2022]
Abstract
Bone morphogenetic protein-7 (BMP-7) counteracts pro-fibrotic effects of TGFβ1 in cultured renal cells and protects from fibrosis in acute and chronic renal injury models. Using the unilateral ureteral obstruction (UUO) model of chronic renal fibrosis, we investigated the effect of exogenous-rhBMP-7 on pro-fibrotic signaling pathways mediated by TGFβ1 and hypoxia. Mice undergoing UUO were treated with vehicle or rhBMP-7 (300μg/kg i.p.) every other day for eight days and kidneys analysed for markers of fibrosis and SMAD, MAPK, and PI3K signaling. In the kidney, collecting duct and tubular epithelial cells respond to BMP-7 via activation of SMAD1/5/8. Phosphorylation of SMAD1/5/8 was reduced in UUO kidneys from vehicle-treated animals yet maintained in UUO kidneys from BMP-7-treated animals, confirming renal bioactivity of exogenous rhBMP-7. BMP-7 inhibited Collagen Iα1 and Collagen IIIα1 gene expression and Collagen I protein accumulation, while increasing expression of Collagen IVα1 in UUO kidneys. Activation of SMAD2, SMAD3, ERK, p38 and PI3K/Akt signaling occurred during fibrogenesis and BMP-7 significantly attenuated SMAD3 and Akt signaling in vivo. Analysis of renal collecting duct (mIMCD) and tubular epithelial (HK-2) cells stimulated with TGFβ1 or hypoxia (1% oxygen) to activate Akt provided further evidence that BMP-7 specifically inhibited PI3K/Akt signaling. PTEN is a negative regulator of PI3K and BMP-7 increased PTEN expression in vivo and in vitro. These data demonstrate an important mechanism by which BMP-7 orchestrates renal protection through Akt inhibition and highlights Akt inhibitors as anti-fibrotic therapeutics.
Collapse
Affiliation(s)
- Debra F Higgins
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Leah M Ewart
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Enda Masterson
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sadhbh Tennant
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Gleb Grebnev
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Marco Prunotto
- F. Hoffmann-La Roche, Dept of Metabolic and Vascular Disease, CH4070 Basel, Switzerland
| | - Sylvia Pomposiello
- F. Hoffmann-La Roche, Dept of Metabolic and Vascular Disease, CH4070 Basel, Switzerland
| | - Karin Conde-Knape
- F. Hoffmann-La Roche, Dept of Metabolic and Vascular Disease, CH4070 Basel, Switzerland
| | - Finian M Martin
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
27
|
Cao Y, Song J, Shen S, Fu H, Li X, Xu Y, Wang A, Li X, Zhang M. Trimedazidine alleviates pulmonary artery banding-induced acute right heart dysfunction and activates PRAS40 in rats. Oncotarget 2017; 8:92064-92078. [PMID: 29190898 PMCID: PMC5696164 DOI: 10.18632/oncotarget.20752] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/08/2017] [Indexed: 02/01/2023] Open
Abstract
The molecular mechanism underlying acute right heart failure (RHF) is poorly understood. We used pulmonary artery banding (PAB) to induce acute RHF characterized by a rapid rise of right ventricular pressure, and then a decrease in right ventricular pressure along with a decrease in blood pressure right after banding. We found higher brain natriuretic peptide (BNP) and beta-myosin heavy chain (βMHC) levels and lower alpha-myosin heavy chain (αMHC) levels in RHF rats than sham-operated rats. Hemodynamic indexes in rats with acute RHF were slightly improved by trimedazidine TMZ, a key inhibitor of fatty acid (FA) oxidation. TMZ also reversed downregulation of peroxisome proliferator-activated receptor gamma coactivator 1-beta (PGC-1β) and peroxisome proliferator-activated receptor alpha (PPARα) by PAB and up-regulates peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), peroxisome proliferator-activated receptor delta (PPARδ) and pyruvate dehydrogenase kinase isoform 4 (PDK4). In addition, TMZ reversed upregulation of phosphorylated Akt by PAB and increased phosphorylated proline-rich Akt-substrate 40 (PRAS40). Autophagy and apoptosis were not modified by PAB or TMZ. An acute RHF model was established in rats through 70% constriction of the pulmonary artery. TMZ treatment alleviated PAB-induced acute RHF by activating PRAS40 and upregulatingPGC-1α, PGC-1β, PPARα, PPARδ, and PDK4.
Collapse
Affiliation(s)
- Yunshan Cao
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou 730000, China.,Department of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Research Center for Translational Medicine, Shanghai 200120, China
| | - Jiyang Song
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Shutong Shen
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Heling Fu
- Animal Core Facility, Nanjing Medical University, Nanjing 210029, China
| | - Xiang Li
- Department of Intensive Care, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Ying Xu
- Intensive Care Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Aqian Wang
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Xinli Li
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Min Zhang
- Department of Pathology, Gansu Provincial Hospital, Lanzhou 730000, China
| |
Collapse
|
28
|
Phosphorylation-Dependent Feedback Inhibition of RIG-I by DAPK1 Identified by Kinome-wide siRNA Screening. Mol Cell 2017; 65:403-415.e8. [PMID: 28132841 DOI: 10.1016/j.molcel.2016.12.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 09/28/2016] [Accepted: 12/20/2016] [Indexed: 12/17/2022]
Abstract
Cell-autonomous induction of type I interferon must be stringently regulated. Rapid induction is key to control virus infection, whereas proper limitation of signaling is essential to prevent immunopathology and autoimmune disease. Using unbiased kinome-wide RNAi screening followed by thorough validation, we identified 22 factors that regulate RIG-I/IRF3 signaling activity. We describe a negative-feedback mechanism targeting RIG-I activity, which is mediated by death associated protein kinase 1 (DAPK1). RIG-I signaling triggers DAPK1 kinase activation, and active DAPK1 potently inhibits RIG-I stimulated IRF3 activity and interferon-beta production. DAPK1 phosphorylates RIG-I in vitro at previously reported as well as other sites that limit 5'ppp-dsRNA sensing and virtually abrogate RIG-I activation.
Collapse
|
29
|
Andreas E, Hoelker M, Neuhoff C, Tholen E, Schellander K, Tesfaye D, Salilew-Wondim D. MicroRNA 17–92 cluster regulates proliferation and differentiation of bovine granulosa cells by targeting PTEN and BMPR2 genes. Cell Tissue Res 2016; 366:219-30. [DOI: 10.1007/s00441-016-2425-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/27/2016] [Indexed: 01/02/2023]
|
30
|
Tao H, Yang JJ, Shi KH, Li J. Wnt signaling pathway in cardiac fibrosis: New insights and directions. Metabolism 2016; 65:30-40. [PMID: 26773927 DOI: 10.1016/j.metabol.2015.10.013] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/19/2015] [Accepted: 10/01/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Wnt signaling pathway significantly participates in cardiac fibrosis and CFs activation. Therefore, we reviewed current evidence on the new perspectives and biological association between Wnt signaling pathway and cardiac fibrosis. DESIGN AND METHODS A PubMed database search was performed for studies of Wnt signaling pathway in cardiac fibrosis and CFs activation. RESULTS Numerous studies have shown that the Wnt signaling pathway significantly participates in cardiac fibrosis pathogenesis. The aim of this review is to describe the present knowledge about the Wnt signaling pathway significantly participating in cardiac fibrosis and CFs activation, and look ahead on new perspectives of Wnt signaling pathway research. Moreover, we will discuss the different insights that interact with the Wnt signaling pathway-regulated cardiac fibrosis. The Wnt proteins are glycoproteins that bind to the Fz receptors on the cell surface, which lead to several important biological functions, such as cell differentiation and proliferation. There are several signals among the characterized pathways of cardiac fibrosis, including Wnt/β-catenin signaling. In this review, new insight into the Wnt signaling pathway in cardiac fibrosis pathogenesis is discussed, with special emphasis on Wnt/β-catenin. CONCLUSION It seems reasonable to suggest the potential targets of Wnt signaling pathway and it can be developed as a therapeutic target for cardiac fibrosis.
Collapse
Affiliation(s)
- Hui Tao
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, China 230601; Cardiovascular Research Center, Anhui Medical University, Hefei, China 230601
| | - Jing-Jing Yang
- Department of Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China 230601.
| | - Kai-Hu Shi
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, China 230601; Cardiovascular Research Center, Anhui Medical University, Hefei, China 230601.
| | - Jun Li
- School of pharmacy, Anhui Medical University, Hefei, China 230032
| |
Collapse
|
31
|
Thies RS, Murry CE. The advancement of human pluripotent stem cell-derived therapies into the clinic. Development 2016; 142:3077-84. [PMID: 26395136 DOI: 10.1242/dev.126482] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human pluripotent stem cells (hPSCs) offer many potential applications for drug screening and 'disease in a dish' assay capabilities. However, a more ambitious goal is to develop cell therapeutics using hPSCs to generate and replace somatic cells that are lost as a result of disease or injury. This Spotlight article will describe the state of progress of some of the hPSC-derived therapeutics that offer the most promise for clinical use. Lessons from developmental biology have been instrumental in identifying signaling molecules that can guide these differentiation processes in vitro, and will be described in the context of these cell therapy programs.
Collapse
Affiliation(s)
- R Scott Thies
- Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Charles E Murry
- Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA Department of Pathology, University of Washington, Seattle, WA 98195, USA Department of Bioengineering, University of Washington, Seattle, WA 98195, USA Department of Medicine/Cardiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|