1
|
Wang J, Nie H. Genome-wide identification and expression analysis of Sox gene family in the Manila clam (Ruditapes philippinarum). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 50:101244. [PMID: 38749209 DOI: 10.1016/j.cbd.2024.101244] [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: 04/03/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/27/2024]
Abstract
Sox transcription factors are vital in numerous fundamental biological processes. In this study, nine Sox gene family members were discovered in the Ruditapes philippinarum genome, classified into the SoxB1, SoxB2, SoxC, SoxD, SoxE, and SoxF groups, marking the first genome-wide identification of this gene family in R. philippinarum. Analyses of phylogeny, exon-intron structures, and domains bolster the support for their categorization and annotation. Furthermore, transcriptomic analyses across various developmental stages revealed that RpSox4, RpSox5, RpSox9, and RpSox11 were significantly expressed in the D-larval stage. Additionally, investigations into transcriptomes of clams with different shell colors indicated that most sox genes exhibited their highest expression levels in orange clams, followed by zebra, white zebra, and white clams, and the results of transcriptomes analysis in different tissues indicated that 8 Sox genes (except RpSox17) were highly expressed in the mantle tissue. Moreover, qPCR was used to detect the expression of Sox gene in R. philippinarum at different developmental periods, different shell colors and different tissues, and the results showed consistency with those of the transcriptomes. This study's findings lay the groundwork for additional exploration into the role of the Sox gene in melanin production in R. philippinarum shells.
Collapse
Affiliation(s)
- Jiadi Wang
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| | - Hongtao Nie
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, Dalian Ocean University, Dalian 116023, China.
| |
Collapse
|
2
|
Liang J, Ul Hassan I, Yee Cheung M, Feng L, Lin YJ, Long Q, Wang C, Ding Y, Wang Z, Zhang Y, Li Y, Guo D, Guo X, Chi Bun Wong T, Kaleem Samma M, Rong Z, Qi X, Cai D, Ngai SM, Zhao H. Mechanistic study of transcription factor Sox18 during heart development. Gen Comp Endocrinol 2024; 350:114472. [PMID: 38373462 DOI: 10.1016/j.ygcen.2024.114472] [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: 10/29/2023] [Revised: 01/18/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Heart development is a delicate and complex process regulated by coordination of various signaling pathways. In this study, we investigated the role of sox18 in heart development by modulating Wnt/β-Catenin signaling pathways. Our spatiotemporal expression analysis revealed that sox18 is mainly expressed in the heart, branchial arch, pharyngeal arch, spinal cord, and intersegmental vessels at the tailbud stage of Xenopus tropicalis embryo. Overexpression of sox18 in the X. tropicalis embryos causes heart edema, while loss-of-function of sox18 can change the signal of developmental heart marker gata4 at different stages, suggesting that sox18 plays an essential role in the development of the heart. Knockdown of SOX18 in human umbilical vein endothelial cells suggests a link between Sox18 and β-CATENIN, a key regulator of the Wnt signaling pathway. Sox18 negatively regulates islet1 and tbx3, the downstream factors of Wnt/β-Catenin signaling, during the linear heart tube formation and the heart looping stage. Taken together, our findings highlight the crucial role of Sox18 in the development of the heart via inhibiting Wnt/β-Catenin signaling.
Collapse
Affiliation(s)
- Jianxin Liang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Imtiaz Ul Hassan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Man Yee Cheung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Lei Feng
- School of Life Sciences and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China; Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yi-Jyun Lin
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Qi Long
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Chengdong Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuyue Ding
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ziqing Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuan Zhang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yulong Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Donghao Guo
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaofang Guo
- School of Life Sciences, Jinan University, Guangzhou, China
| | - Thomas Chi Bun Wong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Muhammad Kaleem Samma
- Department of Biology and Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Zixin Rong
- Department of Gene Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Stockholm 10691, Sweden
| | - Xufeng Qi
- School of Life Sciences, Jinan University, Guangzhou, China
| | - Dongqing Cai
- School of Life Sciences, Jinan University, Guangzhou, China
| | - Sai-Ming Ngai
- School of Life Sciences and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui Zhao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
3
|
Chiang IKN, Humphrey D, Mills RJ, Kaltzis P, Pachauri S, Graus M, Saha D, Wu Z, Young P, Sim CB, Davidson T, Hernandez‐Garcia A, Shaw CA, Renwick A, Scott DA, Porrello ER, Wong ES, Hudson JE, Red‐Horse K, del Monte‐Nieto G, Francois M. Sox7-positive endothelial progenitors establish coronary arteries and govern ventricular compaction. EMBO Rep 2023; 24:e55043. [PMID: 37551717 PMCID: PMC10561369 DOI: 10.15252/embr.202255043] [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: 03/14/2022] [Revised: 07/12/2023] [Accepted: 07/24/2023] [Indexed: 08/09/2023] Open
Abstract
The cardiac endothelium influences ventricular chamber development by coordinating trabeculation and compaction. However, the endothelial-specific molecular mechanisms mediating this coordination are not fully understood. Here, we identify the Sox7 transcription factor as a critical cue instructing cardiac endothelium identity during ventricular chamber development. Endothelial-specific loss of Sox7 function in mice results in cardiac ventricular defects similar to non-compaction cardiomyopathy, with a change in the proportions of trabecular and compact cardiomyocytes in the mutant hearts. This phenotype is paralleled by abnormal coronary artery formation. Loss of Sox7 function disrupts the transcriptional regulation of the Notch pathway and connexins 37 and 40, which govern coronary arterial specification. Upon Sox7 endothelial-specific deletion, single-nuclei transcriptomics analysis identifies the depletion of a subset of Sox9/Gpc3-positive endocardial progenitor cells and an increase in erythro-myeloid cell lineages. Fate mapping analysis reveals that a subset of Sox7-null endothelial cells transdifferentiate into hematopoietic but not cardiomyocyte lineages. Our findings determine that Sox7 maintains cardiac endothelial cell identity, which is crucial to the cellular cross-talk that drives ventricular compaction and coronary artery development.
Collapse
Affiliation(s)
- Ivy KN Chiang
- Centenary Institute, Royal Prince Alfred HospitalThe University of SydneySydneyNSWAustralia
| | - David Humphrey
- The Victor Chang Cardiac Research InstituteDarlinghurstNSWAustralia
| | - Richard J Mills
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Peter Kaltzis
- The Australian Regenerative Medicine InstituteMonash UniversityClaytonVICAustralia
| | - Shikha Pachauri
- Centenary Institute, Royal Prince Alfred HospitalThe University of SydneySydneyNSWAustralia
| | - Matthew Graus
- Centenary Institute, Royal Prince Alfred HospitalThe University of SydneySydneyNSWAustralia
| | - Diptarka Saha
- The Australian Regenerative Medicine InstituteMonash UniversityClaytonVICAustralia
| | - Zhijian Wu
- The Australian Regenerative Medicine InstituteMonash UniversityClaytonVICAustralia
| | - Paul Young
- The Victor Chang Cardiac Research InstituteDarlinghurstNSWAustralia
| | - Choon Boon Sim
- The Murdoch Children's Research InstituteRoyal Children's HospitalMelbourneVICAustralia
| | - Tara Davidson
- Centenary Institute, Royal Prince Alfred HospitalThe University of SydneySydneyNSWAustralia
| | | | - Chad A Shaw
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| | - Alexander Renwick
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| | - Daryl A Scott
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| | - Enzo R Porrello
- The Murdoch Children's Research InstituteRoyal Children's HospitalMelbourneVICAustralia
- Melbourne Centre for Cardiovascular Genomics and Regenerative MedicineThe Royal Children's HospitalMelbourneVICAustralia
- Department of Anatomy and Physiology, School of Biomedical SciencesThe University of MelbourneMelbourneVICAustralia
| | - Emily S Wong
- The Victor Chang Cardiac Research InstituteDarlinghurstNSWAustralia
| | - James E Hudson
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | | | | | - Mathias Francois
- Centenary Institute, Royal Prince Alfred HospitalThe University of SydneySydneyNSWAustralia
| |
Collapse
|
4
|
Hernández-García A, Pendleton KE, Kim S, Li Y, Kim BJ, Zaveri HP, Jordan VK, Berry AM, Ljungberg MC, Chen R, Lanz RB, Scott DA. SOX7 deficiency causes ventricular septal defects through its effects on endocardial-to-mesenchymal transition and the expression of Wnt4 and Bmp2. Hum Mol Genet 2023; 32:2152-2161. [PMID: 37000005 PMCID: PMC10281751 DOI: 10.1093/hmg/ddad050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/09/2023] [Accepted: 03/27/2023] [Indexed: 04/01/2023] Open
Abstract
SOX7 is a transcription factor-encoding gene located in a region on chromosome 8p23.1 that is recurrently deleted in individuals with ventricular septal defects (VSDs). We have previously shown that Sox7-/- embryos die of heart failure around E11.5. Here, we demonstrate that these embryos have hypocellular endocardial cushions with severely reduced numbers of mesenchymal cells. Ablation of Sox7 in the endocardium also resulted in hypocellular endocardial cushions, and we observed VSDs in rare E15.5 Sox7flox/-;Tie2-Cre and Sox7flox/flox;Tie2-Cre embryos that survived to E15.5. In atrioventricular explant studies, we showed that SOX7 deficiency leads to a severe reduction in endocardial-to-mesenchymal transition (EndMT). RNA-seq studies performed on E9.5 Sox7-/- heart tubes revealed severely reduced Wnt4 transcript levels. Wnt4 is expressed in the endocardium and promotes EndMT by acting in a paracrine manner to increase the expression of Bmp2 in the myocardium. Both WNT4 and BMP2 have been previously implicated in the development of VSDs in individuals with 46,XX sex reversal with dysgenesis of kidney, adrenals and lungs (SERKAL) syndrome and in individuals with short stature, facial dysmorphism and skeletal anomalies with or without cardiac anomalies 1 (SSFSC1) syndrome, respectively. We now show that Sox7 and Wnt4 interact genetically in the development of VSDs through their additive effects on endocardial cushion development with Sox7+/-;Wnt4+/- double heterozygous embryos having hypocellular endocardial cushions and perimembranous and muscular VSDs not seen in their Sox7+/- and Wnt4+/- littermates. These results provide additional evidence that SOX7, WNT4 and BMP2 function in the same pathway during mammalian septal development and that their deficiency can contribute to the development of VSDs in humans.
Collapse
Affiliation(s)
- Andrés Hernández-García
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Katherine E Pendleton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sangbae Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yumei Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bum J Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hitisha P Zaveri
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Valerie K Jordan
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Aliska M Berry
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - M Cecilia Ljungberg
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX 77030, USA
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rainer B Lanz
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daryl A Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Dai L, Du L. Genes in pediatric pulmonary arterial hypertension and the most promising BMPR2 gene therapy. Front Genet 2022; 13:961848. [PMID: 36506323 PMCID: PMC9730536 DOI: 10.3389/fgene.2022.961848] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare but progressive and lethal vascular disease of diverse etiologies, mainly caused by proliferation of endothelial cells, smooth muscle cells in the pulmonary artery, and fibroblasts, which ultimately leads to right-heart hypertrophy and cardiac failure. Recent genetic studies of childhood-onset PAH report that there is a greater genetic burden in children than in adults. Since the first-identified pathogenic gene of PAH, BMPR2, which encodes bone morphogenetic protein receptor 2, a receptor in the transforming growth factor-β superfamily, was discovered, novel causal genes have been identified and substantially sharpened our insights into the molecular genetics of childhood-onset PAH. Currently, some newly identified deleterious genetic variants in additional genes implicated in childhood-onset PAH, such as potassium channels (KCNK3) and transcription factors (TBX4 and SOX17), have been reported and have greatly updated our understanding of the disease mechanism. In this review, we summarized and discussed the advances of genetic variants underlying childhood-onset PAH susceptibility and potential mechanism, and the most promising BMPR2 gene therapy and gene delivery approaches to treat childhood-onset PAH in the future.
Collapse
|
6
|
Shi HY, Xie MS, Yang CX, Huang RT, Xue S, Liu XY, Xu YJ, Yang YQ. Identification of SOX18 as a New Gene Predisposing to Congenital Heart Disease. Diagnostics (Basel) 2022; 12:diagnostics12081917. [PMID: 36010266 PMCID: PMC9406965 DOI: 10.3390/diagnostics12081917] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
Congenital heart disease (CHD) is the most frequent kind of birth deformity in human beings and the leading cause of neonatal mortality worldwide. Although genetic etiologies encompassing aneuploidy, copy number variations, and mutations in over 100 genes have been uncovered to be involved in the pathogenesis of CHD, the genetic components predisposing to CHD in most cases remain unclear. We recruited a family with CHD from the Chinese Han population in the present investigation. Through whole-exome sequencing analysis of selected family members, a new SOX18 variation, namely NM_018419.3:c.349A>T; p.(Lys117*), was identified and confirmed to co-segregate with the CHD phenotype in the entire family by Sanger sequencing analysis. The heterozygous variant was absent from the 384 healthy volunteers enlisted as control individuals. Functional exploration via luciferase reporter analysis in cultivated HeLa cells revealed that Lys117*-mutant SOX18 lost transactivation on its target genes NR2F2 and GATA4, two genes responsible for CHD. Moreover, the genetic variation terminated the synergistic activation between SOX18 and NKX2.5, another gene accountable for CHD. The findings strongly indicate SOX18 as a novel gene contributing to CHD, which helps address challenges in the clinical genetic diagnosis and prenatal prophylaxis of CHD.
Collapse
Affiliation(s)
- Hong-Yu Shi
- Department of Cardiology, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai 200940, China
| | - Meng-Shi Xie
- Department of Cardiology, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai 200940, China
| | - Chen-Xi Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
| | - Ri-Tai Huang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xing-Yuan Liu
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Correspondence: (Y.-J.X.); (Y.-Q.Y.)
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Department of Cardiovascular Research Laboratory, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Department of Central Laboratory, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Correspondence: (Y.-J.X.); (Y.-Q.Y.)
| |
Collapse
|
7
|
Afouda BA. Towards Understanding the Gene-Specific Roles of GATA Factors in Heart Development: Does GATA4 Lead the Way? Int J Mol Sci 2022; 23:5255. [PMID: 35563646 PMCID: PMC9099915 DOI: 10.3390/ijms23095255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 02/04/2023] Open
Abstract
Transcription factors play crucial roles in the regulation of heart induction, formation, growth and morphogenesis. Zinc finger GATA transcription factors are among the critical regulators of these processes. GATA4, 5 and 6 genes are expressed in a partially overlapping manner in developing hearts, and GATA4 and 6 continue their expression in adult cardiac myocytes. Using different experimental models, GATA4, 5 and 6 were shown to work together not only to ensure specification of cardiac cells but also during subsequent heart development. The complex involvement of these related gene family members in those processes is demonstrated through the redundancy among them and crossregulation of each other. Our recent identification at the genome-wide level of genes specifically regulated by each of the three family members and our earlier discovery that gata4 and gata6 function upstream, while gata5 functions downstream of noncanonical Wnt signalling during cardiac differentiation, clearly demonstrate the functional differences among the cardiogenic GATA factors. Such suspected functional differences are worth exploring more widely. It appears that in the past few years, significant advances have indeed been made in providing a deeper understanding of the mechanisms by which each of these molecules function during heart development. In this review, I will therefore discuss current evidence of the role of individual cardiogenic GATA factors in the process of heart development and emphasize the emerging central role of GATA4.
Collapse
Affiliation(s)
- Boni A Afouda
- Institute of Medical Sciences, Foresterhill Health Campus, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, UK
| |
Collapse
|
8
|
Huang RT, Guo YH, Yang CX, Gu JN, Qiu XB, Shi HY, Xu YJ, Xue S, Yang YQ. SOX7 loss-of-function variation as a cause of familial congenital heart disease. Am J Transl Res 2022; 14:1672-1684. [PMID: 35422912 PMCID: PMC8991148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 02/08/2022] [Indexed: 06/14/2023]
Abstract
INTRODUCTION As the most frequent type of birth defect in humans, congenital heart disease (CHD) leads to a large amount of morbidity and mortality as well as a tremendous socioeconomic burden. Accumulating studies have convincingly substantiated the pivotal roles of genetic defects in the occurrence of familial CHD, and deleterious variations in a great number of genes have been reported to cause various types of CHD. However, owing to pronounced genetic heterogeneity, the hereditary components underpinning CHD remain obscure in most cases. This investigation aimed to identify novel genetic determinants underlying CHD. METHODS AND RESULTS A four-generation pedigree with high incidence of autosomal-dominant CHD was enrolled from the Chinese Han race population. Using whole-exome sequencing and Sanger sequencing assays of the family members available, a novel SOX7 variation in heterozygous status, NM_031439.4: c.310C>T; p.(Gln104*), was discovered to be in co-segregation with the CHD phenotype in the whole family. The truncating variant was absent in 500 unrelated healthy subjects utilized as control individuals. Functional measurements by dual-luciferase reporter analysis revealed that Gln104*-mutant SOX7 failed to transactivate its two important target genes, GATA4 and BMP2, which are both responsible for CHD. In addition, the nonsense variation invalidated the cooperative transactivation between SOX7 and NKX2.5, which is another recognized CHD-causative gene. CONCLUSION The present study demonstrates for the first time that genetically defective SOX7 predisposes to CHD, which sheds light on the novel molecular mechanism underpinning CHD, and implies significance for precise prevention and personalized treatment in a subset of CHD patients.
Collapse
Affiliation(s)
- Ri-Tai Huang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai 200127, China
| | - Yu-Han Guo
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan UniversityShanghai 200240, China
| | - Chen-Xi Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan UniversityShanghai 200240, China
| | - Jia-Ning Gu
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan UniversityShanghai 200240, China
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai 200030, China
| | - Hong-Yu Shi
- Department of Cardiology, Zhongshan Hospital Wusong Branch, Fudan UniversityShanghai 200940, China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan UniversityShanghai 200240, China
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai 200127, China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan UniversityShanghai 200240, China
- Department of Cardiovascular Research Laboratory, Shanghai Fifth People’s Hospital, Fudan UniversityShanghai 200240, China
- Department of Central Laboratory, Shanghai Fifth People’s Hospital, Fudan UniversityShanghai 200240, China
| |
Collapse
|
9
|
Jiang X, Li T, Li B, Wei W, Li F, Chen S, Xu R, Sun K. SOX7 suppresses endothelial-to-mesenchymal transitions by enhancing VE-cadherin expression during outflow tract development. Clin Sci (Lond) 2021; 135:829-846. [PMID: 33720353 DOI: 10.1042/cs20201496] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/06/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023]
Abstract
The endothelial-to-mesenchymal transition (EndMT) is a critical process that occurs during the development of the outflow tract (OFT). Malformations of the OFT can lead to the occurrence of conotruncal defect (CTD). SOX7 duplication has been reported in patients with congenital CTD, but its specific role in OFT development remains poorly understood. To decipher this, histological analysis showed that SRY-related HMG-box 7 (SOX7) was regionally expressed in the endocardial endothelial cells and in the mesenchymal cells of the OFT, where EndMT occurs. Experiments, using in vitro collagen gel culture system, revealed that SOX7 was a negative regulator of EndMT that inhibited endocardial cell (EC) migration and resulted in decreased number of mesenchymal cells. Forced expression of SOX7 in endothelial cells blocked further migration and improved the expression of the adhesion protein vascular endothelial (VE)-cadherin (VE-cadherin). Moreover, a VE-cadherin knockdown could partly reverse the SOX7-mediated repression of cell migration. Luciferase and electrophoretic mobility shift assay (EMSA) demonstrated that SOX7 up-regulated VE-cadherin by directly binding to the gene's promoter in endothelial cells. The coding exons and splicing regions of the SOX7 gene were also scanned in the 536 sporadic CTD patients and in 300 unaffected controls, which revealed four heterozygous SOX7 mutations. Luciferase assays revealed that two SOX7 variants weakened the transactivation of the VE-cadherin promoter. In conclusion, SOX7 inhibited EndMT during OFT development by directly up-regulating the endothelial-specific adhesion molecule VE-cadherin. SOX7 mutations can lead to impaired EndMT by regulating VE-cadherin, which may give rise to the molecular mechanisms associated with SOX7 in CTD pathogenesis.
Collapse
Affiliation(s)
- Xuechao Jiang
- Scientific Research Center, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Tingting Li
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Bojian Li
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Wei Wei
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Fen Li
- Department of Pediatric Cardiology, Shanghai Children's Medical Center, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Sun Chen
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Rang Xu
- Scientific Research Center, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
10
|
Zhang J, Yu P, Liu T, Qiao D, Hu Q, Su S. Identification and functional analysis of SOX transcription factors in the genome of the Chinese soft-shell turtle (Pelodiscus sinensis). Comp Biochem Physiol B Biochem Mol Biol 2020; 242:110407. [PMID: 31923463 DOI: 10.1016/j.cbpb.2020.110407] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 11/27/2019] [Accepted: 12/10/2019] [Indexed: 11/28/2022]
Abstract
SOX transcription factors play an irreplaceable role in biological developmental processes. Sox genes have been identified in a wide variety of species; however, their identification and functional analysis in the genome of the Chinese soft-shell turtle (Pelodiscus sinensis) have not been performed. In the present study, the Chinese soft-shell turtle genome was found to contain 17 Sox genes, which were categorized into seven groups according to their phylogenetic relationships. Gene structure and protein motif analysis of the Sox genes showed that within the same phylogenetic group, their exon-intron number and motif structure of the Sox family were relatively conserved, but diverged in the comparison between different groups. Sexual dimorphism expression analysis for the Sox genes displayed that Sox8 and Sox9 were upregulated in the testis, while Sox3, Sox7, Sox11, and Sox13 were upregulated in the ovary. A correlation network analysis of SOX transcription factors with their target genes analysis showed that Sox3 correlated negatively with Sox9 and gata4. Sox11 and Sox7 correlated negatively with gata4. Sox8 and Sox9 correlated positively with gata4. Therefore, the genome-wide identification and functional analysis of the Sox gene family will be useful to further reveal the functions of Sox genes in the Chinese soft-shell turtle.
Collapse
Affiliation(s)
- Jun Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.
| | - Peng Yu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Tiantian Liu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Dan Qiao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Qingtao Hu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Shiping Su
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
11
|
Liu F, Guo L, Xin G, Wang Z. miR-452 promotes cell metastasis and the epithelial to mesenchymal by targeting SOX7 in clear-cell renal-cell carcinoma. J Cell Biochem 2019; 120:8401-8408. [PMID: 30506716 DOI: 10.1002/jcb.28125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 10/31/2018] [Indexed: 01/24/2023]
Abstract
Clear-cell renal-cell carcinoma (ccRCC) is the most common renal cell carcinoma (RCC), representing 75%-80% of the cases of RCC, and characterized by a high recurrence rate and poor prognosis. miR-452 acts as a tumor promoter in several tumors, including ccRCC. The purpose of this study was to determine the role of miR-452 in ccRCC. miR-452 and SOX7 messenger RNA and protein levels were calculated by quantitative reverse transcription polymerase chain reaction and Western blot analysis. MTT and Transwell assays were utilized to measure proliferative and invasive abilities. The Kaplan-Meier method was used to evaluate the association between the expression of miR-452 or SOX7 and the overall survival of ccRCC patients. Our results showed that miR-452 was overexpressed in ccRCC tissues and cells, and upregulation of miR-452 predicted a poor 5-year survival in ccRCC patients. In contrast, expression of SOX7 was low and downregulation of SOX7 predicted poor prognosis in ccRCC. In addition, miR-452 promoted cell proliferation, invasion, and the EMT, while SOX7 reversed the function of miR-452 on cell proliferation and invasion in 786-O cells. In conclusion, miR-452 was shown to inhibit cell proliferation, invasion, and the EMT through SOX7 in ccRCC, and the newly identified miR-452/SOX7 axis provided novel insight into the pathogenesis of ccRCC.
Collapse
Affiliation(s)
- Feng Liu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Lihua Guo
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Guangda Xin
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Zhixin Wang
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
12
|
Genetic determinants of risk in pulmonary arterial hypertension: international genome-wide association studies and meta-analysis. THE LANCET. RESPIRATORY MEDICINE 2019; 7:227-238. [PMID: 30527956 PMCID: PMC6391516 DOI: 10.1016/s2213-2600(18)30409-0] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/20/2018] [Accepted: 09/26/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Rare genetic variants cause pulmonary arterial hypertension, but the contribution of common genetic variation to disease risk and natural history is poorly characterised. We tested for genome-wide association for pulmonary arterial hypertension in large international cohorts and assessed the contribution of associated regions to outcomes. METHODS We did two separate genome-wide association studies (GWAS) and a meta-analysis of pulmonary arterial hypertension. These GWAS used data from four international case-control studies across 11 744 individuals with European ancestry (including 2085 patients). One GWAS used genotypes from 5895 whole-genome sequences and the other GWAS used genotyping array data from an additional 5849 individuals. Cross-validation of loci reaching genome-wide significance was sought by meta-analysis. Conditional analysis corrected for the most significant variants at each locus was used to resolve signals for multiple associations. We functionally annotated associated variants and tested associations with duration of survival. All-cause mortality was the primary endpoint in survival analyses. FINDINGS A locus near SOX17 (rs10103692, odds ratio 1·80 [95% CI 1·55-2·08], p=5·13 × 10-15) and a second locus in HLA-DPA1 and HLA-DPB1 (collectively referred to as HLA-DPA1/DPB1 here; rs2856830, 1·56 [1·42-1·71], p=7·65 × 10-20) within the class II MHC region were associated with pulmonary arterial hypertension. The SOX17 locus had two independent signals associated with pulmonary arterial hypertension (rs13266183, 1·36 [1·25-1·48], p=1·69 × 10-12; and rs10103692). Functional and epigenomic data indicate that the risk variants near SOX17 alter gene regulation via an enhancer active in endothelial cells. Pulmonary arterial hypertension risk variants determined haplotype-specific enhancer activity, and CRISPR-mediated inhibition of the enhancer reduced SOX17 expression. The HLA-DPA1/DPB1 rs2856830 genotype was strongly associated with survival. Median survival from diagnosis in patients with pulmonary arterial hypertension with the C/C homozygous genotype was double (13·50 years [95% CI 12·07 to >13·50]) that of those with the T/T genotype (6·97 years [6·02-8·05]), despite similar baseline disease severity. INTERPRETATION This is the first study to report that common genetic variation at loci in an enhancer near SOX17 and in HLA-DPA1/DPB1 is associated with pulmonary arterial hypertension. Impairment of SOX17 function might be more common in pulmonary arterial hypertension than suggested by rare mutations in SOX17. Further studies are needed to confirm the association between HLA typing or rs2856830 genotyping and survival, and to determine whether HLA typing or rs2856830 genotyping improves risk stratification in clinical practice or trials. FUNDING UK NIHR, BHF, UK MRC, Dinosaur Trust, NIH/NHLBI, ERS, EMBO, Wellcome Trust, EU, AHA, ACClinPharm, Netherlands CVRI, Dutch Heart Foundation, Dutch Federation of UMC, Netherlands OHRD and RNAS, German DFG, German BMBF, APH Paris, INSERM, Université Paris-Sud, and French ANR.
Collapse
|
13
|
Zou ZY, Liu J, Chang C, Li JJ, Luo J, Jin Y, Ma Z, Wang TH, Shao JL. Biliverdin administration regulates the microRNA-mRNA expressional network associated with neuroprotection in cerebral ischemia reperfusion injury in rats. Int J Mol Med 2019; 43:1356-1372. [PMID: 30664169 PMCID: PMC6365090 DOI: 10.3892/ijmm.2019.4064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022] Open
Abstract
Inflammatory response has an important role in the outcome of cerebral ischemia reperfusion injury (CIR). Biliverdin (BV) administration can relieve CIR in rats, but the mechanism remains unknown. The aim of the present study was to explore the expressional network of microRNA (miRNA)-mRNA in CIR rats following BV administration. A rat middle cerebral artery occlusion model with BV treatment was established. After neurobehavior was evaluated by neurological severity scores (NSS), miRNA and mRNA expressional profiles were analyzed by microarray technology from the cerebral cortex subjected to ischemia and BV administration. Then, bioinformatics prediction was used to screen the correlation between miRNA and mRNA, and 20 candidate miRNAs and 33 candidate mRNAs were verified by reverse transcription-quantitative polymerase chain reaction. Furthermore, the regulation relationship between ETS proto-oncogene 1 (Ets1) and miRNA204-5p was examined by luciferase assay. A total of 86 miRNAs were differentially expressed in the BV group compared with the other groups. A total of 10 miRNAs and 26 candidate genes were identified as a core 'microRNA-mRNA' regulatory network that was linked with the functional improvement of BV administration in CIR rats. Lastly, the luciferase assay results confirmed that miRNA204-5p directly targeted Ets1. The present findings suggest that BV administration may regulate multiple miRNAs and mRNAs to improve neurobehavior in CIR rats, by influencing cell proliferation, apoptosis, maintaining ATP homeostasis, and angiogenesis.
Collapse
Affiliation(s)
- Zhi-Yao Zou
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Jia Liu
- Experimental Animal Center, Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Cheng Chang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Jun-Jie Li
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Jing Luo
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Yuan Jin
- Experimental Animal Center, Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Zheng Ma
- Experimental Animal Center, Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Ting-Hua Wang
- Experimental Animal Center, Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Jian-Lin Shao
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| |
Collapse
|
14
|
Xu ZJ, Tang CY, Zhou JD, Ma JC, Wen XM, Deng ZQ, Leng JY, Qiu ZY, Qian J, Lin J. SOX7 methylation is an independent prognostic factor in myelodysplastic syndromes. Pathol Res Pract 2018; 215:322-328. [PMID: 30554866 DOI: 10.1016/j.prp.2018.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/14/2018] [Accepted: 12/05/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE SOX7 downregulation caused by its promoter methylation was associated with poor survival in several types of human solid tumors. However, the pattern of SOX7 methylation and its clinical significance are less studied in hematological malignancies. Herein, we evaluated the methylation pattern of SOX7 in myelodysplastic syndrome (MDS) and determined its clinical implication in patients with MDS. METHODS SOX7 methylation was determined by real-time quantitative methylation-specific PCR (RQ-MSP) in 99 MDS patients. Bisulfite sequencing PCR was applied to confirm the results of RQ-MSP. RESULTS SOX7 methylation was detected in 55.6% of 99 patients but not in healthy donors. No correlation was found between SOX7 methylation and clinical parameters including patient age, gender, white blood cell count, hemoglobin, and platelet count. However, patients with SOX7 methylation harbored more U2AF1 mutation than patients without SOX7 methylation (P = 0.015). Kaplan-Meier curves indicated that the patients with SOX7 methylation presented reduced overall survival (OS) (P = 0.034). Furthermore, subgroup analysis indicated that SOX7 methylation was associated with poor OS in male patients (P = 0.034) and in patients older than 60 years (P = 0.019). According to the multivariate analysis, SOX7 methylation remained as an independent prognosis factor in MDS patients both as dichotomous (HR = 2.14, P = 0.041) and as continuous (HR = 1.55, P = 0.042) variable. Importantly, SOX7 methylation was significantly increased during progression from MDS to secondary acute myeloid leukemia (sAML). CONCLUSIONS Our findings demonstrated that SOX7 methylation conferred adverse prognosis in MDS patients and was associated with leukemia progression.
Collapse
Affiliation(s)
- Zi-Jun Xu
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, PR China; Zhenjiang Clinical Research Center of Hematology, Zhenjiang 212002, Jiangsu, P.R. China; The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang 212002, Jiangsu, P.R. China
| | - Chun-Yan Tang
- Department of Nephropathy and Hematology, The First People's Hospital of Aksu Prefecture of Xinjiang, Aksu 843000, Xinjiang, P.R. China
| | - Jing-Dong Zhou
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang 212002, Jiangsu, P.R. China; Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
| | - Ji-Chun Ma
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, PR China; Zhenjiang Clinical Research Center of Hematology, Zhenjiang 212002, Jiangsu, P.R. China; The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang 212002, Jiangsu, P.R. China
| | - Xiang-Mei Wen
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, PR China; Zhenjiang Clinical Research Center of Hematology, Zhenjiang 212002, Jiangsu, P.R. China; The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang 212002, Jiangsu, P.R. China
| | - Zhao-Qun Deng
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, PR China; Zhenjiang Clinical Research Center of Hematology, Zhenjiang 212002, Jiangsu, P.R. China; The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang 212002, Jiangsu, P.R. China.
| | - Jia-Yan Leng
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang 212002, Jiangsu, P.R. China; Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
| | - Zhi-Yuan Qiu
- Department of Oncology, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
| | - Jun Qian
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang 212002, Jiangsu, P.R. China; Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China.
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, PR China; Zhenjiang Clinical Research Center of Hematology, Zhenjiang 212002, Jiangsu, P.R. China; The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang 212002, Jiangsu, P.R. China.
| |
Collapse
|
15
|
Zhang S, Chen X, Wang M, Zhang W, Pan J, Qin Q, Zhong L, Shao J, Sun M, Jiang H, Bian W. Genome-wide identification, phylogeny and expressional profile of the Sox gene family in channel catfish (Ictalurus punctatus). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2018; 28:17-26. [DOI: 10.1016/j.cbd.2018.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 12/11/2017] [Accepted: 03/05/2018] [Indexed: 01/10/2023]
|
16
|
Yang J, Hu F, Fu X, Jiang Z, Zhang W, Chen K. MiR-128/SOX7 alleviates myocardial ischemia injury by regulating IL-33/sST2 in acute myocardial infarction. Biol Chem 2018; 400:533-544. [PMID: 30265647 DOI: 10.1515/hsz-2018-0207] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/18/2018] [Indexed: 12/12/2022]
Abstract
Abstract
Acute myocardial infarction (AMI) induced by ischemia hypoxia severely threatens human life. Cell apoptosis of neurocytes was identified to mediate the pathogenesis, while the potential mechanism was still unclear. Sprague Dawley (SD) rats were used to establish the AMI rat model. Real-time polymerase chain reaction (PCR) and Western blot were performed to detect gene expression in mRNA and protein levels, respectively. A TUNEL assay was carried out to determine cell apoptosis. The relationship between SRY-related HMG-box (SOX7) and miR-128 was verified using luciferase reporter assay. The expression of SOX7 was decreased, while miR-128 was increased in AMI rats and ischemia hypoxia (IH) induced H9c2 cells. Hypoxia induction significantly promoted the expression of interleukin (IL)-33 and soluble ST2 (sST2), and also promoted cell apoptosis. MiR-128 targets SOX7 to regulate its expression. Down-regulated miR-128 reversed the effects of IH on expression of SOX7, sST2 and cell apoptosis, while down-regulated sST2 abolished the effects of miR-128 inhibitor. In addition, overexpressed IL-33 abolished the effects of miR-128 inhibitor that induced by IH on the expression of SOX7 and cell apoptosis. In vivo experiments validated the expression of miR-128 on cell apoptosis. The present study indicated that miR-128 modulated cell apoptosis by targeting SOX7, which was mediated by IL-33/sST2 signaling pathway.
Collapse
Affiliation(s)
- Jinhua Yang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Rd., Zhengzhou 450052, Henan, China
| | - Fudong Hu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Rd., Zhengzhou 450052, Henan, China
| | - Xin Fu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Rd., Zhengzhou 450052, Henan, China
| | - Zhengming Jiang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Rd., Zhengzhou 450052, Henan, China
| | - Wencai Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Rd., Zhengzhou 450052, Henan, China
| | - Kui Chen
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Rd., Zhengzhou 450052, Henan, China
| |
Collapse
|
17
|
Alonso-Martin S, Auradé F, Mademtzoglou D, Rochat A, Zammit PS, Relaix F. SOXF factors regulate murine satellite cell self-renewal and function through inhibition of β-catenin activity. eLife 2018; 7:26039. [PMID: 29882512 PMCID: PMC6021169 DOI: 10.7554/elife.26039] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/07/2018] [Indexed: 12/17/2022] Open
Abstract
Muscle satellite cells are the primary source of stem cells for postnatal skeletal muscle growth and regeneration. Understanding genetic control of satellite cell formation, maintenance, and acquisition of their stem cell properties is on-going, and we have identified SOXF (SOX7, SOX17, SOX18) transcriptional factors as being induced during satellite cell specification. We demonstrate that SOXF factors regulate satellite cell quiescence, self-renewal and differentiation. Moreover, ablation of Sox17 in the muscle lineage impairs postnatal muscle growth and regeneration. We further determine that activities of SOX7, SOX17 and SOX18 overlap during muscle regeneration, with SOXF transcriptional activity requisite. Finally, we show that SOXF factors also control satellite cell expansion and renewal by directly inhibiting the output of β-catenin activity, including inhibition of Ccnd1 and Axin2. Together, our findings identify a key regulatory function of SoxF genes in muscle stem cells via direct transcriptional control and interaction with canonical Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Sonia Alonso-Martin
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Créteil, France.,Université Paris Est, Faculté de Medecine, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Frédéric Auradé
- Sorbonne Université, INSERM U974, Center for Research in Myology, Paris, France
| | - Despoina Mademtzoglou
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Créteil, France.,Université Paris Est, Faculté de Medecine, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Anne Rochat
- Sorbonne Université, INSERM U974, Center for Research in Myology, Paris, France
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Frédéric Relaix
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Créteil, France.,Université Paris Est, Faculté de Medecine, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France.,Etablissement Français du Sang, Creteil, France.,APHP, Hopitaux UniversitairesHenri Mondor, Centre de Référence des Maladies Neuromusculaires GNMH, Créteil, France
| |
Collapse
|
18
|
Afouda BA, Lynch AT, de Paiva Alves E, Hoppler S. Genome-wide transcriptomics analysis of genes regulated by GATA4, 5 and 6 during cardiomyogenesis in Xenopus laevis. Data Brief 2018; 17:559-563. [PMID: 29876429 PMCID: PMC5988223 DOI: 10.1016/j.dib.2018.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/18/2017] [Accepted: 01/04/2018] [Indexed: 11/26/2022] Open
Abstract
The transcription factors GATA4, GATA5 and GATA6 play important roles in heart muscle differentiation. The data presented in this article are related to the research article entitled “Genome-wide transcriptomics analysis identifies sox7 and sox18 as specifically regulated by gata4 in cardiomyogenesis” (Afouda et al., 2017) [1]. The present study identifies genes regulated by these individual cardiogenic GATA factors using genome-wide transcriptomics analysis. We have presented genes that are specifically regulated by each of them, as well those regulated by either of them. The gene ontology terms (GO) associated with the genes differentially affected are also presented. The data set will allow further investigations on the gene regulatory network downstream of individual cardiogenic GATA factors during cardiac muscle formation.
Collapse
Affiliation(s)
- Boni A Afouda
- Institute of Medical Sciences, Foresterhill Health Campus, University of Aberdeen, Scotland, UK
| | - Adam T Lynch
- Institute of Medical Sciences, Foresterhill Health Campus, University of Aberdeen, Scotland, UK
| | - Eduardo de Paiva Alves
- Centre for Genome-Enabled Biology and Medicine, King's College Campus, University of Aberdeen, Scotland, UK
| | - Stefan Hoppler
- Institute of Medical Sciences, Foresterhill Health Campus, University of Aberdeen, Scotland, UK
| |
Collapse
|
19
|
Afouda BA, Lynch AT, de Paiva Alves E, Hoppler S. Genome-wide transcriptomics analysis identifies sox7 and sox18 as specifically regulated by gata4 in cardiomyogenesis. Dev Biol 2017; 434:108-120. [PMID: 29229250 PMCID: PMC5814753 DOI: 10.1016/j.ydbio.2017.11.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/29/2017] [Accepted: 11/29/2017] [Indexed: 01/12/2023]
Abstract
The transcription factors GATA4, GATA5 and GATA6 are important regulators of heart muscle differentiation (cardiomyogenesis), which function in a partially redundant manner. We identified genes specifically regulated by individual cardiogenic GATA factors in a genome-wide transcriptomics analysis. The genes regulated by gata4 are particularly interesting because GATA4 is able to induce differentiation of beating cardiomyocytes in Xenopus and in mammalian systems. Among the specifically gata4-regulated transcripts we identified two SoxF family members, sox7 and sox18. Experimental reinstatement of gata4 restores sox7 and sox18 expression, and loss of cardiomyocyte differentiation due to gata4 knockdown is partially restored by reinstating sox7 or sox18 expression, while (as previously reported) knockdown of sox7 or sox18 interferes with heart muscle formation. In order to test for conservation in mammalian cardiomyogenesis, we confirmed in mouse embryonic stem cells (ESCs) undergoing cardiomyogenesis that knockdown of Gata4 leads to reduced Sox7 (and Sox18) expression and that Gata4 is also uniquely capable of promptly inducing Sox7 expression. Taken together, we identify an important and conserved gene regulatory axis from gata4 to the SoxF paralogs sox7 and sox18 and further to heart muscle cell differentiation. Gata 4, 5 and 6 have redundant and non-redundant functions in heart development. RNA-seq analysis of Gata4, 5 and 6 knockdown experiments was carried out. Genes specifically regulated by Gata4, 5 and 6 were identified. The SoxF genes sox7 and sox18 were identified as specifically regulated by Gata4. Epistasis demonstrates a regulatory axis from Gata4 to Sox7/18 to cardiomyogenesis.
Collapse
Affiliation(s)
- Boni A Afouda
- Institute of Medical Sciences, Foresterhill Health Campus, University of Aberdeen, Scotland, UK
| | - Adam T Lynch
- Institute of Medical Sciences, Foresterhill Health Campus, University of Aberdeen, Scotland, UK
| | - Eduardo de Paiva Alves
- Centre for Genome-Enabled Biology and Medicine, King's College Campus, University of Aberdeen, Scotland, UK
| | - Stefan Hoppler
- Institute of Medical Sciences, Foresterhill Health Campus, University of Aberdeen, Scotland, UK.
| |
Collapse
|
20
|
Owens DA, Butler AM, Aguero TH, Newman KM, Van Booven D, King ML. High-throughput analysis reveals novel maternal germline RNAs crucial for primordial germ cell preservation and proper migration. Development 2017; 144:292-304. [PMID: 28096217 DOI: 10.1242/dev.139220] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 11/25/2016] [Indexed: 01/10/2023]
Abstract
During oogenesis, hundreds of maternal RNAs are selectively localized to the animal or vegetal pole, including determinants of somatic and germline fates. Although microarray analysis has identified localized determinants, it is not comprehensive and is limited to known transcripts. Here, we utilized high-throughput RNA-sequencing analysis to comprehensively interrogate animal and vegetal pole RNAs in the fully grown Xenopus laevis oocyte. We identified 411 (198 annotated) and 27 (15 annotated) enriched mRNAs at the vegetal and animal pole, respectively. Ninety were novel mRNAs over 4-fold enriched at the vegetal pole and six were over 10-fold enriched at the animal pole. Unlike mRNAs, microRNAs were not asymmetrically distributed. Whole-mount in situ hybridization confirmed that all 17 selected mRNAs were localized. Biological function and network analysis of vegetally enriched transcripts identified protein-modifying enzymes, receptors, ligands, RNA-binding proteins, transcription factors and co-factors with five defining hubs linking 47 genes in a network. Initial functional studies of maternal vegetally localized mRNAs show that sox7 plays a novel and important role in primordial germ cell (PGC) development and that ephrinB1 (efnb1) is required for proper PGC migration. We propose potential pathways operating at the vegetal pole that highlight where future investigations might be most fruitful.
Collapse
Affiliation(s)
- Dawn A Owens
- Department of Cell Biology, University of Miami Miller School of Medicine, 1011 NW 15th St, Miami, FL 33136, USA
| | - Amanda M Butler
- Department of Cell Biology, University of Miami Miller School of Medicine, 1011 NW 15th St, Miami, FL 33136, USA
| | - Tristan H Aguero
- Department of Cell Biology, University of Miami Miller School of Medicine, 1011 NW 15th St, Miami, FL 33136, USA
| | - Karen M Newman
- Department of Cell Biology, University of Miami Miller School of Medicine, 1011 NW 15th St, Miami, FL 33136, USA
| | - Derek Van Booven
- The Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, 1011 NW 15th St, Miami, FL 33136, USA
| | - Mary Lou King
- Department of Cell Biology, University of Miami Miller School of Medicine, 1011 NW 15th St, Miami, FL 33136, USA
| |
Collapse
|
21
|
Rajgara RF, Lala-Tabbert N, Marchildon F, Lamarche É, MacDonald JK, Scott DA, Blais A, Skerjanc IS, Wiper-Bergeron N. SOX7 Is Required for Muscle Satellite Cell Development and Maintenance. Stem Cell Reports 2017; 9:1139-1151. [PMID: 28943254 PMCID: PMC5639291 DOI: 10.1016/j.stemcr.2017.08.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 01/26/2023] Open
Abstract
Satellite cells are skeletal-muscle-specific stem cells that are activated by injury to proliferate, differentiate, and fuse to enable repair. SOX7, a member of the SRY-related HMG-box family of transcription factors is expressed in quiescent satellite cells. To elucidate SOX7 function in skeletal muscle, we knocked down Sox7 expression in embryonic stem cells and primary myoblasts and generated a conditional knockout mouse in which Sox7 is excised in PAX3+ cells. Loss of Sox7 in embryonic stem cells reduced Pax3 and Pax7 expression. In vivo, conditional knockdown of Sox7 reduced the satellite cell population from birth, reduced myofiber caliber, and impaired regeneration after acute injury. Although Sox7-deficient primary myoblasts differentiated normally, impaired myoblast fusion and increased sensitivity to apoptosis in culture and in vivo were observed. Taken together, these results indicate that SOX7 is dispensable for myogenesis but is necessary to promote satellite cell development and survival.
Collapse
Affiliation(s)
- Rashida F Rajgara
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Graduate Program in Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Neena Lala-Tabbert
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - François Marchildon
- Graduate Program in Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Émilie Lamarche
- Graduate Program in Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jennifer K MacDonald
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Daryl A Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexandre Blais
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Ilona S Skerjanc
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Nadine Wiper-Bergeron
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
22
|
Charney RM, Paraiso KD, Blitz IL, Cho KWY. A gene regulatory program controlling early Xenopus mesendoderm formation: Network conservation and motifs. Semin Cell Dev Biol 2017; 66:12-24. [PMID: 28341363 DOI: 10.1016/j.semcdb.2017.03.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/12/2017] [Accepted: 03/20/2017] [Indexed: 02/08/2023]
Abstract
Germ layer formation is among the earliest differentiation events in metazoan embryos. In triploblasts, three germ layers are formed, among which the endoderm gives rise to the epithelial lining of the gut tube and associated organs including the liver, pancreas and lungs. In frogs (Xenopus), where early germ layer formation has been studied extensively, the process of endoderm specification involves the interplay of dozens of transcription factors. Here, we review the interactions between these factors, summarized in a transcriptional gene regulatory network (GRN). We highlight regulatory connections conserved between frog, fish, mouse, and human endodermal lineages. Especially prominent is the conserved role and regulatory targets of the Nodal signaling pathway and the T-box transcription factors, Vegt and Eomes. Additionally, we highlight network topologies and motifs, and speculate on their possible roles in development.
Collapse
Affiliation(s)
- Rebekah M Charney
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Kitt D Paraiso
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Ira L Blitz
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Ken W Y Cho
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
23
|
Lilly AJ, Lacaud G, Kouskoff V. SOXF transcription factors in cardiovascular development. Semin Cell Dev Biol 2017; 63:50-57. [PMID: 27470491 DOI: 10.1016/j.semcdb.2016.07.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/19/2016] [Accepted: 07/23/2016] [Indexed: 12/24/2022]
Abstract
Cardiovascular development during embryogenesis involves complex changes in gene regulatory networks regulated by a variety of transcription factors. In this review we discuss the various reported roles of the SOXF factors: SOX7, SOX17 and SOX18 in cardiac, vascular and lymphatic development. SOXF factors have pleiotropic roles during these processes, and there is significant redundancy and functional compensation between SOXF family members. Despite this, evidence suggests that there is some specificity in the transcriptional programs they regulate which is necessary to control the differentiation and behaviour of endothelial subpopulations. Furthermore, SOXF factors appear to have an indirect role in regulating cardiac mesoderm specification and differentiation. Understanding how SOXF factors are regulated, as well as their downstream transcriptional target genes, will be important for unravelling their roles in cardiovascular development and related diseases.
Collapse
Affiliation(s)
- Andrew J Lilly
- Cancer Research UK, Stem Cell Hematopoiesis, The University of Manchester, Wilmslow road, M20 4BX, UK
| | - Georges Lacaud
- Cancer Research UK, Stem Cell Biology group Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow road, M20 4BX, UK.
| | - Valerie Kouskoff
- Cancer Research UK, Stem Cell Hematopoiesis, The University of Manchester, Wilmslow road, M20 4BX, UK.
| |
Collapse
|
24
|
Xiuju C, Zhen W, Yanchao S. SOX7 inhibits tumor progression of glioblastoma and is regulated by miRNA-24. Open Med (Wars) 2016; 11:133-137. [PMID: 28352781 PMCID: PMC5329813 DOI: 10.1515/med-2016-0026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/04/2016] [Indexed: 01/01/2023] Open
Abstract
Objective Sex-determining region Y-box 7 (SOX7) is a putative tumor suppressor in various types of human cancers. In the present study, the expression and function of SOX7 was investigated in human glioblastoma (GBM) cells. Methods Real-time PCR and western blot were carried out to reveal the expression of SOX7 in GBM specimens and cultured cell lines. A short interfering RNA (siRNA) targeting SOX7 was synthesized and transfected into U87 cells. 3-(4,5-Dimethylthiazol-2-yl)- 2,5-diphenyltetrazolium bromide (MTT) assay was performed to valuate the cell proliferation ability in U87 cells. Bioinformatics analysis further predicted its regulation by microRNA-24 (miR-24). Luciferase reporter assay was performed to prove this regulation. Results SOX7 was downregulated in GBM specimens and cell lines. Inhibition of SOX7 in cultured U87 cells resulted in a slower growth rate. Mechanically, SOX7 was a target of miR-24, demonstrated by reporter assay. Conclusion SOX7 was a strong tumor suppressor regulated by miR-24 in human GBM cells.
Collapse
Affiliation(s)
- Chen Xiuju
- Department of Neurology, Tianjin Nankai Hospital, Tianjin PR China 300100
| | - Wang Zhen
- Cardiology, Tianjin Nankai Hospital, Tianjin PR China 300100
| | - Shi Yanchao
- Department of Neurology, Tianjin Port Hospital, Tianjin PR China 300456
| |
Collapse
|
25
|
Costain G, Lionel AC, Ogura L, Marshall CR, Scherer SW, Silversides CK, Bassett AS. Genome-wide rare copy number variations contribute to genetic risk for transposition of the great arteries. Int J Cardiol 2015; 204:115-21. [PMID: 26655555 DOI: 10.1016/j.ijcard.2015.11.127] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/10/2015] [Accepted: 11/20/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Transposition of the great arteries (TGA) is an uncommon but severe congenital heart malformation of unknown etiology. Rare copy number variations (CNVs) have been implicated in other, more common conotruncal heart defects like tetralogy of Fallot (TOF), but there are as yet no CNV studies dedicated to TGA. METHODS Using high-resolution genome-wide microarrays and rigorous methods, we investigated CNVs in a group of prospectively recruited adults with TGA (n=101) from a single center. We compared rare CNV burden to well-matched cohorts of controls and TOF cases, adjudicating rarity using 10,113 independent population-based controls and excluding all subjects with 22q11.2 deletions. We identified candidate genes for TGA based on rare CNVs that overlapped the same gene in unrelated individuals, and pre-existing evidence suggesting a role in cardiac development. RESULTS The TGA group was significantly enriched for large rare CNVs (2.3-fold increase, p=0.04) relative to controls, to a degree comparable with the TOF group. Extra-cardiac features were not reliable predictors of rare CNV burden. Smaller rare CNVs helped to narrow critical regions for conotruncal defects at chromosomes 10q26 and 13q13. Established and novel candidate susceptibility genes identified included ACKR3, IFT57, ITGB8, KL, NF1, NKX1-2, RERE, SLC8A1, SOX18, and ULK1. CONCLUSIONS These data demonstrate a genome-wide role for rare CNVs in genetic risk for TGA. The findings provide further support for a genetically-related spectrum of congenital heart disease that includes TGA and TOF.
Collapse
Affiliation(s)
- Gregory Costain
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Medical Genetics Residency Training Program, University of Toronto, and Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Anath C Lionel
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada; McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Lucas Ogura
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Christian R Marshall
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada; McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Candice K Silversides
- The Toronto Congenital Cardiac Centre for Adults & Division of Cardiology in the Department of Medicine, University Health Network, Toronto, Ontario, Canada.
| | - Anne S Bassett
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; The Toronto Congenital Cardiac Centre for Adults & Division of Cardiology in the Department of Medicine, University Health Network, Toronto, Ontario, Canada; Department of Psychiatry, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; The Dalglish Family Hearts and Minds Clinic for 22q11.2 Deletion Syndrome, University Health Network, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| |
Collapse
|
26
|
8p23.1 Interstitial Deletion in a Patient with Congenital Cardiopathy, Neurobehavioral Disorders, and Minor Signs Suggesting 22q11.2 Deletion Syndrome. J Dev Behav Pediatr 2015; 36:544-8. [PMID: 26263419 DOI: 10.1097/dbp.0000000000000197] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Copy number variation studies of known disorders have the potential to improve the characterization of clinical phenotypes and may help identifying candidate genes and their pathways. The authors described a child with congenital heart disease, microcephaly, facial dysmorphisms, developmental delay, learning difficulties, and behavioral problems. There was initially a clinical suspicion of 22q11.2 deletion syndrome (22q11.2 DS), but molecular cytogenetic analysis (array genomic hybridization [aGH]) showed the presence of a de novo 3.6-Mb interstitial microdeletion in 8p23.1. The main features of 8p23.1 DS include congenital heart disease and behavioral problems, in addition to minor dysmorphisms and mental delay. Therefore, this article highlights the application of aGH to investigate 8p23.1 deletion in nonconfirmed 22q11.2 DS patients presenting neurobehavioral disorders, congenital cardiopathy, and minor dysmorphisms.
Collapse
|
27
|
Yang J, Aguero T, King ML. The Xenopus Maternal-to-Zygotic Transition from the Perspective of the Germline. Curr Top Dev Biol 2015; 113:271-303. [PMID: 26358876 DOI: 10.1016/bs.ctdb.2015.07.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In Xenopus, the germline is specified by the inheritance of germ-plasm components synthesized at the beginning of oogenesis. Only the cells in the early embryo that receive germ plasm, the primordial germ cells (PGCs), are competent to give rise to the gametes. Thus, germ-plasm components continue the totipotent potential exhibited by the oocyte into the developing embryo at a time when most cells are preprogrammed for somatic differentiation as dictated by localized maternal determinants. When zygotic transcription begins at the mid-blastula transition, the maternally set program for somatic differentiation is realized. At this time, genetic control is ceded to the zygotic genome, and developmental potential gradually becomes more restricted within the primary germ layers. PGCs are a notable exception to this paradigm and remain transcriptionally silent until the late gastrula. How the germ-cell lineage retains full potential while somatic cells become fate restricted is a tale of translational repression, selective degradation of somatic maternal determinants, and delayed activation of zygotic transcription.
Collapse
Affiliation(s)
- Jing Yang
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Tristan Aguero
- Department of Cell Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Mary Lou King
- Department of Cell Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
28
|
She ZY, Yang WX. SOX family transcription factors involved in diverse cellular events during development. Eur J Cell Biol 2015; 94:547-63. [PMID: 26340821 DOI: 10.1016/j.ejcb.2015.08.002] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 08/11/2015] [Accepted: 08/11/2015] [Indexed: 12/22/2022] Open
Abstract
In metazoa, SOX family transcription factors play many diverse roles. In vertebrate, they are well-known regulators of numerous developmental processes. Wide-ranging studies have demonstrated the co-expression of SOX proteins in various developing tissues and that they occur in an overlapping manner and show functional redundancy. In particular, studies focusing on the HMG box of SOX proteins have revealed that the HMG box regulates DNA-binding properties, and mediates both the nucleocytoplasmic shuttling of SOX proteins and their physical interactions with partner proteins. Posttranslational modifications are further implicated in the regulation of the transcriptional activities of SOX proteins. In this review, we discuss the underlying molecular mechanisms involved in the SOX-partner factor interactions and the functional modes of SOX-partner complexes during development. We particularly emphasize the representative roles of the SOX group proteins in major tissues during developmental and physiological processes.
Collapse
Affiliation(s)
- Zhen-Yu She
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China.
| |
Collapse
|
29
|
Man CH, Fung TK, Wan H, Cher CY, Fan A, Ng N, Ho C, Wan TSK, Tanaka T, So CWE, Kwong YL, Leung AYH. Suppression of SOX7 by DNA methylation and its tumor suppressor function in acute myeloid leukemia. Blood 2015; 125:3928-36. [PMID: 25940713 DOI: 10.1182/blood-2014-06-580993] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 03/29/2015] [Indexed: 12/15/2022] Open
Abstract
SOX7 belongs to the SOX (Sry-related high-mobility group [HMG] box) gene family, a group of transcription factors containing in common a HMG box domain. Its role in hematologic malignancies and, in particular, acute myeloid leukemia (AML) is completely unknown. Here, we showed that SOX7 expression was regulated by DNA hypermethylation in AML but not in acute lymphoblastic leukemia or normal bone marrow cells. In cell lines (KG1, ML2, and K562) and in primary CD34(+) AML samples, SOX7 expression could be induced by the DNA demethylating agent 5-aza-2'-deoxycytidine. Overexpression of SOX7 in K562 cells inhibited cell proliferation, with cell cycle delay in S/G2/M phases and reduced clonogenic activity. Apoptosis was unaffected. Ectopic expression of SOX7 in K562 and THP-1 cells, as well as primary CD33(+)CD34(+) AML cells, abrogated leukemia engraftment in xenogeneic transplantation. SOX7 expression inhibited the Wnt/β-catenin pathway through direct protein binding to β-catenin, and the antileukemia effects of SOX7 in THP-1 cells were significantly reduced by deletion of its β-catenin binding site. The results provided unequivocal evidence for a novel tumor suppressor role of SOX7 in AML via a negative modulatory effect on the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Cheuk Him Man
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Tsz Kan Fung
- Leukaemia and Stem Cell Biology Laboratory, Department of Haematological Medicine, King's College London, London, United Kingdom
| | - Haixia Wan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chae Yin Cher
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - August Fan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Nelson Ng
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Christa Ho
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Thomas S K Wan
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; and
| | - Toshiyuki Tanaka
- Laboratory of Immunobiology, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Japan
| | - Chi Wai Eric So
- Leukaemia and Stem Cell Biology Laboratory, Department of Haematological Medicine, King's College London, London, United Kingdom
| | - Yok Lam Kwong
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Anskar Y H Leung
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
30
|
Chibby functions in Xenopus ciliary assembly, embryonic development, and the regulation of gene expression. Dev Biol 2014; 395:287-98. [PMID: 25220153 DOI: 10.1016/j.ydbio.2014.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 09/03/2014] [Accepted: 09/05/2014] [Indexed: 12/21/2022]
Abstract
Wnt signaling and ciliogenesis are core features of embryonic development in a range of metazoans. Chibby (Cby), a basal-body associated protein, regulates β-catenin-mediated Wnt signaling in the mouse but not Drosophila. Here we present an analysis of Cby's embryonic expression and morphant phenotypes in Xenopus laevis. Cby RNA is supplied maternally, negatively regulated by Snail2 but not Twist1, preferentially expressed in the neuroectoderm, and regulates β-catenin-mediated gene expression. Reducing Cby levels reduced the density of multiciliated cells, the number of basal bodies per multiciliated cell, and the numbers of neural tube primary cilia; it also led to abnormal development of the neural crest, central nervous system, and pronephros, all defects that were rescued by a Cby-GFP chimera. Reduction of Cby led to an increase in Wnt8a and decreases in Gli2, Gli3, and Shh RNA levels. Many, but not all, morphant phenotypes were significantly reversed by the Wnt inhibitor SFRP2. These observations extend our understanding of Cby's role in mediating the network of interactions between ciliogenesis, signaling systems and tissue patterning.
Collapse
|
31
|
Bazil JN, Stamm KD, Li X, Thiagarajan R, Nelson TJ, Tomita-Mitchell A, Beard DA. The inferred cardiogenic gene regulatory network in the mammalian heart. PLoS One 2014; 9:e100842. [PMID: 24971943 PMCID: PMC4074065 DOI: 10.1371/journal.pone.0100842] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 05/31/2014] [Indexed: 12/22/2022] Open
Abstract
Cardiac development is a complex, multiscale process encompassing cell fate adoption, differentiation and morphogenesis. To elucidate pathways underlying this process, a recently developed algorithm to reverse engineer gene regulatory networks was applied to time-course microarray data obtained from the developing mouse heart. Approximately 200 genes of interest were input into the algorithm to generate putative network topologies that are capable of explaining the experimental data via model simulation. To cull specious network interactions, thousands of putative networks are merged and filtered to generate scale-free, hierarchical networks that are statistically significant and biologically relevant. The networks are validated with known gene interactions and used to predict regulatory pathways important for the developing mammalian heart. Area under the precision-recall curve and receiver operator characteristic curve are 9% and 58%, respectively. Of the top 10 ranked predicted interactions, 4 have already been validated. The algorithm is further tested using a network enriched with known interactions and another depleted of them. The inferred networks contained more interactions for the enriched network versus the depleted network. In all test cases, maximum performance of the algorithm was achieved when the purely data-driven method of network inference was combined with a data-independent, functional-based association method. Lastly, the network generated from the list of approximately 200 genes of interest was expanded using gene-profile uniqueness metrics to include approximately 900 additional known mouse genes and to form the most likely cardiogenic gene regulatory network. The resultant network supports known regulatory interactions and contains several novel cardiogenic regulatory interactions. The method outlined herein provides an informative approach to network inference and leads to clear testable hypotheses related to gene regulation.
Collapse
Affiliation(s)
- Jason N. Bazil
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Karl D. Stamm
- Biotechnology and Bioengineering Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Xing Li
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Raghuram Thiagarajan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Timothy J. Nelson
- Departments of Medicine, Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic Center for Regenerative Medicine, Rochester, Minnesota, United States of America
| | - Aoy Tomita-Mitchell
- Biotechnology and Bioengineering Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Daniel A. Beard
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
32
|
Behrens AN, Zierold C, Shi X, Ren Y, Koyano-Nakagawa N, Garry DJ, Martin CM. Sox7 is regulated by ETV2 during cardiovascular development. Stem Cells Dev 2014; 23:2004-13. [PMID: 24762086 DOI: 10.1089/scd.2013.0525] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Vasculogenesis/angiogenesis is one of the earliest processes that occurs during embryogenesis. ETV2 and SOX7 were previously shown to play a role in endothelial development; however, their mechanistic interaction has not been defined. In the present study, concomitant expression of Etv2 and Sox7 in endothelial progenitor cells was verified. ETV2 was shown to be a direct upstream regulator of Sox7 that binds to ETV2 binding elements in the Sox7 upstream regulatory region and activates transcription. We observed that SOX7 over-expression can mimic ETV2 and increase endothelial progenitor cells in embryonic bodies (EBs), while knockdown of Sox7 is able to block ETV2-induced increase in endothelial progenitor cell formation. Angiogenic sprouting was increased by ETV2 over-expression in EBs, and it was significantly decreased in the presence of Sox7 shRNA. Collectively, these studies support the conclusion that ETV2 directly regulates Sox7, and that ETV2 governs endothelial development by regulating transcriptional networks which include Sox7.
Collapse
Affiliation(s)
- Ann N Behrens
- Lillehei Heart Institute, University of Minnesota , Minneapolis, Minnesota
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Cerebral cavernous malformation is a clinically well-defined microvascular disorder predisposing to stroke; however, the major phenotype observed in zebrafish is the cardiac defect, specifically an enlarged heart. Less effort has been made to explore this phenotypic discrepancy between human and zebrafish. Given the fact that the gene products from Ccm1/Ccm2 are nearly identical between the two species, the common sense has dictated that the zebrafish animal model would provide a great opportunity to dissect the detailed molecular function of Ccm1/Ccm2 during angiogenesis. We recently reported on the cellular role of the Ccm1 gene in biochemical processes that permit proper angiogenic microvascular development in the zebrafish model. In the course of this experimentation, we encountered a vast amount of recent research on the relationship between dysfunctional angiogenesis and cardiovascular defects in zebrafish. Here we compile the findings of our research with the most recent contributions in this field and glean conclusions about the effect of defective angiogenesis on the developing cardiovascular system. Our conclusion also serves as a bridge for the phenotypic discrepancy between humans and animal models, which might provide some insights into future translational research on human stroke.
Collapse
|
34
|
Stovall DB, Cao P, Sui G. SOX7: from a developmental regulator to an emerging tumor suppressor. Histol Histopathol 2013; 29:439-45. [PMID: 24288056 DOI: 10.14670/hh-29.10.439] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
SOX7 belongs to the SOX (SRY-related HMG-box) family of transcription factors that have been shown to regulate multiple biological processes, such as hematopoiesis, vasculogenesis and cardiogenesis during embryonic development. Recent studies indicate that several SOX family members play important roles in tumorigenesis. In this review, we introduce SOX7 gene and protein structures, and discuss its expression and functional role in cancer development and progression. SOX7 is frequently downregulated in many human cancers and its reduced expression correlates with poor prognoses of several cancers. Functional studies reveal many tumor suppressive properties of SOX7 in prostate, colon, lung, and breast cancers. To date, although a few target genes of SOX7 have been identified, SOX7-mediated gene expression has not been investigated in a cancer-relevant context. Our recent studies not only for the first time demonstrate a tumor suppressive role of SOX7 in a xenograft mouse model, but also unravel that many genes regulating cell death, growth and apoptosis are affected by SOX7, strongly supporting a pivotal role of SOX7 in tumorigenesis. Thus, currently available data clearly indicate a tumor suppressive role of SOX7, but the mechanisms underlying its gene expression and tumor suppressive activity remain undetermined. The research of SOX7 in cancers remains a fertile area to be explored.
Collapse
Affiliation(s)
- Daniel B Stovall
- Department of Cancer Biology and Comprehensive Cancer Center, and Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Paul Cao
- Department of Cancer Biology and Comprehensive Cancer Center, and Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Guangchao Sui
- Department of Cancer Biology and Comprehensive Cancer Center, and Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
35
|
Osoegawa K, Iovannisci DM, Lin B, Parodi C, Schultz K, Shaw GM, Lammer EJ. Identification of novel candidate gene loci and increased sex chromosome aneuploidy among infants with conotruncal heart defects. Am J Med Genet A 2013; 164A:397-406. [PMID: 24127225 DOI: 10.1002/ajmg.a.36291] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 09/10/2013] [Indexed: 12/17/2022]
Abstract
Congenital heart defects (CHDs) are common malformations, affecting four to eight per 1,000 total births. Conotruncal defects are an important pathogenetic subset of CHDs, comprising nearly 20% of the total. Although both environmental and genetic factors are known to contribute to the occurrence of conotruncal defects, the causes remain unknown for most. To identify novel candidate genes/loci, we used array comparative genomic hybridization to detect chromosomal microdeletions/duplications. From a population base of 974,579 total births born during 1999-2004, we screened 389 California infants born with tetralogy of Fallot or d-transposition of the great arteries. We found that 1.7% (5/288) of males with a conotruncal defect had sex chromosome aneuploidy, a sevenfold increased frequency (relative risk = 7.0; 95% confidence interval 2.9-16.9). We identified eight chromosomal microdeletions/duplications for conotruncal defects. From these duplications and deletions, we found five high priority candidate genes (GATA4, CRKL, BMPR1A, SNAI2, and ZFHX4). This is the initial report that sex chromosome aneuploidy is associated with conotruncal defects among boys. These chromosomal microduplications/deletions provide evidence that GATA4, SNAI2, and CRKL are highly dosage sensitive genes involved in outflow tract development. Genome wide screening for copy number variation can be productive for identifying novel genes/loci contributing to non-syndromic common malformations.
Collapse
Affiliation(s)
- Kazutoyo Osoegawa
- Center for Genetics, Children's Hospital Oakland Research Institute, Children's Hospital Research Center Oakland, Oakland, California
| | | | | | | | | | | | | |
Collapse
|
36
|
The regulation of SOX7 and its tumor suppressive role in breast cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1645-1653. [PMID: 24012678 DOI: 10.1016/j.ajpath.2013.07.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/16/2013] [Accepted: 07/10/2013] [Indexed: 12/23/2022]
Abstract
Both epigenetic silencing and genetic deletion of tumor suppressors contribute to the development and progression of breast cancer. SOX7 is a transcription factor important to development, and its down-regulation has been reported in tumor tissues and cell lines of prostate, colon, and lung cancers. However, the regulation of SOX7 expression and its functional role in breast cancer have not been reported. The current study demonstrates that SOX7 mRNA and protein expression are down-regulated in breast cancer tissues and cell lines compared with adjacent normal tissues and nontumorigenic cells, respectively. The SOX7 promoter is hypermethylated in breast cancer cell lines compared with nontumorigenic cells, and the inhibition of DNA methylation increases SOX7 mRNA levels. With shRNA-mediated SOX7 silencing, nontumorigenic immortal breast cells display increased proliferation, migration, and invasion and form structures that resemble that of breast cancer cells in a three-dimensional culture system. Conversely, ectopic SOX7 expression inhibits proliferation, migration, and invasion of breast cancer cells in vitro and tumor growth in vivo. Importantly, we discovered that SOX7 transcript levels positively correlated with clinical outcome of 674 breast cancer patients. Overall, our data suggest that SOX7 acts as a tumor suppressor in breast cancer. SOX7 expression is likely regulated by multiple mechanisms and potentially serves as a prognostic marker for breast cancer patients.
Collapse
|
37
|
Long F, Wang X, Fang S, Xu Y, Sun K, Chen S, Xu R. A potential relationship among beta-defensins haplotype, SOX7 duplication and cardiac defects. PLoS One 2013; 8:e72515. [PMID: 24009689 PMCID: PMC3757027 DOI: 10.1371/journal.pone.0072515] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 07/11/2013] [Indexed: 01/30/2023] Open
Abstract
OBJECTIVE To determine the pathogenesis of a patient born with congenital heart defects, who had appeared normal in prenatal screening. METHODS In routine prenatal screening, G-banding was performed to analyse the karyotypes of the family and fluorescence in situ hybridization was used to investigate the 22q11.2 deletion in the fetus. After birth, the child was found to be suffering from heart defects by transthoracic echocardiography. In the following study, sequencing was used to search for potential mutations in pivotal genes. SNP-array was employed for fine mapping of the aberrant region and quantitative real-time PCR was used to confirm the results. Furthermore, other patients with a similar phenotype were screened for the same genetic variations. To compare with a control, these variations were also assessed in the general population. RESULTS The child and his mother each had a region that was deleted in the beta-defensin repeats, which are usually duplicated in the general population. Besides, the child carried a SOX7-gene duplication. While this duplication was not detected in his mother, it was found in two other patients with cardiac defects who also had the similar deletion in the beta-defensin repeats. CONCLUSION The congenital heart defects of the child were probably caused by a SOX7-gene duplication, which may be a consequence of the partial haplotype of beta-defensin regions at 8p23.1. To our knowledge, this is the first congenital heart defect case found to have the haplotype of beta-defensin and the duplication of SOX7.
Collapse
Affiliation(s)
- Fei Long
- Scientific Research Center, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Xike Wang
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Shaohai Fang
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Yuejuan Xu
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Sun Chen
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
- * E-mail: (SC); (RX)
| | - Rang Xu
- Scientific Research Center, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
- * E-mail: (SC); (RX)
| |
Collapse
|
38
|
Barber JCK, Rosenfeld JA, Foulds N, Laird S, Bateman MS, Thomas NS, Baker S, Maloney VK, Anilkumar A, Smith WE, Banks V, Ellingwood S, Kharbutli Y, Mehta L, Eddleman KA, Marble M, Zambrano R, Crolla JA, Lamb AN. 8p23.1 duplication syndrome; common, confirmed, and novel features in six further patients. Am J Med Genet A 2013; 161A:487-500. [PMID: 23345203 DOI: 10.1002/ajmg.a.35767] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 10/14/2012] [Indexed: 01/07/2023]
Abstract
The 8p23.1 duplication syndrome is a relatively rare genomic condition that has been confirmed with molecular cytogenetic methods in only 11 probands and five family members. Here, we describe another prenatal and five postnatal patients with de novo 8p23.1 duplications analyzed with oligonucleotide array comparative genomic hybridization (oaCGH). Of the common features, mild or moderate developmental delays and/or learning difficulties have been found in 11/12 postnatal probands, a variable degree of mild dysmorphism in 8/12 and congenital heart disease (CHD) in 4/5 prenatal and 3/12 postnatal probands. Behavioral problems, cleft lip and/or palate, macrocephaly, and seizures were confirmed as additional features among the new patients, and novel features included neonatal respiratory distress, attention deficit hyperactivity disorder (ADHD), ocular anomalies, balance problems, hypotonia, and hydrocele. The core duplication of 3.68 Mb contains 31 genes and microRNAs of which only GATA4, TNKS, SOX7, and XKR6 are likely to be dosage sensitive genes and MIR124-1 and MIR598 have been implicated in neurocognitive phenotypes. A combination of the duplication of GATA4, SOX7, and related genes may account for the variable penetrance of CHD. Two of the duplications were maternal and intrachromosomal in origin with maternal heterozygosity for the common inversion between the repeats in 8p23.1. These additional patients and the absence of the 8p23.1 duplications in published controls, indicate that the 8p23.1 duplication syndrome may now be considered a pathogenic copy number variation (pCNV) with an estimated population prevalence of 1 in 58,000.
Collapse
Affiliation(s)
- John C K Barber
- Faculty of Medicine, Department of Human Genetics and Genomic Medicine, University of Southampton, Southampton General Hospital, Southampton, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
The heart as a functional organ first appeared in bilaterians as a single peristaltic pump and evolved through arthropods, fish, amphibians, and finally mammals into a four-chambered engine controlling blood-flow within the body. The acquisition of cardiac complexity in the evolving heart was a product of gene duplication events and the co-option of novel signaling pathways to an ancestral cardiac-specific gene network. T-box factors belong to an evolutionary conserved family of transcriptional regulators with diverse roles in development. Their regulatory functions are integral in the initiation and potentiation of heart development, and mutations in these genes are associated with congenital heart defects. In this review we will discuss the evolutionary conserved cardiac regulatory functions of this family as well as their implication in disease in an aim to facilitate future gene-targeted and regenerative therapeutic remedies.
Collapse
Affiliation(s)
- Fadi Hariri
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, C.P. 6128, Succursale, Centre-ville Montréal, Quebec, H3C3J7, Canada
| | | | | |
Collapse
|
40
|
Lalani SR, Shaw C, Wang X, Patel A, Patterson LW, Kolodziejska K, Szafranski P, Ou Z, Tian Q, Kang SHL, Jinnah A, Ali S, Malik A, Hixson P, Potocki L, Lupski JR, Stankiewicz P, Bacino CA, Dawson B, Beaudet AL, Boricha FM, Whittaker R, Li C, Ware SM, Cheung SW, Penny DJ, Jefferies JL, Belmont JW. Rare DNA copy number variants in cardiovascular malformations with extracardiac abnormalities. Eur J Hum Genet 2012; 21:173-81. [PMID: 22929023 DOI: 10.1038/ejhg.2012.155] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Clinically significant cardiovascular malformations (CVMs) occur in 5-8 per 1000 live births. Recurrent copy number variations (CNVs) are among the known causes of syndromic CVMs, accounting for an important fraction of cases. We hypothesized that many additional rare CNVs also cause CVMs and can be detected in patients with CVMs plus extracardiac anomalies (ECAs). Through a genome-wide survey of 203 subjects with CVMs and ECAs, we identified 55 CNVs >50 kb in length that were not present in children without known cardiovascular defects (n=872). Sixteen unique CNVs overlapping these variants were found in an independent CVM plus ECA cohort (n=511), which were not observed in 2011 controls. The study identified 12/16 (75%) novel loci including non-recurrent de novo 16q24.3 loss (4/714) and de novo 2q31.3q32.1 loss encompassing PPP1R1C and PDE1A (2/714). The study also narrowed critical intervals in three well-recognized genomic disorders of CVM, such as the cat-eye syndrome region on 22q11.1, 8p23.1 loss encompassing GATA4 and SOX7 and 17p13.3-p13.2 loss. An analysis of protein-interaction databases shows that the rare inherited and de novo CNVs detected in the combined cohort are enriched for genes encoding proteins that are direct or indirect partners of proteins known to be required for normal cardiac development. Our findings implicate rare variants such as 16q24.3 loss and 2q31.3-q32.1 loss, and delineate regions within previously reported structural variants known to cause CVMs.
Collapse
Affiliation(s)
- Seema R Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Costa G, Mazan A, Gandillet A, Pearson S, Lacaud G, Kouskoff V. SOX7 regulates the expression of VE-cadherin in the haemogenic endothelium at the onset of haematopoietic development. Development 2012; 139:1587-98. [PMID: 22492353 DOI: 10.1242/dev.071282] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024]
Abstract
At early stages of vertebrate ontogeny, blood and endothelial cells develop from a common mesodermal progenitor, the haemangioblast. Upon haematopoietic commitment, the haemangioblast generates blood precursors through populations of endothelial cells with haemogenic properties. Although several transcription factors have been implicated in haemangioblast differentiation, the precise mechanisms governing cell fate decisions towards the generation of haemogenic endothelium precursors remain largely unknown. Under defined conditions, embryonic stem (ES) cells can be differentiated into haemangioblast-like progenitors that faithfully recapitulate early embryonic haematopoiesis. Here, we made use of mouse ES cells as a model system to understand the role of SOX7, a member of a large family of transcription factors involved in a wide range of developmental processes. During haemangioblast differentiation, SOX7 is expressed in haemogenic endothelium cells and is downregulated in nascent blood precursors. Gain-of-function assays revealed that the enforced expression of Sox7 in haemangioblast-derived blast colonies blocks further differentiation and sustains the expression of endothelial markers. Thus, to explore the transcriptional activity of SOX7, we focused on the endothelial-specific adhesion molecule VE-cadherin. Similar to SOX7, VE-cadherin is expressed in haemogenic endothelium and is downregulated during blood cell formation. We show that SOX7 binds and activates the promoter of VE-cadherin, demonstrating that this gene is a novel downstream transcriptional target of SOX7. Altogether, our findings suggest that SOX7 is involved in the transcriptional regulation of genes expressed in the haemogenic endothelium and provide new clues to decipher the molecular pathways that drive early embryonic haematopoiesis.
Collapse
Affiliation(s)
- Guilherme Costa
- Cancer Research UK Stem Cell Research Group, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | | | | | | | | | | |
Collapse
|
42
|
Hoeth M, Niederleithner H, Hofer-Warbinek R, Bilban M, Mayer H, Resch U, Lemberger C, Wagner O, Hofer E, Petzelbauer P, de Martin R. The transcription factor SOX18 regulates the expression of matrix metalloproteinase 7 and guidance molecules in human endothelial cells. PLoS One 2012; 7:e30982. [PMID: 22292085 PMCID: PMC3264645 DOI: 10.1371/journal.pone.0030982] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 12/29/2011] [Indexed: 11/18/2022] Open
Abstract
Background Mutations in the transcription factor SOX18 are responsible for specific cardiovascular defects in humans and mice. In order to gain insight into the molecular basis of its action, we identified target genes of SOX18 and analyzed one, MMP7, in detail. Methodology/Principal Findings SOX18 was expressed in HUVEC using a recombinant adenoviral vector and the altered gene expression profile was analyzed using microarrays. Expression of several regulated candidate SOX18 target genes was verified by real-time PCR. Knock-down of SOX18 using RNA interference was then used to confirm the effect of the transcription factor on selected genes that included the guidance molecules ephrin B2 and semaphorin 3G. One gene, MMP7, was chosen for further analysis, including detailed promoter studies using reporter gene assays, electrophoretic mobility shift analysis and chromatin-immunoprecipitation, revealing that it responds directly to SOX18. Immunohistochemical analysis demonstrated the co-expression of SOX18 and MMP7 in blood vessels of human skin. Conclusions/Significance The identification of MMP7 as a direct SOX18 target gene as well as other potential candidates including guidance molecules provides a molecular basis for the proposed function of this transcription factor in the regulation of vessel formation.
Collapse
Affiliation(s)
- Martina Hoeth
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | | | - Renate Hofer-Warbinek
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Martin Bilban
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Vienna, Vienna, Austria
| | - Herbert Mayer
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Ulrike Resch
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Christof Lemberger
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Oswald Wagner
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Vienna, Vienna, Austria
| | - Erhard Hofer
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Peter Petzelbauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Rainer de Martin
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
43
|
M. A, M. H. S, M. KS, B. G. Differential expression pattern of the human endoderm-specific transcription factor sox17 in various tissues and cells. Int J Organ Transplant Med 2012; 3:183-7. [PMID: 25013644 PMCID: PMC4089296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Sox17 is a member of the Sry-related high mobility group (HMG) of transcription factors that is necessary for endodermal formation and liver development in multiple species. Sox17 gene expression is required for formation of definitive endoderm that gives rise to various tissues. OBJECTIVE To examine the expression of Sox17 in various human tissues and cells. METHODS Semiquantitative polymerase chain reaction (RT-PCR) was used to evaluate the expression of Sox17 in adult liver, small intestine, spleen, placenta, fetal liver as well as embryonic stem cells (ESCs), and human HepG2 hepatoma cell line. RESULTS Low Sox17 gene expression was observed in ESCs. However, there was no expression of Sox17 in human placental tissue, small intestine, adult liver, spleen, and HepG2 cells. But its expression in human fetal liver was very high. CONCLUSION The data presented in this study reflect the differential expression pattern of Sox17 in the fetal development during early mammalian endodermal formation which is temporal and tightly regulated.
Collapse
Affiliation(s)
- Ayatollahi M.
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran,Correspondence: Maryam Ayatollahi, Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran, Tel/fax: +98-711-647-4331, E-mail:
| | - Sanati M. H.
- National Institute of Genetic Engineering and Biotechnology, Tehran, IranAbstract
| | - Kabir Salmani M.
- National Institute of Genetic Engineering and Biotechnology, Tehran, IranAbstract
| | - Geramizaeh B.
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
44
|
UY BENJAMINR, SIMOES-COSTA MARCOS, SAUKA-SPENGLER TATJANA, BRONNER MARIANNEE. Expression of Sox family genes in early lamprey development. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2012; 56:377-83. [PMID: 22811271 PMCID: PMC4118928 DOI: 10.1387/ijdb.113416bu] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Members of the Sox (Sry-related high mobility group box) family of transcription factors play a variety of roles during development of both vertebrates and invertebrates. A marked expansion in gene number occurred during the emergence of vertebrates, apparently via gene duplication events that are thought to have facilitated new functions. By screening a macroarrayed library as well as the lamprey genome, we have isolated genes of the Sox B, D, E and F subfamilies in the basal jawless vertebrate, lamprey. The expression patterns of all identified Sox genes were examined from gastrulation through early organogenesis (embryonic day 4-14), with particular emphasis on the neural crest, a vertebrate innovation. Coupled with phylogenetic analysis of these Sox genes, the results provide insight into gene duplication and di-vergence in paralog deployment occurring during early vertebrate evolution.
Collapse
Affiliation(s)
- BENJAMIN R. UY
- Division of Biology 139-74, California Institute of Technology, Pasadena, CA 91125 USA
| | - MARCOS SIMOES-COSTA
- Division of Biology 139-74, California Institute of Technology, Pasadena, CA 91125 USA
| | | | - MARIANNE E. BRONNER
- Division of Biology 139-74, California Institute of Technology, Pasadena, CA 91125 USA
| |
Collapse
|
45
|
Abstract
The study of gene function in developmental biology has been significantly furthered by advances in antisense technology made in the early 2000s. This was achieved, in particular, by the introduction of morpholino (MO) oligonucleotides. The introduction of antisense MO oligonucleotides into cells enables researchers to readily reduce the levels of their protein of interest without investing huge financial or temporal resources, in both in vivo and in vitro model systems. Historically, the African clawed frog Xenopus has been used to study vertebrate embryological development, due to its ability to produce vast numbers of offspring that develop rapidly, in synchrony, and can be cultured in buffers with ease. The developmental progress of Xenopus embryos has been extensively characterized and this model organism is very easy to maintain. It is these attributes that enable MO-based knockdown strategies to be so effective in Xenopus. In this chapter, we will detail the methods of microinjecting MO oligonucleotides into early embryos of X. laevis and X. tropicalis. We will discuss how MOs can be used to prevent either pre-mRNA splicing or translation of the specific gene of interest resulting in abrogation of that gene's function and advise on what control experiments should be undertaken to verify their efficacy.
Collapse
Affiliation(s)
- Panna Tandon
- Department of Genetics, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | | |
Collapse
|
46
|
Eom BW, Jo MJ, Kook MC, Ryu KW, Choi IJ, Nam BH, Kim YW, Lee JH. The lymphangiogenic factor SOX 18: A key indicator to stage gastric tumor progression. Int J Cancer 2011; 131:41-8. [DOI: 10.1002/ijc.26325] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 07/13/2011] [Indexed: 01/22/2023]
|
47
|
Chung MIS, Ma ACH, Fung TK, Leung AYH. Characterization of Sry-related HMG box group F genes in zebrafish hematopoiesis. Exp Hematol 2011; 39:986-998.e5. [PMID: 21726513 DOI: 10.1016/j.exphem.2011.06.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Revised: 06/18/2011] [Accepted: 06/22/2011] [Indexed: 02/08/2023]
Abstract
OBJECTIVE The roles of Sry-related HMG box (Sox) genes in zebrafish hematopoiesis are not clearly defined. In this study, we have characterized the sequence homology, gene expression, hematopoietic functions, and regulation of sox genes in F group (SoxF) in zebrafish embryos. MATERIALS AND METHODS Expression of zebrafish SoxF genes were analyzed by whole-mount in situ hybridization, reverse transcription polymerase chain reaction, and real-time reverse transcription polymerase chain reaction of erythroid cells obtained from Tg(gata1:GFP) embryos by fluorescence-activated cell sorting. Roles of SoxF genes were analyzed in zebrafish embryos using morpholino knockdown and analyzed by whole-mount in situ hybridization and real-time reverse transcription polymerase chain reaction. Embryo patterning and vascular development were analyzed. RESULTS All members, except sox17, contained a putative β-catenin binding site. sox7 and 18 expressed primarily in the vasculature. sox17 expressed in the intermediate cell mass and its knockdown significantly reduced primitive erythropoiesis at 18 hours post-fertilization (hpf). Definitive hematopoiesis was unaffected. Concomitant sox7 and sox18 knockdown disrupted vasculogenesis and angiogenesis, but not hematopoiesis. sox32 knockdown delayed medial migration of hematopoietic and endothelial progenitors at 18 hpf and abolished cmyb expression at the caudal hematopoietic tissue at 48 hpf. These defects could be prevented by delaying its knockdown using a caged sox32 morpholino uncaged at 10 hpf. Knockdown of SoxF genes significantly upregulated their own expression and that of sox32 also upregulated sox18 expression. CONCLUSIONS sox17 helped to maintain primitive hematopoiesis, whereas sox7 and sox18 regulated angiogenesis and vasculogenesis. sox32 affected both vascular and hematopoietic development through its effects on medial migration of the hematopoietic and endothelial progenitors.
Collapse
Affiliation(s)
- Martin In Shing Chung
- Division of Haematology and Bone Marrow Transplantation, Department of Medicine, Queen Mary Hospital, University of Hong Kong, China
| | | | | | | |
Collapse
|
48
|
Aranguren XL, Beerens M, Vandevelde W, Dewerchin M, Carmeliet P, Luttun A. Transcription factor COUP-TFII is indispensable for venous and lymphatic development in zebrafish and Xenopus laevis. Biochem Biophys Res Commun 2011; 410:121-6. [PMID: 21641336 DOI: 10.1016/j.bbrc.2011.05.117] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 05/20/2011] [Indexed: 11/16/2022]
Abstract
Transcription factors play a central role in cell fate determination. Gene targeting in mice revealed that Chicken Ovalbumin Upstream Promoter-Transcription Factor II (COUP-TFII, also known as Nuclear Receptor 2F2 or NR2F2) induces a venous phenotype in endothelial cells (ECs). More recently, NR2F2 was shown to be required for initiating the expression of Prox1, responsible for lymphatic commitment of venous ECs. Small animal models like zebrafish embryos and Xenopus laevis tadpoles have been very useful to elucidate mechanisms of (lymph) vascular development. Therefore, the role of NR2F2 in (lymph) vascular development was studied by eliminating its expression in these models. Like in mice, absence of NR2F2 in zebrafish resulted in distinct vascular defects including loss of venous marker expression, major trunk vessel fusion and vascular leakage. Both in zebrafish and Xenopus the development of the main lymphatic structures was severely hampered. NR2F2 knockdown significantly decreased prox1 expression in zebrafish ECs and the same manipulation affected lymphatic (L)EC commitment, migration and function in Xenopus tadpoles. Therefore, the role of NR2F2 in EC fate determination is evolutionary conserved.
Collapse
Affiliation(s)
- Xabier L Aranguren
- Center for Molecular and Vascular Biology, Katholieke Universiteit Leuven, Campus Gasthuisberg, Onderwijs & Navorsing 1, Herestraat 49, B-3000 Leuven, Belgium.
| | | | | | | | | | | |
Collapse
|
49
|
Kaltenbrun E, Tandon P, Amin NM, Waldron L, Showell C, Conlon FL. Xenopus: An emerging model for studying congenital heart disease. ACTA ACUST UNITED AC 2011; 91:495-510. [PMID: 21538812 DOI: 10.1002/bdra.20793] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 01/18/2011] [Accepted: 01/28/2011] [Indexed: 02/02/2023]
Abstract
Congenital heart defects affect nearly 1% of all newborns and are a significant cause of infant death. Clinical studies have identified a number of congenital heart syndromes associated with mutations in genes that are involved in the complex process of cardiogenesis. The African clawed frog, Xenopus, has been instrumental in studies of vertebrate heart development and provides a valuable tool to investigate the molecular mechanisms underlying human congenital heart diseases. In this review, we discuss the methodologies that make Xenopus an ideal model system to investigate heart development and disease. We also outline congenital heart conditions linked to cardiac genes that have been well studied in Xenopus and describe some emerging technologies that will further aid in the study of these complex syndromes.
Collapse
Affiliation(s)
- Erin Kaltenbrun
- University of North Carolina McAllister Heart Institute, Chapel Hill, NC 27599, USA
| | | | | | | | | | | |
Collapse
|
50
|
Affiliation(s)
- Michela Noseda
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Tessa Peterkin
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Filipa C. Simões
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Roger Patient
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Michael D. Schneider
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| |
Collapse
|