1
|
Guo S, Hu X, Cotton JL, Ma L, Li Q, Cui J, Wang Y, Thakare RP, Tao Z, Ip YT, Wu X, Wang J, Mao J. VGLL2 and TEAD1 fusion proteins identified in human sarcoma drive YAP/TAZ-independent tumorigenesis by engaging EP300. eLife 2025; 13:RP98386. [PMID: 40338073 PMCID: PMC12061476 DOI: 10.7554/elife.98386] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025] Open
Abstract
Studies on Hippo pathway regulation of tumorigenesis largely center on YAP and TAZ, the transcriptional co-regulators of TEADs. Here, we present an oncogenic mechanism involving VGLL and TEAD fusions that is Hippo pathway-related but YAP/TAZ-independent. We characterize two recurrent fusions, VGLL2-NCOA2 and TEAD1-NCOA2, recently identified in human spindle cell rhabdomyosarcoma. We demonstrate that in contrast to VGLL2 and TEAD1 the fusion proteins are potent activators of TEAD-dependent transcription, and the function of these fusion proteins does not require YAP/TAZ. Furthermore, we identify that VGLL2 and TEAD1 fusions engage specific epigenetic regulation by recruiting histone acetyltransferase EP300 to control TEAD-mediated transcriptional and epigenetic landscapes. We show that small-molecule EP300 inhibition can suppress fusion protein-induced oncogenic transformation both in vitro and in vivo in mouse models. Overall, our study reveals a molecular basis for VGLL involvement in cancer and provides a framework for targeting tumors carrying VGLL, TEAD, or NCOA translocations.
Collapse
Affiliation(s)
- Susu Guo
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaodi Hu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Jennifer L Cotton
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Lifang Ma
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qi Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Jiangtao Cui
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yongjie Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ritesh P Thakare
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Zhipeng Tao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Y Tony Ip
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Xu Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| |
Collapse
|
2
|
Chen Y, Liu B, Tao S, Liu L, Gao J, Liang Y, Dong W, Zhou D. CITED2 Binding to EP300 Regulates Human Spermatogonial Stem Cell Proliferation and Survival Through HSPA6. Stem Cells Int 2025; 2025:2362489. [PMID: 40313859 PMCID: PMC12045681 DOI: 10.1155/sci/2362489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/22/2025] [Indexed: 05/03/2025] Open
Abstract
Spermatogonial stem cells (SSCs) are essential for the initiation and continuation of spermatogenesis, a process fundamental to male fertility. Despite extensive studies on mouse SSCs, the mechanisms governing self-renewal and differentiation in human SSCs remain to be elucidated. This study investigated the regulatory mechanisms of SSCs by analyzing single-cell sequencing data from the GEO dataset of human testis. Analysis revealed dominant expression of CITED2 in human SSCs. Reduction of CITED2 levels in hSSC lines significantly inhibited proliferation and increased apoptosis. Protein interaction prediction and immunoprecipitation identified interactions between CITED2 and EP300 in SSC lines. RNA sequencing results indicated that CITED2 knockdown significantly affected the MAPK pathway and the HSPA6 gene. Overexpression of HSPA6 mitigated the proliferative and apoptotic changes provoked by CITED2 downregulation. These findings provide novel insights into the regulatory and functional mechanisms of CITED2-mediated hSSC development.
Collapse
Affiliation(s)
- Yongzhe Chen
- Gynecology and Obstetrics and Reproductive Medical Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- MOE Key Lab of Rare Pediatric Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Bang Liu
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Sisi Tao
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Lvjun Liu
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Jianxin Gao
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Ying Liang
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Weilei Dong
- Gynecology and Obstetrics and Reproductive Medical Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- MOE Key Lab of Rare Pediatric Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Dai Zhou
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Research Department, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| |
Collapse
|
3
|
Chu Y, Setayesh J, Dumontet T, Krumeich L, Werner J, Moretti IF, De Sousa K, Kennedy C, La Pensee C, Lerario AM, Hammer GD. Adrenocortical stem cells in health and disease. Nat Rev Endocrinol 2025:10.1038/s41574-025-01091-2. [PMID: 40065108 DOI: 10.1038/s41574-025-01091-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 04/13/2025]
Abstract
The adrenal cortex is the major site of production of steroid hormones, which are essential for life. The normal development and homeostatic renewal of the adrenal cortex depend on capsular stem cells and cortical progenitor cells. These cell populations are highly plastic and support adaptation to physiological demands, injury and disease, linking steroid production and adrenal (organ) homeostasis with systemic endocrine cues and organismal homeostasis. This Review integrates findings from the past decade, outlining the mechanisms that govern the establishment and maintenance of the adrenal stem cell niche under different physiological and pathological conditions. The sophisticated regulation of the stem cell niche by gene regulatory networks, coordinated through paracrine and endocrine signalling, is highlighted in a context-dependent and sex-specific manner. We discuss how dysregulation of this intricate regulatory network is implicated in a wide range of adrenal diseases, and how emerging knowledge from adrenal stem cell research is inspiring the future development of gene-based and cell-based therapeutic strategies.
Collapse
Affiliation(s)
- Yulan Chu
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jordan Setayesh
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
| | - Typhanie Dumontet
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Lauren Krumeich
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Johanna Werner
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Isabele F Moretti
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Kelly De Sousa
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Christopher Kennedy
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Christopher La Pensee
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Antonio M Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Gary D Hammer
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
- Endocrine Oncology Program, Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
4
|
Guo S, Hu X, Cotton JL, Ma L, Li Q, Cui J, Wang Y, Thakare RP, Tao Z, Ip YT, Wu X, Wang J, Mao J. VGLL2 and TEAD1 fusion proteins drive YAP/TAZ-independent tumorigenesis by engaging p300. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.01.592016. [PMID: 38746415 PMCID: PMC11092657 DOI: 10.1101/2024.05.01.592016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Studies on Hippo pathway regulation of tumorigenesis largely center on YAP and TAZ, the transcriptional co-regulators of TEAD. Here, we present an oncogenic mechanism involving VGLL and TEAD fusions that is Hippo pathway-related but YAP/TAZ-independent. We characterize two recurrent fusions, VGLL2-NCOA2 and TEAD1-NCOA2, recently identified in spindle cell rhabdomyosarcoma. We demonstrate that in contrast to VGLL2 and TEAD1, the fusion proteins are strong activators of TEAD-dependent transcription, and their function does not require YAP/TAZ. Furthermore, we identify that VGLL2 and TEAD1 fusions engage specific epigenetic regulation by recruiting histone acetyltransferase p300 to control TEAD-mediated transcriptional and epigenetic landscapes. We showed that small molecule p300 inhibition can suppress fusion proteins-induced oncogenic transformation both in vitro and in vivo. Overall, our study reveals a molecular basis for VGLL involvement in cancer and provides a framework for targeting tumors carrying VGLL, TEAD, or NCOA translocations.
Collapse
Affiliation(s)
- Susu Guo
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No 241, West Huaihai Road, Shanghai, P. R., 200030, China
| | - Xiaodi Hu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
| | - Jennifer L. Cotton
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
| | - Lifang Ma
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No 241, West Huaihai Road, Shanghai, P. R., 200030, China
| | - Qi Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
| | - Jiangtao Cui
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No 241, West Huaihai Road, Shanghai, P. R., 200030, China
| | - Yongjie Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No 241, West Huaihai Road, Shanghai, P. R., 200030, China
| | - Ritesh P. Thakare
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
| | - Zhipeng Tao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, 01605, USA
| | - Y. Tony Ip
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
| | - Xu Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, 01605, USA
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No 241, West Huaihai Road, Shanghai, P. R., 200030, China
| | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
| |
Collapse
|
5
|
Forman-Rubinsky R, Feng W, Schlegel BT, Paul A, Zuppo D, Kedziora K, Stoltz D, Watkins S, Rajasundaram D, Li G, Tsang M. Cited4a limits cardiomyocyte dedifferentiation and proliferation during zebrafish heart regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.626917. [PMID: 39713454 PMCID: PMC11661073 DOI: 10.1101/2024.12.05.626917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Cardiac regeneration involves the interplay of complex interactions between many different cell types, including cardiomyocytes. The exact mechanism that enables cardiomyocytes to undergo dedifferentiation and proliferation to replace lost cells has been intensely studied. Here we report a single nuclear RNA sequencing profile of the injured zebrafish heart and identify distinct cardiomyocyte populations in the injured heart. These cardiomyocyte populations have diverse functions, including stress response, myofibril assembly, proliferation and contraction. The contracting cardiomyocyte population also involves the activation of maturation pathways as an early response to injury. This intriguing finding suggests that constant maintenance of a distinctive terminally differentiated cardiomyocyte population is important for cardiac function during regeneration. To test this hypothesis, we determined that cited4a, a p300/CBP transcriptional coactivator, is induced after injury in the mature cardiomyocyte population. Moreover, loss-of-cited4a mutants presented increased dedifferentiation, proliferation and accelerated heart regeneration. Thus, suppressing cardiomyocyte maturation pathway activity in injured hearts could be an approach to promote heart regeneration.
Collapse
Affiliation(s)
- Rachel Forman-Rubinsky
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Wei Feng
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Brent T Schlegel
- Department of Pediatrics, Division of Health Informatics, Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Angela Paul
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Daniel Zuppo
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Current address: Department of Medicine, University of Rochester Medical Center Rochester, NY
| | - Katarzyna Kedziora
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Biological Imaging, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Donna Stoltz
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Biological Imaging, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Simon Watkins
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Biological Imaging, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Division of Health Informatics, Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Guang Li
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Michael Tsang
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| |
Collapse
|
6
|
Zhang Z, Qin Y, Huang J, Wang Y, Zeng L, Wang Y, Zhuyun F, Wang L. Oestrogen promotes the progression of adenomyosis by inhibiting CITED2 through miR-145. Reprod Biomed Online 2024; 49:104108. [PMID: 39293195 DOI: 10.1016/j.rbmo.2024.104108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/08/2024] [Accepted: 05/04/2024] [Indexed: 09/20/2024]
Abstract
RESEARCH QUESTION Is the microRNA miR-145 involved in adenomyosis, and by what mechanisms does it affect disease development and is itself regulated? DESIGN Fluorescence in-situ hybridization was used to observe the expression pattern of miR-145 in adenomyosis ectopic endometrium (n = 13), adenomyosis eutopic endometrium (n = 15) and non-adenomyosis eutopic endometrium (n = 14). RNA sequencing was used to screen target genes as well as downstream pathways of miR-145, which were validated by reporter gene assay, quantitative polymerase chain reaction and western blot, and further analysed using cell migration assay and chromatin immunoprecipitation assay. RESULTS The fluorescence in-situ hybridization assay revealed a noteworthy elevation in miR-145 expression in adenomyosis tissue compared with non-adenomyosis tissue. Furthermore, RNA sequencing analysis revealed that overexpression of miR-145 resulted in heightened expression of genes associated with the cytokine signalling pathway, nucleotide-binding and oligomerization domain-like pathway and adhesion pathway, including IL-1β and IL-6. Our study has identified CITED2 as a downstream direct target gene of miR-145, which is implicated in the inhibition of stromal cell migration induced by miR-145. Moreover, chromatin immunoprecipitation was used to validate the direct effect of oestradiol on the promoter region of miR-145, mediated by oestrogen receptor α, which facilitates the upregulation of miR-145 expression. CONCLUSION Our findings provide evidence supporting the role of oestradiol, acting through its receptor α, in modulating the discovered miR-145-CITED2 signalling axis, thereby promoting the progression of adenomyosis.
Collapse
Affiliation(s)
- Ziyu Zhang
- Department of Pathology, Jiangxi Maternal and Child Health Hospital, 330006 Nanchang, Jiangxi, PR China; The Subcenter of National Clinical Research Center for Obstetrics and Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, PR China; Clinical Research Center for Obstetrics and Gynecology of Jiangxi province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, PR China; Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, 330006 Nanchang, Jiangxi, PR China.
| | - Yunna Qin
- Department of Pathology, Jiangxi Maternal and Child Health Hospital, 330006 Nanchang, Jiangxi, PR China
| | - Jia Huang
- Department of Gynaecology, Jiangxi Maternal and Child Health Hospital, 330006 Nanchang, Jiangxi, PR China
| | - Yaoqing Wang
- Department of Gynaecology, Jiangxi Maternal and Child Health Hospital, 330006 Nanchang, Jiangxi, PR China
| | - Liqin Zeng
- Department of Gynaecology, Jiangxi Maternal and Child Health Hospital, 330006 Nanchang, Jiangxi, PR China
| | - Yuanqin Wang
- Department of Gynaecology, Jiangxi Maternal and Child Health Hospital, 330006 Nanchang, Jiangxi, PR China
| | - Fu Zhuyun
- Jiujiang Blood Central, Jiujiang, Jiangxi, PR China.
| | - Liqun Wang
- Department of Gynaecology, Jiangxi Maternal and Child Health Hospital, 330006 Nanchang, Jiangxi, PR China.
| |
Collapse
|
7
|
Zeng M, Yang X, Chen Y, Fan J, Cao L, Wang M, Xiao P, Ling Z, Yin Y, Chen Y. A Network and Pathway Analysis of Genes Associated With Atrial Fibrillation. Cardiovasc Ther 2024; 2024:7054039. [PMID: 39742001 PMCID: PMC11470814 DOI: 10.1155/2024/7054039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/09/2024] [Indexed: 01/03/2025] Open
Abstract
Background: Atrial fibrillation (AF) is affected by both environmental and genetic factors. Previous genetic association studies, especially genome-wide association studies, revealed a large group of AF-associated genes. However, little is known about the functions and interactions of these genes. Moreover, established genetic variants of AF contribute modestly to AF variance, implying that numerous additional AF-associated genetic variations need to be identified. Hence, a systematic network and pathway analysis is needed. Methods: We retrieved all AF-associated genes from genetic association studies in various databases and performed integrative analyses including pathway enrichment analysis, pathway crosstalk analysis, network analysis, and microarray meta-analysis. Results: We collected 254 AF-associated genes from genetic association studies in various databases. Pathway enrichment analysis revealed the top biological pathways that were enriched in the AF-associated genes related to cardiac electromechanical activity. Pathway crosstalk analysis showed that numerous neuro-endocrine-immune pathways connected AF with various diseases including cancers, inflammatory diseases, and cardiovascular diseases. Furthermore, an AF-specific subnetwork was constructed with the prize-collecting Steiner forest algorithm based on the AF-associated genes, and 24 novel genes that were potentially associated with AF were inferred by the subnetwork. In the microarray meta-analysis, six of the 24 novel genes (APLP1, CREB1, CREBBP, PRMT1, IRAK1, and PLXND1) were expressed differentially in patients with AF and sinus rhythm. Conclusions: AF is not only an isolated disease with abnormal electrophysiological activity but might also share a common genetic basis and biological process with tumors and inflammatory diseases as well as cardiovascular diseases. Moreover, the six novel genes inferred from network analysis might help detect the missing AF risk loci.
Collapse
Affiliation(s)
- Mengying Zeng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Xian Yang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | | | - Jinqi Fan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Li Cao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Menghao Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peilin Xiao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Zhiyu Ling
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Yuehui Yin
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Yunlin Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| |
Collapse
|
8
|
Carreon CK, Ronai C, Hoffmann JK, Tworetzky W, Morton SU, Wilkins-Haug LE. Maternal Vascular Malperfusion and Anatomic Cord Abnormalities Are Prevalent in Pregnancies With Fetal Congenital Heart Disease. Prenat Diagn 2024. [PMID: 39215461 DOI: 10.1002/pd.6650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE Impairments in the maternal-fetal environment are associated with adverse postnatal outcomes among infants with congenital heart disease. Therefore, we sought to investigate placental anomalies as they related to various forms of fetal congenital heart disease (FCHD). METHODS We reviewed the placental pathology in singleton pregnancies with and without FCHD. FCHD was divided into separate categories (transposition physiology, obstructive left, obstructive right, biventricular without obstruction, and others). Exclusion criteria included other prenatally known structural malformations and/or aneuploidy. The significance threshold was set at p < 0.05 or False Discovery rate q < 0.05 when multiple tests were performed. RESULTS The cohort included 215 FCHD and 122 non-FCHD placentas. FCHD placentas showed increased rates of maternal vascular malperfusion (24% vs. 5%, q < 0.001) and cord anomalies (27% vs. 1%, q < 0.001). Placentas with fetal TGA demonstrated a lower rate of hypoplasia when compared with other FCHD types (1/39 vs. 51/176, Fisher's exact p = 0.015). CONCLUSION Placental maternal vascular malperfusion is increased in FCHD. The prevalence of vascular malperfusion did not differ by FCHD type, indicating that CHD type does not predict the likelihood of placental vascular dysfunction. Further investigation of the placental-fetal heart axis in FCHD is warranted given the importance of placental health.
Collapse
Affiliation(s)
- Chrystalle Katte Carreon
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Christina Ronai
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Julia K Hoffmann
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatric Cardiology and Pediatric Intensive Care, Ludwig Maximilian University, Munich, Germany
| | - Wayne Tworetzky
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Sarah U Morton
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Louise E Wilkins-Haug
- Division of Maternal Fetal Medicine and Reproductive Genetics, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Kuna M, Soares MJ. Cited2 is a key regulator of placental development and plasticity. Bioessays 2024; 46:e2300118. [PMID: 38922923 PMCID: PMC11331489 DOI: 10.1002/bies.202300118] [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: 06/30/2023] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
The biology of trophoblast cell lineage development and placentation is characterized by the involvement of several known transcription factors. Central to the action of a subset of these transcriptional regulators is CBP-p300 interacting transactivator with Glu/Asp-rich carboxy-terminal domain 2 (CITED2). CITED2 acts as a coregulator modulating transcription factor activities and affecting placental development and adaptations to physiological stressors. These actions of CITED2 on the trophoblast cell lineage and placentation are conserved across the mouse, rat, and human. Thus, aspects of CITED2 biology in hemochorial placentation can be effectively modeled in the mouse and rat. In this review, we present information on the conserved role of CITED2 in the biology of placentation and discuss the use of CITED2 as a tool to discover new insights into regulatory mechanisms controlling placental development.
Collapse
Affiliation(s)
- Marija Kuna
- Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Michael J. Soares
- Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy, Kansas City, MO
| |
Collapse
|
10
|
Imam R, Aizezi M, Yan F, Zhu T, Zhang W. Sequence variations in GATA4 and CITED2 gene among patients with cardiac septation defects from Xinjiang, China. Cardiol Young 2024; 34:1506-1513. [PMID: 38456293 DOI: 10.1017/s1047951124000192] [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] [Indexed: 03/09/2024]
Abstract
Studies have shown that genetic factors play an important role in CHD's development. The mutations in GATA4 and CITED2 genes result in the failure of the heart to develop normally, thereby leading to septal defects. The present study investigated the underlying molecular aetiology of patients with cardiac septation defects from Xinjiang. We investigated variants of the GATA4 and CITED2 gene coding regions in 172 patients with cardiac septation defects by sequencing. Healthy controls (n = 200) were included. Three heterozygous variations (p.V380M, p.P394T, and p.P407Q) of the GATA4 gene were identified in three patients. p.V380M was discovered in a patient with atrial septal defect. p.P394T was noted in a patient with atrial septal defect. p.V380M and p.P407Q of the GATA4 gene were detected in one patient with ventricular septal defect. A novel homozygous variation (p. Sl92G) of the CITED2 gene was found in one patient with ventricular septal defect. Other patients and healthy individuals were normal. The limited prevalence of genetic variations observed in individuals with cardiac septal defects from Xinjiang provides evidence in favour of the hypothesis that CHD is a polygenic hereditary disorder. It is plausible that mutations in the GATA4 and CITED2 genes could potentially underlie the occurrence of idiopathic CHD in affected patients.
Collapse
Affiliation(s)
- Renati Imam
- Department of Cardiac Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Maimaitiaili Aizezi
- Department of Cardiac Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Fei Yan
- Department of Cardiac Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Tao Zhu
- Department of Cardiac Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Weimin Zhang
- Department of Cardiac Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
11
|
Jank M, Schwartz J, Miyake Y, Ozturk Aptekmann A, Patel D, Boettcher M, Keijzer R. Dysregulation of CITED2 in abnormal lung development in the nitrofen rat model. Pediatr Surg Int 2024; 40:43. [PMID: 38291157 DOI: 10.1007/s00383-023-05607-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/22/2023] [Indexed: 02/01/2024]
Abstract
PURPOSE CITED2 both modulates lung, heart and diaphragm development. The role of CITED2 in the pathogenesis of congenital diaphragmatic hernia (CDH) is unknown. We aimed to study CITED2 during abnormal lung development in the nitrofen model. METHODS Timed-pregnant rats were given nitrofen on embryonic day (E) 9 to induce CDH. Fetal lungs were harvested on E15, 18 and 21. We performed RT-qPCR, RNAscope™ in situ hybridization and immunofluorescence staining for CITED2. RESULTS We observed no difference in RT-qPCR (control: 1.09 ± 0.22 and nitrofen: 0.95 ± 0.18, p = 0.64) and in situ hybridization (1.03 ± 0.03; 1.04 ± 0.03, p = 0.97) for CITED2 expression in E15 nitrofen and control pups. At E18, CITED2 expression was reduced in in situ hybridization of nitrofen lungs (1.47 ± 0.05; 1.14 ± 0.07, p = 0.0006), but not altered in RT-qPCR (1.04 ± 0.16; 0.81 ± 0.13, p = 0.33). In E21 nitrofen lungs, CITED2 RNA expression was increased in RT-qPCR (1.04 ± 0.11; 1.52 ± 0.17, p = 0.03) and in situ hybridization (1.08 ± 0.07, 1.29 ± 0.04, p = 0.02). CITED2 protein abundance was higher in immunofluorescence staining of E21 nitrofen lungs (2.96 × 109 ± 0.13 × 109; 4.82 × 109 ± 0.25 × 109, p < 0.0001). CONCLUSION Our data suggest that dysregulation of CITED2 contributes to abnormal lung development of CDH, as demonstrated by the distinct spatial-temporal distribution in nitrofen-induced lungs.
Collapse
MESH Headings
- Animals
- Female
- Pregnancy
- Rats
- 2,4-Dinitrophenol
- Disease Models, Animal
- Gene Expression Regulation, Developmental
- Hernias, Diaphragmatic, Congenital/chemically induced
- Hernias, Diaphragmatic, Congenital/genetics
- Hernias, Diaphragmatic, Congenital/metabolism
- Lung/abnormalities
- Lung Diseases/metabolism
- Phenyl Ethers/toxicity
- Rats, Sprague-Dawley
- Respiratory System Abnormalities
Collapse
Affiliation(s)
- Marietta Jank
- Department of Surgery, Division of Pediatric Surgery, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, and Children's Hospital Research Institute of Manitoba, AE402-820 Sherbrook Street, Winnipeg, MB, R3A 1S1, Canada
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Jacquelyn Schwartz
- Department of Surgery, Division of Pediatric Surgery, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, and Children's Hospital Research Institute of Manitoba, AE402-820 Sherbrook Street, Winnipeg, MB, R3A 1S1, Canada
| | - Yuichiro Miyake
- Department of Surgery, Division of Pediatric Surgery, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, and Children's Hospital Research Institute of Manitoba, AE402-820 Sherbrook Street, Winnipeg, MB, R3A 1S1, Canada
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Arzu Ozturk Aptekmann
- Department of Surgery, Division of Pediatric Surgery, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, and Children's Hospital Research Institute of Manitoba, AE402-820 Sherbrook Street, Winnipeg, MB, R3A 1S1, Canada
| | - Daywin Patel
- Department of Surgery, Division of Pediatric Surgery, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, and Children's Hospital Research Institute of Manitoba, AE402-820 Sherbrook Street, Winnipeg, MB, R3A 1S1, Canada
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Richard Keijzer
- Department of Surgery, Division of Pediatric Surgery, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, and Children's Hospital Research Institute of Manitoba, AE402-820 Sherbrook Street, Winnipeg, MB, R3A 1S1, Canada.
| |
Collapse
|
12
|
Gill E, Bamforth SD. Molecular Pathways and Animal Models of Semilunar Valve and Aortic Arch Anomalies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:777-796. [PMID: 38884748 DOI: 10.1007/978-3-031-44087-8_46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The great arteries of the vertebrate carry blood from the heart to the systemic circulation and are derived from the pharyngeal arch arteries. In higher vertebrates, the pharyngeal arch arteries are a symmetrical series of blood vessels that rapidly remodel during development to become the asymmetric aortic arch arteries carrying oxygenated blood from the left ventricle via the outflow tract. At the base of the aorta, as well as the pulmonary trunk, are the semilunar valves. These valves each have three leaflets and prevent the backflow of blood into the heart. During development, the process of aortic arch and valve formation may go wrong, resulting in cardiovascular defects, and these may, at least in part, be caused by genetic mutations. In this chapter, we will review models harboring genetic mutations that result in cardiovascular defects affecting the great arteries and the semilunar valves.
Collapse
Affiliation(s)
- Eleanor Gill
- Newcastle University Biosciences Institute, Newcastle upon Tyne, UK
| | - Simon D Bamforth
- Newcastle University Biosciences Institute, Newcastle upon Tyne, UK.
| |
Collapse
|
13
|
Gill E, Bamforth SD. Molecular Pathways and Animal Models of Truncus Arteriosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:853-865. [PMID: 38884754 DOI: 10.1007/978-3-031-44087-8_52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
In normal cardiovascular development in birds and mammals, the outflow tract of the heart is divided into two distinct channels to separate the oxygenated systemic blood flow from the deoxygenated pulmonary circulation. When the process of outflow tract septation fails, a single common outflow vessel persists resulting in a serious clinical condition known as persistent truncus arteriosus or common arterial trunk. In this chapter, we will review molecular pathways and the cells that are known to play a role in the formation and development of the outflow tract and how genetic manipulation of these pathways in animal models can result in common arterial trunk.
Collapse
Affiliation(s)
- Eleanor Gill
- Newcastle University Biosciences Institute, Newcastle, UK
| | | |
Collapse
|
14
|
Bojórquez Martínez CA, García Murillo IM, Segón Mora S, López Mereles A. Tetralogy of Fallot: Hypoxia, the villain of the story? Birth Defects Res 2024; 116:e2279. [PMID: 38277413 DOI: 10.1002/bdr2.2279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Tetralogy of Fallot (ToF) is a cyanotic congenital heart disease, composed of four malformations: persistent communication between the right and the left ventricle, pulmonary stenosis, overriding aorta, and right ventricle hypertrophy. The etiology of this disease is not entirely known as yet, but it has been proposed that the pathology has genetic components. During embryonic development, the fetus is exposed to a physiological hypoxia to facilitate the formation of blood vessels and blood cells through de novo processes. METHODS After researching scientific databases on the implications of oxygen on the normal and abnormal development of organs, especially the heart, we were able to propose that oxygen deprivation may be the cause of the disease. RESULTS During this period, the hypoxia-inducible factor is activated and triggers transcriptional responses that enable adaptation to the hypoxic environment through angiogenic activation. High levels of this protein can alter certain physiological pathways, such as those related to the vascular endothelial growth factor. Research has shown that prolonged oxygen deprivation during embryological development can lead to the occurrence of congenital heart diseases, such as ToF. CONCLUSIONS Studies using animal models have demonstrated that the deficiency or disruption of a protein called "CITED2," which plays an important role in cardiac morphogenesis and its loss, results in the alteration of pluripotent, cardiac, and neural lineage differentiation, thereby disrupting the normal development of the heart and other tissues.
Collapse
Affiliation(s)
| | | | - Santiago Segón Mora
- Faculty of Medicine, Facultad Mexicana de Medicina-La Salle University, Mexico City, Tlalpan, Mexico
| | - Andrea López Mereles
- Faculty of Medicine, Facultad Mexicana de Medicina-La Salle University, Mexico City, Tlalpan, Mexico
| |
Collapse
|
15
|
Gill E, Bamforth SD. Molecular Pathways and Animal Models of d-Transposition of the Great Arteries. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:683-696. [PMID: 38884742 DOI: 10.1007/978-3-031-44087-8_40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
During normal cardiovascular development, the outflow tract becomes septated and rotates so that the separate aorta and pulmonary trunk are correctly aligned with the left and right ventricles, respectively. However, when this process goes wrong, the aorta and pulmonary trunk are incorrectly positioned, resulting in oxygenated blood being directly returned to the lungs, with deoxygenated blood being delivered to the systemic circulation. This is termed transposition of the great arteries (TGA). The precise etiology of TGA is not known, but the use of animal models has elucidated that genes involved in determination of the left- embryonic body axis play key roles. Other factors such as retinoic acid levels are also crucial. This chapter reviews the animal models presenting with TGA that have been generated by genetic manipulation or with exogenous agents.
Collapse
Affiliation(s)
- Eleanor Gill
- Newcastle University Biosciences Institute, Newcastle, UK
| | | |
Collapse
|
16
|
Chen Z, Chen HX, Hou HT, Yin XY, Yang Q, He GW. Identification and Functional Verification of CITED2 Gene Promoter Region in Patients with Patent Ductus Arteriosus. Int J Mol Sci 2023; 24:16204. [PMID: 38003393 PMCID: PMC10671043 DOI: 10.3390/ijms242216204] [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: 07/03/2023] [Revised: 10/08/2023] [Accepted: 10/18/2023] [Indexed: 11/26/2023] Open
Abstract
Patent ductus arteriosus (PDA) is a common congenital heart disease. CITED2 plays an important role in the development of the heart, and genetic variants in its coding region are significantly associated with cardiac malformations. However, the role of variants in the promoter region of CITED2 in the development of PDA remains unclear. We extracted the peripheral blood of 646 subjects (including 353 PDA patients and 293 unrelated healthy controls) for sequencing. We identified 13 promoter variants of the CITED2 gene (including 2 novel heterozygous variants). Of the 13 variants, 10 were found only in PDA patients. In mouse cardiomyocytes (HL-1) and rat cardiac myocytes (RCM), the transcriptional activity of the CITED2 gene promoter was significantly changed by the variants (p < 0.05). The results of the experiments of electrophoretic mobility indicated that these variants may affect the transcription of the CITED2 gene by influencing the binding ability of transcription factors. These results, combined with the JASPAR database analysis, showed that the destruction/production of transcription factor binding sites due to the variants in the promoter region of the CITED2 gene may directly or indirectly affect the binding ability of transcription factors. Our results suggest for the first time that variants at the CITED2 promoter region may cause low expression of CITED2 protein related to the formation of PDA.
Collapse
Affiliation(s)
| | | | | | | | | | - Guo-Wei He
- The Institute of Cardiovascular Diseases & Department Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin 300457, China; (Z.C.); (H.-X.C.); (H.-T.H.); (X.-Y.Y.); (Q.Y.)
| |
Collapse
|
17
|
Taha M, Awny N, Ismail S, Ashaat EA, Senousy MA. Screening and evaluation of TBX20 and CITED2 mutations in children with congenital cardiac septal defects: Correlation with cardiac troponin T and caspase-3. Gene 2023; 882:147660. [PMID: 37481008 DOI: 10.1016/j.gene.2023.147660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/07/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
Congenital cardiac septal defect (CCSD) is the main type of congenital heart disease and owns a very high mortality rate among newborns. CCSD is controlled by specific transcription factors, including T-box transcription factor 20 (TBX20) and Cbp/P300 interacting transactivator with Glu/Asp rich carboxy-terminal domain 2 (CITED2) which are key molecular actors in heart development. Here, we screened for mutations in TBX20 and CITED2 genes in Egyptian children with CCSD and assessed their association with CCSD susceptibility and with cardiac troponin T (cTnT) and the apoptotic marker caspase-3 as biochemical markers for CCSD. Thirty unrelated newborns and children affected with CCSD and 30 matched healthy controls with no personal history of cardiac diseases were recruited. Selection criteria were children (<18 years) with any age diagnosed with CCSD using ECHO. Mutational analysis and genotyping were done using PCR-Sanger DNA sequencing technique. Serum cTnT and caspase-3 were analyzed using ELISA. Sequencing analysis identified 2 TBX20 variants (c.766T>C and c.39T>C) in the CCSD and control groups and 2 CITED2 variants (c.12T>C and c.9C>T) in one CCSD patient, while were absent in controls. In silico analysis identified TBX20 c.766T>C (rs3999941) as a missense (F256L) pathogenic variant and the other three variants as synonymous and benign. Compared with controls, TBX20 c.766T>C TC genotype and minor C allele were candidate high-risk factors for CCSD. Besides, serum cTnT and caspase-3 were dramatically elevated in CCSD children compared to controls. TBX20 c.766T>C TC genotype was associated with high cTnT in CCSD children. Conclusively, we advocate TBX20 c.766T>C variant as a potential genetic marker for CCSD which might associate with high cTnT levels. CITED2 genetic variants might have rare incidence among Egyptian CCSD children. Serum cTnT and caspase-3 are useful markers for ascertaining CCSD in children. These data could be exploited in prenatal genetic counseling, pre-implantation genotyping, and therapy of CCSD.
Collapse
Affiliation(s)
- Mohamed Taha
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Nourhan Awny
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Giza, Egypt
| | - Somaia Ismail
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Giza, Egypt
| | - Engy A Ashaat
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Giza, Egypt
| | - Mahmoud A Senousy
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo 11786, Egypt
| |
Collapse
|
18
|
Du C, Li Z, Zou B, Li X, Chen F, Liang Y, Luo X, Shu S. Novel heterozygous variants in the EP300 gene cause Rubinstein-Taybi syndrome 2: Reports from two Chinese children. Mol Genet Genomic Med 2023; 11:e2192. [PMID: 37162176 PMCID: PMC10496081 DOI: 10.1002/mgg3.2192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/22/2023] [Accepted: 04/25/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Rubinstein-Taybi syndrome (RSTS) is a rare autosomal-dominant genetic disease caused by variants of CREBBP (RSTS1) or EP300 (RSTS2) gene. RSTS2 is much less common, with less than 200 reported cases worldwide to date. More reports are still needed to increase the understanding of its clinical manifestations and genetic characteristics. METHODS The clinical data of two children with RSTS2 were analyzed retrospectively, and their clinical manifestations, auxiliary examinations, and mutational spectrum were summarized. Liquid chromatography-tandem mass spectrometer (LC-MS/MS) technology was used to detect the levels of steroid hormones if possible. RESULTS After analyzing the clinical and genetic characteristics of two boys with RSTS2 (0.7 and 10.4 years old, respectively) admitted in our hospital, we identified two novel heterozygous variants in the EP300 exon 22 (c.3750C > A, p. Cys1250*, pathogenic; c.1889A > G, p. Tyr630Cys, likely pathogenic), which could account for their phenotype. In addition to common clinical manifestations such as special facial features, microcephaly, growth retardation, intellectual disability, speech delay, congenital heart defect, recurrent respiratory infections, and immunodeficiency, we found one of them had a rare feature of adrenal insufficiency, and LC-MS/MS detection showed an overall decrease in steroid hormones. CONCLUSION In our study, we identified two novel variants in the EP300 exon 22, and for the first time, we reported a case of RSTS2 associated with adrenal insufficiency, which will enrich the clinical and mutational spectrum of this syndrome.
Collapse
Affiliation(s)
- Caiqi Du
- Department of Pediatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhuoguang Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of EndocrinologyShenzhen Children's HospitalShenzhenChina
| | - Biao Zou
- Department of Pediatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xuesong Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Fan Chen
- Department of Pediatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yan Liang
- Department of Pediatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Sainan Shu
- Department of Pediatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
19
|
Liu K, Xiao Y, Gan L, Li W, Zhang J, Min J. Structural basis for specific DNA sequence motif recognition by the TFAP2 transcription factors. Nucleic Acids Res 2023; 51:8270-8282. [PMID: 37409559 PMCID: PMC10450164 DOI: 10.1093/nar/gkad583] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/01/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023] Open
Abstract
The TFAP2 family regulates gene expression during differentiation, development, and organogenesis, and includes five homologs in humans. They all possess a highly conserved DNA binding domain (DBD) followed by a helix-span-helix (HSH) domain. The DBD-HSH tandem domain specifically binds to a GCC(N3)GGC consensus sequence, but the precise recognition mechanisms remain unclear. Here, we found that TFAP2 preferred binding to the GCC(N3)GGC sequence, and the pseudo-palindromic GCC and GGC motifs and the length of the central spacer between the two motifs determined their binding specificity. Structural studies revealed that the two flat amphipathic α-helical HSH domains of TFAP2A stacked with each other to form a dimer via hydrophobic interactions, while the stabilized loops from both DBD domains inserted into two neighboring major grooves of the DNA duplex to form base-specific interactions. This specific DNA binding mechanism controlled the length of the central spacer and determined the DNA sequence specificity of TFAP2. Mutations of the TFAP2 proteins are implicated in various diseases. We illustrated that reduction or disruption of the DNA binding ability of the TFAP2 proteins is the primary cause of TFAP2 mutation-associated diseases. Thus, our findings also offer valuable insights into the pathogenesis of disease-associated mutations in TFAP2 proteins.
Collapse
Affiliation(s)
- Ke Liu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, PR China
| | - Yuqing Xiao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, PR China
| | - Linyao Gan
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, PR China
| | - Weifang Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, PR China
| | - Jin Zhang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, PR China
| | - Jinrong Min
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, PR China
| |
Collapse
|
20
|
Bragança J, Pinto R, Silva B, Marques N, Leitão HS, Fernandes MT. Charting the Path: Navigating Embryonic Development to Potentially Safeguard against Congenital Heart Defects. J Pers Med 2023; 13:1263. [PMID: 37623513 PMCID: PMC10455635 DOI: 10.3390/jpm13081263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Congenital heart diseases (CHDs) are structural or functional defects present at birth due to improper heart development. Current therapeutic approaches to treating severe CHDs are primarily palliative surgical interventions during the peri- or prenatal stages, when the heart has fully developed from faulty embryogenesis. However, earlier interventions during embryonic development have the potential for better outcomes, as demonstrated by fetal cardiac interventions performed in utero, which have shown improved neonatal and prenatal survival rates, as well as reduced lifelong morbidity. Extensive research on heart development has identified key steps, cellular players, and the intricate network of signaling pathways and transcription factors governing cardiogenesis. Additionally, some reports have indicated that certain adverse genetic and environmental conditions leading to heart malformations and embryonic death may be amendable through the activation of alternative mechanisms. This review first highlights key molecular and cellular processes involved in heart development. Subsequently, it explores the potential for future therapeutic strategies, targeting early embryonic stages, to prevent CHDs, through the delivery of biomolecules or exosomes to compensate for faulty cardiogenic mechanisms. Implementing such non-surgical interventions during early gestation may offer a prophylactic approach toward reducing the occurrence and severity of CHDs.
Collapse
Affiliation(s)
- José Bragança
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Rute Pinto
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Bárbara Silva
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- PhD Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve, 8005-139 Faro, Portugal
| | - Nuno Marques
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- School of Health, University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
21
|
Wang S, Wu X, Wang H, Song S, Hu Y, Guo Y, Chang S, Cheng Y, Zeng S. Role of FBXL5 in redox homeostasis and spindle assembly during oocyte maturation in mice. FASEB J 2023; 37:e23080. [PMID: 37462473 DOI: 10.1096/fj.202300244rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/01/2023] [Accepted: 06/26/2023] [Indexed: 07/21/2023]
Abstract
As an E3 ubiquitin ligase, F-box and leucine-rich repeat protein 5 (FBXL5) participates in diverse biologic processes. However, the role of Fbxl5 in mouse oocyte meiotic maturation has not yet been fully elucidated. The present study revealed that mouse oocytes depleted of Fbxl5 were unable to complete meiosis, as Fbxl5 silencing led to oocyte meiotic failure with reduced rates of GVBD and polar body extrusion. In addition, Fbxl5 depletion induced aberrant mitochondrial dynamics as we noted the overproduction of reactive oxygen species (ROS) and the accumulation of phosphorylated γH2AX with Fbxl5 knockdown. We also found that Fbxl5-KD led to the abnormal accumulation of CITED2 proteins in mouse oocytes. Our in vitro ubiquitination assay showed that FBXL5 interacted with CITED2 and that it mediated the degradation of CITED2 protein through the ubiquitination-proteasome pathway. Collectively, our data revealed critical functions of FBXL5 in redox hemostasis and spindle assembly during mouse oocyte maturation.
Collapse
Affiliation(s)
- Shiwei Wang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xuan Wu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Han Wang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shuang Song
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yuling Hu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yajun Guo
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Siyu Chang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yuanweilu Cheng
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shenming Zeng
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
22
|
Kang Y, Laprocina K, Zheng HS, Huang CCJ. Current insight into the transient X-zone in the adrenal gland cortex. VITAMINS AND HORMONES 2023; 124:297-339. [PMID: 38408801 PMCID: PMC11023618 DOI: 10.1016/bs.vh.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Mouse models have been widely used in the study of adrenal gland development and diseases. The X-zone is a unique structure of the mouse adrenal gland and lineage-tracing studies show that the X-zone is a remnant of the fetal adrenal cortex. Although the X-zone is considered analogous to the fetal zone in the human adrenal cortex, the functional significance of the X-zone has remained comparatively more obscure. The X-zone forms during the early postnatal stages of adrenal development and regresses later in a remarkable sexually dimorphic fashion. The formation and regression of the X-zone can be different in mice with different genetic backgrounds. Mouse models with gene mutations, hormone/chemical treatments, and/or gonadectomy can also display an aberrant development of the X-zone or alternatively a dysregulated X-zone regression. These models have shed light on the molecular mechanisms regulating the development and regression of these unique adrenocortical cells. This review paper briefly describes the development of the adrenal gland including the formation and regression processes of the X-zone. It also summarizes and lists mouse models that demonstrate different X-zone phenotypes.
Collapse
Affiliation(s)
- Yuan Kang
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Karly Laprocina
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Huifei Sophia Zheng
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Chen-Che Jeff Huang
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.
| |
Collapse
|
23
|
Abstract
The adrenal cortex undergoes multiple structural and functional rearrangements to satisfy the systemic needs for steroids during fetal life, postnatal development, and adulthood. A fully functional adrenal cortex relies on the proper subdivision in regions or 'zones' with distinct but interconnected functions, which evolve from the early embryonic stages to adulthood, and rely on a fine-tuned gene network. In particular, the steroidogenic activity of the fetal adrenal is instrumental in maintaining normal fetal development and growth. Here, we review and discuss the most recent advances in our understanding of embryonic and fetal adrenal development, including the known causes for adrenal dys-/agenesis, and the steroidogenic pathways that link the fetal adrenal with the hormone system of the mother through the fetal-placental unit. Finally, we discuss what we think are the major open questions in the field, including, among others, the impact of osteocalcin, thyroid hormone, and other hormone systems on adrenal development and function, and the reliability of rodents as models of adrenal pathophysiology.
Collapse
Affiliation(s)
- Emanuele Pignatti
- Department of Pediatrics, Division of Endocrinology, Diabetology and Metabolism, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland.
- Department for BioMedical Research, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland.
| | - Therina du Toit
- Department for BioMedical Research, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland.
| | - Christa E Flück
- Department of Pediatrics, Division of Endocrinology, Diabetology and Metabolism, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland
- Department for BioMedical Research, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland
| |
Collapse
|
24
|
Lerario AM, Mohan DR, Hammer GD. Update on Biology and Genomics of Adrenocortical Carcinomas: Rationale for Emerging Therapies. Endocr Rev 2022; 43:1051-1073. [PMID: 35551369 PMCID: PMC9695111 DOI: 10.1210/endrev/bnac012] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Indexed: 11/19/2022]
Abstract
The adrenal glands are paired endocrine organs that produce steroid hormones and catecholamines required for life. Adrenocortical carcinoma (ACC) is a rare and often fatal cancer of the peripheral domain of the gland, the adrenal cortex. Recent research in adrenal development, homeostasis, and disease have refined our understanding of the cellular and molecular programs controlling cortical growth and renewal, uncovering crucial clues into how physiologic programs are hijacked in early and late stages of malignant neoplasia. Alongside these studies, genome-wide approaches to examine adrenocortical tumors have transformed our understanding of ACC biology, and revealed that ACC is composed of distinct molecular subtypes associated with favorable, intermediate, and dismal clinical outcomes. The homogeneous transcriptional and epigenetic programs prevailing in each ACC subtype suggest likely susceptibility to any of a plethora of existing and novel targeted agents, with the caveat that therapeutic response may ultimately be limited by cancer cell plasticity. Despite enormous biomedical research advances in the last decade, the only potentially curative therapy for ACC to date is primary surgical resection, and up to 75% of patients will develop metastatic disease refractory to standard-of-care adjuvant mitotane and cytotoxic chemotherapy. A comprehensive, integrated, and current bench-to-bedside understanding of our field's investigations into adrenocortical physiology and neoplasia is crucial to developing novel clinical tools and approaches to equip the one-in-a-million patient fighting this devastating disease.
Collapse
Affiliation(s)
- Antonio Marcondes Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | - Dipika R Mohan
- Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| |
Collapse
|
25
|
Transgenic Mouse Models to Study the Development and Maintenance of the Adrenal Cortex. Int J Mol Sci 2022; 23:ijms232214388. [PMID: 36430866 PMCID: PMC9693478 DOI: 10.3390/ijms232214388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
The cortex of the adrenal gland is organized into concentric zones that produce distinct steroid hormones essential for body homeostasis in mammals. Mechanisms leading to the development, zonation and maintenance of the adrenal cortex are complex and have been studied since the 1800s. However, the advent of genetic manipulation and transgenic mouse models over the past 30 years has revolutionized our understanding of these mechanisms. This review lists and details the distinct Cre recombinase mouse strains available to study the adrenal cortex, and the remarkable progress total and conditional knockout mouse models have enabled us to make in our understanding of the molecular mechanisms regulating the development and maintenance of the adrenal cortex.
Collapse
|
26
|
Chen Z, Chen HX, Hou HT, Yin XY, Yang Q, He GW. Pathophysiological Role of Variants of the Promoter Region of CITED2 Gene in Sporadic Tetralogy of Fallot Patients with Cellular Function Verification. Biomolecules 2022; 12:1644. [PMID: 36358994 PMCID: PMC9687598 DOI: 10.3390/biom12111644] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/26/2023] Open
Abstract
Tetralogy of Fallot (TOF) is a common congenital heart malformation. Genetic variants in the CITED2 coding region are known to be significantly associated with cardiac malformation, but the role of variants in the CITED2 promoter region in the development of TOF remains unclear. In this study, we investigated CITED2 promoter variants in the DNA of 605 subjects, including 312 TOF patients and 293 unrelated healthy controls, by Sanger sequencing. We identified nine CITED2 gene promoter variants (including one novel heterozygous variant). Six were found only in patients with TOF and none in the control group. The transcriptional activity of the CITED2 gene promoter in mouse cardiomyocyte (HL-1) cells was significantly altered by the six variants (p < 0.05). The results of the electrophoretic mobility change assay and JASPAR database analysis showed that these variants generated or destroyed a series of possible transcription factor binding sites, resulting in changes in the CITED2 protein expression. We conclude that CITED2 promoter variants in TOF patients affect transcriptional activity and may be involved in the occurrence and progression of TOF. These findings may provide new insights into molecular pathogenesis and potential therapeutic insights in patients with TOF.
Collapse
Affiliation(s)
- Zhuo Chen
- The Institute of Cardiovascular Diseases & Department Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin 300457, China
- School of Pharmacy, Drug Research & Development Center, Wannan Medical College, Wuhu 241002, China
| | - Huan-Xin Chen
- The Institute of Cardiovascular Diseases & Department Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Hai-Tao Hou
- The Institute of Cardiovascular Diseases & Department Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Xiu-Yun Yin
- The Institute of Cardiovascular Diseases & Department Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin 300457, China
- School of Pharmacy, Drug Research & Development Center, Wannan Medical College, Wuhu 241002, China
| | - Qin Yang
- The Institute of Cardiovascular Diseases & Department Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Guo-Wei He
- The Institute of Cardiovascular Diseases & Department Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin 300457, China
| |
Collapse
|
27
|
Lu M, Liu Y, Xian Z, Yu X, Chen J, Tan S, Zhang P, Guo Y. VEGF to CITED2 ratio predicts the collateral circulation of acute ischemic stroke. Front Neurol 2022; 13:1000992. [PMID: 36247751 PMCID: PMC9563238 DOI: 10.3389/fneur.2022.1000992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Objective The research objective was to evaluate the predicting role of the vascular endothelial growth factor to CBP/P300-interacting transactivator with Glu/Asp-rich C-terminal domain 2 Ratio (VEGF/CITED2) from peripheral blood mononuclear cells (PBMCs) in the collateral circulation of acute ischemic stroke (AIS). Methods In an observational study of patients with AIS, the western blot was applied to test the protein expression of VEGF and CITED2. Then, we calculated the VEGF/CITED2 and collected other clinical data. Binary logistic regression analysis between collateral circulation and clinical data was performed. Finally, receiver operating characteristic (ROC) curve analysis was used to explore the predictive value of VEGF/CITED2. Results A total of 67 patients with AIS were included in the study. Binary logistic regression analysis indicated the VEGF/CITED2 (OR 165.79, 95%CI 7.25–3,791.54, P = 0.001) was an independent protective factor. The ROC analyses showed an area under the ROC curve of the VEGF/CITED2 was 0.861 (95%CI 0.761–0.961). The optimal cutoff value of 1.013 for VEGF/CITED2 had a sensitivity of 89.1% and a specificity of 85.7%. Conclusion In patients with AIS, the VEGF/CITED2 was related to the establishment of collateral circulation. The VEGF/CITED2 is a potentially valuable biomarker for predicting collateral circulation. Clinical trial registration ClinicalTrials.gov, identifier: NCT05345366.
Collapse
Affiliation(s)
- Minyi Lu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuben Liu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiqiang Xian
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoyao Yu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Chen
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Sheng Tan
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peidong Zhang
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Peidong Zhang
| | - Yang Guo
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Yang Guo
| |
Collapse
|
28
|
Chen Z, Chen HX, Hou HT, Yin XY, Yang Q, Han J, He GW. Genetic Variants of CITED2 Gene Promoter in Human Atrial Septal Defects: Case-Control Study and Cellular Functional Verification. J Cardiovasc Dev Dis 2022; 9:321. [PMID: 36286273 PMCID: PMC9604052 DOI: 10.3390/jcdd9100321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Atrial septal defect (ASD) is one of the most common forms of congenital heart disease (CHD). Genetic variants in the coding region of the CITED2 gene are known to be significantly correlated with CHD, but the role of variants in the promoter region of CITED2 is unknown. We investigated variants in the promoter of the CITED2 gene in 625 subjects (332 ASD and 293 healthy controls) through Sanger sequencing. Four variants in the CITED2 gene promoter were found only in eight ASD patients with zero occurrence in the control subjects (one case of g.4078A>C(rs1165649373), one case of g.4240C>A(rs1235857801), four cases of g.4935C>T(rs111470468), two cases of g.5027C>T(rs112831934)). Cellular functional analysis showed that these four variants significantly changed the transcriptional activity of the CITED2 gene promoter in HEK-293 and HL-1 cells. Electrophoretic mobility change assay results and JASPAR database analysis demonstrated that these variants created or destroyed a series of possible transcription factor binding sites, resulting in changes in the expression of CITED2 protein. We conclude that the variants of CITED2 promoter in ASD patients affect the transcriptional activity and are likely involved in the occurrence and development of ASD. These findings provide new perspectives on the pathogenesis and potential therapeutic insights of ASD.
Collapse
Affiliation(s)
- Zhuo Chen
- School of Pharmacy, Drug Research & Development Center, Wannan Medical College, Wuhu, Anhui 241002, China and The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Huan-Xin Chen
- The Institute of Cardiovascular Diseases and Department Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University and Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Hai-Tao Hou
- The Institute of Cardiovascular Diseases and Department Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University and Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Xiu-Yun Yin
- School of Pharmacy, Drug Research & Development Center, Wannan Medical College, Wuhu, Anhui 241002, China and The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Qin Yang
- The Institute of Cardiovascular Diseases and Department Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University and Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Jun Han
- School of Pharmacy, Drug Research & Development Center, Wannan Medical College, Wuhu, Anhui 241002, China
| | - Guo-Wei He
- School of Pharmacy, Drug Research & Development Center, Wannan Medical College, Wuhu, Anhui 241002, China and The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin 300457, China
- The Institute of Cardiovascular Diseases and Department Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University and Chinese Academy of Medical Sciences, Tianjin 300457, China
| |
Collapse
|
29
|
Lin JM, Mitchell TA, Rothstein M, Pehl A, Taroc EZM, Katreddi RR, Parra KE, Zuloaga DG, Simoes-Costa M, Forni PE. Sociosexual behavior requires both activating and repressive roles of Tfap2e/AP-2ε in vomeronasal sensory neurons. eLife 2022; 11:e77259. [PMID: 36111787 PMCID: PMC9525060 DOI: 10.7554/elife.77259] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Neuronal identity dictates the position in an epithelium, and the ability to detect, process, and transmit specific signals to specified targets. Transcription factors (TFs) determine cellular identity via direct modulation of genetic transcription and recruiting chromatin modifiers. However, our understanding of the mechanisms that define neuronal identity and their magnitude remain a critical barrier to elucidate the etiology of congenital and neurodegenerative disorders. The rodent vomeronasal organ provides a unique system to examine in detail the molecular mechanisms underlying the differentiation and maturation of chemosensory neurons. Here, we demonstrated that the identity of postmitotic/maturing vomeronasal sensory neurons (VSNs), and vomeronasal-dependent behaviors can be reprogrammed through the rescue of Tfap2e/AP-2ε expression in the Tfap2eNull mice, and partially reprogrammed by inducing ectopic Tfap2e expression in mature apical VSNs. We suggest that the TF Tfap2e can reprogram VSNs bypassing cellular plasticity restrictions, and that it directly controls the expression of batteries of vomeronasal genes.
Collapse
Affiliation(s)
- Jennifer M Lin
- Department of Biological Sciences, University at Albany, State University of New YorkAlbanyUnited States
- The RNA Institute, University at AlbanyAlbanyUnited States
| | - Tyler A Mitchell
- Department of Biological Sciences, University at Albany, State University of New YorkAlbanyUnited States
- The RNA Institute, University at AlbanyAlbanyUnited States
| | - Megan Rothstein
- Department of Molecular Biology and Genetics, Cornell UniversityIthacaUnited States
| | - Alison Pehl
- Department of Biological Sciences, University at Albany, State University of New YorkAlbanyUnited States
- The RNA Institute, University at AlbanyAlbanyUnited States
| | - Ed Zandro M Taroc
- Department of Biological Sciences, University at Albany, State University of New YorkAlbanyUnited States
- The RNA Institute, University at AlbanyAlbanyUnited States
| | - Raghu R Katreddi
- Department of Biological Sciences, University at Albany, State University of New YorkAlbanyUnited States
- The RNA Institute, University at AlbanyAlbanyUnited States
| | - Katherine E Parra
- Department of Psychology, University at Albany, State University of New YorkAlbanyUnited States
| | - Damian G Zuloaga
- Department of Psychology, University at Albany, State University of New YorkAlbanyUnited States
| | - Marcos Simoes-Costa
- Department of Molecular Biology and Genetics, Cornell UniversityIthacaUnited States
| | - Paolo Emanuele Forni
- Department of Biological Sciences, University at Albany, State University of New YorkAlbanyUnited States
- The RNA Institute, University at AlbanyAlbanyUnited States
| |
Collapse
|
30
|
Cui G, Gao Z, Chang S, Narwade N, Chen Y, Poudel B, Lei KMK, Zhang W, Li G, Poon TCW, Cheung E. TRIM37 Augments AP-2γ Transcriptional Activity and Cellular Localization via K63-linked Ubiquitination to Drive Breast Cancer Progression. Int J Biol Sci 2022; 18:4316-4328. [PMID: 35864973 PMCID: PMC9295074 DOI: 10.7150/ijbs.69466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/09/2022] [Indexed: 11/26/2022] Open
Abstract
Activator Protein 2 gamma (AP-2γ) is a master transcription factor that plays a critical role in the development and progression of breast cancer. However, the underlying mechanism is still unclear. Herein, using a proteomics approach, we identified Tripartite motif-containing 37 (TRIM37) as a novel coactivator of AP-2γ-mediated transcription in breast cancer cells. We demonstrate that TRIM37 facilitates AP-2γ chromatin binding to directly regulate the AP-2γ mediated transcriptional program. We also show that TRIM37 achieves this by stimulating K63 chain-linked ubiquitination of AP-2γ, promoting protein localization from the cytoplasm to the nucleus. In clinical analyses, we find TRIM37 is upregulated in multiple breast cancer datasets, supporting our findings that the TRIM37-AP-2γ interaction is essential for breast cancer tumor growth. Overall, our work reveals that TRIM37 is an oncogenic coactivator of AP-2γ in breast cancer and provides a novel therapeutic target for treating the disease.
Collapse
Affiliation(s)
- Guimei Cui
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Zhuoran Gao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Shiehong Chang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Nitin Narwade
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Yitian Chen
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Barun Poudel
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Kate M K Lei
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.,Pilot Laboratory, University of Macau, Macau SAR, China.,Institute of Translational Medicine, University of Macau, Macau SAR, China.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Weibo Zhang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Gang Li
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Terence C W Poon
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.,Pilot Laboratory, University of Macau, Macau SAR, China.,Institute of Translational Medicine, University of Macau, Macau SAR, China.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Edwin Cheung
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| |
Collapse
|
31
|
Kenny C, Dilshat R, Seberg HE, Van Otterloo E, Bonde G, Helverson A, Franke CM, Steingrímsson E, Cornell RA. TFAP2 paralogs facilitate chromatin access for MITF at pigmentation and cell proliferation genes. PLoS Genet 2022; 18:e1010207. [PMID: 35580127 PMCID: PMC9159589 DOI: 10.1371/journal.pgen.1010207] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 06/01/2022] [Accepted: 04/19/2022] [Indexed: 12/13/2022] Open
Abstract
In developing melanocytes and in melanoma cells, multiple paralogs of the Activating-enhancer-binding Protein 2 family of transcription factors (TFAP2) contribute to expression of genes encoding pigmentation regulators, but their interaction with Microphthalmia transcription factor (MITF), a master regulator of these cells, is unclear. Supporting the model that TFAP2 facilitates MITF's ability to activate expression of pigmentation genes, single-cell seq analysis of zebrafish embryos revealed that pigmentation genes are only expressed in the subset of mitfa-expressing cells that also express tfap2 paralogs. To test this model in SK-MEL-28 melanoma cells we deleted the two TFAP2 paralogs with highest expression, TFAP2A and TFAP2C, creating TFAP2 knockout (TFAP2-KO) cells. We then assessed gene expression, chromatin accessibility, binding of TFAP2A and of MITF, and the chromatin marks H3K27Ac and H3K27Me3 which are characteristic of active enhancers and silenced chromatin, respectively. Integrated analyses of these datasets indicate TFAP2 paralogs directly activate enhancers near genes enriched for roles in pigmentation and proliferation, and directly repress enhancers near genes enriched for roles in cell adhesion. Consistently, compared to WT cells, TFAP2-KO cells proliferate less and adhere to one another more. TFAP2 paralogs and MITF co-operatively activate a subset of enhancers, with the former necessary for MITF binding and chromatin accessibility. By contrast, TFAP2 paralogs and MITF do not appear to co-operatively inhibit enhancers. These studies reveal a mechanism by which TFAP2 profoundly influences the set of genes activated by MITF, and thereby the phenotype of pigment cells and melanoma cells.
Collapse
Affiliation(s)
- Colin Kenny
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Ramile Dilshat
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Hannah E. Seberg
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Eric Van Otterloo
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Gregory Bonde
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Annika Helverson
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Christopher M. Franke
- Department of Surgery, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Eiríkur Steingrímsson
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Robert A. Cornell
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
32
|
Enomoto Y, Yokoi T, Tsurusaki Y, Murakami H, Tominaga M, Minatogawa M, Abe-Hatano C, Kuroda Y, Ohashi I, Ida K, Shiiya S, Kumaki T, Naruto T, Mitsui J, Harada N, Kido Y, Kurosawa K. Divergent variant patterns among 19 patients with Rubinstein-Taybi syndrome uncovered by comprehensive genetic analysis including whole genome sequencing. Clin Genet 2021; 101:335-345. [PMID: 34958122 DOI: 10.1111/cge.14103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 12/01/2022]
Abstract
Rubinstein-Taybi syndrome (RSTS) is characterized by dysmorphic facial features, broad thumbs, and intellectual disability. CREBBP or EP300 are causative genes. To elucidate the underlying genetic and genomic architecture related to the RSTS phenotype, we performed comprehensive genetic analysis targeting CREBBP and/or EP300 in 22 clinically diagnosed patients. During the 11-year study period, we used several analysis methods including high resolution melting, array-based comparative genomic hybridization, panel-based exome sequencing, whole exome sequencing, and whole genome sequencing (WGS). We identified the causative variants in 19 patients (86.3%), but they were variable and complex, so we must combine multiple analysis methods. Notably, we found genetic alterations in the non-coding regions of two patients (10.5%, 2/19): scattered deletions including a partial 5'-untranslated region of CREBBP in one patient (all coding exons were intact), and a deep 229-bp intronic deletion in another patient, resulting in a splicing error. Furthermore, we identified rare clinical findings: two patients with an EP300 variant showed abnormal development of the neural tube, and one patient with a CREBBP variant had anorectal atresia with a cloaca. Our findings expand the allelic heterogeneity of RSTS, underscore the utility of comprehensive genetic analysis, and suggest that WGS may be a practical diagnostic strategy.
Collapse
Affiliation(s)
- Yumi Enomoto
- Clinical Research Institute, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Takayuki Yokoi
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yoshinori Tsurusaki
- Clinical Research Institute, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hiroaki Murakami
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Makiko Tominaga
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Mari Minatogawa
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Chihiro Abe-Hatano
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yukiko Kuroda
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Ikuko Ohashi
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Kazumi Ida
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Shizuka Shiiya
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Tatsuro Kumaki
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Takuya Naruto
- Clinical Research Institute, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Jun Mitsui
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noriaki Harada
- Department of Clinical Laboratory, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yasuhiro Kido
- Department of Pediatrics, Saitama Medical Center, Dokkyo Medical University, Koshigaya, Japan
| | - Kenji Kurosawa
- Clinical Research Institute, Kanagawa Children's Medical Center, Yokohama, Japan.,Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| |
Collapse
|
33
|
Lawson H, van de Lagemaat LN, Barile M, Tavosanis A, Durko J, Villacreces A, Bellani A, Mapperley C, Georges E, Martins-Costa C, Sepulveda C, Allen L, Campos J, Campbell KJ, O'Carroll D, Göttgens B, Cory S, Rodrigues NP, Guitart AV, Kranc KR. CITED2 coordinates key hematopoietic regulatory pathways to maintain the HSC pool in both steady-state hematopoiesis and transplantation. Stem Cell Reports 2021; 16:2784-2797. [PMID: 34715054 PMCID: PMC8581166 DOI: 10.1016/j.stemcr.2021.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 10/26/2022] Open
Abstract
Hematopoietic stem cells (HSCs) reside at the apex of the hematopoietic differentiation hierarchy and sustain multilineage hematopoiesis. Here, we show that the transcriptional regulator CITED2 is essential for life-long HSC maintenance. While hematopoietic-specific Cited2 deletion has a minor impact on steady-state hematopoiesis, Cited2-deficient HSCs are severely depleted in young mice and fail to expand upon aging. Moreover, although they home normally to the bone marrow, they fail to reconstitute hematopoiesis upon transplantation. Mechanistically, CITED2 is required for expression of key HSC regulators, including GATA2, MCL-1, and PTEN. Hematopoietic-specific expression of anti-apoptotic MCL-1 partially rescues the Cited2-deficient HSC pool and restores their reconstitution potential. To interrogate the Cited2→Pten pathway in HSCs, we generated Cited2;Pten compound heterozygous mice, which had a decreased number of HSCs that failed to reconstitute the HSC compartment. In addition, CITED2 represses multiple pathways whose elevated activity causes HSC exhaustion. Thus, CITED2 promotes pathways necessary for HSC maintenance and suppresses those detrimental to HSC integrity.
Collapse
Affiliation(s)
- Hannah Lawson
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Louie N van de Lagemaat
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Melania Barile
- Department of Haematology, Wellcome and Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0AW, UK
| | - Andrea Tavosanis
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jozef Durko
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Arnaud Villacreces
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Aarushi Bellani
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Christopher Mapperley
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Elise Georges
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | | | - Catarina Sepulveda
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Lewis Allen
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Joana Campos
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | | | - Dónal O'Carroll
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Berthold Göttgens
- Department of Haematology, Wellcome and Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0AW, UK
| | - Suzanne Cory
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Neil P Rodrigues
- European Cancer Stem Cell Research Institute, Cardiff University, School of Biosciences, Cardiff CF24 4HQ, UK
| | - Amelie V Guitart
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK; Université de Bordeaux, Institut National de la Santé et de la Recherche Médicale INSERM U1035, 33000 Bordeaux, France.
| | - Kamil R Kranc
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| |
Collapse
|
34
|
Khasawneh RR, Kist R, Queen R, Hussain R, Coxhead J, Schneider JE, Mohun TJ, Zaffran S, Peters H, Phillips HM, Bamforth SD. Msx1 haploinsufficiency modifies the Pax9-deficient cardiovascular phenotype. BMC DEVELOPMENTAL BIOLOGY 2021; 21:14. [PMID: 34615475 PMCID: PMC8493722 DOI: 10.1186/s12861-021-00245-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/23/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND Successful embryogenesis relies on the coordinated interaction between genes and tissues. The transcription factors Pax9 and Msx1 genetically interact during mouse craniofacial morphogenesis, and mice deficient for either gene display abnormal tooth and palate development. Pax9 is expressed specifically in the pharyngeal endoderm at mid-embryogenesis, and mice deficient for Pax9 on a C57Bl/6 genetic background also have cardiovascular defects affecting the outflow tract and aortic arch arteries giving double-outlet right ventricle, absent common carotid arteries and interruption of the aortic arch. RESULTS In this study we have investigated both the effect of a different genetic background and Msx1 haploinsufficiency on the presentation of the Pax9-deficient cardiovascular phenotype. Compared to mice on a C57Bl/6 background, congenic CD1-Pax9-/- mice displayed a significantly reduced incidence of outflow tract defects but aortic arch defects were unchanged. Pax9-/- mice with Msx1 haploinsufficiency, however, have a reduced incidence of interrupted aortic arch, but more cases with cervical origins of the right subclavian artery and aortic arch, than seen in Pax9-/- mice. This alteration in arch artery defects was accompanied by a rescue in third pharyngeal arch neural crest cell migration and smooth muscle cell coverage of the third pharyngeal arch arteries. Although this change in phenotype could theoretically be compatible with post-natal survival, using tissue-specific inactivation of Pax9 to maintain correct palate development whilst inducing the cardiovascular defects was unable to prevent postnatal death in the mutant mice. Hyoid bone and thyroid cartilage formation were abnormal in Pax9-/- mice. CONCLUSIONS Msx1 haploinsufficiency mitigates the arch artery defects in Pax9-/- mice, potentially by maintaining the survival of the 3rd arch artery through unimpaired migration of neural crest cells to the third pharyngeal arches. With the neural crest cell derived hyoid bone and thyroid cartilage also being defective in Pax9-/- mice, we speculate that the pharyngeal endoderm is a key signalling centre that impacts on neural crest cell behaviour highlighting the ability of cells in different tissues to act synergistically or antagonistically during embryo development.
Collapse
Affiliation(s)
- Ramada R. Khasawneh
- grid.419328.50000 0000 9225 6820Newcastle University Biosciences Institute, Centre for Life, Newcastle, NE1 3BZ UK ,grid.14440.350000 0004 0622 5497Present Address: Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Ralf Kist
- grid.419328.50000 0000 9225 6820Newcastle University Biosciences Institute, Centre for Life, Newcastle, NE1 3BZ UK ,grid.1006.70000 0001 0462 7212School of Dental Sciences, Newcastle University, Newcastle, NE2 4BW UK
| | - Rachel Queen
- grid.1006.70000 0001 0462 7212Bioinformatics Support Unit, Newcastle University, Newcastle, NE1 3BZ UK
| | - Rafiqul Hussain
- grid.1006.70000 0001 0462 7212Genomics Core Facility, Newcastle University, Newcastle, NE1 3BZ UK
| | - Jonathan Coxhead
- grid.1006.70000 0001 0462 7212Genomics Core Facility, Newcastle University, Newcastle, NE1 3BZ UK
| | - Jürgen E. Schneider
- grid.9909.90000 0004 1936 8403Biomedical Imaging, University of Leeds, Leeds, LS2 9JT UK
| | - Timothy J. Mohun
- grid.451388.30000 0004 1795 1830The Francis Crick Institute, London, NW1 1AT UK
| | - Stéphane Zaffran
- grid.5399.60000 0001 2176 4817INSERM, Marseille Medical Genetics, U1251, Aix Marseille University, Marseille, France
| | - Heiko Peters
- grid.419328.50000 0000 9225 6820Newcastle University Biosciences Institute, Centre for Life, Newcastle, NE1 3BZ UK
| | - Helen M. Phillips
- grid.419328.50000 0000 9225 6820Newcastle University Biosciences Institute, Centre for Life, Newcastle, NE1 3BZ UK
| | - Simon D. Bamforth
- grid.419328.50000 0000 9225 6820Newcastle University Biosciences Institute, Centre for Life, Newcastle, NE1 3BZ UK
| |
Collapse
|
35
|
Knutson AK, Williams AL, Boisvert WA, Shohet RV. HIF in the heart: development, metabolism, ischemia, and atherosclerosis. J Clin Invest 2021; 131:137557. [PMID: 34623330 DOI: 10.1172/jci137557] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The heart forms early in development and delivers oxygenated blood to the rest of the embryo. After birth, the heart requires kilograms of ATP each day to support contractility for the circulation. Cardiac metabolism is omnivorous, utilizing multiple substrates and metabolic pathways to produce this energy. Cardiac development, metabolic tuning, and the response to ischemia are all regulated in part by the hypoxia-inducible factors (HIFs), central components of essential signaling pathways that respond to hypoxia. Here we review the actions of HIF1, HIF2, and HIF3 in the heart, from their roles in development and metabolism to their activity in regeneration and preconditioning strategies. We also discuss recent work on the role of HIFs in atherosclerosis, the precipitating cause of myocardial ischemia and the leading cause of death in the developed world.
Collapse
|
36
|
Pignatti E, Flück CE. Adrenal cortex development and related disorders leading to adrenal insufficiency. Mol Cell Endocrinol 2021; 527:111206. [PMID: 33607267 DOI: 10.1016/j.mce.2021.111206] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023]
Abstract
The adult human adrenal cortex produces steroid hormones that are crucial for life, supporting immune response, glucose homeostasis, salt balance and sexual maturation. It consists of three histologically distinct and functionally specialized zones. The fetal adrenal forms from mesodermal material and produces predominantly adrenal C19 steroids from its fetal zone, which involutes after birth. Transition to the adult cortex occurs immediately after birth for the formation of the zona glomerulosa and fasciculata for aldosterone and cortisol production and continues through infancy until the zona reticularis for adrenal androgen production is formed with adrenarche. The development of this indispensable organ is complex and not fully understood. This article gives an overview of recent knowledge gained of adrenal biology from two perspectives: one, from basic science studying adrenal development, zonation and homeostasis; and two, from adrenal disorders identified in persons manifesting with various isolated or syndromic forms of primary adrenal insufficiency.
Collapse
Affiliation(s)
- Emanuele Pignatti
- Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern and Department of BioMedical Research, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland.
| | - Christa E Flück
- Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern and Department of BioMedical Research, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland.
| |
Collapse
|
37
|
The role of DNA methylation in syndromic and non-syndromic congenital heart disease. Clin Epigenetics 2021; 13:93. [PMID: 33902696 PMCID: PMC8077695 DOI: 10.1186/s13148-021-01077-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023] Open
Abstract
Congenital heart disease (CHD) is a common structural birth defect worldwide, and defects typically occur in the walls and valves of the heart or enlarged blood vessels. Chromosomal abnormalities and genetic mutations only account for a small portion of the pathogenic mechanisms of CHD, and the etiology of most cases remains unknown. The role of epigenetics in various diseases, including CHD, has attracted increased attention. The contributions of DNA methylation, one of the most important epigenetic modifications, to CHD have not been illuminated. Increasing evidence suggests that aberrant DNA methylation is related to CHD. Here, we briefly introduce DNA methylation and CHD and then review the DNA methylation profiles during cardiac development and in CHD, abnormalities in maternal genome-wide DNA methylation patterns are also described. Whole genome methylation profile and important differentially methylated genes identified in recent years are summarized and clustered according to the sample type and methodologies. Finally, we discuss the novel technology for and prospects of CHD-related DNA methylation.
Collapse
|
38
|
Majumdar U, Yasuhara J, Garg V. In Vivo and In Vitro Genetic Models of Congenital Heart Disease. Cold Spring Harb Perspect Biol 2021; 13:cshperspect.a036764. [PMID: 31818859 DOI: 10.1101/cshperspect.a036764] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Congenital cardiovascular malformations represent the most common type of birth defect and encompass a spectrum of anomalies that range from mild to severe. The etiology of congenital heart disease (CHD) is becoming increasingly defined based on prior epidemiologic studies that supported the importance of genetic contributors and technological advances in human genome analysis. These have led to the discovery of a growing number of disease-contributing genetic abnormalities in individuals affected by CHD. The ever-growing population of adult CHD survivors, which are the result of reductions in mortality from CHD during childhood, and this newfound genetic knowledge have led to important questions regarding recurrence risks, the mechanisms by which these defects occur, the potential for novel approaches for prevention, and the prediction of long-term cardiovascular morbidity in adult CHD survivors. Here, we will review the current status of genetic models that accurately model human CHD as they provide an important tool to answer these questions and test novel therapeutic strategies.
Collapse
Affiliation(s)
- Uddalak Majumdar
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, Ohio 43205, USA
| | - Jun Yasuhara
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, Ohio 43205, USA
| | - Vidu Garg
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio 43205, USA.,Department of Molecular Genetics, The Ohio State University, Columbus, Ohio 43205, USA
| |
Collapse
|
39
|
A gain-of-function mutation in CITED2 is associated with congenital heart disease. Mutat Res 2021; 822:111741. [PMID: 33706167 DOI: 10.1016/j.mrfmmm.2021.111741] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/26/2021] [Indexed: 12/20/2022]
Abstract
CITED2 is a transcription co-activator that interacts with TFAP2 and CBP/ P300 transcription factors to regulate the proliferation and differentiation of the cardiac progenitor cells. It acts upstream to NODAL-PITX2 pathways and regulates the left-right asymmetry. Both human genetic and model organism studies have shown that altered expression of CITED2 causes various forms of congenital heart disease. Therefore, we sought to screen the coding region of CITED2 to identify rare genetic variants and assess their impact on the structure and function of the protein. Here, we have screened 271 non-syndromic, sporadic CHD cases by Sanger's sequencing method and detected a non-synonymous variant (c.301C>T, p.P101S) and two synonymous variants (c.21C>A, p.A7A; c.627C>G, p.P209P). The non-synonymous variant c.301C>T (rs201639244) is a rare variant with a minor allele frequency of 0.00011 in the gnomAD browser and 0.0018 in the present study. in vitro analysis has demonstrated that p.P101S mutation upregulates the expression of downstream target genes Gata4, Mef2c, Nfatc1&2, Nodal, Pitx2, and Tbx5 in P19 cells. Luciferase reporter assay also demonstrates enhanced activation of downstream target promoters. Further, in silico analyses implicate that increased activity of mutant CITED2 is possibly due to phosphorylation of Serine residue by proline-directed kinases. Homology modeling and alignment analysis have also depicted differences in hydrogen bonding and tertiary structures of wild-type versus mutant protein. The impact of synonymous variations on the mRNA structure of CITED2has been analyzed by Mfold and relative codon bias calculations. Mfold results have revealed that both the synonymous variants can alter the mRNA structure and stability. Relative codon usage analysis has suggested that the rate of translation is attenuated due to these variations. Altogether, our results from genetic screening as well as in vitro and in silico studies support a possible role of nonsynonymous and synonymous mutations in CITED2contributing to pathogenesis of CHD.
Collapse
|
40
|
Tsukasaki M, Huynh NCN, Okamoto K, Muro R, Terashima A, Kurikawa Y, Komatsu N, Pluemsakunthai W, Nitta T, Abe T, Kiyonari H, Okamura T, Sakai M, Matsukawa T, Matsumoto M, Kobayashi Y, Penninger JM, Takayanagi H. Stepwise cell fate decision pathways during osteoclastogenesis at single-cell resolution. Nat Metab 2020; 2:1382-1390. [PMID: 33288951 DOI: 10.1038/s42255-020-00318-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 11/04/2020] [Indexed: 12/16/2022]
Abstract
Osteoclasts are the exclusive bone-resorbing cells, playing a central role in bone metabolism, as well as the bone damage that occurs under pathological conditions1,2. In postnatal life, haematopoietic stem-cell-derived precursors give rise to osteoclasts in response to stimulation with macrophage colony-stimulating factor and receptor activator of nuclear factor-κB ligand, both of which are produced by osteoclastogenesis-supporting cells such as osteoblasts and osteocytes1-3. However, the precise mechanisms underlying cell fate specification during osteoclast differentiation remain unclear. Here, we report the transcriptional profiling of 7,228 murine cells undergoing in vitro osteoclastogenesis, describing the stepwise events that take place during the osteoclast fate decision process. Based on our single-cell transcriptomic dataset, we find that osteoclast precursor cells transiently express CD11c, and deletion of receptor activator of nuclear factor-κB specifically in CD11c-expressing cells inhibited osteoclast formation in vivo and in vitro. Furthermore, we identify Cbp/p300-interacting transactivator with Glu/Asp-rich carboxy-terminal domain 2 (Cited2) as the molecular switch triggering terminal differentiation of osteoclasts, and deletion of Cited2 in osteoclast precursors in vivo resulted in a failure to commit to osteoclast fate. Together, the results of this study provide a detailed molecular road map of the osteoclast differentiation process, refining and expanding our understanding of the molecular mechanisms underlying osteoclastogenesis.
Collapse
Affiliation(s)
- Masayuki Tsukasaki
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nam Cong-Nhat Huynh
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryunosuke Muro
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Asuka Terashima
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshitaka Kurikawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noriko Komatsu
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Warunee Pluemsakunthai
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Nitta
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mashito Sakai
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Toshiya Matsukawa
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Michihiro Matsumoto
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yasuhiro Kobayashi
- Division of Hard Tissue Research, Institute for Oral Science, Matsumoto Dental University, Shiojiri, Japan
| | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
- Life Science Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
41
|
Jing F, Zhang SW, Zhang S. Prediction of enhancer-promoter interactions using the cross-cell type information and domain adversarial neural network. BMC Bioinformatics 2020; 21:507. [PMID: 33160328 PMCID: PMC7648314 DOI: 10.1186/s12859-020-03844-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/27/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Enhancer-promoter interactions (EPIs) play key roles in transcriptional regulation and disease progression. Although several computational methods have been developed to predict such interactions, their performances are not satisfactory when training and testing data from different cell lines. Currently, it is still unclear what extent a across cell line prediction can be made based on sequence-level information. RESULTS In this work, we present a novel Sequence-based method (called SEPT) to predict the enhancer-promoter interactions in new cell line by using the cross-cell information and Transfer learning. SEPT first learns the features of enhancer and promoter from DNA sequences with convolutional neural network (CNN), then designing the gradient reversal layer of transfer learning to reduce the cell line specific features meanwhile retaining the features associated with EPIs. When the locations of enhancers and promoters are provided in new cell line, SEPT can successfully recognize EPIs in this new cell line based on labeled data of other cell lines. The experiment results show that SEPT can effectively learn the latent import EPIs-related features between cell lines and achieves the best prediction performance in terms of AUC (the area under the receiver operating curves). CONCLUSIONS SEPT is an effective method for predicting the EPIs in new cell line. Domain adversarial architecture of transfer learning used in SEPT can learn the latent EPIs shared features among cell lines from all other existing labeled data. It can be expected that SEPT will be of interest to researchers concerned with biological interaction prediction.
Collapse
Affiliation(s)
- Fang Jing
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, 127 West Youyi Road, Xi’an, 710072 Shaanxi China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, 127 West Youyi Road, Xi’an, 710072 Shaanxi China
| | - Shihua Zhang
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 55 Zhongguancun East Road, Beijing, 10090 China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, 100049 China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223 China
| |
Collapse
|
42
|
Frei T, Cella F, Tedeschi F, Gutiérrez J, Stan GB, Khammash M, Siciliano V. Characterization and mitigation of gene expression burden in mammalian cells. Nat Commun 2020; 11:4641. [PMID: 32934213 PMCID: PMC7492461 DOI: 10.1038/s41467-020-18392-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/18/2020] [Indexed: 12/19/2022] Open
Abstract
Despite recent advances in circuit engineering, the design of genetic networks in mammalian cells is still painstakingly slow and fraught with inexplicable failures. Here, we demonstrate that transiently expressed genes in mammalian cells compete for limited transcriptional and translational resources. This competition results in the coupling of otherwise independent exogenous and endogenous genes, creating a divergence between intended and actual function. Guided by a resource-aware mathematical model, we identify and engineer natural and synthetic miRNA-based incoherent feedforward loop (iFFL) circuits that mitigate gene expression burden. The implementation of these circuits features the use of endogenous miRNAs as elementary components of the engineered iFFL device, a versatile hybrid design that allows burden mitigation to be achieved across different cell-lines with minimal resource requirements. This study establishes the foundations for context-aware prediction and improvement of in vivo synthetic circuit performance, paving the way towards more rational synthetic construct design in mammalian cells.
Collapse
Affiliation(s)
- Timothy Frei
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Federica Cella
- Istituto Italiano di Tecnologia-IIT, Largo Barsanti e Matteucci, Naples, 80125, Italy
- University of Genoa, Genoa, 16132, Italy
| | - Fabiana Tedeschi
- Istituto Italiano di Tecnologia-IIT, Largo Barsanti e Matteucci, Naples, 80125, Italy
| | - Joaquín Gutiérrez
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Guy-Bart Stan
- Department of Bioengineering and Centre for Synthetic Biology, Imperial College London, London, SW7 2AZ, UK
| | - Mustafa Khammash
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland.
| | - Velia Siciliano
- Istituto Italiano di Tecnologia-IIT, Largo Barsanti e Matteucci, Naples, 80125, Italy.
| |
Collapse
|
43
|
Abstract
Cbp/P300 interacting transactivator with Glu/Asp-rich carboxy-terminal domain 2 (CITED2) is a transcription co-factor that interacts with several other transcription factors and co-factors, and serves critical roles in fundamental cell processes, including proliferation, apoptosis, differentiation, migration and autophagy. The interacting transcription factors or co-factors of CITED2 include LIM homeobox 2, transcription factor AP-2, SMAD2/3, peroxisome proliferator-activated receptor γ, oestrogen receptor, MYC, Nucleolin and p300/CBP, which regulate downstream gene expression, and serve important roles in the aforementioned fundamental cell processes. Emerging evidence has demonstrated that CITED2 serves an essential role in embryonic and adult tissue stem cells, including hematopoietic stem cells and tendon-derived stem/progenitor cells. Additionally, CITED2 has been reported to function in different types of cancer. Although the functions of CITED2 in different tissues vary depending on the interaction partner, altered CITED2 expression or altered interactions with transcription factors or co-factors result in alterations of fundamental cell processes, and may affect stem cell maintenance or cancer cell survival. The aim of this review is to summarize the molecular mechanisms of CITED2 function and how it serves a role in stem cells and different types of cancer based on the currently available literature.
Collapse
|
44
|
Johnson AL, Schneider JE, Mohun TJ, Williams T, Bhattacharya S, Henderson DJ, Phillips HM, Bamforth SD. Early Embryonic Expression of AP-2α Is Critical for Cardiovascular Development. J Cardiovasc Dev Dis 2020; 7:jcdd7030027. [PMID: 32717817 PMCID: PMC7570199 DOI: 10.3390/jcdd7030027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 12/17/2022] Open
Abstract
Congenital cardiovascular malformation is a common birth defect incorporating abnormalities of the outflow tract and aortic arch arteries, and mice deficient in the transcription factor AP-2α (Tcfap2a) present with complex defects affecting these structures. AP-2α is expressed in the pharyngeal surface ectoderm and neural crest at mid-embryogenesis in the mouse, but the precise tissue compartment in which AP-2α is required for cardiovascular development has not been identified. In this study we describe the fully penetrant AP-2α deficient cardiovascular phenotype on a C57Bl/6J genetic background and show that this is associated with increased apoptosis in the pharyngeal ectoderm. Neural crest cell migration into the pharyngeal arches was not affected. Cre-expressing transgenic mice were used in conjunction with an AP-2α conditional allele to examine the effect of deleting AP-2α from the pharyngeal surface ectoderm and the neural crest, either individually or in combination, as well as the second heart field. This, surprisingly, was unable to fully recapitulate the global AP-2α deficient cardiovascular phenotype. The outflow tract and arch artery phenotype was, however, recapitulated through early embryonic Cre-mediated recombination. These findings indicate that AP-2α has a complex influence on cardiovascular development either being required very early in embryogenesis and/or having a redundant function in many tissue layers.
Collapse
Affiliation(s)
- Amy-Leigh Johnson
- Newcastle University Biosciences Institute, Centre for Life, Newcastle NE1 3BZ, UK; (A.-L.J.); (D.J.H.); (H.M.P.)
| | | | | | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado Anshutz Medical Campus, Aurora, CO 80045, USA;
| | - Shoumo Bhattacharya
- Department of Cardiovascular Medicine, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford OX3 7BN, UK;
| | - Deborah J. Henderson
- Newcastle University Biosciences Institute, Centre for Life, Newcastle NE1 3BZ, UK; (A.-L.J.); (D.J.H.); (H.M.P.)
| | - Helen M. Phillips
- Newcastle University Biosciences Institute, Centre for Life, Newcastle NE1 3BZ, UK; (A.-L.J.); (D.J.H.); (H.M.P.)
| | - Simon D. Bamforth
- Newcastle University Biosciences Institute, Centre for Life, Newcastle NE1 3BZ, UK; (A.-L.J.); (D.J.H.); (H.M.P.)
- Correspondence: ; Tel.: +44-191-241-8764
| |
Collapse
|
45
|
Pong Ng H, Kim GD, Ricky Chan E, Dunwoodie SL, Mahabeleshwar GH. CITED2 limits pathogenic inflammatory gene programs in myeloid cells. FASEB J 2020; 34:12100-12113. [PMID: 32697413 PMCID: PMC7496281 DOI: 10.1096/fj.202000864r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/18/2020] [Accepted: 06/29/2020] [Indexed: 12/24/2022]
Abstract
Monocyte‐derived macrophages are the major innate immune cells that provide the first line of cellular defense against infections or injuries. These recruited macrophages at the site of inflammation are exposed to a broad range of cytokines that categorically incite a robust pro‐inflammatory response. However, macrophage pro‐inflammatory activation must be under exquisite control to avert unbridled inflammation. Thus, endogenous mechanisms must exist that rigorously preserve macrophage quiescence and yet, allow nimble pro‐inflammatory macrophage response with precise spatiotemporal control. Herein, we identify the CBP/p300‐interacting transactivator with glutamic acid/aspartic acid‐rich carboxyl‐terminal domain 2 (CITED2) as a critical intrinsic negative regulator of inflammation, which broadly attenuates pro‐inflammatory gene programs in macrophages. Our in vivo studies revealed that myeloid‐CITED2 deficiency significantly heightened macrophages and neutrophils recruitment to the site of inflammation. Our integrated transcriptomics and gene set enrichment analysis (GSEA) studies uncovered that CITED2 deficiency broadly enhances NFκB targets, IFNγ/IFNα responses, and inflammatory response gene expression in macrophages. Using complementary gain‐ and loss‐of‐function studies, we observed that CITED2 overexpression attenuate and CITED2 deficiency elevate LPS‐induced NFκB transcriptional activity and NFκB‐p65 recruitment to target gene promoter in macrophages. More importantly, blockade of NFκB signaling completely reversed elevated pro‐inflammatory gene expression in macrophages. Collectively, our findings show that CITED2 restrains NFκB activation and curtails broad pro‐inflammatory gene programs in myeloid cells.
Collapse
Affiliation(s)
- Hang Pong Ng
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Gun-Dong Kim
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sally L Dunwoodie
- Victor Chang Cardiac Research Institute, Sydney, Australia.,UNSW Sydney, Sydney, Australia
| | - Ganapati H Mahabeleshwar
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
46
|
Fernandes MT, Calado SM, Mendes-Silva L, Bragança J. CITED2 and the modulation of the hypoxic response in cancer. World J Clin Oncol 2020; 11:260-274. [PMID: 32728529 PMCID: PMC7360518 DOI: 10.5306/wjco.v11.i5.260] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/13/2020] [Accepted: 05/12/2020] [Indexed: 02/06/2023] Open
Abstract
CITED2 (CBP/p300-interacting transactivator with Glu/Asp-rich C-terminal domain, 2) is a ubiquitously expressed protein exhibiting a high affinity for the CH1 domain of the transcriptional co-activators CBP/p300, for which it competes with hypoxia-inducible factors (HIFs). CITED2 is particularly efficient in the inhibition of HIF-1α-dependent transcription in different contexts, ranging from organ development and metabolic homeostasis to tissue regeneration and immunity, being also potentially involved in various other physiological processes. In addition, CITED2 plays an important role in inhibiting HIF in some diseases, including kidney and heart diseases and type 2-diabetes. In the particular case of cancer, CITED2 either functions by promoting or suppressing cancer development depending on the context and type of tumors. For instance, CITED2 overexpression promotes breast and prostate cancers, as well as acute myeloid leukemia, while its expression is downregulated to sustain colorectal cancer and hepatocellular carcinoma. In addition, the role of CITED2 in the maintenance of cancer stem cells reveals its potential as a target in non-small cell lung carcinoma and acute myeloid leukemia, for example. But besides the wide body of evidence linking both CITED2 and HIF signaling to carcinogenesis, little data is available regarding CITED2 role as a negative regulator of HIF-1α specifically in cancer. Therefore, comprehensive studies exploring further the interactions of these two important mediators in cancer-specific models are sorely needed and this can potentially lead to the development of novel targeted therapies.
Collapse
Affiliation(s)
- Mónica T Fernandes
- School of Health, Universidade do Algarve, Campus of Gambelas, Faro 8005-139, Portugal
- Centre for Biomedical Research, Universidade do Algarve, Campus of Gambelas, Faro 8005-139, Portugal
- Algarve Biomedical Centre, Faro 8005-139, Portugal
| | - Sofia M Calado
- Centre for Biomedical Research, Universidade do Algarve, Campus of Gambelas, Faro 8005-139, Portugal
- Algarve Biomedical Centre, Faro 8005-139, Portugal
| | - Leonardo Mendes-Silva
- Centre for Biomedical Research, Universidade do Algarve, Campus of Gambelas, Faro 8005-139, Portugal
- Algarve Biomedical Centre, Faro 8005-139, Portugal
- Department of Biomedical Sciences and Medicine, Universidade do Algarve, Campus of Gambelas, Faro 8005-139, Portugal
| | - José Bragança
- Centre for Biomedical Research, Universidade do Algarve, Campus of Gambelas, Faro 8005-139, Portugal
- Algarve Biomedical Centre, Faro 8005-139, Portugal
- Department of Biomedical Sciences and Medicine, Universidade do Algarve, Campus of Gambelas, Faro 8005-139, Portugal
| |
Collapse
|
47
|
Single-Cell Transcriptome Analysis Maps the Developmental Track of the Human Heart. Cell Rep 2020; 26:1934-1950.e5. [PMID: 30759401 DOI: 10.1016/j.celrep.2019.01.079] [Citation(s) in RCA: 313] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/14/2018] [Accepted: 01/22/2019] [Indexed: 02/06/2023] Open
Abstract
The heart is the central organ of the circulatory system, and its proper development is vital for maintaining human life. Here, we used single-cell RNA sequencing to profile the gene expression landscapes of ∼4,000 cardiac cells from human embryos and identified four major types of cells: cardiomyocytes (CMs), cardiac fibroblasts, endothelial cells (ECs), and valvar interstitial cells (VICs). Atrial and ventricular CMs acquired distinct features early in heart development. Furthermore, both CMs and fibroblasts show stepwise changes in gene expression. As development proceeds, VICs may be involved in the remodeling phase, and ECs display location-specific characteristics. Finally, we compared gene expression profiles between humans and mice and identified a series of unique features of human heart development. Our study lays the groundwork for elucidating the mechanisms of in vivo human cardiac development and provides potential clues to understand cardiac regeneration.
Collapse
|
48
|
Wei H. Construction of a hierarchical gene regulatory network centered around a transcription factor. Brief Bioinform 2020; 20:1021-1031. [PMID: 29186304 DOI: 10.1093/bib/bbx152] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/11/2017] [Indexed: 12/24/2022] Open
Abstract
We have modified a multitude of transcription factors (TFs) in numerous plant species and some animal species, and obtained transgenic lines that exhibit phenotypic alterations. Whenever we observe phenotypic changes in a TF's transgenic lines, we are always eager to identify its target genes, collaborative regulators and even upstream high hierarchical regulators. This issue can be addressed by establishing a multilayered hierarchical gene regulatory network (ML-hGRN) centered around a given TF. In this article, a practical approach for constructing an ML-hGRN centered on a TF using a combined approach of top-down and bottom-up network construction methods is described. Strategies for constructing ML-hGRNs are vitally important, as these networks provide key information to advance our understanding of how biological processes are regulated.
Collapse
Affiliation(s)
- Hairong Wei
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, Heilongjiang, China.,School of Forest Resources and Environmental Science, Michigan Technological University, Houghton, MI, USA
| |
Collapse
|
49
|
Dianatpour S, Khatami M, Heidari MM, Hadadzadeh M. Novel Point Mutations of CITED2 Gene Are Associated with Non-familial Congenital Heart Disease (CHD) in Sporadic Pediatric Patients. Appl Biochem Biotechnol 2019; 190:896-906. [PMID: 31515672 DOI: 10.1007/s12010-019-03125-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/25/2019] [Indexed: 12/20/2022]
Abstract
CITED2 is a cardiac transcription factor that plays a critical role in cardiac development. Gene mutations in CITED2 lead to a series of cardiac malformations and congenital heart defects (CHD). Congenital heart disease generally refers to defects in the heart's structure or function and often seen in many forms such as ventricular septal defects (VSDs), atrial septal defects (ASDs), and tetralogy of Fallot (TOF). However, the mechanisms involved in these mutations are poorly understood. The aim of the present study was to evaluate the mutations of the CITED2 gene in pediatric patients with congenital heart defects. We studied the potential impact of sequence variations of the CITED2 gene in a cohort of 150 patients with non-familial CHD and 98 control individuals by polymerase chain reaction-single-stranded conformation polymorphism (PCR-SSCP) and subsequently direct sequencing. We identified seven novel CITED2 nucleotide changes. Four of these alterations were found in the coding region (c.716insG, c.389A>G, c.450G>C and c.512-538del27) and were only seen in our patients, and not detected in the control group. These mutations are leading to changes in the amino acid sequence in the position of p.Gly236fs, p.Asn125Ser, p.Gln145His, and p.Ser170-Gly178del, respectively. Other variations are located in the 5'UTR region of the gene (c.-43C>T, c.-64C>T and c.-90A>G). CITED2 gene mutations in control subjects were not observed. Our Bioinformatics assay results showed that these novel mutations alter the RNA folding, protein structure, and, therefore, probable effect on the protein function and may play a significant role in the development of congenital heart diseases.
Collapse
Affiliation(s)
- Sima Dianatpour
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran
| | - Mehri Khatami
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran.
| | | | - Mehdi Hadadzadeh
- Department of Cardiac Surgery, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
50
|
Santos JMA, Mendes-Silva L, Afonso V, Martins G, Machado RSR, Lopes JA, Cancela L, Futschik ME, Sachinidis A, Gavaia P, Bragança J. Exogenous WNT5A and WNT11 proteins rescue CITED2 dysfunction in mouse embryonic stem cells and zebrafish morphants. Cell Death Dis 2019; 10:582. [PMID: 31378782 PMCID: PMC6680046 DOI: 10.1038/s41419-019-1816-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/06/2019] [Accepted: 07/11/2019] [Indexed: 02/07/2023]
Abstract
Mutations and inadequate methylation profiles of CITED2 are associated with human congenital heart disease (CHD). In mouse, Cited2 is necessary for embryogenesis, particularly for heart development, and its depletion in embryonic stem cells (ESC) impairs cardiac differentiation. We have now determined that Cited2 depletion in ESC affects the expression of transcription factors and cardiopoietic genes involved in early mesoderm and cardiac specification. Interestingly, the supplementation of the secretome prepared from ESC overexpressing CITED2, during the onset of differentiation, rescued the cardiogenic defects of Cited2-depleted ESC. In addition, we demonstrate that the proteins WNT5A and WNT11 held the potential for rescue. We also validated the zebrafish as a model to investigate cited2 function during development. Indeed, the microinjection of morpholinos targeting cited2 transcripts caused developmental defects recapitulating those of mice knockout models, including the increased propensity for cardiac defects and severe death rate. Importantly, the co-injection of anti-cited2 morpholinos with either CITED2 or WNT5A and WNT11 recombinant proteins corrected the developmental defects of Cited2-morphants. This study argues that defects caused by the dysfunction of Cited2 at early stages of development, including heart anomalies, may be remediable by supplementation of exogenous molecules, offering the opportunity to develop novel therapeutic strategies aiming to prevent CHD.
Collapse
Affiliation(s)
- João M A Santos
- Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, room 2.22, 8005-139, Faro, Portugal
| | - Leonardo Mendes-Silva
- Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, room 2.22, 8005-139, Faro, Portugal
| | - Vanessa Afonso
- Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, room 2.22, 8005-139, Faro, Portugal
| | - Gil Martins
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139, Faro, Portugal
| | - Rui S R Machado
- Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, room 2.22, 8005-139, Faro, Portugal
| | - João A Lopes
- Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, room 2.22, 8005-139, Faro, Portugal
| | - Leonor Cancela
- Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139, Faro, Portugal
- ABC-Algarve Biomedical Centre, 8005-139, Faro, Portugal
| | - Matthias E Futschik
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, room 2.22, 8005-139, Faro, Portugal
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139, Faro, Portugal
- School of Biomedical Sciences, Faculty of Medicine and Dentistry, Institute of Translational and Stratified Medicine (ITSMED), University of Plymouth, Plymouth, PL6 8BU, UK
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Paulo Gavaia
- Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139, Faro, Portugal
| | - José Bragança
- Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal.
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, room 2.22, 8005-139, Faro, Portugal.
- ABC-Algarve Biomedical Centre, 8005-139, Faro, Portugal.
| |
Collapse
|