1
|
Fan J, Wang J, Ning J, Wu S, Wang C, Wang YC. Genome-wide identification and expression analysis of the Sox gene family in bivalves. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101489. [PMID: 40139063 DOI: 10.1016/j.cbd.2025.101489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/06/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Since the discovery of the Sox gene family in 1990, research on its distribution, classification, characterization, and function across various species has been significantly deepened. However, the Sox gene family has not yet been systematically and comprehensively analyzed in bivalves. In this study, 254 Sox genes were identified in 51 bivalves (covering 20 orders and 37 families). The Sox gene numbers ranged from 1 and 10 in most bivalves but no Sox gene was identified in the transcriptomes of Poromya illevis (Poromyoidea), Thracia phaseolina (Thracioidea), Solen vaginoides (Solenoidea), Lamychaena hians (Gastrochaenoidea), and Limopsis sp. and Solemya velesiana (Limopsoidea). The phylogenetic analyses revealed that Sox genes in bivalves are divided into 7 primary groups: SoxB1, SoxB2, SoxC, SoxD, SoxE, SoxF, and SoxH, with different groups exhibiting distinct conserved motif patterns. Notably, SoxA and SoxG found in most vertebrates were not identified in bivalves. Moreover, through spatiotemporal expression profiling in 6 distinct bivalve species, it was determined that the SoxH genes exhibit male-biased expression mainly in non-hermaphroditic bivalves, while SoxB1 and SoxC genes demonstrate female-biased expression, and these two Sox genes may serve a pivotal role in embryonic development stage and SoxB2, SoxC and SoxE may play a significant impact in neural development in bivalves. Sox family members also appear to possess disparate functions across different species and tissues. Overall, this study may provide a basis for future investigations into the functions and evolution of Sox genes in bivalves, and offer new perspectives on their roles in development in bivalves.
Collapse
Affiliation(s)
- Jiawei Fan
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinjing Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junhao Ning
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Shaoxuan Wu
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Chunde Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Yin-Chu Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; National Basic Science Data Center, Beijing 100190, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China.
| |
Collapse
|
2
|
Feng W, Hong N, Wu Y, Huang J, Zhang Q, Liu G, Qian Z, Chen Y, Jin L, Ding X, Zhao P, Chen AF, Yu Y. Deficiency of Sox7 leads to congenital aortic stenosis via abnormal valve remodeling. J Mol Cell Cardiol 2025; 199:81-94. [PMID: 39746830 DOI: 10.1016/j.yjmcc.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 11/20/2024] [Accepted: 11/30/2024] [Indexed: 01/04/2025]
Abstract
Abnormal valve development is the most common congenital heart malformation. The transcription factor Sox7 plays a critical role in the development of vascular and cardiac septation. However, it remains unclear whether Sox7 is required for heart valve development. In the present study, Sox7 was strongly expressed in the endocardial and mesenchymal cells of the developing aortic valve in mice and humans, and that endocardial cell specific deletion of Sox7 (Nfatc1 Cre;Sox7fl/fl) in mice leads to congenital aortic stenosis basing on our echocardiography data and multiple staining results. Mechanistically, Sox7 influences extracellular matrix (ECM) remodeling of the valve through regulating MMP9. Meanwhile, Sox7 also affects other valvular remodeling processes, including apoptosis and proliferation of valvular cells in Sox7 deficiency mice. Similarly, in valvular interstitial cells (VICs), Sox7 overexpression increased the protein levels of cleaved caspase3 and TUNEL-positive VICs, while Ki67-positive VICs decreased. The reverse trend was observed in VICs with Sox7 deficiency. Significant enhancement of Rbm25 transcriptional levels was observed in the Sox7 overexpression group, and the mRNA and protein levels of calcification markers such as Osterix, Osteopontin and Runx2 were reduced. The reverse trend was observed in VICs with Sox7 deficiency. Von Kossa staining and Alizarin Red staining also demonstrated that sever calcification in Nfatc1 Cre;Sox7fl/fl mice. Moreover, we detected the Sox7 protein expression in human fetal aortic valves in patients with aortic stenosis, in which Sox7 positive mesenchymal cells were decreased. Taken together, these findings identify Sox7 as a potential pathogenic gene responsible for congenital aortic stenosis in human. Our study provides novel strategies for the diagnosis and treatment of congenital valvular malformation.
Collapse
Affiliation(s)
- Weiqi Feng
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Nanchao Hong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Yizhuo Wu
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Junxin Huang
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Qi Zhang
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Guizhu Liu
- Wuxi School of Medicine, Jiang Nan University, Wuxi 214122, China
| | - Ziling Qian
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Yinghui Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Lihui Jin
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Xiaowei Ding
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Pengjun Zhao
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| | - Yu Yu
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| |
Collapse
|
3
|
Niharika, Ureka L, Roy A, Patra SK. Dissecting SOX2 expression and function reveals an association with multiple signaling pathways during embryonic development and in cancer progression. Biochim Biophys Acta Rev Cancer 2024; 1879:189136. [PMID: 38880162 DOI: 10.1016/j.bbcan.2024.189136] [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: 05/09/2023] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
SRY (Sex Determining Region) box 2 (SOX2) is an essential transcription factor that plays crucial roles in activating genes involved in pre- and post-embryonic development, adult tissue homeostasis, and lineage specifications. SOX2 maintains the self-renewal property of stem cells and is involved in the generation of induced pluripotency stem cells. SOX2 protein contains a particular high-mobility group domain that enables SOX2 to achieve the capacity to participate in a broad variety of functions. The information about the involvement of SOX2 with gene regulatory elements, signaling networks, and microRNA is gradually emerging, and the higher expression of SOX2 is functionally relevant to various cancer types. SOX2 facilitates the oncogenic phenotype via cellular proliferation and enhancement of invasive tumor properties. Evidence are accumulating in favor of three dimensional (higher order) folding of chromatin and epigenetic control of the SOX2 gene by chromatin modifications, which implies that the expression level of SOX2 can be modulated by epigenetic regulatory mechanisms, specifically, via DNA methylation and histone H3 modification. In view of this, and to focus further insights into the roles SOX2 plays in physiological functions, involvement of SOX2 during development, precisely, the advances of our knowledge in pre- and post-embryonic development, and interactions of SOX2 in this scenario with various signaling pathways in tumor development and cancer progression, its potential as a therapeutic target against many cancers are summarized and discussed in this article.
Collapse
Affiliation(s)
- Niharika
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Lina Ureka
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India.
| |
Collapse
|
4
|
Vachon L, Jean G, Milasan A, Babran S, Lacroix E, Guadarrama Bello D, Villeneuve L, Rak J, Nanci A, Mihalache-Avram T, Tardif JC, Finnerty V, Ruiz M, Boilard E, Tessier N, Martel C. Platelet extracellular vesicles preserve lymphatic endothelial cell integrity and enhance lymphatic vessel function. Commun Biol 2024; 7:975. [PMID: 39128945 PMCID: PMC11317532 DOI: 10.1038/s42003-024-06675-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 08/02/2024] [Indexed: 08/13/2024] Open
Abstract
Lymphatic vessels are essential for preventing the accumulation of harmful components within peripheral tissues, including the artery wall. Various endogenous mechanisms maintain adequate lymphatic function throughout life, with platelets being essential for preserving lymphatic vessel integrity. However, since lymph lacks platelets, their impact on the lymphatic system has long been viewed as restricted to areas where lymphatics intersect with blood vessels. Nevertheless, platelets can also exert long range effects through the release of extracellular vesicles (EVs) upon activation. We observed that platelet EVs (PEVs) are present in lymph, a compartment to which they could transfer regulatory effects of platelets. Here, we report that PEVs in lymph exhibit a distinct signature enabling them to interact with lymphatic endothelial cells (LECs). In vitro experiments show that the internalization of PEVs by LECs maintains their functional integrity. Treatment with PEVs improves lymphatic contraction capacity in atherosclerosis-prone mice. We suggest that boosting lymphatic pumping with exogenous PEVs offers a novel therapeutic approach for chronic inflammatory diseases characterized by defective lymphatics.
Collapse
Affiliation(s)
- Laurent Vachon
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Gabriel Jean
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Andreea Milasan
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Sara Babran
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Elizabeth Lacroix
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | | | | | - Janusz Rak
- McGill University and Research, Institute of the McGill University Health Centre, Montreal, Canada
- Department of Experimental Medicine, McGill University, Montreal, Canada
| | - Antonio Nanci
- Department of Stomatology, Faculty of Dental Medicine, Université de Montréal, Montreal, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | | | - Jean-Claude Tardif
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | | | - Matthieu Ruiz
- Department of Nutrition, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Metabolomics platform, Montreal, Canada
| | - Eric Boilard
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Université Laval, Québec, Québec, Canada
- Infectious and Immune Diseases Axis, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
| | - Nolwenn Tessier
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada.
- Montreal Heart Institute, Montreal, Canada.
| |
Collapse
|
5
|
Ream MW, Randolph LN, Jiang Y, Chang Y, Bao X, Lian XL. Direct programming of human pluripotent stem cells into endothelial progenitors with SOX17 and FGF2. Stem Cell Reports 2024; 19:579-595. [PMID: 38518781 PMCID: PMC11096437 DOI: 10.1016/j.stemcr.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 03/24/2024] Open
Abstract
Transcription factors (TFs) are pivotal in guiding stem cell behavior, including their maintenance and differentiation. Using single-cell RNA sequencing, we investigated TFs expressed in endothelial progenitors (EPs) derived from human pluripotent stem cells (hPSCs) and identified upregulated expression of SOXF factors SOX7, SOX17, and SOX18 in the EP population. To test whether overexpression of these factors increases differentiation efficiency, we established inducible hPSC lines for each SOXF factor and found only SOX17 overexpression robustly increased the percentage of cells expressing CD34 and vascular endothelial cadherin (VEC). Conversely, SOX17 knockdown via CRISPR-Cas13d significantly compromised EP differentiation. Intriguingly, we discovered SOX17 overexpression alone was sufficient to generate CD34+VEC+CD31- cells, and, when combined with FGF2 treatment, more than 90% of CD34+VEC+CD31+ EP was produced. These cells are capable of further differentiating into endothelial cells. These findings underscore an undiscovered role of SOX17 in programming hPSCs toward an EP lineage, illuminating pivotal mechanisms in EP differentiation.
Collapse
Affiliation(s)
- Michael W Ream
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Lauren N Randolph
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Yuqian Jiang
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Yun Chang
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Xiaoping Bao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Xiaojun Lance Lian
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA; Department of Biology, Pennsylvania State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
6
|
Payne S, Neal A, De Val S. Transcription factors regulating vasculogenesis and angiogenesis. Dev Dyn 2024; 253:28-58. [PMID: 36795082 PMCID: PMC10952167 DOI: 10.1002/dvdy.575] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Transcription factors (TFs) play a crucial role in regulating the dynamic and precise patterns of gene expression required for the initial specification of endothelial cells (ECs), and during endothelial growth and differentiation. While sharing many core features, ECs can be highly heterogeneous. Differential gene expression between ECs is essential to pattern the hierarchical vascular network into arteries, veins and capillaries, to drive angiogenic growth of new vessels, and to direct specialization in response to local signals. Unlike many other cell types, ECs have no single master regulator, instead relying on differing combinations of a necessarily limited repertoire of TFs to achieve tight spatial and temporal activation and repression of gene expression. Here, we will discuss the cohort of TFs known to be involved in directing gene expression during different stages of mammalian vasculogenesis and angiogenesis, with a primary focus on development.
Collapse
Affiliation(s)
- Sophie Payne
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| | - Alice Neal
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| | - Sarah De Val
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| |
Collapse
|
7
|
Chiang IKN, Humphrey D, Mills RJ, Kaltzis P, Pachauri S, Graus M, Saha D, Wu Z, Young P, Sim CB, Davidson T, Hernandez‐Garcia A, Shaw CA, Renwick A, Scott DA, Porrello ER, Wong ES, Hudson JE, Red‐Horse K, del Monte‐Nieto G, Francois M. Sox7-positive endothelial progenitors establish coronary arteries and govern ventricular compaction. EMBO Rep 2023; 24:e55043. [PMID: 37551717 PMCID: PMC10561369 DOI: 10.15252/embr.202255043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/12/2023] [Accepted: 07/24/2023] [Indexed: 08/09/2023] Open
Abstract
The cardiac endothelium influences ventricular chamber development by coordinating trabeculation and compaction. However, the endothelial-specific molecular mechanisms mediating this coordination are not fully understood. Here, we identify the Sox7 transcription factor as a critical cue instructing cardiac endothelium identity during ventricular chamber development. Endothelial-specific loss of Sox7 function in mice results in cardiac ventricular defects similar to non-compaction cardiomyopathy, with a change in the proportions of trabecular and compact cardiomyocytes in the mutant hearts. This phenotype is paralleled by abnormal coronary artery formation. Loss of Sox7 function disrupts the transcriptional regulation of the Notch pathway and connexins 37 and 40, which govern coronary arterial specification. Upon Sox7 endothelial-specific deletion, single-nuclei transcriptomics analysis identifies the depletion of a subset of Sox9/Gpc3-positive endocardial progenitor cells and an increase in erythro-myeloid cell lineages. Fate mapping analysis reveals that a subset of Sox7-null endothelial cells transdifferentiate into hematopoietic but not cardiomyocyte lineages. Our findings determine that Sox7 maintains cardiac endothelial cell identity, which is crucial to the cellular cross-talk that drives ventricular compaction and coronary artery development.
Collapse
Affiliation(s)
- Ivy KN Chiang
- Centenary Institute, Royal Prince Alfred HospitalThe University of SydneySydneyNSWAustralia
| | - David Humphrey
- The Victor Chang Cardiac Research InstituteDarlinghurstNSWAustralia
| | - Richard J Mills
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Peter Kaltzis
- The Australian Regenerative Medicine InstituteMonash UniversityClaytonVICAustralia
| | - Shikha Pachauri
- Centenary Institute, Royal Prince Alfred HospitalThe University of SydneySydneyNSWAustralia
| | - Matthew Graus
- Centenary Institute, Royal Prince Alfred HospitalThe University of SydneySydneyNSWAustralia
| | - Diptarka Saha
- The Australian Regenerative Medicine InstituteMonash UniversityClaytonVICAustralia
| | - Zhijian Wu
- The Australian Regenerative Medicine InstituteMonash UniversityClaytonVICAustralia
| | - Paul Young
- The Victor Chang Cardiac Research InstituteDarlinghurstNSWAustralia
| | - Choon Boon Sim
- The Murdoch Children's Research InstituteRoyal Children's HospitalMelbourneVICAustralia
| | - Tara Davidson
- Centenary Institute, Royal Prince Alfred HospitalThe University of SydneySydneyNSWAustralia
| | | | - Chad A Shaw
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| | - Alexander Renwick
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| | - Daryl A Scott
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| | - Enzo R Porrello
- The Murdoch Children's Research InstituteRoyal Children's HospitalMelbourneVICAustralia
- Melbourne Centre for Cardiovascular Genomics and Regenerative MedicineThe Royal Children's HospitalMelbourneVICAustralia
- Department of Anatomy and Physiology, School of Biomedical SciencesThe University of MelbourneMelbourneVICAustralia
| | - Emily S Wong
- The Victor Chang Cardiac Research InstituteDarlinghurstNSWAustralia
| | - James E Hudson
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | | | | | - Mathias Francois
- Centenary Institute, Royal Prince Alfred HospitalThe University of SydneySydneyNSWAustralia
| |
Collapse
|
8
|
Oda Y, Wong CT, Oh DH, Meyer MB, Pike JW, Bikle DD. Vitamin D receptor cross-talk with p63 signaling promotes epidermal cell fate. J Steroid Biochem Mol Biol 2023; 232:106352. [PMID: 37330071 PMCID: PMC11634282 DOI: 10.1016/j.jsbmb.2023.106352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/12/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023]
Abstract
The vitamin D receptor with its ligand 1,25 dihydroxy vitamin D3 (1,25D3) regulates epidermal stem cell fate, such that VDR removal from Krt14 expressing keratinocytes delays re-epithelialization of epidermis after wound injury in mice. In this study we deleted Vdr from Lrig1 expressing stem cells in the isthmus of the hair follicle then used lineage tracing to evaluate the impact on re-epithelialization following injury. We showed that Vdr deletion from these cells prevents their migration to and regeneration of the interfollicular epidermis without impairing their ability to repopulate the sebaceous gland. To pursue the molecular basis for these effects of VDR, we performed genome wide transcriptional analysis of keratinocytes from Vdr cKO and control littermate mice. Ingenuity Pathway analysis (IPA) pointed us to the TP53 family including p63 as a partner with VDR, a transcriptional factor that is essential for proliferation and differentiation of epidermal keratinocytes. Epigenetic studies on epidermal keratinocytes derived from interfollicular epidermis showed that VDR is colocalized with p63 within the specific regulatory region of MED1 containing super-enhancers of epidermal fate driven transcription factor genes such as Fos and Jun. Gene ontology analysis further implicated that Vdr and p63 associated genomic regions regulate genes involving stem cell fate and epidermal differentiation. To demonstrate the functional interaction between VDR and p63, we evaluated the response to 1,25(OH)2D3 of keratinocytes lacking p63 and noted a reduction in epidermal cell fate determining transcription factors such as Fos, Jun. We conclude that VDR is required for the epidermal stem cell fate orientation towards interfollicular epidermis. We propose that this role of VDR involves cross-talk with the epidermal master regulator p63 through super-enhancer mediated epigenetic dynamics.
Collapse
Affiliation(s)
- Yuko Oda
- Departments of Medicine and Endocrinology, United States
| | - Christian T Wong
- Department of Dermatology, University of California San Francisco, United States; San Francisco VA Health Care system, United States
| | - Dennis H Oh
- Department of Dermatology, University of California San Francisco, United States; San Francisco VA Health Care system, United States
| | - Mark B Meyer
- Department of Nutritional Sciences, University of Wisconsin-Madison, United States
| | - J Wesley Pike
- Department of Biochemistry, University of Wisconsin-Madison, United States
| | - Daniel D Bikle
- Departments of Medicine and Endocrinology, United States.
| |
Collapse
|
9
|
Chen Q, Xi X, Ma J, Wang X, Xia Y, Xi W, Deng Y, Li Y. The mechanism by which crocetin regulates the lncRNA NEAT1/miR-125b-5p/SOX7 molecular axis to inhibit high glucose-induced diabetic retinopathy. Exp Eye Res 2022; 222:109157. [PMID: 35718188 DOI: 10.1016/j.exer.2022.109157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/27/2022] [Accepted: 06/13/2022] [Indexed: 11/25/2022]
Abstract
Diabetic retinopathy (DR) is a high-incidence microvascular complication with retinal neovascularization that generates irreversible visual impairment. However, the mechanism of DR is unclear and needs to be further explored. To explore the expression of NEAT1 and miR-125b-5p and the proliferation activity, migration ability, and angiogenesis ability of human retinal microvascular endothelial cells (hRMECs), RT-qPCR, CCK-8, Transwell, and tube formation assays were performed. Additionally, western blotting was used to detect the expression of SOX7, VEGFA and CD31. Furthermore, a dual-luciferase reporter gene was used to verify the targeting connection. The DR mouse model was constructed by STZ. The effect of crocetin on DR angiogenesis was detected by hematoxylin-eosin (HE) staining, immunohistochemistry (IHC), retinal digest preparations and Western blotting. The results showed that crocetin inhibited the high-glucose (Hg)-induced upregulation of NEAT1 and SOX7 and the downregulation of miR-125b-5p. Crocetin inhibited Hg-induced proliferation, migration and angiogenesis by upregulating the targeted inhibition of SOX7 by miR-125b-5p through the inhibition of NEAT1. To summarize, our study revealed that crocetin has a protective effect against Hg-induced DR by regulating the lncRNA NEAT1/miR-125b-5p/SOX7 molecular axis.
Collapse
Affiliation(s)
- Qianbo Chen
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Xichang Road 295, Kunming, 650031, Yunnan, China
| | - Xiaoting Xi
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Xichang Road 295, Kunming, 650031, Yunnan, China
| | - Jia Ma
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Xichang Road 295, Kunming, 650031, Yunnan, China
| | - Xuewei Wang
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Xichang Road 295, Kunming, 650031, Yunnan, China
| | - Yuan Xia
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Xichang Road 295, Kunming, 650031, Yunnan, China
| | - Wang Xi
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Xichang Road 295, Kunming, 650031, Yunnan, China
| | - Yachun Deng
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Xichang Road 295, Kunming, 650031, Yunnan, China
| | - Yan Li
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Xichang Road 295, Kunming, 650031, Yunnan, China.
| |
Collapse
|
10
|
Predisposition to atrioventricular septal defects may be caused by SOX7 variants that impair interaction with GATA4. Mol Genet Genomics 2022; 297:671-687. [PMID: 35260939 DOI: 10.1007/s00438-022-01859-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 01/12/2022] [Indexed: 10/18/2022]
Abstract
Atrioventricular septal defects (AVSD) are a complicated subtype of congenital heart defects for which the genetic basis is poorly understood. Many studies have demonstrated that the transcription factor SOX7 plays a pivotal role in cardiovascular development. However, whether SOX7 single nucleotide variants are involved in AVSD pathogenesis is unclear. To explore the potential pathogenic role of SOX7 variants, we recruited a total of 100 sporadic non-syndromic AVSD Chinese Han patients and screened SOX7 variants in the patient cohort by targeted sequencing. Functional assays were performed to evaluate pathogenicity of nonsynonymous variants of SOX7. We identified three rare SOX7 variants, c.40C > G, c.542G > A, and c.743C > T, in the patient cohort, all of which were found to be highly conserved in mammals. Compared to the wild type, these SOX7 variants had increased mRNA expression and decreased protein expression. In developing hearts, SOX7 and GATA4 were highly expressed in the region of atrioventricular cushions. Moreover, SOX7 overexpression promoted the expression of GATA4 in human umbilical vein endothelial cells. A chromatin immunoprecipitation assay revealed that SOX7 could directly bind to the GATA4 promoter and luciferase assays demonstrated that SOX7 activated the GATA4 promoter. The SOX7 variants had impaired transcriptional activity relative to wild-type SOX7. Furthermore, the SOX7 variants altered the ability of GATA4 to regulate its target genes. In conclusion, our findings showed that deleterious SOX7 variants potentially contribute to human AVSD by impairing its interaction with GATA4. This study provides novel insights into the etiology of AVSD and contributes new strategies to the prenatal diagnosis of AVSD.
Collapse
|
11
|
Prenatal Diagnosis of 8p23 Deletion Syndrome by Single Nucleotide Polymorphism Microarray. JOURNAL OF FETAL MEDICINE 2021. [DOI: 10.1007/s40556-021-00322-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
12
|
Okur V, Hamm L, Kavus H, Mebane C, Robinson S, Levy B, Chung WK. Clinical and genomic characterization of 8p cytogenomic disorders. Genet Med 2021; 23:2342-2351. [PMID: 34282301 DOI: 10.1038/s41436-021-01270-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 11/09/2022] Open
Abstract
PURPOSE To provide a detailed clinical and cytogenomic summary of individuals with chromosome 8p rearrangements of invdupdel(8p), del(8p), and dup(8p). METHODS We enrolled 97 individuals with invdupdel(8p), del(8p), and dup(8p). Clinical and molecular data were collected to delineate and compare the clinical findings and rearrangement breakpoints. We included additional 5 individuals with dup(8p) from the literature for a total of 102 individuals. RESULTS Eighty-one individuals had recurrent rearrangements of invdupdel(8p) (n = 49), del(8p)_distal (n = 4), del(8p)_proximal (n = 9), del(8p)_proximal&distal (n = 12), and dup(8p)_proximal (n = 7). Twenty-one individuals had nonrecurrent rearrangements. While all individuals had neurodevelopmental features, the frequency and severity of clinical findings were higher in individuals with invdupdel(8p), and with larger duplications. All individuals with GATA4 deletion had structural congenital heart defects; however, the presence of structural heart defects in some individuals with normal GATA4 copy number suggests there are other potentially contributing gene(s) on 8p. CONCLUSION Our study may inform families and health-care providers about the associated clinical findings and severity in individuals with chromosome 8p rearrangements, and guide researchers in investigating the underlying molecular and biological mechanisms by providing detailed clinical and cytogenomic information about individuals with distinct 8p rearrangements.
Collapse
Affiliation(s)
- Volkan Okur
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Medical Center, New York, NY, USA.,Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Laura Hamm
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Haluk Kavus
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Caroline Mebane
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Scott Robinson
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Brynn Levy
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Wendy K Chung
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Medical Center, New York, NY, USA. .,Department of Medicine, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
13
|
Li B, Tian Y, Wen H, Qi X, Wang L, Zhang J, Li J, Dong X, Zhang K, Li Y. Systematic identification and expression analysis of the Sox gene family in spotted sea bass (Lateolabrax maculatus). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 38:100817. [PMID: 33677158 DOI: 10.1016/j.cbd.2021.100817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 10/22/2022]
Abstract
The Sox gene family encodes a set of transcription factors characterized by a conserved Sry-related high mobility group (HMG)-box domain, which performs a series of essential biological functions in diverse tissues and developmental processes. In this study, the Sox gene family was systematically characterized in spotted sea bass (Lateolabrax maculatus). A total of 26 Sox genes were identified and classified into eight subfamilies, namely, SoxB1, SoxB2, SoxC, SoxD, SoxE, SoxF, SoxH and SoxK. The phylogenetic relationship, exon-intron and domain structure analyses supported their annotation and classification. Comparison of gene copy numbers and chromosome locations among different species indicated that except tandem duplicated paralogs of Sox17/Sox32, duplicated Sox genes in spotted sea bass were generated from teleost-specific whole genome duplication during evolution. In addition, qRT-PCR was performed to detect the expression profiles of Sox genes during development and adulthood. The results showed that the expression of 16 out of 26 Sox genes was induced dramatically at different starting points after the multicellular stage, which is consistent with embryogenesis. At the early stage of sex differentiation, 9 Sox genes exhibited sexually dimorphic expression patterns, among which Sox3, Sox19 and Sox6b showed the most significant ovary-biased expression. Moreover, the distinct expression pattern of Sox genes was observed in different adult tissues. Our results provide a fundamental resource for further investigating the functions of Sox genes in embryonic processes, sex determination and differentiation as well as controlling the homeostasis of adult tissues in spotted sea bass.
Collapse
Affiliation(s)
- Bingyu Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Yuan Tian
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Haishen Wen
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Xin Qi
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Lingyu Wang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Jingru Zhang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Jinku Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Ximeng Dong
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Kaiqiang Zhang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Yun Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China.
| |
Collapse
|
14
|
Gonçalves AN, Correia-Pinto J, Nogueira-Silva C. ROBO2 signaling in lung development regulates SOX2/SOX9 balance, branching morphogenesis and is dysregulated in nitrofen-induced congenital diaphragmatic hernia. Respir Res 2020; 21:302. [PMID: 33208157 PMCID: PMC7672875 DOI: 10.1186/s12931-020-01568-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/09/2020] [Indexed: 11/23/2022] Open
Abstract
Background Characterized by abnormal lung growth or maturation, congenital diaphragmatic hernia (CDH) affects 1:3000 live births. Cellular studies report proximal (SOX2+) and distal (SOX9+) progenitor cells as key modulators of branching morphogenesis and epithelial differentiation, whereas transcriptome studies demonstrate ROBO/SLIT as potential therapeutic targets for diaphragm defect repair in CDH. In this study, we tested the hypothesis that (a) experimental-CDH could changes the expression profile of ROBO1, ROBO2, SOX2 and SOX9; and (b) ROBO1 or ROBO2 receptors are regulators of branching morphogenesis and SOX2/SOX9 balance. Methods The expression profile for receptors and epithelial progenitor markers were assessed by Western blot and immunohistochemistry in a nitrofen-induced CDH rat model. Immunohistochemistry signals by pulmonary structure were also quantified from embryonic-to-saccular stages in normal and hypoplastic lungs. Ex vivo lung explant cultures were harvested at E13.5, cultures during 4 days and treated with increasing doses of recombinant rat ROBO1 or human ROBO2 Fc Chimera proteins for ROBO1 and ROBO2 inhibition, respectively. The lung explants were analyzed morphometrically and ROBO1, ROBO2, SOX2, SOX9, BMP4, and β-Catenin were quantified by Western blot. Results Experimental-CDH induces distinct expression profiles by pulmonary structure and developmental stage for both receptors (ROBO1 and ROBO2) and epithelial progenitor markers (SOX2 and SOX9) that provide evidence of the impairment of proximodistal patterning in experimental-CDH. Ex vivo functional studies showed unchanged branching morphogenesis after ROBO1 inhibition; increased fetal lung growth after ROBO2 inhibition in a mechanism-dependent on SOX2 depletion and overexpression of SOX9, non-phospho β-Catenin, and BMP4. Conclusions These studies provided evidence of receptors and epithelial progenitor cells which are severely affected by CDH-induction from embryonic-to-saccular stages and established the ROBO2 inhibition as promoter of branching morphogenesis through SOX2/SOX9 balance.
Collapse
Affiliation(s)
- Ana N Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Gualtar, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Jorge Correia-Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Gualtar, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.,Department of Pediatric Surgery, Hospital de Braga, Braga, Portugal
| | - Cristina Nogueira-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Gualtar, Braga, Portugal. .,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal. .,Department of Obstetrics and Gynecology, Hospital de Braga, Braga, Portugal.
| |
Collapse
|
15
|
Jiang M, Liu T, Zhang J, Gao S, Tao B, Cao R, Qiu Y, Liu J, Li Y, Wang Y, Cao F. Rapamycin Promotes Cardiomyocyte Differentiation of Human Induced Pluripotent Stem Cells in a Stage-Dependent Manner. Stem Cells Dev 2020; 29:1229-1239. [PMID: 32693734 DOI: 10.1089/scd.2020.0025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) are a promising source for cardiac regenerative therapy, and ideal for in vitro cell modeling of cardiovascular diseases and drug screening. Recent studies have shown that rapamycin can promote cardiomyocyte differentiation in various stem cells. However, how rapamycin affects cardiomyocyte differentiation of iPSCs is still not fully understood. This study aimed to investigate the effect of rapamycin on cardiomyocyte differentiation based on embryoid body (EB) method. First, to determine the autophagy induction protocol, different concentrations of rapamycin were applied in hEBs on day 6. The autophagy was most significant when applying rapamycin at 1 μM for 48 h, demonstrating by the LC3II/LC3I ratio and p62 expression. Then, 1 μM rapamycin was applied for 48 h at different time points of cardiomyocyte differentiation to investigate the role of rapamycin in this process. Compared with control, rapamycin applied on days 0-4 of differentiation significantly decreased the proportion of beating EBs and expression of cardiomyocyte-specific genes, while rapamycin applied on days 4-14 significantly increased them. Among all groups, rapamycin applied on days 4-6 achieved highest cardiomyocyte differentiation efficiency. Furthermore, using autophagy inhibitor NH4Cl and GSK-3β inhibitor CHIR-99021, we found rapamycin-induced autophagy promoted cardiomyocyte differentiation at middle stage by negatively regulating the Wnt/β-catenin signaling pathway. These results suggest that rapamycin regulates EB-based cardiomyocyte differentiation in a stage-dependent manner, and the negative regulation of Wnt/β-catenin signaling pathway by autophagy was involved in the prodifferentiation effect of rapamycin at middle stage.
Collapse
Affiliation(s)
- Min Jiang
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital & Medical School of Chinese PLA, Beijing, China
| | - Tong Liu
- Department of Cardiology, The Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, China
| | - Jibin Zhang
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital & Medical School of Chinese PLA, Beijing, China
| | - Shan Gao
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital & Medical School of Chinese PLA, Beijing, China
| | - Bo Tao
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital & Medical School of Chinese PLA, Beijing, China
| | - Ruihua Cao
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital & Medical School of Chinese PLA, Beijing, China
| | - Ya Qiu
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital & Medical School of Chinese PLA, Beijing, China
| | - Junsong Liu
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital & Medical School of Chinese PLA, Beijing, China
| | - Yanhua Li
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital & Medical School of Chinese PLA, Beijing, China
| | - Yabin Wang
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital & Medical School of Chinese PLA, Beijing, China
| | - Feng Cao
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital & Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
16
|
Oh KS, Febres-Aldana CA, Pelaez L, Alexis J. 8p23.1 Microdeletion syndrome and obstructing myxomatous heart valve nodules. AUTOPSY AND CASE REPORTS 2020; 10:e2020168. [PMID: 33344285 PMCID: PMC7703449 DOI: 10.4322/acr.2020.168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Kei Shing Oh
- Mount Sinai Medical Center, AM Rywlin Department of Pathology and Laboratory Medicine. Miami Beach, FL, USA
| | | | - Liset Pelaez
- Mount Sinai Medical Center, AM Rywlin Department of Pathology and Laboratory Medicine. Miami Beach, FL, USA.,Nicklaus Children's Hospital, Department of Pathology and Clinical Laboratories. Miami, FL, USA
| | - John Alexis
- Mount Sinai Medical Center, AM Rywlin Department of Pathology and Laboratory Medicine. Miami Beach, FL, USA.,Florida International University, Herbert Wertheim College of Medicine, Department of Pathology. Miami, FL, USA
| |
Collapse
|