1
|
Han L, Miao J, Ding M, Fan Q, Wang X, Pan L. Role of NR5A2 in regulating sex differentiation, steroidogenesis, and gonadal development in Chlamys farreri. J Steroid Biochem Mol Biol 2025; 248:106690. [PMID: 39914682 DOI: 10.1016/j.jsbmb.2025.106690] [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: 11/03/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
Chlamys farreri is a commercially important bivalve species in global aquaculture. However, research on the mechanisms regulating its sex differentiation and reproduction remains relatively sparse. In this study, the role of nuclear receptor subfamily 5 group A member 2 (NR5A2) in sex differentiation, steroidogenesis, and gonadal development in C. farreri was investigated using a 28-day RNA interference experiment. RNA-seq data analysis revealed differentially expressed genes between males and females following NR5A2 knockdown. Weighted gene co-expression network analysis (WGCNA) further identified gene modules closely associated with reproductive development, with the yellow module demonstrating a significant correlation with the sex phenotype. Gene set enrichment analysis (GSEA) identified several signaling pathways related to reproduction that were suppressed, including ovarian follicle development, cholesterol metabolism, and ovarian steroidogenesis. Based on the above analysis, we identified 25 differentially expressed genes linked to these processes. Histological observations revealed that NR5A2 knockdown significantly delayed gonadal development in both sexes of scallops, as indicated by a notable decrease in follicular cell number and size. Taken together, NR5A2 knockdown significantly affected signaling pathways related to cholesterol metabolism, ovarian steroidogenesis, sex differentiation and gonadal development, providing a novel theoretical basis for understanding sex differentiation and reproductive development in invertebrates.
Collapse
Affiliation(s)
- Lianxue Han
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| | - Jingjing Miao
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China.
| | - Min Ding
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China; Qinhuangdao Marine Environmental Monitoring Central Station of SOA, Qinhuangdao 066002, PR China
| | - Qichao Fan
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| | - Xuening Wang
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| | - Luqing Pan
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| |
Collapse
|
2
|
Ura K, Tsue S, Sato T, Hagihara S, Satoh M, Higuchi I, Wang H, Nishimiya O, Ijiri S, Takagi Y. Identification and expression of nuclear receptor genes during nutritive phagocyte development in sea urchin Mesocentrotus nudus gonads. Gen Comp Endocrinol 2025; 366:114715. [PMID: 40139329 DOI: 10.1016/j.ygcen.2025.114715] [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: 01/02/2025] [Revised: 02/21/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
The role of the endocrine system during synthesis of the nutrients (e.g., proteins, lipids, polysaccharides) in the gonad in sea urchins is imperfectly understood. We identify genes encoding nuclear receptors (NRs) involved in the development of nutritive phagocytes of the sea urchin Mesocentrotus nudus, and investigate their gene expression patterns during this time. RNA-seq analysis was performed on immature gonads using a next-generation sequencer. A total of 7,651,421 quality-controlled reads obtained using an Ion PGM sequencer were assembled into 175,092 contigs. BLASTn analysis identified 20 NR genes potentially involved in the development of nutritive phagocytes. Expression analyses revealed levels of four NR genes to increase during the development of nutritive phagocytes. These results indicate that these 20 NR genes have physiological functions in gonadal growth, and that 4 NR genes mainly control the expression of genes involved in the synthesis and metabolism of proteins, lipids, and polysaccharides during cellular proliferation in sea urchin gonads.
Collapse
Affiliation(s)
- Kazuhiro Ura
- Laboratory of Comparative Physiology, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Japan.
| | - Shiori Tsue
- Laboratory of Comparative Physiology, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Japan.
| | - Takushi Sato
- Laboratory of Comparative Physiology, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Japan.
| | - Seishi Hagihara
- Laboratory of Reproductive Physiology, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Japan.
| | - Mizuha Satoh
- Laboratory of Comparative Physiology, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Japan.
| | - Ichiro Higuchi
- Laboratory of Comparative Physiology, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Japan.
| | - Heng Wang
- Laboratory of Comparative Physiology, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Japan; Key Laboratory of Mariculture and Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China.
| | - Osamu Nishimiya
- Laboratory of Comparative Physiology, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Japan.
| | - Shigeho Ijiri
- Laboratory of Reproductive Physiology, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Japan.
| | - Yasuaki Takagi
- Laboratory of Comparative Physiology, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Japan.
| |
Collapse
|
3
|
Hao R, Ao X, Xu Y, Gao M, Jia C, Dong X, Cirenluobu, Shang P, Ye Y, Wei Z. Enhancing oxygen utilization and mitigating oxidative stress in Tibetan chickens for adaptation to high-altitude hypoxia. Poult Sci 2025; 104:104893. [PMID: 40014967 PMCID: PMC11910141 DOI: 10.1016/j.psj.2025.104893] [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: 11/20/2024] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 03/01/2025] Open
Abstract
Tibetan chicken (TBC) is one of the native poultry species that is well adapted to the high-altitude environment of the Qinghai-Tibet Plateau. To elucidate the genetic mechanisms underlying adaptation, the transcriptomes of five tissues (heart (HE), lung (LU), liver (LI), ovary (OV), and abdominal fat (AB)) were compared between TBCs and Roman chickens (RMCs) inhabiting the plateau for one year. Moreover, weighted gene co-expression network analysis (WGCNA) was applied to detect tissue-associated modules and hub genes. A total of 1105, 239, 400, 483, and 275 differentially expressed genes (DEGs) were identified in the LI, HE, LU, AB, and OV tissues, respectively. Fifteen tissue-specific modules were identified in TBC and thirteen in RMC. Analysis of transcription factor (TF) binding sites revealed nineteen hub TFs in TBC and twenty in RMC across the pool of hub genes in these two breeds. Functional enrichment analyses demonstrated that TBC exhibited robust capacity for oxygen transport, heme binding, oxidative phosphorylation, and antioxidant responses in high-altitude regions. Further investigation of the function of hub TFs indicated the involvement of ATF4, CEBPA, TCF7L1, and GFI1B in improving oxygen transport in TBCs. These hub TFs were associated with angiogenesis or hematopoiesis and likely linked to various regulatory functions and facilitate communication across multiple tissues. In conclusion, TBCs have developed a systemic adaptive mechanism to cope with high altitudes, involving the coordinated transcriptional regulation in multi-tissues to enhance oxygen transport and utilization, along with amelioration of oxidative stress.
Collapse
Affiliation(s)
- Ruidong Hao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xianpei Ao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Yijing Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Mengyu Gao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Cunling Jia
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xianggui Dong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Cirenluobu
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa 860000, PR China
| | - Peng Shang
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Nyingchi, Tibet 860000, PR China
| | - Yourong Ye
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Nyingchi, Tibet 860000, PR China
| | - Zehui Wei
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
4
|
Wang Y, Bendre SV, Krauklis SA, Steelman AJ, Nelson ER. Role of Protein Regulators of Cholesterol Homeostasis in Immune Modulation and Cancer Pathophysiology. Endocrinology 2025; 166:bqaf031. [PMID: 39951497 PMCID: PMC11878532 DOI: 10.1210/endocr/bqaf031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/30/2025] [Accepted: 02/12/2025] [Indexed: 02/16/2025]
Abstract
Cholesterol metabolism and homeostasis have emerged as important factors governing various aspects of cancer biology. Clinical associations between circulating cholesterol and poor prognosis or use of cholesterol-lowering medication and improved prognosis have been noted for several different solid tumors. Mechanistically, cholesterol has many different direct and indirect effects on cancer cells themselves but is also critically involved in shaping the function of other cells of the tumor microenvironment, especially immune cells. There are 2 major feedback loops regulating cholesterol homeostasis. Here we highlight the major proteins involved in the so-called oxysterol-bile acid feedback loop and discuss how each has been implicated in cancer biology. We focus on roles within the immune system with implications for cancer. Given that many of these proteins are enzymes or nuclear receptors, both of which are amenable to small molecule intervention, we posit that this axis may represent a promising area for therapeutic intervention.
Collapse
Affiliation(s)
- Yu Wang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Shruti V Bendre
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Steven A Krauklis
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Andrew J Steelman
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People (ERN) and Regenerative Biology & Tissue Engineering (AJS), University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People (ERN) and Regenerative Biology & Tissue Engineering (AJS), University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
5
|
Wang C, Zhang S, Shao Z, Sun P, Zhang J, Zhang S, Kong J, Zhi H, Li L, Li M, Yu J, Liu Z, Lu X, Peng H, Tang S. Reproductive toxicity of bisphenol A and nitro-bisphenol A in male zebrafish at environmentally relevant concentrations. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 958:177905. [PMID: 39674150 DOI: 10.1016/j.scitotenv.2024.177905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/05/2024] [Accepted: 12/01/2024] [Indexed: 12/16/2024]
Abstract
Bisphenol A (BPA) is a well-known endocrine-disrupting pollutant that poses significant environmental challenges globally. However, the toxicity of nitro-BPA (NBPA), the primary transformation product of BPA, remains poorly understood. This study employs a multi-omics approach, integrating in silico and bioinformatics analyses, to investigate and compare the male reproductive toxicity of BPA and NBPA in male zebrafish exposed to environmentally relevant concentrations. After 21 days of exposure, we observed a significant increase in cumulative egg production over five days in the NBPA 200 nM group compared to pre-exposure levels. Conversely, the gonadosomatic index of NBPA 200 nM group was significantly reduced by approximately 41.65 %. Our findings indicate that the activation of ESRRγ and inhibition of NR5A2 are critical molecular initiating events linked to male reproductive toxicity. Additionally, both BPA and NBPA were found to disrupt several key events within the steroid hormone biosynthesis pathway. This disruption includes the downregulation of genes encoding cytochrome P450 (CYP450) and hydroxysteroid dehydrogenase enzymes, as well as alterations in the levels of steroid hormones such as cholesterol and 25-hydroxycholesterol. Our study identifies biomolecular targets of BPA and NBPA at environmentally relevant concentrations that induce reproductive toxicity, enhancing our understanding of NBPA toxicity and are anticipated to inform the development of effective mitigation strategies.
Collapse
Affiliation(s)
- Chao Wang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shuyi Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Beijing Key Lab for Source Control Technology of Water Pollution, College of Environmental Sciences & Engineering, Beijing Forestry University, Beijing, China
| | - Zijin Shao
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Peijie Sun
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Jiran Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Shaoping Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jian Kong
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hong Zhi
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Li Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Mingshu Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jie Yu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiaobo Lu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China.
| | - Hui Peng
- Department of Chemistry, School of Environment, University of Toronto, Toronto, Ontario, Canada
| | - Song Tang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
6
|
Douville NJ, Bastarache L, Bertucci-Richter E, Patil S, Jewell ES, Freundlich RE, Kertai MD, Engoren MC. Genetic variants associated with sepsis-associated acute kidney injury. PLoS One 2024; 19:e0311318. [PMID: 39636799 PMCID: PMC11620412 DOI: 10.1371/journal.pone.0311318] [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: 05/17/2024] [Accepted: 09/17/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Kidney dysfunction is a common complication in septic patients. Studies have identified numerous risk factors for sepsis-associated acute kidney injury (S-AKI), yet there is wide variability in the incidence even among patients with similar risk factors, suggesting the presence of additional uncharacterized risk factors, including genetic differences. The expansion of biobanks, advances in genotyping, and standardized diagnostic criteria have enabled large-scale, hypothesis-generating studies into the genetic mechanisms underlying S-AKI. We hypothesize that the genetic pathway behind S-AKI has overlapping mechanisms with key differences based upon the specific subtype of acute kidney injury (AKI). METHODS To test this hypothesis, we performed a genome-wide association study (GWAS) of S-AKI in three logistic regression models. Model 1, controlled for 1) age, 2) sex, 3) genotyping chip, and 4) the first five principal components. In Model 2, pre-sepsis baseline serum creatinine was added to the variables in Model 1. Finally, in Model 3, we controlled for the full range of patient, clinical, and ICU-related risk factors. Each of the 3-models were repeated in a pre-specified sensitivity analysis of higher severity S-AKI, defined as KDIGO Stage 2 or 3. We then compare associated variants and genes from our GWAS with previously published AKI sub-types and model other factors associated with S-AKI in our dataset. FINDINGS 3,348 qualifying Sepsis-3 patients have been genotyped in our dataset. Of these patients, 383 (11.4%) developed Stage 1, 2, or 3 AKI (primary outcome) and 181 (5.4%) developed Stage 2 or 3 AKI (sensitivity analysis). The median age was 61 years (interquartile range (IQR): 51,69), 42% were female, and the increase in SOFA score (between 48-hours before to 24-hours after the onset of suspected infection) was 2 (2-3). No variants exceeded our threshold for genome-wide significance (P<5x10-8), however, a total of 13 variants exceeded the suggestive (P<1x10-6) threshold. Notably, rs184516290 (chr1:199814965:G:A), near the NR5A2 gene, chr1:199805801:T:TA, also near the NR5A2 gene, and rs117313146 (chr15:31999784:G:C), near the CHRNA7 gene, were associated with S-AKI at the suggestive level in all three models presented. Variants in the suppressor of fused homolog (SUFU) gene, previously shown to be correlated with renal function in bacteremic patients, consistently exceeded the P<0.05 threshold in our models. CONCLUSIONS While failing to identify any novel association for S-AKI at the level of genome-wide significance, our study did suggest multiple variants in previously characterized pathways for S-AKI including CHRNA7, NR5A2, and SUFU. We failed to replicate associations from multiple prior studies which may result from differences in how the phenotype was defined or, alternatively, limited genetic contribution and low heritability.
Collapse
Affiliation(s)
- Nicholas J. Douville
- Department of Anesthesiology, Michigan Medicine, Ann Arbor, Michigan, United States of America
- Institute of Healthcare Policy & Innovation, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | | | - Snehal Patil
- Precision Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Elizabeth S. Jewell
- Department of Anesthesiology, Michigan Medicine, Ann Arbor, Michigan, United States of America
| | - Robert E. Freundlich
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Miklos D. Kertai
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Milo C. Engoren
- Department of Anesthesiology, Michigan Medicine, Ann Arbor, Michigan, United States of America
| |
Collapse
|
7
|
Wu T, Lu ZF, Yu HN, Wu XS, Liu Y, Xu Y. Liver receptor homolog-1: structures, related diseases, and drug discovery. Acta Pharmacol Sin 2024; 45:1571-1581. [PMID: 38632319 PMCID: PMC11272790 DOI: 10.1038/s41401-024-01276-x] [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: 02/06/2024] [Accepted: 03/24/2024] [Indexed: 04/19/2024]
Abstract
Liver receptor homolog-1 (LRH-1), a member of the nuclear receptor superfamily, is a ligand-regulated transcription factor that plays crucial roles in metabolism, development, and immunity. Despite being classified as an 'orphan' receptor due to the ongoing debate surrounding its endogenous ligands, recent researches have demonstrated that LRH-1 can be modulated by various synthetic ligands. This highlights the potential of LRH-1 as an attractive drug target for the treatment of inflammation, metabolic disorders, and cancer. In this review, we provide an overview of the structural basis, functional activities, associated diseases, and advancements in therapeutic ligand research targeting LRH-1.
Collapse
Affiliation(s)
- Tong Wu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory of Biomedicine and Health, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
| | - Zhi-Fang Lu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory of Biomedicine and Health, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
| | - Hao-Nan Yu
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory of Biomedicine and Health, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
| | - Xi-Shan Wu
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory of Biomedicine and Health, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
| | - Yang Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Yong Xu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory of Biomedicine and Health, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Wu L, Chen X, Zeng Q, Lai Z, Fan Z, Ruan X, Li X, Yan J. NR5A2 gene affects the overall survival of LUAD patients by regulating the activity of CSCs through SNP pathway by OCLR algorithm and immune score. Heliyon 2024; 10:e28282. [PMID: 38601554 PMCID: PMC11004709 DOI: 10.1016/j.heliyon.2024.e28282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/12/2024] Open
Abstract
Objective Differentially expressed genes (DEGs) in lung adenocarcinoma (LUAD) tumor stem cells were screened, and the biological characteristics of NR5A2 gene were investigated. Methods The expression and prognosis of NR5A2 in human LUAD were predicted and analyzed through bioinformatics analysis from a human cancer database. Gene expression and clinical data of LUAD tumor and normal lung tissues were obtained from The Cancer Genome Atlas (TCGA) database, and DEGs associated with lung cancer tumor stem cells (CSCs) were screened. Univariate and multivariate Cox regression models were used to screen and establish prognostic risk prediction models. The immune function of the patients was scored according to the model, and the relative immune functions of the high- and low-risk groups were compared to determine the difference in survival prognosis between the two groups. In addition, we calculated the index of stemness based on the transcriptome of the samples using one-class linear regression (OCLR). Results Bioinformatics analysis of a clinical cancer database showed that NR5A2 was significantly decreased in human LUAD tissues than in normal lung tissues, and the decrease in NR5A2 gene expression shortened the overall survival and progression-free survival of patients with LUAD. Conclusion The NR5A2 gene may regulate LUAD tumor stem cells through selective splicing mutations, thereby affecting the survival and prognosis of patients with lung cancer, and the NR5A2 gene may regulate CSCs through single nucleotide polymorphism.
Collapse
Affiliation(s)
- Liusheng Wu
- School of Medicine, Tsinghua University, Beijing, 100084, China
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Xiaofan Chen
- Department of Traditional Chinese Medicine, Affiliated Sanming First Hospital of Fujian Medical University, Sanming, 365000, China
| | - Qi Zeng
- Department of Information Technology, Union College of Fujian Normal University, Fuzhou, 350116, China
| | - Zelin Lai
- Department of Information and Computational Sciences, School of Mathematics, Liaoning Normal University, Liaoning, 116029, China
| | - Zhengyang Fan
- Department of Graduate School, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | - Xin Ruan
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Xiaoqiang Li
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Jun Yan
- School of Medicine, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
9
|
Olszewska M, Malcher A, Stokowy T, Pollock N, Berman AJ, Budkiewicz S, Kamieniczna M, Jackowiak H, Suszynska-Zajczyk J, Jedrzejczak P, Yatsenko AN, Kurpisz M. Effects of Tcte1 knockout on energy chain transportation and spermatogenesis: implications for male infertility. Hum Reprod Open 2024; 2024:hoae020. [PMID: 38650655 PMCID: PMC11035007 DOI: 10.1093/hropen/hoae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/08/2024] [Indexed: 04/25/2024] Open
Abstract
STUDY QUESTION Is the Tcte1 mutation causative for male infertility? SUMMARY ANSWER Our collected data underline the complex and devastating effect of the single-gene mutation on the testicular molecular network, leading to male reproductive failure. WHAT IS KNOWN ALREADY Recent data have revealed mutations in genes related to axonemal dynein arms as causative for morphology and motility abnormalities in spermatozoa of infertile males, including dysplasia of fibrous sheath (DFS) and multiple morphological abnormalities in the sperm flagella (MMAF). The nexin-dynein regulatory complex (N-DRC) coordinates the dynein arm activity and is built from the DRC1-DRC7 proteins. DRC5 (TCTE1), one of the N-DRC elements, has already been reported as a candidate for abnormal sperm flagella beating; however, only in a restricted manner with no clear explanation of respective observations. STUDY DESIGN SIZE DURATION Using the CRISPR/Cas9 genome editing technique, a mouse Tcte1 gene knockout line was created on the basis of the C57Bl/6J strain. The mouse reproductive potential, semen characteristics, testicular gene expression levels, sperm ATP, and testis apoptosis level measurements were then assessed, followed by visualization of N-DRC proteins in sperm, and protein modeling in silico. Also, a pilot genomic sequencing study of samples from human infertile males (n = 248) was applied for screening of TCTE1 variants. PARTICIPANTS/MATERIALS SETTING METHODS To check the reproductive potential of KO mice, adult animals were crossed for delivery of three litters per caged pair, but for no longer than for 6 months, in various combinations of zygosity. All experiments were performed for wild-type (WT, control group), heterozygous Tcte1+/- and homozygous Tcte1-/- male mice. Gross anatomy was performed on testis and epididymis samples, followed by semen analysis. Sequencing of RNA (RNAseq; Illumina) was done for mice testis tissues. STRING interactions were checked for protein-protein interactions, based on changed expression levels of corresponding genes identified in the mouse testis RNAseq experiments. Immunofluorescence in situ staining was performed to detect the N-DRC complex proteins: Tcte1 (Drc5), Drc7, Fbxl13 (Drc6), and Eps8l1 (Drc3) in mouse spermatozoa. To determine the amount of ATP in spermatozoa, the luminescence level was measured. In addition, immunofluorescence in situ staining was performed to check the level of apoptosis via caspase 3 visualization on mouse testis samples. DNA from whole blood samples of infertile males (n = 137 with non-obstructive azoospermia or cryptozoospermia, n = 111 samples with a spectrum of oligoasthenoteratozoospermia, including n = 47 with asthenozoospermia) was extracted to perform genomic sequencing (WGS, WES, or Sanger). Protein prediction modeling of human-identified variants and the exon 3 structure deleted in the mouse knockout was also performed. MAIN RESULTS AND THE ROLE OF CHANCE No progeny at all was found for the homozygous males which were revealed to have oligoasthenoteratozoospermia, while heterozygous animals were fertile but manifested oligozoospermia, suggesting haploinsufficiency. RNA-sequencing of the testicular tissue showed the influence of Tcte1 mutations on the expression pattern of 21 genes responsible for mitochondrial ATP processing or linked with apoptosis or spermatogenesis. In Tcte1-/- males, the protein was revealed in only residual amounts in the sperm head nucleus and was not transported to the sperm flagella, as were other N-DRC components. Decreased ATP levels (2.4-fold lower) were found in the spermatozoa of homozygous mice, together with disturbed tail:midpiece ratios, leading to abnormal sperm tail beating. Casp3-positive signals (indicating apoptosis) were observed in spermatogonia only, at a similar level in all three mouse genotypes. Mutation screening of human infertile males revealed one novel and five ultra-rare heterogeneous variants (predicted as disease-causing) in 6.05% of the patients studied. Protein prediction modeling of identified variants revealed changes in the protein surface charge potential, leading to disruption in helix flexibility or its dynamics, thus suggesting disrupted interactions of TCTE1 with its binding partners located within the axoneme. LARGE SCALE DATA All data generated or analyzed during this study are included in this published article and its supplementary information files. RNAseq data are available in the GEO database (https://www.ncbi.nlm.nih.gov/geo/) under the accession number GSE207805. The results described in the publication are based on whole-genome or exome sequencing data which includes sensitive information in the form of patient-specific germline variants. Information regarding such variants must not be shared publicly following European Union legislation, therefore access to raw data that support the findings of this study are available from the corresponding author upon reasonable request. LIMITATIONS REASONS FOR CAUTION In the study, the in vitro fertilization performance of sperm from homozygous male mice was not checked. WIDER IMPLICATIONS OF THE FINDINGS This study contains novel and comprehensive data concerning the role of TCTE1 in male infertility. The TCTE1 gene is the next one that should be added to the 'male infertility list' because of its crucial role in spermatogenesis and proper sperm functioning. STUDY FUNDING/COMPETING INTERESTS This work was supported by National Science Centre in Poland, grants no.: 2015/17/B/NZ2/01157 and 2020/37/B/NZ5/00549 (to M.K.), 2017/26/D/NZ5/00789 (to A.M.), and HD096723, GM127569-03, NIH SAP #4100085736 PA DoH (to A.N.Y.). The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.
Collapse
Affiliation(s)
- Marta Olszewska
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Agnieszka Malcher
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Tomasz Stokowy
- Scientific Computing Group, IT Division, University of Bergen, Bergen, Norway
| | - Nijole Pollock
- Department of OB/GYN and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrea J Berman
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sylwia Budkiewicz
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | - Hanna Jackowiak
- Department of Histology and Embryology, Poznan University of Life Sciences, Poznan, Poland
| | | | - Piotr Jedrzejczak
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Alexander N Yatsenko
- Department of OB/GYN and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maciej Kurpisz
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
10
|
Ye W, Ya‐xuan C, Shan‐shan T, Qiu L, Ting M, Shao‐jie C, Yu C. NR5A2 promotes malignancy progression and mediates the effect of cisplatin in cutaneous squamous cell carcinoma. Immun Inflamm Dis 2024; 12:e1172. [PMID: 38358044 PMCID: PMC10868143 DOI: 10.1002/iid3.1172] [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: 08/03/2023] [Revised: 12/26/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024] Open
Abstract
INTRODUCTION Nuclear receptor subfamily five group A member two (NR5A2) plays a key role in the development of many tumor types, while it is uncertain in cutaneous squamous cell carcinoma (cSCC). The aim of this work was to determine the role of NR5A2 in cSCC proliferation, and to determine whether NR5A2 mediates the effect of cisplatin in cSCC. METHODS We performed a systematic study of existing data and conducted a preliminary bioinformatics analysis of NR5A2 expression in cSCC using bioinformatics databases. Immunohistochemical staining was performed on cSCC tissues of seven patients to study NR5A2 expression. NR5A2 expression was examined in human keratin-forming cells (HaCaT) and human cSCC cells (A431, Colo-16, SCL-1, SCL-2, and HSC-5). Stable A431 and SCL-2 cell lines consisting of sh-RNA-NR5A2 were constructed to detect changes in cell proliferation, cell cycle, apoptosis, and to determine the key proteins in the Wnt/β-catenin pathway. We also investigated changes in the effects of cisplatin on cSCC cells by CCK-8, clone formation assay, and Flow apoptosis assay after NR5A2 knockdown. RESULTS NR5A2 showed enhanced expression in cSCC tissues than in healthy tissues. Downregulation of NR5A2 in cSCC cells led to the formation of a less malignant phenotype. In contrast, the proliferative capacity of the cSCC cells was enhanced posttreatment with RJW100, an NR5A2 agonist. Additionally, NR5A2 knockdown led to a decrease in the expression level of the proteins in the Wnt/β-catenin pathway, and this inhibition was reversed by LiCl and recombinant antibody, Wnt3a. Moreover, NR5A2 knockdown resulted in diminished proliferative capacity and increased apoptotic cells after the addition of cisplatin. CONCLUSION NR5A2 plays a crucial role in the progression of cSCC, and the Wnt/β-catenin signaling pathway may be involved in the regulation of NR5A2-mediated cSCC. Knockdown of NR5A2 enhanced both the proliferation inhibiting and apoptosis promoting effects of cisplatin on cSCC.
Collapse
Affiliation(s)
- Wang Ye
- School of Clinical MedicineGuizhou Medical UniversityGuiyangChina
| | - Cao Ya‐xuan
- Department of DermatologyAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Tang Shan‐shan
- School of Clinical MedicineGuizhou Medical UniversityGuiyangChina
| | - Long Qiu
- School of Clinical MedicineGuizhou Medical UniversityGuiyangChina
| | - Ma Ting
- School of Clinical MedicineGuizhou Medical UniversityGuiyangChina
| | - Chen Shao‐jie
- School of Clinical MedicineGuizhou Medical UniversityGuiyangChina
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Cao Yu
- School of Clinical MedicineGuizhou Medical UniversityGuiyangChina
- Department of DermatologyAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| |
Collapse
|
11
|
Manchester AC, Chow L, Wheat W, Dow S. Modulation of In Vitro Macrophage Responses via Primary and Secondary Bile Acids in Dogs. Animals (Basel) 2023; 13:3714. [PMID: 38067065 PMCID: PMC10705343 DOI: 10.3390/ani13233714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 04/07/2024] Open
Abstract
Bile acids (BA) are important metabolites secreted into the intestinal lumen and impacted by luminal microbes and dietary intake. Prior studies in humans and rodents have shown that BAs are immunologically active and that primary and secondary BAs have distinct immune properties. Therefore, the composition of the gut BA pool may influence GI inflammatory responses. The current study investigated the relative immune modulatory properties of primary (cholic acid, CA) and secondary BAs (lithocholic acid, LCA) by assessing their effects on canine macrophage cytokine secretion and BA receptor (TGR5) expression. In addition, RNA sequencing was used to further interrogate how CA and LCA differentially modulated macrophage responses to LPS (lipopolysaccharide). We found that exposure to either CA or LCA influenced LPS-induced cytokine production via macrophages similarly, with suppression of TNF-α secretion and enhancement of IL-10 secretion. Neither BA altered the expression of the BA receptor TGR5. Transcriptomic analysis revealed that CA activated inflammatory signaling pathways in macrophages involving type II interferon signaling and the aryl hydrocarbon receptor, whereas LCA activated pathways related to nitric oxide signaling and cell cycle regulation. Thus, we concluded that both primary and secondary BAs are active modulators of macrophage responses in dogs, with differential and shared effects evident with sequencing analysis.
Collapse
Affiliation(s)
- Alison C. Manchester
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA (S.D.)
| | - Lyndah Chow
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA (S.D.)
| | - William Wheat
- Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Steven Dow
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA (S.D.)
| |
Collapse
|
12
|
Meng L, Dong F, Deng J. NR5A2 as a potential target for exercise to improve metabolic syndrome. Aging (Albany NY) 2023; 15:2485-2502. [PMID: 37053002 PMCID: PMC10120892 DOI: 10.18632/aging.204606] [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: 11/16/2022] [Accepted: 03/04/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND Metabolic syndrome is a syndrome of a variety of metabolic disorders. Exercise is beneficial to the human body. However, the association of NR5A2 and exercise with metabolic syndrome remains unclear. METHODS Download the GSE10540 and GSE12385 from GEO database. Bioinformatics analysis was used to screen the hub molecular of the metabolic syndrome. Forty 3-week-old C57BL/6J male mice were used in this study. The mean body weight was (17.5 ± 2.1) g. After 10 days of adaptive feeding, they were randomly divided into 4 groups according to the random number table method: Model + Exercise (n = 10), Model (n = 10), Model/NR5A2-OE (n = 10), Model/NR5A2-KO (n = 10). Western Blotting was performed to detect the expression of hub genes and signaling pathway. RESULTS There were 349 DEGs in GSE10540 and 49 DEGs in GSE12385. 10 core genes were obtained. GO showed that differentially expressed genes were mainly enriched in vascular morphogenesis, contractile fiber fraction, chemotaxis, and MAPK cascade regulation. KEGG showed that MAPK signaling pathway was a significant section in the metabolic syndrome. PIK3R2, STRA8, FLT1, DMRT1, FGF22, NR5A2, and FLT were up-regulated and PRDM14, POU5F1, and KDR were down-regulated in metabolic syndrome after exercise. CONCLUSION The expression of NR5A2 is down-regulated in metabolic syndrome, and exercise can increase the expression level of NR5A2. NR5A2 might be used as a potential target for exercise to improve metabolic syndrome.
Collapse
Affiliation(s)
- Lingxiu Meng
- Department of Cardiology, Qinhuangdao Second Hospital, Qinhuangdao, Hebei 066600, PR China
| | - Fusheng Dong
- Department of Anesthesiology, Qinhuangdao Second Hospital, Qinhuangdao, Hebei 066600, PR China
| | - Junguo Deng
- Department of Cardiology, Qinhuangdao Second Hospital, Qinhuangdao, Hebei 066600, PR China
| |
Collapse
|
13
|
Hecht I, Weiner C, Kotlyar A, Shoshany N, Pras E. Micro chromosomal deletions at the NYS7 locus and autosomal dominant nystagmus. Exp Eye Res 2023; 230:109459. [PMID: 37001852 DOI: 10.1016/j.exer.2023.109459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/13/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023]
Abstract
Nystagmus is an ocular condition characterized by bilateral involuntary ocular oscillation which can severely affect vision. When not associated with other ocular or systemic diseases, it is referred to as idiopathic or congenital motor nystagmus (CMN). Genome-wide linkage studies have previously identified several loci associated with CMN, however the genes responsible for some of these loci have yet to be identified. We have examined a large, five-generation family with autosomal dominant CMN. Our purpose was to characterize the clinical manifestations and reveal the molecular basis of the disease in this family. In addition to full ophthalmic examination and imaging, molecular analysis included copy number variation analysis, linkage studies, and Sanger sequencing. Expression analyses of candidate genes was done by real-time PCR. Of the 68 family members, 27 subjects in five-generations had CMN, in line with an autosomal dominant inheritance pattern. Molecular analysis was performed on 27 members, 15 of them affected by CMN. Copy number variation analysis using array comparative genomic hybridization (aCGH) revealed a novel deletion located on 1q32 (NYS7) among affected individuals. Linkage analysis using polymorphic markers demonstrated full segregation with a heterozygous haplotype in all affected patients, with a LOD score of >5. Sanger sequencing of affected subjects revealed a novel deletion of 732,526 bp in the linkage interval. No protein-coding genes exist within the deleted region; however, the deletion disrupts topologically associated domains encompassing the gene NR5A2 and the non-protein coding MIR181A. Both are strongly associated with other genes expressed in the retina such as PROX1, which in turn is also associated with genes related to nystagmus such as PAX6. We therefore hypothesized that the deletion might affect NR5A2 and MIR181A expression, causing CMN. Expression analysis by real-time PCR showed significantly lower expression of NR5A2, and significantly higher expression of PROX1 among patients compared with controls. To conclude, among a large five-generation family with autosomal dominant CMN, a large deletion in the interval of NYS7 was linked with the disease. No protein-coding genes exist inside the deleted region, and so the exact mechanism in which CMN is caused is uncertain. Based on topological association and expression analyses we suggest a possible mechanism for the pathogenesis.
Collapse
Affiliation(s)
- Idan Hecht
- Department of Ophthalmology, Shamir Medical Center (formerly Assaf-Harofeh), Tzrifin, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Matlow's Ophthalmo-Genetics Laboratory, Department of Ophthalmology, Shamir Medical Center (formerly Assaf-Harofeh), Tzrifin, Israel.
| | - Chen Weiner
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Matlow's Ophthalmo-Genetics Laboratory, Department of Ophthalmology, Shamir Medical Center (formerly Assaf-Harofeh), Tzrifin, Israel
| | - Alina Kotlyar
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Matlow's Ophthalmo-Genetics Laboratory, Department of Ophthalmology, Shamir Medical Center (formerly Assaf-Harofeh), Tzrifin, Israel
| | - Nadav Shoshany
- Department of Ophthalmology, Shamir Medical Center (formerly Assaf-Harofeh), Tzrifin, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Matlow's Ophthalmo-Genetics Laboratory, Department of Ophthalmology, Shamir Medical Center (formerly Assaf-Harofeh), Tzrifin, Israel
| | - Eran Pras
- Department of Ophthalmology, Shamir Medical Center (formerly Assaf-Harofeh), Tzrifin, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Matlow's Ophthalmo-Genetics Laboratory, Department of Ophthalmology, Shamir Medical Center (formerly Assaf-Harofeh), Tzrifin, Israel
| |
Collapse
|