1
|
Malla WA, Singh K, Ayman N, Boro C, Devi NC, Mech P, Siddiqui N, Anvikar AR, Bharti PK. Genome-wide cataloging and orthology analysis of long noncoding RNA expression in three species of Anopheles mosquito. BMC Genomics 2025; 26:510. [PMID: 40394474 PMCID: PMC12090406 DOI: 10.1186/s12864-025-11687-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/09/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are versatile regulatory molecules that affect cellular phenotypes through context-specific expression. While their role in controlling cellular pathways is well-established in insects, investigating lncRNA expression in Anopheles in a tissue- and species-specific manner could add to our understanding of malaria transmission by this important vector. METHODOLOGY We performed de novo transcriptome assembly of Anopheles minimus, Anopheles albimanus, and Anopheles arabiensis utilising publicly available RNA-Seq datasets of male reproductive tissues, male carcasses, female reproductive tissues, and female carcasses. Various bioinformatics tools were subsequently used for lncRNA identification, conservation analysis, and differential expression analysis across sexes and tissues. RESULTS We identified 9331, 5372, and 5256 lncRNA transcripts in An. albimanus, An. arabiensis, and An. minimus, respectively. Compared with An. albimanus lncRNAs, conservation analysis revealed that a total of 1964 and 1400 lncRNAs were conserved in An. arabiensis and An. minimus; however, only 283 and 253 lncRNAs presented sequence-level conservation. Differential expression (DE) analysis revealed that the carcasses presented the lowest difference in lncRNA expression, whereas in each comparison, the reproductive tissues (whether male or female) presented relatively high levels of differential expression. Additionally, 69 lncRNAs were found to be conserved at the sequence level in all 3 species. These lncRNAs were almost exclusively upregulated in males (reproductive tissue as well as carcasses) and exclusively downregulated in female carcasses. CONCLUSIONS The genes in the vicinity of differentially expressed, conserved lncRNAs were found to be involved in critical pathways such as nuclear structure, chromatin remodelling, protein and RNA metabolism, and cell cycle in each of these species. Future studies on these lncRNAs can provide useful insights into how these functions control sexually-dimorphic physiological phenomena such as host-seeking and biting behaviour of female mosquitoes, blood meal metabolism, reproductive behaviour, and disease-carrying capacity.
Collapse
Affiliation(s)
- Waseem Akram Malla
- ICMR-National Institute of Malaria Research, Field Unit Guwahati, Guwahati, Assam, 781036, India.
| | - Kuldeep Singh
- ICMR-National Institute of Malaria Research, New Delhi, 110077, India
- Faculty of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Niha Ayman
- Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Srinagar, Shuhama, Jammu and Kashmir, 190006, India
| | - Chandini Boro
- ICMR-National Institute of Malaria Research, Field Unit Guwahati, Guwahati, Assam, 781036, India
| | - Naorem Chaoba Devi
- ICMR-National Institute of Malaria Research, Field Unit Guwahati, Guwahati, Assam, 781036, India
| | - Priyanka Mech
- ICMR-National Institute of Malaria Research, Field Unit Guwahati, Guwahati, Assam, 781036, India
| | - Nida Siddiqui
- ICMR-National Institute of Malaria Research, Field Unit Guwahati, Guwahati, Assam, 781036, India
| | - Anupkumar R Anvikar
- ICMR-National Institute of Malaria Research, New Delhi, 110077, India
- Faculty of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Praveen Kumar Bharti
- ICMR-National Institute of Malaria Research, New Delhi, 110077, India.
- Faculty of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India.
| |
Collapse
|
2
|
Murcia-Belmonte V, Liu Y, Shamsi S, Shaw S, Collie-Duguid E, Herrera E, Collinson JM, Vargesson N, Erskine L. Identification of lens-regulated genes driving anterior eye development. Dev Biol 2025; 520:91-107. [PMID: 39814158 DOI: 10.1016/j.ydbio.2025.01.004] [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: 08/13/2024] [Revised: 12/13/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025]
Abstract
Signals from the lens regulate multiple aspects of eye development, including establishment of eye size, patterning of the presumptive iris and ciliary body in the anterior optic cup and migration and differentiation of neural crest cells. To advance understanding of the molecular mechanism by which the lens regulates eye development, we performed transcriptome profiling of embryonic chicken retinas after lens removal. Genes associated with nervous system development were upregulated in lens-removed eyes, but the presumptive ciliary body and iris region did not adopt a neural retina identity following lens removal. Lens-regulated genes implicated in periocular mesenchyme, cornea and anterior optic cup development were identified, including factors not previously implicated in eye development. Unexpectedly, transcriptomic differences were identified in retinas from male versus female chicken embryos, suggesting sexual dimorphism from early stages. In situ hybridisation of embryonic chicken eyes and analyses of datasets from embryonic mouse and adult human eyes confirmed expression of candidate genes, including multiple WNT genes, in tissues important for anterior eye development and function. Remarkably, pharmacological activation of canonical WNT signalling restored eye development and size in the absence of the lens. These analyses have identified candidate genes and biological pathways involved in eye development, providing avenues for new research in this area.
Collapse
Affiliation(s)
- Verónica Murcia-Belmonte
- University of Aberdeen, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, UK; Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Av. Ramón y Cajal S/n, Alicante, 03550, Spain
| | - Yanlin Liu
- University of Aberdeen, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Sadia Shamsi
- University of Aberdeen, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Sophie Shaw
- University of Aberdeen, Centre for Genome Enabled Biology and Medicine, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, UK; Current Address: All Wales Medical Genomics Service, Cardiff and Vale University Health Board, University Hospital of Wales, CF14 4XW, UK
| | - Elaina Collie-Duguid
- University of Aberdeen, Centre for Genome Enabled Biology and Medicine, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Eloisa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Av. Ramón y Cajal S/n, Alicante, 03550, Spain
| | - J Martin Collinson
- University of Aberdeen, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Neil Vargesson
- University of Aberdeen, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Lynda Erskine
- University of Aberdeen, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
3
|
Farina R, Simonelli A, Tomasi C, Ioannidou E, Trombelli L. Sexual dimorphism in periodontal inflammation: A cross-sectional study. J Periodontol 2025; 96:346-354. [PMID: 39903662 PMCID: PMC12062729 DOI: 10.1002/jper.24-0466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/04/2024] [Accepted: 12/18/2024] [Indexed: 02/06/2025]
Abstract
BACKGROUND The purpose of the present study was to evaluate the role of sexual dimorphism (SD) in the clinical manifestation of plaque-induced periodontal inflammation by analyzing the association between patient-related factors and the full-mouth prevalence of bleeding on probing (BOP%) within 2 cohorts of male and female individuals. METHODS Data on BOP (dichotomously recorded as present/absent after the assessment of probing depth [PD]) were retrospectively obtained from the files of adult patients undergoing a first periodontal visit at a University center. Two multiple regression models (1 for males, 1 for females) were built with BOP% as the dependent variable and patient-related factors (i.e., age; smoking status; daily cigarette consumption; history of diabetes diagnosis; number of teeth present; proportion of sites with PD ≥ 5 mm around teeth) as independent variables. RESULTS In males (n = 212), BOP% was 5.9% lower in smokers compared to non-smokers (p = 0.021). In females (n = 389), BOP% increased by 1.6% for each 10-year increase in age (p = 0.046). The proportion of sites with PD ≥ 5 mm showed a strongly significant, positive association with BOP% irrespective of biological sex (p < 0.001). CONCLUSION SD manifested as a sex-dependent diversity in the association between patient-related factors and periodontal inflammation expressed as BOP%. While smoking determined a lower BOP% only in males, aging was associated with increased BOP% only in females. PLAIN LANGUAGE SUMMARY Gingival bleeding upon mechanical stimulation of the bottom of the gingival sulcus/pocket with a periodontal probe (bleeding on probing [BOP]) is suggestive of the presence of an inflammatory infiltrate induced by dental plaque within the gingival tissue. The prevalence of BOP within the dentition (BOP%) has a diagnostic relevance, being one of the main parameters to discriminate between periodontal health and disease. Also, BOP% informs the probability for a patient to either develop destructive form of periodontal disease (i.e., periodontitis) or manifest periodontitis progression. Based on the documented influence of biological sex on the incidence, traits, and/or progression rate of several diseases, which goes under the name of sexual dimorphism (SD), the effect of SD was investigated in relation to the factors that were previously associated with BOP% in a cohort of patients with heterogeneous periodontal conditions undergoing their first periodontal visit. Interestingly, SD manifested as a sex-dependent diversity in the association between patient-related factors and periodontal inflammation expressed by BOP%. While smoking determined a lower BOP% only in males, aging was associated with increased BOP% only in females. The present findings may find potential applications in personalized periodontal medicine and inspire future studies in this field.
Collapse
Affiliation(s)
- Roberto Farina
- Research Center for the Study of Periodontal and Peri‐Implant DiseasesUniversity of FerraraFerraraItaly
- Operative Unit of DentistryUniversity‐Hospital of FerraraFerraraItaly
| | - Anna Simonelli
- Research Center for the Study of Periodontal and Peri‐Implant DiseasesUniversity of FerraraFerraraItaly
- Operative Unit of DentistryUniversity‐Hospital of FerraraFerraraItaly
| | - Cristiano Tomasi
- Department of PeriodontologyInstitute of OdontologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Effie Ioannidou
- Department of Orofacial SciencesSchool of DentistryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Leonardo Trombelli
- Research Center for the Study of Periodontal and Peri‐Implant DiseasesUniversity of FerraraFerraraItaly
- Operative Unit of DentistryUniversity‐Hospital of FerraraFerraraItaly
| |
Collapse
|
4
|
Wang DH, He DW, Lv TT, Zhang XK, Li ZJ, Wang ZY. Estrogen receptor α suppresses hepatocellular carcinoma by restricting M2 macrophage infiltration through the YAP-CCL2 axis. BMC Cancer 2025; 25:550. [PMID: 40148834 PMCID: PMC11948847 DOI: 10.1186/s12885-025-13676-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 02/06/2025] [Indexed: 03/29/2025] Open
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide, with significant differences in incidence and outcomes between men and women. Estrogen receptor alpha (ERα) expression is associated with sex-based differences and poor prognostic outcomes in HCC. However, the detailed function of ERα in the tumor microenvironment of HCC remains unclear. METHODS Bioinformatics analysis of differentially expressed genes in HCC samples was performed from publicly available databases, and ERα was selected. The function of ERα was examined in the cell experiments. A co-culture system was built to study function of ERα-treated liver cells on macrophages in vitro. The precise mechanism was determined using quantitative real-time PCR, western blotting, immunohistochemistry, mass spectrometry, co-immunoprecipitation, and dual-luciferase reporter assay. RESULTS ERα played an important role in the pathogenesis of sexual dimorphism in HCC. ERα mainly acted on macrophages in the tumor microenvironment (TME) of HCC and reduced M2 macrophage infiltration through CCL2. By acting on NF2 and 14-3-3theta, ERα enhanced YAP phosphorylation and attenuated the nuclear translocation of YAP, thereby suppressing CCL2 expression. It also acted as a transcription factor that regulated CCL2 expression at the transcriptional level. CONCLUSION ERα/YAP/CCL2 signaling reduced M2 macrophages infiltration to inhibit HCC progression, revealing the effect of ERα in cancer cells on immune cells in HCC microenvironment.
Collapse
Affiliation(s)
- De-Hua Wang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, P. R. China
- Division of Liver Disease, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, Hebei, 050023, P. R. China
| | - Dong-Wei He
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, P. R. China
| | - Ting-Ting Lv
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, P. R. China
| | - Xiao-Kuan Zhang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, P. R. China
| | - Zi-Jie Li
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, P. R. China
| | - Zhi-Yu Wang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, P. R. China.
- , 12, Jiankang Road, Chang'an District, Shijiazhuang City, Hebei Province, China.
| |
Collapse
|
5
|
Zhang Y, Qing J, Li Y, Gao X, Lu D, Wang Y, Gu L, Zhang H, Li Z, Wang X, Zhou Y, Zhang P. PRMT7-Mediated PTEN Activation Enhances Bone Regeneration in Female Mice. Int J Mol Sci 2025; 26:2981. [PMID: 40243588 PMCID: PMC11988880 DOI: 10.3390/ijms26072981] [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: 02/18/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Epigenetic regulation provides new insights into the mechanisms of osteogenic differentiation and identifies potential targets for treating bone-related diseases. However, the specific regulatory networks and mechanisms involved still need further investigation. In this study, we identify PRMT7 as a novel epigenetic regulator of mesenchymal stem cells (MSCs) osteogenic commitment. Conditional knockout of Prmt7 in mice reveals a significant impairment in osteogenesis and bone regeneration, specifically in females, affecting both femurs and mandibles, with no noticeable effect in males. Mechanistically, PRMT7 modulates MSCs osteogenic differentiation by activating PTEN. Specifically, PRMT7 enhances PTEN transcription by increasing H3R2me1 levels at the PTEN promoter. Additionally, PRMT7 interacts with the PTEN protein and stabilizes nuclear PTEN, revealing an unprecedented pathway. Notably, overexpression of PTEN alleviates the osteogenic deficits observed in Prmt7-deficient mice. This research establishes PRMT7 as a potential therapeutic target for promoting bone formation/regeneration and offers novel molecular insights into the PRMT7-PTEN regulatory axis, underscoring its significance in bone biology and regenerative medicine.
Collapse
Affiliation(s)
- Yingfei Zhang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Jia Qing
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Yang Li
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Xin Gao
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Dazhuang Lu
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Yiyang Wang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Lanxin Gu
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Hui Zhang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Zechuan Li
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Xu Wang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| |
Collapse
|
6
|
Lin H, Wei S, Huang S, Tang Z, Mo Z. Gender difference in the association of OSBPL8 polymorphisms with nephrolithiasis within a Chinese cohort. Gene 2025; 942:149218. [PMID: 39761801 DOI: 10.1016/j.gene.2025.149218] [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: 11/13/2024] [Revised: 12/17/2024] [Accepted: 01/02/2025] [Indexed: 01/21/2025]
Abstract
BACKGROUND Kidney stone disease (KSD) is a common disorder of the urinary system and is closely related to genetic polymorphisms. However, the relationship between OSBPL8 polymorphisms and kidney stones has not been thoroughly investigated. METHODS Six OSBPL8 polymorphisms (rs17042391,rs17042409,rs4761431,rs7303892,rs4761434, and rs17042390) were analyzed in a Chinese case-control cohort containing 923 nephrolithiasis patients and 945 healthy controls.The association of these OSBPL8 gene polymorphisms with KSD susceptibility was analyzed using logistic regression, and examined by calculating the odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS The OSBPL8 polymorphisms (GG for rs17042391, rs17042409, rs4761431, rs7303892; AA for rs4761434; and G for rs17042390) were significantly associated with a decreased risk of KSD in females.The protected alleles (G allele of rs17042391, G allele of rs17042409, G allele of rs4761431, A allele of rs4761434, and G allele of rs17042390) were related to decreased BMI levels in KSD patients; female patients with these alleles also exhibited lower BMI, HDL, and LDL levels,the G allele of rs7303892 was linked to reduced serum cholesterol levels in these females.Additionally, the haplotype ACAAGA was associated with decreased KSD risk in females, but haplotype GGGGAG presented an opposing effect. CONCLUSION Our research shows that the OSBPL8 gene polymorphisms reduced the risk of KSD in females, and were also associated with lipid-related metabolic traits.
Collapse
Affiliation(s)
- Haisong Lin
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi 530021, China; Departments of Urology, The Second Nanning People's Hospital, Nanning, Guangxi 530021, China; Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Suchun Wei
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi 530021, China; Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; The First Affiliated Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi Zhuang Autonomous Region, 545005, China
| | - Shengzhu Huang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhen Tang
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi 530021, China; Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Departments of Urology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi Zhuang Autonomous Region, 545005, China
| | - Zengnan Mo
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi 530021, China; Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
7
|
Zhang Z, Su V, Wiese CB, Cheng L, Wang D, Cui Y, Kallapur A, Kim J, Wu X, Tran PH, Zhou Z, Casero D, Li W, Hevener AL, Reue K, Sallam T. A genome-wide ATLAS of liver chromatin accessibility reveals that sex dictates diet-induced nucleosome dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.13.623052. [PMID: 40161732 PMCID: PMC11952359 DOI: 10.1101/2024.11.13.623052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The three-dimensional organization of the genome plays an important role in cellular function. Alterations between open and closed chromatin states contributes to DNA binding, collaborative transcriptional activities and informs post-transcriptional processing. The liver orchestrates systemic metabolic control and has the ability to mount a rapid adaptive response to environmental challenges. We interrogated the chromatin architecture in liver under different dietary cues. Using ATAC-seq, we mapped over 120,000 nucleosome peaks, revealing a remarkably preserved hepatic chromatin landscape across feeding conditions. Stringent analysis of nucleosome rearrangements in response to diet revealed that sex is the dominant factor segregating changes in chromatin accessibility. A lipid-rich diet led to a more accessible chromatin confirmation at promoter regions in female mice along with enrichment of promoter binding CCAAT-binding domain proteins. Male liver exhibited stronger binding for nutrient sensing nuclear receptors. Integrative analysis with gene expression corroborated a role for chromatin states in informing functional differences in metabolic traits. We distinguished the impact of gonadal sex and chromosomal sex as determinants of chromatin modulation by diet using the Four Core Genotypes mouse model. Our data provide mechanistic evidence underlying the regulation for the critical sex-dimorphic GWAS gene, Pnpla3 . In summary, we provide a comprehensive epigenetic resource in murine liver that uncovers the complexity of chromatin dynamics in response to diet and sex. Highlights ATAC-Seq, RNA-Seq, and FCG model-integrated analysis unravel sex differences in chromatin accessibility and transcriptome responses to dietary challenges.Lipid-rich diet led to sex-biased chromatin confirmation at promoter regions.Gonadal sex emerged as the most prevalent determinant of the sex bias hepatic chromatin modulation by lipid-rich diets. The critical sex-dimorphic GWAS gene Pnpla3 is suppressed by testosterone, which underlies hepatic differences in expression between the sexes.
Collapse
|
8
|
Seah C, Karabacak M, Margetis K. Transcriptomic imputation identifies tissue-specific genes associated with cervical myelopathy. Spine J 2025; 25:588-596. [PMID: 39491753 DOI: 10.1016/j.spinee.2024.10.014] [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: 05/31/2024] [Revised: 09/25/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND CONTEXT Degenerative cervical myelopathy (DCM) is a progressive spinal condition that can lead to severe neurological dysfunction. Despite its degenerative pathophysiology, family history has shown to be a largely important factor in incidence and progression, suggesting that inherent genetic predisposition may play a role in pathophysiology. PURPOSE To determine the tissue-specific, functional genetic basis of hereditary predisposition to cervical myelopathy. STUDY DESIGN Retrospective case-control study using patient genetics and matched EHR from the Mount Sinai BioMe Biobank. METHODS In a large, diverse, urban biobank of 32,031 individuals, with 558 individuals with cervical myopathy, we applied transcriptomic imputation to identify genetically regulated gene expression signatures associated with DCM. We performed drug-repurposing analysis using the CMAP database to identify candidate therapeutic interventions to reverse the cervical myelopathy-associated gene signature. RESULTS We identified 16 genes significantly associated with DCM across 5 different tissues, suggesting tissue-specific manifestations of inherited genetic risk (upregulated: HES6, PI16, TMEM183A, BDH2, LINC00937, CLEC4D, USP43, SPATA1; downregulated: TTC12, CDK5, PAFAH1B2, RCSD1, KLHL29, PTPRG, RP11-620J15.3, C1RL). Drug repurposing identified 22 compounds with the potential to reverse the DCM-associated signature, suggesting points of therapeutic intervention. CONCLUSIONS The inherited genetic risk for cervical myelopathy is functionally associated with genes involved in tissue-specific nociceptive and proliferative processes. These signatures may be reversed by candidate therapeutics with nociceptive, calcium channel modulating, and antiproliferative effects. CLINICAL SIGNIFICANCE Understanding the genetic basis of DCM provides critical insights into the hereditary factors contributing to the disease, allowing for more personalized and targeted therapeutic approaches. The identification of candidate drugs through transcriptomic imputation and drug repurposing analysis offers potential new treatments that could significantly improve patient outcomes and quality of life by addressing the underlying genetic mechanisms of DCM.
Collapse
Affiliation(s)
- Carina Seah
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neurosurgery, Mount Sinai Health System, New York, NY, USA.
| | - Mert Karabacak
- Department of Neurosurgery, Mount Sinai Health System, New York, NY, USA
| | | |
Collapse
|
9
|
Zhang JW, Zhang N, Lyu Y, Zhang XF. Influence of Sex in the Development of Liver Diseases. Semin Liver Dis 2025; 45:15-32. [PMID: 39809453 DOI: 10.1055/a-2516-0261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The liver is a sexually dimorphic organ. Sex differences in prevalence, progression, prognosis, and treatment prevail in most liver diseases, and the mechanism of how liver diseases act differently among male versus female patients has not been fully elucidated. Biological sex differences in normal physiology and disease arise principally from sex hormones and/or sex chromosomes. Sex hormones contribute to the development and progression of most liver diseases, with estrogen- and androgen-mediated signaling pathways mechanistically involved. In addition, genetic factors in sex chromosomes have recently been found to contribute to the sex disparity of many liver diseases, which might explain, to some extent, the difference in gene expression pattern, immune response, and xenobiotic metabolism between men and women. Although increasing evidence suggests that sex is one of the most important modulators of disease prevalence and outcomes, at present, basic and clinical studies have long been sex unbalanced, with female subjects underestimated. As such, this review focuses on sex disparities of liver diseases and summarizes the current understanding of sex-specific mechanisms, including sex hormones, sex chromosomes, etc. We anticipate that understanding sex-specific pathogenesis will aid in promoting personalized therapies for liver disease among male versus female patients.
Collapse
Affiliation(s)
- Jie-Wen Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Nan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Yi Lyu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Xu-Feng Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| |
Collapse
|
10
|
Thangaraj SV, Bellingham M, Lea R, Evans N, Sinclair K, Padmanabhan V. Developmental programming: Sex-specific effects of prenatal exposure to a real-life mixture of environmental chemicals on liver function and transcriptome in sheep. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 367:125630. [PMID: 39756566 PMCID: PMC11813678 DOI: 10.1016/j.envpol.2025.125630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/01/2024] [Accepted: 01/01/2025] [Indexed: 01/07/2025]
Abstract
Humans are chronically exposed to a mixture of environmental chemicals (ECs), many with metabolic and endocrine disrupting potential, contributing to non-communicable disease burden. Understanding the effects of chronic exposure to low-level mixtures of ECs requires an animal model that reflects real-world conditions, lags behind studies on single ECs. Biosolids, from wastewater treatment, offers a real-life model to investigate the developmental health risks from EC mixtures. Prenatal biosolids exposure studies have documented metabolic perturbations including heavier thyroid glands in male fetuses and reduced bodyweight in prepubertal male lambs followed by catchup growth. We hypothesized that maternal preconceptional and gestational exposure of sheep to biosolids programs sex-specific transcriptional and functional changes in the offspring liver. Ewes (F0) were grazed on either inorganic fertilizer (C) or biosolids-treated pastures (BTP) preconception till parturition. All lambs (n = 15/group with male n = 7/group and females n = 8/group) were raised on Control pastures until euthanasia at 9.5 weeks. Next generation sequencing of liver RNA and DESeq2 was used to identify exposure-specific differentially expressed genes (DEG) and sex-differentially expressed genes (SDG). Liver function was assessed with markers of oxidative stress, triglyceride and fibrosis markers. Control lambs exhibited 647 SDGs confirming the inherent sexual dimorphism in hepatic gene expression. A sex-stratified analysis identified 10 DEG, mostly affecting metabolism, in male and none in female lambs. Biosolids exposure diminished the sexual dimorphism in hepatic gene expression barring 41 genes, potentially due to the increase in androgenic steroids found in F0 maternal circulation. Additionally, BTP male lambs showed elevated plasma triglyceride and a trend towards increased liver triglyceride concentrations. The identified effects of prenatal exposure to low-dose mixture of ECs via biosolids, in a precocial species paralleling human developmental patterns holds translational importance for understanding the sexually dimorphic origin of non-communicable diseases.
Collapse
Affiliation(s)
| | - Michelle Bellingham
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Richard Lea
- Schools of Biosciences and Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Neil Evans
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Kevin Sinclair
- Schools of Biosciences and Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | | |
Collapse
|
11
|
Du M, Wang C, Jiang Z, Cong R, Li A, Wang W, Zhang G, Li L. Genotype-by-Environment Effects of Cis-Variations in the Atgl Promoter Mediate the Divergent Pattern of Phenotypic Plasticity for Temperature Adaptation in Two Congeneric Oyster Species. Mol Ecol 2025; 34:e17623. [PMID: 39718158 DOI: 10.1111/mec.17623] [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: 01/30/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/25/2024]
Abstract
Phenotypic plasticity plays an essential role in adaptive evolution. However, the molecular mechanisms of how genotype-by-environment interaction (G × E) effects shape phenotypic plasticity in marine organisms remain poorly understood. The crucial temperature-responsive trait triacylglycerol (TAG) content and its major gene adipose triglyceride lipase (Atgl) expression have divergent plastic patterns in two congeneric oyster species (Crassostrea gigas and Crassostrea angulata) to adapt to relative-cold/northern and relative-warm/southern habitats, respectively. In this study, eight putative loci were identified in the Atgl promoter region (cis-variations) between wild C. gigas and C. angulata that exhibited differential environmental responsiveness (G × E). The G and G × E effects of each locus were further dissected by measuring the Atgl gene expression of different genotypes in response to temperature changes at the cellular and organismal levels. Two transcription factors, non-environmentally responsive non-POU domain-containing octamer-binding protein (Nono) and environmentally responsive heterogeneous nuclear ribonucleoprotein K (Hnrnpk), were screened for binding to g.-1804 (G locus) and g.-1919 (G + G × E locus), respectively. The specificity of Nono binding to the C. angulata allele mediated the G effects of g.-1804, and the lower environmental sensitivity of Hnrnpk in C. angulata mediated the G × E effects of g.-1919, jointly regulating the trade-offs between higher constitutive and lower plastic expression of Atgl gene expression in C. angulata. This study served as an experimental case to reveal how the genetic variations with G and (or) G × E effects propagate into the divergent pattern of plasticity in environmental adaptive traits, which provides new insights into predicting the adaptability of marine organisms to future climate changes.
Collapse
Affiliation(s)
- Mingyang Du
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chaogang Wang
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
| | - Zhuxiang Jiang
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Rihao Cong
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Ao Li
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- Shandong Center of Technology Innovation for Oyster Seed Industry, Qingdao, China
| | - Wei Wang
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
| | - Guofan Zhang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- Shandong Center of Technology Innovation for Oyster Seed Industry, Qingdao, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Li Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- Shandong Center of Technology Innovation for Oyster Seed Industry, Qingdao, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| |
Collapse
|
12
|
Kim YH, Jeong S, Cho KA, Woo SY, Han SH, Ryu KH. Reduction of Low-Density Lipoprotein Cholesterol by Mesenchymal Stem Cells in a Mouse Model of Exogenous Cushing's Syndrome. Tissue Eng Regen Med 2025; 22:237-248. [PMID: 39873947 PMCID: PMC11794754 DOI: 10.1007/s13770-024-00697-3] [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: 10/18/2024] [Revised: 12/11/2024] [Accepted: 12/22/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Exogenous Cushing's syndrome, which results from prolonged glucocorticoid treatment, is associated with metabolic abnormalities. Previously, we reported the inhibitory effect of tonsil-derived mesenchymal stem cell conditioned medium (T-MSC CM) on glucocorticoid signal transduction. In this study, we investigated the therapeutic efficacy of T-MSCs in a mouse model of exogenous Cushing's syndrome. METHODS Exogenous Cushing's syndrome model mice was generated by corticosterone administration in the drinking water for 5 weeks, and T-MSCs were injected intraperitoneally twice during the third week. Serum lipid profiles were measured using a chemistry analyzer. HepG2 cells were treated with dexamethasone and co-cultured with T-MSCs. Expression levels of genes involved in cholesterol metabolism were examined using real-time PCR. Low-density lipoprotein receptor (LDLR) protein levels were determined using western blotting and immunohistochemistry. Liver RNA extracted from the CORT and CORT + MSC mouse groups was used for transcriptome sequencing analysis and protein-protein interaction analysis. RESULTS Weight reduction and improvements in dyslipidemia by T-MSC administration were observed only in female mice. T-MSCs reduce circulating LDL cholesterol levels by downregulating liver X receptor α (LXRα) and inducible degrader of LDLR (IDOL) expression, thereby stabilizing LDLRs in the liver. Transcriptome analysis of liver tissue revealed pathways that are regulated by T-MSCs administration. CONCLUSION Administration of MSCs to female mice receiving chronic corticosterone treatment reduced the circulating LDL cholesterol level by downregulating the LXRα-IDOL axis in hepatocytes. These results suggest that T-MSCs may offer a novel therapeutic strategy for managing exogenous Cushing's syndrome by regulating cholesterol metabolism.
Collapse
Affiliation(s)
- Yu-Hee Kim
- Advance Biomedical Research Institute, Ewha Womans University Seoul Hospital, Seoul, South Korea
| | - Seonghee Jeong
- Ewha Medical Academy, Ewha Womans University Medical Center, Seoul, South Korea
| | - Kyung-Ah Cho
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, South Korea
| | - So-Youn Woo
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, South Korea
| | - Seung-Ho Han
- Ewha Medical Academy, Ewha Womans University Medical Center, Seoul, South Korea
- Department of Anatomy, College of Medicine, Ewha Womans University, Seoul, South Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul, 07804, South Korea.
| |
Collapse
|
13
|
Fodor Duric L, Belčić V, Oberiter Korbar A, Ćurković S, Vujicic B, Gulin T, Muslim J, Gulin M, Grgurević M, Catic Cuti E. The Role of SHBG as a Marker in Male Patients with Metabolic-Associated Fatty Liver Disease: Insights into Metabolic and Hormonal Status. J Clin Med 2024; 13:7717. [PMID: 39768643 PMCID: PMC11677371 DOI: 10.3390/jcm13247717] [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: 11/12/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/03/2025] Open
Abstract
Background: Metabolic-associated fatty liver disease (MAFLD) is a spectrum of liver diseases linked to insulin resistance (IR), type 2 diabetes, and metabolic disorders. IR accelerates fat accumulation in the liver, worsening MAFLD. Regular physical activity and weight loss can improve liver function, reduce fat, and lower cardiovascular risk. This study examines the role of sex hormone-binding globulin (SHBG) in MAFLD, focusing on its potential as a biomarker and its relationship with insulin resistance. Methods: The study included 98 male patients (ages 30-55) with MAFLD, identified through systematic examinations, and 74 healthy male controls. All participants underwent abdominal ultrasound and blood tests after fasting, assessing markers such as glucose, liver enzymes (AST, ALT, γGT), lipids (cholesterol, triglycerides), insulin, SHBG, estradiol, and testosterone. SHBG levels were analyzed in relation to body mass index (BMI) and age. Results: A significant association was found between low SHBG levels and the presence of fatty liver. Individuals with MAFLD had lower SHBG levels compared to controls. BMI and age were key factors influencing SHBG, with higher BMI linked to lower SHBG in younger men, while SHBG remained stable in older individuals regardless of BMI. Conclusion: SHBG may serve as a valuable biomarker for early detection and risk assessment of MAFLD. The complex relationship between SHBG, BMI, and age highlights the importance of considering both hormonal and metabolic factors when assessing fatty liver risk. Our findings support the need for comprehensive metabolic evaluations in clinical practice.
Collapse
Affiliation(s)
- Ljiljana Fodor Duric
- School of Medicine, University of Catholica Croatica, 10000 Zagreb, Croatia
- Department of Nephrology and Arterial Hypertension, Medikol Polyclinic, 10000 Zagreb, Croatia
| | - Velimir Belčić
- Medikol Polyclinic, 10000 Zagreb, Croatia; (V.B.); (J.M.)
| | | | - Sanja Ćurković
- Faculty of Kinesiology, University of Zagreb, 10000 Zagreb, Croatia;
- Faculty of Agriculture, University of Zagreb, 10000 Zagreb, Croatia
| | - Bozidar Vujicic
- School of Medicine, University of Rijeka, 10000 Rijeka, Croatia;
- Department of Nephrology, Dialysis and Transplantation, University Hospital Center Rijeka, 10000 Rijeka, Croatia
| | - Tonko Gulin
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
- Department of Nephrology and Arterial Hypertension, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia
| | - Jelena Muslim
- Medikol Polyclinic, 10000 Zagreb, Croatia; (V.B.); (J.M.)
| | - Matko Gulin
- Department of Radiology, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia;
| | - Mladen Grgurević
- Department of Diabetes, Endocrinology and Metabolic Diseases Vuk Vrhovac, Merkur University Hospital, 10000 Zagreb, Croatia;
| | | |
Collapse
|
14
|
Aljabali SM, Pai S, Teperino R. Paternal impact on the developmental programming of sexual dimorphism. Front Cell Dev Biol 2024; 12:1520783. [PMID: 39712575 PMCID: PMC11659275 DOI: 10.3389/fcell.2024.1520783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
Sexual dimorphism involves distinct anatomical, physiological, behavioral, and developmental differences between males and females of the same species, influenced by factors prior to conception and during early development. These sex-specific traits contribute to varied phenotypes and individual disease risks within and across generations and understanding them is essential in mammalian studies. Hormones, sex chromosomes, and imprinted genes drive this dimorphism, with over half of quantitative traits in wildtype mice showing sex-based variation. This review focuses on the impact of paternal non-genetic factors on sexual dimorphism. We synthesize current research on how paternal health before conception affects offspring phenotypes in a sex-specific manner, examining mechanisms such as DNA methylation, paternally imprinted genes, sperm RNA, and seminal plasma. Additionally, we explore how paternal influences indirectly shape offspring through maternal behavior, uterine environment, and placental changes, affecting males and females differently. We propose mechanisms modulating sexual dimorphism during development, underscoring the need for sex-specific documentation in animal studies.
Collapse
Affiliation(s)
- Shefa’ M. Aljabali
- Institute of Experimental Genetics, Helmholtz Munich GmbH, German Research Center for Environmental Health, Neuherberg, Germany
- DZD – German Center for Diabetes Research, Neuherberg, Germany
| | - Shruta Pai
- Institute of Experimental Genetics, Helmholtz Munich GmbH, German Research Center for Environmental Health, Neuherberg, Germany
- DZD – German Center for Diabetes Research, Neuherberg, Germany
| | - Raffaele Teperino
- Institute of Experimental Genetics, Helmholtz Munich GmbH, German Research Center for Environmental Health, Neuherberg, Germany
- DZD – German Center for Diabetes Research, Neuherberg, Germany
| |
Collapse
|
15
|
Dezső K, Paku S, Juhász M, Kóbori L, Nagy P. Evolutionary View of Liver Pathology. Evol Appl 2024; 17:e70059. [PMID: 39717436 PMCID: PMC11664044 DOI: 10.1111/eva.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/23/2024] [Accepted: 11/28/2024] [Indexed: 12/25/2024] Open
Abstract
Evolutionary medicine emerged in the late twentieth century, integrating principles of natural selection and adaptation with the health sciences. Today, with a rapidly widening gap between the biology of Homo sapiens and its environment, maladaptation or maladaptive disorders can be detected in almost all diseases, including liver dysfunction. However, in hepatology, as in most medical specialties, evolutionary considerations are neglected because the majority of the medical community is not familiar with evolutionary principles. The aim of this brief review is to highlight an evolutionary approach that may facilitate understanding various liver diseases.
Collapse
Affiliation(s)
- Katalin Dezső
- Department of Pathology and Experimental Cancer ResearchSemmelweis UniversityBudapestHungary
| | - Sándor Paku
- Department of Pathology and Experimental Cancer ResearchSemmelweis UniversityBudapestHungary
| | - Mária‐Manuela Juhász
- Department of Pathology and Experimental Cancer ResearchSemmelweis UniversityBudapestHungary
| | - László Kóbori
- Department of Surgery, Transplantation and GastroenterologySemmelweis UniversityBudapestHungary
| | - Péter Nagy
- Department of Pathology and Experimental Cancer ResearchSemmelweis UniversityBudapestHungary
| |
Collapse
|
16
|
Dadam FM, Gonzalez L, Vivas L, Godino A, Caeiro XE. Vasopressinergic sexual dimorphism: Sex chromosome complement and organizational hormonal effects. Mol Cell Endocrinol 2024; 594:112390. [PMID: 39427963 DOI: 10.1016/j.mce.2024.112390] [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: 07/10/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
This study aimed to analyze the role of the sex chromosomes (SCC: XX/XY) and the interaction with organizational hormonal effects on Avp gene expression at the supraoptic (SON) and paraventricular nuclei (PVN) due to water deprivation, as well as on the vasopressinergic sexually dimorphic antidiuretic and pressor responses. For this purpose, we used gonadectomized (GDX) transgenic mice of the "four core genotypes" model, in which the effect of gonadal sex and SCC are dissociated. A significant interaction between treatment and SCC on Avp gene expression at the SON was observed. Regardless of sex, XX mice showed higher basal expression than those with XY; however after water deprivation no changes in mRNA Avp expression were observed in the XX group, while an increase for XY was reported. At the PVN an interaction of SCC, organizational hormonal, and treatment factors was observed, revealing an increase in Avp gene expression in the XY-GDX male DEP group. Although no SCC or organizational hormonal effects were observed on the demopressin-antidiuretic response and renal Avpr2 mRNA expression, an interplay of organizational hormonal and SCC factors in short and medium-term vasopressin-blood pressure regulation were reported. XX-GDX females showed a facilitated vasopressin-bradycardic baroreflex response when compared to the other genotypes. Furthermore, although vasopressin continuous infusion resulted initially in the expected increase in the percentage change in MAP in all genotypes, in XX-GDX male and female this increase was sustained until the 30-min infusion, while in XY-GDX male and in XY-GDX female mice a decrease in MAP was observed.
Collapse
Affiliation(s)
- Florencia María Dadam
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Lihue Gonzalez
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Laura Vivas
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Andrea Godino
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ximena E Caeiro
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
17
|
Nascimento JDF, de Oliveira KA, de Freitas PA, Falci JDAM, Vasconcelos RP, Magalhães SC, Farias TM, Alonso-Vale MIC, Loureiro ACC, de Carvalho DP, Fortunato RS, de Oliveira AC. Increased NOX-dependent ROS production and proportionally enhanced antioxidant response in white adipose tissue of male rats. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e240136. [PMID: 39876964 PMCID: PMC11771750 DOI: 10.20945/2359-4292-2024-0136] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/06/2024] [Indexed: 01/31/2025]
Abstract
Objective This study aimed to investigate the redox balance in subcutaneous and retroperitoneal fat pads of male and female Wistar rats. Materials and methods The study analyzed the activity and gene expression of the antioxidant enzymes superoxide dismutase, catalase, and glutathione peroxidase, along with the production of NADPH oxidases dependent on H2O2 and gene expression of NOX1, NOX2, and NOX4. Results The retroperitoneal fat pad in males compared with females had greater NOX2 and NOX4 expression, along with higher superoxide dismutase activity. Additionally, their subcutaneous fat pad had greater NOX4 expression and higher intracellular H2O2 production, together with greater expression and activity of both superoxide dismutase and catalase. Conclusion The white adipose tissue of male rats had greater reactive oxygen species (ROS) production compared with that of female rats, but also a proportionally greater antioxidant response. These findings are important for ongoing investigations into how sex differences may be linked to the development of metabolic diseases and the unique susceptibilities of each sex.
Collapse
Affiliation(s)
- Jessica de Freitas Nascimento
- Universidade Estadual do CearáInstituto Superior de Ciências BiomédicasLaboratório de Fisiologia Endócrina e MetabolismoFortalezaCEBrasilLaboratório de Fisiologia Endócrina e Metabolismo, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, CE, Brasil
| | - Keciany Alves de Oliveira
- Universidade Estadual do CearáPrograma de Pós-graduação em Nutrição e SaúdeFortalezaCEBrasilPrograma de Pós-graduação em Nutrição e Saúde, Universidade Estadual do Ceará, Fortaleza, CE, Brasil
| | - Paula Alexandre de Freitas
- Universidade Estadual do CearáInstituto Superior de Ciências BiomédicasLaboratório de Fisiologia Endócrina e MetabolismoFortalezaCEBrasilLaboratório de Fisiologia Endócrina e Metabolismo, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, CE, Brasil
| | - Júlia de Araújo Marques Falci
- Universidade Federal do Rio de JaneiroInstituto de Biofísica Carlos Chagas FilhoCentro de Ciências da SaúdeRio de JaneiroRJBrasilCentro de Ciências da Saúde, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Renata Prado Vasconcelos
- Universidade Estadual do CearáInstituto Superior de Ciências BiomédicasLaboratório de Fisiologia Endócrina e MetabolismoFortalezaCEBrasilLaboratório de Fisiologia Endócrina e Metabolismo, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, CE, Brasil
| | - Saulo Chaves Magalhães
- Universidade Estadual do CearáInstituto Superior de Ciências BiomédicasLaboratório de Fisiologia Endócrina e MetabolismoFortalezaCEBrasilLaboratório de Fisiologia Endócrina e Metabolismo, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, CE, Brasil
| | - Talita Mendes Farias
- Universidade Federal de São PauloDepartamento de Ciências BiológicasPiracicabaSPBrasilDepartamento de Ciências Biológicas, Universidade Federal de São Paulo, Piracicaba, SP, Brasil
| | - Maria Isabel Cardoso Alonso-Vale
- Universidade Federal de São PauloDepartamento de Ciências BiológicasPiracicabaSPBrasilDepartamento de Ciências Biológicas, Universidade Federal de São Paulo, Piracicaba, SP, Brasil
| | - Adriano Cesar Carneiro Loureiro
- Universidade Estadual do CearáInstituto Superior de Ciências BiomédicasLaboratório de Fisiologia Endócrina e MetabolismoFortalezaCEBrasilLaboratório de Fisiologia Endócrina e Metabolismo, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, CE, Brasil
| | - Denise Pires de Carvalho
- Universidade Federal do Rio de JaneiroInstituto de Biofísica Carlos Chagas FilhoCentro de Ciências da SaúdeRio de JaneiroRJBrasilCentro de Ciências da Saúde, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Rodrigo Soares Fortunato
- Universidade Federal do Rio de JaneiroInstituto de Biofísica Carlos Chagas FilhoCentro de Ciências da SaúdeRio de JaneiroRJBrasilCentro de Ciências da Saúde, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Ariclécio Cunha de Oliveira
- Universidade Estadual do CearáInstituto Superior de Ciências BiomédicasLaboratório de Fisiologia Endócrina e MetabolismoFortalezaCEBrasilLaboratório de Fisiologia Endócrina e Metabolismo, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, CE, Brasil
- Universidade Estadual do CearáPrograma de Pós-graduação em Nutrição e SaúdeFortalezaCEBrasilPrograma de Pós-graduação em Nutrição e Saúde, Universidade Estadual do Ceará, Fortaleza, CE, Brasil
| |
Collapse
|
18
|
Zhang R, Qiu C, Filippova G, Li G, Shendure J, Vert JP, Deng X, Disteche C, Noble WS. Multi-condition and multi-modal temporal profile inference during mouse embryonic development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.03.583179. [PMID: 38496477 PMCID: PMC10942306 DOI: 10.1101/2024.03.03.583179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The emergence of single-cell time-series datasets enables modeling of changes in various types of cellular profiles over time. However, due to the disruptive nature of single-cell measurements, it is impossible to capture the full temporal trajectory of a particular cell. Furthermore, single-cell profiles can be collected at mismatched time points across different conditions (e.g., sex, batch, disease) and data modalities (e.g., scRNA-seq, scATAC-seq), which makes modeling challenging. Here we propose a joint modeling framework, Sunbear, for integrating multi-condition and multi-modal single-cell profiles across time. Sunbear can be used to impute single-cell temporal profile changes, align multi-dataset and multi-modal profiles across time, and extrapolate single-cell profiles in a missing modality. We applied Sunbear to reveal sex-biased transcription during mouse embryonic development and predict dynamic relationships between epigenetic priming and transcription for cells in which multi-modal profiles are unavailable. Sunbear thus enables the projection of single-cell time-series snapshots to multi-modal and multi-condition views of cellular trajectories.
Collapse
Affiliation(s)
- Ran Zhang
- Department of Genome Sciences, University of Washington
- eScience Institute, University of Washington
| | | | | | - Gang Li
- Department of Genome Sciences, University of Washington
- eScience Institute, University of Washington
| | - Jay Shendure
- Brotman Baty Institute for Precision Medicine, University of Washington
- Howard Hughes Medical Institute
- Allen Center for Cell Lineage Tracing
| | | | - Xinxian Deng
- Department of Pathology, University of Washington
| | | | - William Stafford Noble
- Department of Genome Sciences, University of Washington
- Paul G. Allen School of Computer Science and Engineering, University of Washington
| |
Collapse
|
19
|
Soto Sauza KA, Ryan KK. FGF21 mediating the Sex-dependent Response to Dietary Macronutrients. J Clin Endocrinol Metab 2024; 109:e1689-e1696. [PMID: 38801670 PMCID: PMC11319005 DOI: 10.1210/clinem/dgae363] [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: 03/01/2024] [Revised: 05/15/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Sex is key variable influencing body composition and substrate utilization. At rest, females maintain greater adiposity than males and resist the mobilization of fat. Males maintain greater lean muscle mass and mobilize fat readily. Determining the mechanisms that direct these sex-dependent effects is important for both reproductive and metabolic health. Here, we highlight the fundamental importance of sex in shaping metabolic physiology and assess growing evidence that the hepatokine fibroblast growth factor-21 (FGF21) plays a mechanistic role to facilitate sex-dependent responses to a changing nutritional environment. First, we examine the importance of sex in modulating body composition and substrate utilization. We summarize new data that point toward sex-biased effects of pharmacologic FGF21 administration on these endpoints. When energy is not limited, metabolic responses to FGF21 mirror broader sex differences; FGF21-treated males conserve lean mass at the expense of increased lipid catabolism, whereas FGF21-treated females conserve fat mass at the expense of reduced lean mass. Next, we examine the importance of sex in modulating the endogenous secretion of FGF21 in response to changing macronutrient and energy availability. During the resting state when energy is not limited, macronutrient imbalance increases the secretion of FGF21 more so in males than females. When energy is limited, the effect of sex on both the secretion of FGF21 and its metabolic actions may be reversed. Altogether, we argue that a growing literature supports FGF21 as a plausible mechanism contributing to the sex-dependent mobilization vs preservation of lipid storage and highlight the need for further research.
Collapse
Affiliation(s)
- Karla A Soto Sauza
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| | - Karen K Ryan
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| |
Collapse
|
20
|
Seidemann L, Lippold CP, Rohm CM, Eckel JC, Schicht G, Matz-Soja M, Berg T, Seehofer D, Damm G. Sex hormones differently regulate lipid metabolism genes in primary human hepatocytes. BMC Endocr Disord 2024; 24:135. [PMID: 39090659 PMCID: PMC11292922 DOI: 10.1186/s12902-024-01663-9] [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: 01/15/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is higher in men than in women. Hormonal and genetic causes may account for the sex differences in MASLD. Current human in vitro liver models do not sufficiently take the influence of biological sex and sex hormones into consideration. METHODS Primary human hepatocytes (PHHs) were isolated from liver specimen of female and male donors and cultured with sex hormones (17β-estradiol, testosterone and progesterone) for up to 72 h. mRNA expression levels of 8 hepatic lipid metabolism genes were analyzed by RT-qPCR. Sex hormones and their metabolites were determined in cell culture supernatants by LC-MS analyses. RESULTS A sex-specific expression was observed for LDLR (low density lipoprotein receptor) with higher mRNA levels in male than female PHHs. All three sex hormones were metabolized by PHHs and the effects of hormones on gene expression levels varied depending on hepatocyte sex. Only in female PHHs, 17β-estradiol treatment affected expression levels of PPARA (peroxisome proliferator-activated receptor alpha), LIPC (hepatic lipase) and APOL2 (apolipoprotein L2). Further changes in mRNA levels of female PHHs were observed for ABCA1 (ATP-binding cassette, sub-family A, member 1) after testosterone and for ABCA1, APOA5 (apolipoprotein A-V) and PPARA after progesterone treatment. Only the male PHHs showed changing mRNA levels for LDLR after 17β-estradiol and for APOA5 after testosterone treatment. CONCLUSIONS Male and female PHHs showed differences in their expression levels of hepatic lipid metabolism genes and their responsiveness towards sex hormones. Thus, cellular sex should be considered, especially when investigating the pathophysiological mechanisms of MASLD.
Collapse
Affiliation(s)
- Lena Seidemann
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, 04103, Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103, Leipzig, Germany
| | - Clara Paula Lippold
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, 04103, Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103, Leipzig, Germany
| | - Carolin Marie Rohm
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, 04103, Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103, Leipzig, Germany
| | - Julian Connor Eckel
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, 04103, Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103, Leipzig, Germany
| | - Gerda Schicht
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, 04103, Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103, Leipzig, Germany
| | - Madlen Matz-Soja
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, 04103, Leipzig, Germany
| | - Thomas Berg
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, 04103, Leipzig, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, 04103, Leipzig, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, 04103, Leipzig, Germany.
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103, Leipzig, Germany.
| |
Collapse
|
21
|
Booijink R, Ramachandran P, Bansal R. Implications of innate immune sexual dimorphism for MASLD pathogenesis and treatment. Trends Pharmacol Sci 2024; 45:614-627. [PMID: 38853100 DOI: 10.1016/j.tips.2024.05.004] [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: 04/10/2024] [Revised: 04/18/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
Growing evidence suggests that metabolic dysfunction-associated steatotic liver disease (MASLD) is significantly higher in men versus women. Increased prevalence is observed in postmenopausal women, suggesting that age and sex (hormones) influence MASLD development and progression. Molecular data further reveal that sex regulates the innate immune responses with an essential role in MASLD progression. To date, there has been limited focus on the role of innate immune sexual dimorphism in MASLD, and differences between men and women are not considered in the current drug discovery landscape. In this review, we summarize the sex disparities and innate immune sexual dimorphism in MASLD pathogenesis. We further highlight the importance of harnessing sexual dimorphism in identifying therapeutic targets, developing pharmacological therapies, and designing (pre-) clinical studies for the personalized treatment for MASLD.
Collapse
Affiliation(s)
- Richell Booijink
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Prakash Ramachandran
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, Edinburgh, UK
| | - Ruchi Bansal
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
22
|
Su L, Luo H, Yan Y, Yang Z, Lu J, Xu D, Du L, Liu J, Yang G, Chi H. Exploiting gender-based biomarkers and drug targets: advancing personalized therapeutic strategies in hepatocellular carcinoma. Front Pharmacol 2024; 15:1433540. [PMID: 38966543 PMCID: PMC11222576 DOI: 10.3389/fphar.2024.1433540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 07/06/2024] Open
Abstract
This review systematically examines gender differences in hepatocellular carcinoma (HCC), identifying the influence of sex hormones, genetic variance, and environmental factors on the disease's epidemiology and treatment outcomes. Recognizing the liver as a sexually dimorphic organ, we highlight how gender-specific risk factors, such as alcohol consumption and obesity, contribute differently to hepatocarcinogenesis in men and women. We explore molecular mechanisms, including the differential expression of androgen and estrogen receptors, which mediate diverse pathways in tumor biology such as cell proliferation, apoptosis, and DNA repair. Our analysis underscores the critical need for gender-specific research in liver cancer, from molecular studies to clinical trials, to improve diagnostic accuracy and therapeutic effectiveness. By incorporating a gender perspective into all facets of liver cancer research, we advocate for a more precise and personalized approach to cancer treatment that acknowledges gender as a significant factor in both the progression of HCC and its response to treatment. This review aims to foster a deeper understanding of the biological and molecular bases of gender differences in HCC and to promote the development of tailored interventions that enhance outcomes for all patients.
Collapse
Affiliation(s)
- Lanqian Su
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Huanyu Luo
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Yalan Yan
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Zhongqiu Yang
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Jiaan Lu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Danqi Xu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Linjuan Du
- Department of Oncology, Dazhou Central Hospital, Dazhou, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| |
Collapse
|
23
|
Younes S. The relationship between gender and pharmacology. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 7:100192. [PMID: 39101002 PMCID: PMC11295939 DOI: 10.1016/j.crphar.2024.100192] [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: 04/14/2024] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 08/05/2024] Open
Abstract
The part of sexuality in pharmacology research was not acknowledged, and it was not thought-out to be a determinant that could impact strength and disease. For decades research has mainly contained male, women and animals, leading to a lack of news about syndromes in females. Still, it is critical to guarantee equal likeness so that determine the security, influence, and resistance of healing agents for all individuals. The underrepresentation of female models in preclinical studies over various decades has surpassed to disparities in the understanding, disease, and treatment of ailments 'tween genders. The closeness of sexuality bias has happened recognized as a contributing determinant to the restricted interpretation and replicability of preclinical research. Many demands operation have stressed the significance of including sexuality as a organic changeable, and this view is acquire growing support. Regardless of important progress in incorporating more female models into preclinical studies, differences prevail contemporary. The current review focuses on the part of sexuality and common in biomedical research and, therefore, their potential function in pharmacology and analyze the potential risks guide.
Collapse
Affiliation(s)
- Samer Younes
- Department of Pharmacy, Tartous University, Syria
| |
Collapse
|
24
|
Wilson CA, Postlethwait JH. A maternal-to-zygotic-transition gene block on the zebrafish sex chromosome. G3 (BETHESDA, MD.) 2024; 14:jkae050. [PMID: 38466753 PMCID: PMC11075544 DOI: 10.1093/g3journal/jkae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/22/2024] [Accepted: 03/01/2024] [Indexed: 03/13/2024]
Abstract
Wild zebrafish (Danio rerio) have a ZZ/ZW chromosomal sex-determination system with the major sex locus on the right arm of chromosome-4 (Chr4R) near the largest heterochromatic block in the genome, suggesting that Chr4R transcriptomics might differ from the rest of the genome. To test this hypothesis, we conducted an RNA-seq analysis of adult ZW ovaries and ZZ testes in the Nadia strain and identified 4 regions of Chr4 with different gene expression profiles. Unique in the genome, protein-coding genes in a 41.7 Mb section (Region-2) were expressed in testis but silent in ovary. The AB lab strain, which lacks sex chromosomes, verified this result, showing that testis-biased gene expression in Region-2 depends on gonad biology, not on sex-determining mechanism. RNA-seq analyses in female and male brains and livers validated reduced transcripts from Region-2 in somatic cells, but without sex specificity. Region-2 corresponds to the heterochromatic portion of Chr4R and its content of genes and repetitive elements distinguishes it from the rest of the genome. Region-2 lacks protein-coding genes with human orthologs; has zinc finger genes expressed early in zygotic genome activation; has maternal 5S rRNA genes, maternal spliceosome genes, a concentration of tRNA genes, and a distinct set of repetitive elements. The colocalization of (1) genes silenced in ovaries but not in testes that are (2) expressed in embryos briefly at the onset of zygotic genome activation; (3) maternal-specific genes for translation machinery; (4) maternal-specific spliceosome components; and (5) adjacent genes encoding miR-430, which mediates maternal transcript degradation, suggest that this is a maternal-to-zygotic-transition gene regulatory block.
Collapse
|
25
|
Li F, Xing X, Jin Q, Wang XM, Dai P, Han M, Shi H, Zhang Z, Shao X, Peng Y, Zhu Y, Xu J, Li D, Chen Y, Wu W, Wang Q, Yu C, Chen L, Bai F, Gao D. Sex differences orchestrated by androgens at single-cell resolution. Nature 2024; 629:193-200. [PMID: 38600383 DOI: 10.1038/s41586-024-07291-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
Sex differences in mammalian complex traits are prevalent and are intimately associated with androgens1-7. However, a molecular and cellular profile of sex differences and their modulation by androgens is still lacking. Here we constructed a high-dimensional single-cell transcriptomic atlas comprising over 2.3 million cells from 17 tissues in Mus musculus and explored the effects of sex and androgens on the molecular programs and cellular populations. In particular, we found that sex-biased immune gene expression and immune cell populations, such as group 2 innate lymphoid cells, were modulated by androgens. Integration with the UK Biobank dataset revealed potential cellular targets and risk gene enrichment in antigen presentation for sex-biased diseases. This study lays the groundwork for understanding the sex differences orchestrated by androgens and provides important evidence for targeting the androgen pathway as a broad therapeutic strategy for sex-biased diseases.
Collapse
Affiliation(s)
- Fei Li
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xudong Xing
- Biomedical Pioneering Innovation Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics (ICG), School of Life Sciences, Peking University, Beijing, China
| | - Qiqi Jin
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiang-Ming Wang
- Biomedical Pioneering Innovation Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics (ICG), School of Life Sciences, Peking University, Beijing, China
| | - Pengfei Dai
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ming Han
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Huili Shi
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ze Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Xianlong Shao
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yunyi Peng
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yiqin Zhu
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Jiayi Xu
- Shanghai Normal University, Shanghai, China
| | - Dan Li
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wei Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiao Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Yu
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China.
| | - Luonan Chen
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics (ICG), School of Life Sciences, Peking University, Beijing, China.
| | - Dong Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
26
|
Astafev AA, Mezhnina V, Poe A, Jiang P, Kondratov RV. Sexual dimorphism of circadian liver transcriptome. iScience 2024; 27:109483. [PMID: 38550984 PMCID: PMC10973666 DOI: 10.1016/j.isci.2024.109483] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/14/2024] [Accepted: 03/08/2024] [Indexed: 11/12/2024] Open
Abstract
Sexual dimorphism affects various aspects of physiology, metabolism and longevity. Circadian clock is a master regulator of metabolism. Anti-aging dietary interventions reprogram circadian transcriptome in the liver and other tissues, but little is known about sexual dimorphism of circadian transcriptome. We compared circadian transcriptomes in the liver of male and female mice on ad libitum (AL) and 30% caloric restriction (CR) diets. We found that AL female mice had a larger number of oscillating genes than male mice, and the portion of the transcriptome with sex-specific rhythms displayed phase difference. We found that CR increased the number of oscillating genes in both sexes and strongly synchronized the transcriptome without complete elimination of sex dimorphism in rhythms. Sex also had an effect on the response of the rhythms to CR. Gene ontology analysis revealed sex-specific signatures in metabolic pathways, which suggests a complex interaction of sex, circadian rhythms, and diet.
Collapse
Affiliation(s)
- Artem A. Astafev
- Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, Cleveland, OH 44115, USA
- Department of Biological Geological and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Volha Mezhnina
- Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, Cleveland, OH 44115, USA
- Department of Biological Geological and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Allan Poe
- Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, Cleveland, OH 44115, USA
- Department of Biological Geological and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Peng Jiang
- Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, Cleveland, OH 44115, USA
- Department of Biological Geological and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
- Center for Applied Data Analysis and Modeling (ADAM), Cleveland State University, Cleveland, OH 44115, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Roman V. Kondratov
- Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, Cleveland, OH 44115, USA
- Department of Biological Geological and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| |
Collapse
|
27
|
Hauth A, Panten J, Kneuss E, Picard C, Servant N, Rall I, Pérez-Rico YA, Clerquin L, Servaas N, Villacorta L, Jung F, Luong C, Chang HY, Zaugg JB, Stegle O, Odom DT, Loda A, Heard E. Escape from X inactivation is directly modulated by levels of Xist non-coding RNA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.22.581559. [PMID: 38559194 PMCID: PMC10979913 DOI: 10.1101/2024.02.22.581559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
In placental females, one copy of the two X chromosomes is largely silenced during a narrow developmental time window, in a process mediated by the non-coding RNA Xist1. Here, we demonstrate that Xist can initiate X-chromosome inactivation (XCI) well beyond early embryogenesis. By modifying its endogenous level, we show that Xist has the capacity to actively silence genes that escape XCI both in neuronal progenitor cells (NPCs) and in vivo, in mouse embryos. We also show that Xist plays a direct role in eliminating TAD-like structures associated with clusters of escapee genes on the inactive X chromosome, and that this is dependent on Xist's XCI initiation partner, SPEN2. We further demonstrate that Xist's function in suppressing gene expression of escapees and topological domain formation is reversible for up to seven days post-induction, but that sustained Xist up-regulation leads to progressively irreversible silencing and CpG island DNA methylation of facultative escapees. Thus, the distinctive transcriptional and regulatory topologies of the silenced X chromosome is actively, directly - and reversibly - controlled by Xist RNA throughout life.
Collapse
Affiliation(s)
- Antonia Hauth
- European Molecular Biology Laboratory, Directors' Research, 69117 Heidelberg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Germany
| | - Jasper Panten
- Division of Regulatory Genomics and Cancer Evolution, German Cancer Research Centre (DKFZ), 69120, Heidelberg, Germany
- Division of Computational Genomics and Systems Genetics, German Cancer Research Centre (DKFZ), 69120, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69117, Heidelberg, Germany
| | - Emma Kneuss
- European Molecular Biology Laboratory, Directors' Research, 69117 Heidelberg, Germany
| | - Christel Picard
- European Molecular Biology Laboratory, Directors' Research, 69117 Heidelberg, Germany
- Present address: Institute of Molecular Genetics of Montpellier University of Montpellier, CNRS, 34090 Montpellier, France
| | - Nicolas Servant
- Bioinformatics and Computational Systems Biology of Cancer, INSERM U900, Paris 75005, France
| | - Isabell Rall
- European Molecular Biology Laboratory, Directors' Research, 69117 Heidelberg, Germany
- Present address: Institute of Human Biology (IHB), Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Yuvia A Pérez-Rico
- European Molecular Biology Laboratory, Directors' Research, 69117 Heidelberg, Germany
| | - Lena Clerquin
- European Molecular Biology Laboratory, Directors' Research, 69117 Heidelberg, Germany
| | - Nila Servaas
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117 Heidelberg, Germany
| | - Laura Villacorta
- European Molecular Biology Laboratory, Genomics Core Facility, 69117 Heidelberg, Germany
| | - Ferris Jung
- European Molecular Biology Laboratory, Genomics Core Facility, 69117 Heidelberg, Germany
| | - Christy Luong
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Judith B Zaugg
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117 Heidelberg, Germany
- Molecular Medicine Partnership Unit, EMBL-University of Heidelberg, Heidelberg, Germany
| | - Oliver Stegle
- European Molecular Biology Laboratory, Genome Biology Unit, 69117 Heidelberg, Germany
- Division of Computational Genomics and Systems Genetics, German Cancer Research Centre (DKFZ), 69120, Heidelberg, Germany
| | - Duncan T Odom
- Division of Regulatory Genomics and Cancer Evolution, German Cancer Research Centre (DKFZ), 69120, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69117, Heidelberg, Germany
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Agnese Loda
- European Molecular Biology Laboratory, Directors' Research, 69117 Heidelberg, Germany
| | - Edith Heard
- European Molecular Biology Laboratory, Directors' Research, 69117 Heidelberg, Germany
- Collège de France, Paris 75005, France
| |
Collapse
|
28
|
Brie B, Sarmento-Cabral A, Pascual F, Cordoba-Chacon J, Kineman RD, Becu-Villalobos D. Modifications of the GH Axis Reveal Unique Sexually Dimorphic Liver Signatures for Lcn13, Asns, Hamp2, Hao2, and Pgc1a. J Endocr Soc 2024; 8:bvae015. [PMID: 38370444 PMCID: PMC10872697 DOI: 10.1210/jendso/bvae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Indexed: 02/20/2024] Open
Abstract
Growth hormone (GH) modifies liver gene transcription in a sexually dimorphic manner to meet liver metabolic demands related to sex; thus, GH dysregulation leads to sex-biased hepatic disease. We dissected the steps of the GH regulatory cascade modifying GH-dependent genes involved in metabolism, focusing on the male-predominant genes Lcn13, Asns, and Cyp7b1, and the female-predominant genes Hao2, Pgc1a, Hamp2, Cyp2a4, and Cyp2b9. We explored mRNA expression in 2 settings: (i) intact liver GH receptor (GHR) but altered GH and insulin-like growth factor 1 (IGF1) levels (NeuroDrd2KO, HiGH, aHepIGF1kd, and STAT5bCA mouse lines); and (ii) liver loss of GHR, with or without STAT5b reconstitution (aHepGHRkd, and aHepGHRkd + STAT5bCA). Lcn13 was downregulated in males in most models, while Asns and Cyp7b1 were decreased in males by low GH levels or action, or constant GH levels, but unexpectedly upregulated in both sexes by the loss of liver Igf1 or constitutive Stat5b expression. Hao, Cyp2a4, and Cyp2b9 were generally decreased in female mice with low GH levels or action (NeuroDrd2KO and/or aHepGHRkd mice) and increased in HiGH females, while in contrast, Pgc1a was increased in female NeuroDrd2KO but decreased in STAT5bCA and aHepIGF1kd females. Bioinformatic analysis of RNAseq from aHepGHRkd livers stressed the greater impact of GHR loss on wide gene expression in males and highlighted that GH modifies almost completely different gene signatures in each sex. Concordantly, we show that altering different steps of the GH cascade in the liver modified liver expression of Lcn13, Asns, Cyp7b1, Hao2, Hamp2, Pgc1a, Cyp2a4, and Cyp2b9 in a sex- and gene-specific manner.
Collapse
Affiliation(s)
- Belen Brie
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1428 Ciudad de Buenos Aires, Argentina
| | - Andre Sarmento-Cabral
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Florencia Pascual
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1428 Ciudad de Buenos Aires, Argentina
| | - Jose Cordoba-Chacon
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rhonda Denise Kineman
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL 60612, USA
- Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| | - Damasia Becu-Villalobos
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1428 Ciudad de Buenos Aires, Argentina
| |
Collapse
|
29
|
Christians JK, Reue K. The role of gonadal hormones and sex chromosomes in sex-dependent effects of early nutrition on metabolic health. Front Endocrinol (Lausanne) 2023; 14:1304050. [PMID: 38189044 PMCID: PMC10770830 DOI: 10.3389/fendo.2023.1304050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Early-life conditions such as prenatal nutrition can have long-term effects on metabolic health, and these effects may differ between males and females. Understanding the biological mechanisms underlying sex differences in the response to early-life environment will improve interventions, but few such mechanisms have been identified, and there is no overall framework for understanding sex differences. Biological sex differences may be due to chromosomal sex, gonadal sex, or interactions between the two. This review describes approaches to distinguish between the roles of chromosomal and gonadal sex, and summarizes findings regarding sex differences in metabolism. The Four Core Genotypes (FCG) mouse model allows dissociation of the sex chromosome genotype from gonadal type, whereas the XY* mouse model can be used to distinguish effects of X chromosome dosage vs the presence of the Y chromosome. Gonadectomy can be used to distinguish between organizational (permanent) and activational (reversible) effects of sex hormones. Baseline sex differences in a variety of metabolic traits are influenced by both activational and organizational effects of gonadal hormones, as well as sex chromosome complement. Thus far, these approaches have not been widely applied to examine sex-dependent effects of prenatal conditions, although a number of studies have found activational effects of estradiol to be protective against the development of hypertension following early-life adversity. Genes that escape X chromosome inactivation (XCI), such as Kdm5c, contribute to baseline sex-differences in metabolism, while Ogt, another XCI escapee, leads to sex-dependent responses to prenatal maternal stress. Genome-wide approaches to the study of sex differences include mapping genetic loci influencing metabolic traits in a sex-dependent manner. Seeking enrichment for binding sites of hormone receptors among genes showing sexually-dimorphic expression can elucidate the relative roles of hormones. Using the approaches described herein to identify mechanisms underlying sex-dependent effects of early nutrition on metabolic health may enable the identification of fundamental mechanisms and potential interventions.
Collapse
Affiliation(s)
- Julian K. Christians
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
- Women’s Health Research Institute, BC Women’s Hospital and Health Centre, Vancouver, BC, Canada
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
30
|
Wilson CA, Postlethwait JH. A maternal-to-zygotic-transition gene block on the zebrafish sex chromosome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.06.570431. [PMID: 38106184 PMCID: PMC10723407 DOI: 10.1101/2023.12.06.570431] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Wild zebrafish (Danio rerio) have a ZZ/ZW chromosomal sex determination system with the major sex locus on the right arm of chromosome-4 (Chr4R) near the largest heterochromatic block in the genome, suggesting the hypothesis that the Chr4R transcriptome might be different from the rest of the genome. We conducted an RNA-seq analysis of adult ZW ovaries and ZZ testes and identified four regions of Chr4 with different gene expression profiles. Unique in the genome, protein-coding genes in a 41.7 Mb section (Region-2) were expressed in testis but silent in ovary. The AB lab strain, which lacks sex chromosomes, verified this result, showing that testis-biased gene expression in Region-2 depends on gonad biology, not on sex-determining mechanism. RNA-seq analyses in female and male brain and liver validated few transcripts from Region-2 in somatic cells, but without sex-specificity. Region-2 corresponds to the heterochromatic portion of Chr4R and its content of genes and repetitive elements distinguishes it from the rest of the genome. In Region-2, protein-coding genes lack human orthologs; it has zinc finger genes expressed early in zygotic genome activation; it has maternal 5S rRNA genes, maternal spliceosome genes, a concentration of tRNA genes, and an distinct set of repetitive elements. The colocalization of 1) genes silenced in ovaries but not in testes that are 2) expressed in embryos briefly at the onset of zygotic genome activation; 3) maternal-specific genes for translation machinery; 4) maternal-specific spliceosome components; and 4) adjacent genes encoding miR-430, which mediates maternal transcript degradation, suggest that this is a Maternal-to-Zygotic-Transition Gene Regulatory Block.
Collapse
|
31
|
Bahijri S, Eldakhakhny B, Enani S, Ajabnoor G, Al-Mowallad AS, Alsheikh L, Alhozali A, Alamoudi AA, Borai A, Tuomilehto J. Fibroblast Growth Factor 21: A More Effective Biomarker Than Free Fatty Acids and Other Insulin Sensitivity Measures for Predicting Non-alcoholic Fatty Liver Disease in Saudi Arabian Type 2 Diabetes Patients. Cureus 2023; 15:e50524. [PMID: 38222178 PMCID: PMC10787595 DOI: 10.7759/cureus.50524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2023] [Indexed: 01/16/2024] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is more prevalent among individuals with type 2 diabetes (T2DM), elevating their risk of cardiovascular diseases (CVDs) and premature mortality. There is a need to modify treatment strategies to prevent or delay these adverse outcomes. Currently, there are no sensitive or specific biomarkers for predicting NAFLD in Saudi T2DM patients. Therefore, we aimed to explore the possibility of using fibroblast growth factor 21 (FGF-21), free fatty acids (FFAs), homeostatic model assessment for insulin resistance (HOMA-IR), and quantitative insulin sensitivity check index (QUICKI) as possible markers. Methodology In this study, a total of 67 T2DM patients were recruited. NAFLD was detected by ultrasonography in 28 patients. Plasma glucose, FFAs, FGF-21, and serum insulin were measured in fasting blood samples. HOMA-IR and QUICKI were calculated. The means of the two groups with and without NAFLD were statistically compared. The receiver operating characteristics (ROC) curve and the area under the curve (AUC) were used to assess the ability to identify NAFLD. Results The mean levels of FGF-21 and HOMA-IR were significantly higher and that of QUICKI was significantly lower in patients with NAFLD than in those without (p < 0.001, p = 0.023, and p = 0.018, respectively). FGF-21 had the highest AUC to identify NAFLD (AUC = 0.981, 95% confidence interval = 0.954-1, P < 0.001). The AUCs for HOMA-IR, QUICKI, and FFA were <0.7. The highest sensitivity, specificity, positive likelihood ratio, and the lowest negative likelihood ratio were found when FGF-21 was used to predict NAFLD. Conclusions FGF-21 may be used as a biomarker to predict NAFLD in people with T2DM due to its high sensitivity and specificity compared to the other markers.
Collapse
Affiliation(s)
- Suhad Bahijri
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, SAU
| | - Basmah Eldakhakhny
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, SAU
| | - Sumia Enani
- Department of Food and Nutrition, Faculty of Human Sciences and Design, King Abdulaziz University, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, SAU
| | - Ghada Ajabnoor
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, SAU
| | - Alaa S Al-Mowallad
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Lubna Alsheikh
- Department of Biochemistry, King Abdulaziz University, Jeddah, SAU
| | - Amani Alhozali
- Department of Internal Medicine, King Abdulaziz University Hospital, Jeddah, SAU
| | - Aliaa A Alamoudi
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
| | - Anwar Borai
- King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
| | - Jaakko Tuomilehto
- Department of Public Health, University of Helsinki, Helsinki, FIN
- Public Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, FIN
| |
Collapse
|
32
|
Chiodi V, Rappa F, Lo Re O, Chaldakov GN, Lelouvier B, Micale V, Domenici MR, Vinciguerra M. Deficiency of histone variant macroH2A1.1 is associated with sexually dimorphic obesity in mice. Sci Rep 2023; 13:19123. [PMID: 37926763 PMCID: PMC10625986 DOI: 10.1038/s41598-023-46304-8] [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: 05/08/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023] Open
Abstract
Obesity has a major socio-economic health impact. There are profound sex differences in adipose tissue deposition and obesity-related conditions. The underlying mechanisms driving sexual dimorphism in obesity and its associated metabolic disorders remain unclear. Histone variant macroH2A1.1 is a candidate epigenetic mechanism linking environmental and dietary factors to obesity. Here, we used a mouse model genetically depleted of macroH2A1.1 to investigate its potential epigenetic role in sex dimorphic obesity, metabolic disturbances and gut dysbiosis. Whole body macroH2A1 knockout (KO) mice, generated with the Cre/loxP technology, and their control littermates were fed a high fat diet containing 60% of energy derived from fat. The diet was administered for three months starting from 10 to 12 weeks of age. We evaluated the progression in body weight, the food intake, and the tolerance to glucose by means of a glucose tolerance test. Gut microbiota composition, visceral adipose and liver tissue morphology were assessed. In addition, adipogenic gene expression patterns were evaluated in the visceral adipose tissue. Female KO mice for macroH2A1.1 had a more pronounced weight gain induced by high fat diet compared to their littermates, while the increase in body weight in male mice was similar in the two genotypes. Food intake was generally increased upon KO and decreased by high fat diet in both sexes, with the exception of KO females fed a high fat diet that displayed the same food intake of their littermates. In glucose tolerance tests, glucose levels were significantly elevated upon high fat diet in female KO compared to a standard diet, while this effect was absent in male KO. There were no differences in hepatic histology. Upon a high fat diet, in female adipocyte cross-sectional area was larger in KO compared to littermates: activation of proadipogenic genes (ACACB, AGT, ANGPT2, FASN, RETN, SLC2A4) and downregulation of antiadipogenic genes (AXIN1, E2F1, EGR2, JUN, SIRT1, SIRT2, UCP1, CCND1, CDKN1A, CDKN1B, EGR2) was detected. Gut microbiota profiling showed increase in Firmicutes and a decrease in Bacteroidetes in females, but not males, macroH2A1.1 KO mice. MacroH2A1.1 KO mice display sexual dimorphism in high fat diet-induced obesity and in gut dysbiosis, and may represent a useful model to investigate epigenetic and metabolic differences associated to the development of obesity-associated pathological conditions in males and females.
Collapse
Affiliation(s)
- Valentina Chiodi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanita', Rome, Italy
| | - Francesca Rappa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Oriana Lo Re
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
- International Clinical Research Center (FNUSA-ICRC), St'Anne University Hospital, Brno, Czech Republic
| | - George N Chaldakov
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
- Department of Anatomy and Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
| | | | - Vincenzo Micale
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Maria Rosaria Domenici
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanita', Rome, Italy
| | - Manlio Vinciguerra
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria.
- International Clinical Research Center (FNUSA-ICRC), St'Anne University Hospital, Brno, Czech Republic.
- Liverpool Centre for Cardiovascular Science (LCCS), Liverpool John Moores University, Liverpool, UK.
| |
Collapse
|
33
|
Rodríguez-Montes L, Ovchinnikova S, Yuan X, Studer T, Sarropoulos I, Anders S, Kaessmann H, Cardoso-Moreira M. Sex-biased gene expression across mammalian organ development and evolution. Science 2023; 382:eadf1046. [PMID: 37917687 PMCID: PMC7615307 DOI: 10.1126/science.adf1046] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 09/18/2023] [Indexed: 11/04/2023]
Abstract
Sexually dimorphic traits are common among mammals and are specified during development through the deployment of sex-specific genetic programs. Because little is known about these programs, we investigated them using a resource of gene expression profiles in males and females throughout the development of five organs in five mammals (human, mouse, rat, rabbit, and opossum) and a bird (chicken). We found that sex-biased gene expression varied considerably across organs and species and was often cell-type specific. Sex differences increased abruptly around sexual maturity instead of increasing gradually during organ development. Finally, sex-biased gene expression evolved rapidly at the gene level, with differences between organs in the evolutionary mechanisms used, but more slowly at the cellular level, with the same cell types being sexually dimorphic across species.
Collapse
Affiliation(s)
- Leticia Rodríguez-Montes
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | | | - Xuefei Yuan
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Tania Studer
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Ioannis Sarropoulos
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Simon Anders
- BioQuant, Heidelberg University, D-69120 Heidelberg, Germany
| | - Henrik Kaessmann
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | | |
Collapse
|
34
|
Willekens J, Mosca P, Burt-Oberecken N, Laugeais E, Kaoma T, Bernardin F, Vallar L, Dimofski P, Renaud M, Lambert L, Leheup B, Guéant JL, Leininger-Muller B, Dreumont N. Cross-Talk between miRNAs from the Dlk1-Dio3 Locus and Histone Methylation to Protect Male Cerebellum from Methyl Donor Deficiency. Mol Nutr Food Res 2023; 67:e2300040. [PMID: 37672803 DOI: 10.1002/mnfr.202300040] [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: 01/24/2023] [Revised: 05/12/2023] [Indexed: 09/08/2023]
Abstract
SCOPE Disruption of the one carbon metabolism during development, i.e., following a gestational vitamin B9 and B12 deficiencies, is involved in birth defects and brain development delay. Using a rat nutritional model, consisting of pups born to dams fed a vitamin B9 and B12 deficient diet (MDD), the study previously reports molecular and cellular alterations in the brain, in a sex dependent manner, with females being more affected than males. The study hypothesizes that epigenetic modifications could participate in the sex differences is observed. METHODS AND RESULTS The study investigates lysine methylation of histones and expression of microRNAs in the cerebellum of MDD male and female pups. The study reports a differential regulation of H3K36Me2 and H4K20Me3 between males and females, in response to MDD. Moreover, distinct regulation of Kmt5b and Kdm2a expression by miR-134-5p and miR-369-5p from the Dlk1-Dio3 locus, contributes to the maintenance of expression of genes involved in synaptic plasticity. CONCLUSION These results could explain the neuroprotection to MDD that male pups display. The work will contribute to the understanding of the consequences of vitamin starvation on brain development, as well as how the epigenome is affected by one carbon metabolism disruption.
Collapse
Affiliation(s)
- Jeremy Willekens
- Université de Lorraine, Inserm, NGERE, Nancy, F-54000, France
- CINJ, Rutgers Cancer Institute of New Jersey, 195 Little Albany St, New Brunswick, NJ 08901, USA
| | - Pauline Mosca
- Université de Lorraine, Inserm, NGERE, Nancy, F-54000, France
| | | | - Edgar Laugeais
- Université de Lorraine, Inserm, NGERE, Nancy, F-54000, France
| | - Tony Kaoma
- Luxembourg Institute of Health, Bioinformatics Platform, 1 A-B, Luxembourg, L-1445, Luxembourg
| | - François Bernardin
- Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, L-1210, Luxembourg
| | - Laurent Vallar
- Luxembourg Institute of Health, Bioinformatics Platform, 1 A-B, Luxembourg, L-1445, Luxembourg
| | | | - Mathilde Renaud
- Université de Lorraine, Inserm, NGERE, Nancy, F-54000, France
- CHRU Nancy, Hôpital d'enfants, Service de Génétique Clinique, Nancy, F-54000, France
| | - Laetitia Lambert
- Université de Lorraine, Inserm, NGERE, Nancy, F-54000, France
- CHRU Nancy, Hôpital d'enfants, Service de Génétique Clinique, Nancy, F-54000, France
| | - Bruno Leheup
- Université de Lorraine, Inserm, NGERE, Nancy, F-54000, France
- CHRU Nancy, Hôpital d'enfants, Service de Génétique Clinique, Nancy, F-54000, France
| | | | | | | |
Collapse
|
35
|
Teefy BB, Lemus AJJ, Adler A, Xu A, Bhala R, Hsu K, Benayoun BA. Widespread sex dimorphism across single-cell transcriptomes of adult African turquoise killifish tissues. Cell Rep 2023; 42:113237. [PMID: 37837621 PMCID: PMC10842523 DOI: 10.1016/j.celrep.2023.113237] [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: 05/05/2023] [Revised: 08/18/2023] [Accepted: 09/25/2023] [Indexed: 10/16/2023] Open
Abstract
The African turquoise killifish (Nothobranchius furzeri), the shortest-lived vertebrate that can be bred in captivity, is an emerging model organism for aging research. Here, we describe a multitissue, single-cell gene expression atlas of female and male blood, kidney, liver, and spleen. We annotate 22 cell types, define marker genes, and infer differentiation trajectories. We find pervasive sex-dimorphic gene expression across cell types. Sex-dimorphic genes tend to be linked to lipid metabolism, consistent with clear differences in lipid storage in female vs. male turquoise killifish livers. We use machine learning to predict sex using single-cell gene expression and identify potential markers for molecular sex identity. As a proof of principle, we show that our atlas can be used to deconvolute existing bulk RNA sequencing (RNA-seq) data to obtain accurate estimates of cell type proportions. This atlas can be a resource to the community that could be leveraged to develop cell-type-specific expression in transgenic animals.
Collapse
Affiliation(s)
- Bryan B Teefy
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Aaron J J Lemus
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts, and Sciences, Los Angeles, CA 90089, USA
| | - Ari Adler
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Alan Xu
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; Quantitative & Computational Biology Department, USC Dornsife College of Letters, Arts, and Sciences, Los Angeles, CA 90089, USA
| | - Rajyk Bhala
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Katelyn Hsu
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts, and Sciences, Los Angeles, CA 90089, USA
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts, and Sciences, Los Angeles, CA 90089, USA; Biochemistry and Molecular Medicine Department, USC Keck School of Medicine, Los Angeles, CA 90089, USA; Epigenetics and Gene Regulation, USC Norris Comprehensive Cancer Center, Los Angeles, CA 90089, USA; USC Stem Cell Initiative, Los Angeles, CA 90089, USA.
| |
Collapse
|
36
|
Asbjornsdottir B, Sigurdsson S, Miranda-Ribera A, Fiorentino M, Konno T, Lan J, Gudmundsson LS, Gottfredsson M, Lauth B, Birgisdottir BE, Fasano A. Evaluating Prophylactic Effect of Bovine Colostrum on Intestinal Barrier Function in Zonulin Transgenic Mice: A Transcriptomic Study. Int J Mol Sci 2023; 24:14730. [PMID: 37834178 PMCID: PMC10572565 DOI: 10.3390/ijms241914730] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
The intestinal barrier comprises a single layer of epithelial cells tightly joined to form a physical barrier. Disruption or compromise of the intestinal barrier can lead to the inadvertent activation of immune cells, potentially causing an increased risk of chronic inflammation in various tissues. Recent research has suggested that specific dietary components may influence the function of the intestinal barrier, potentially offering a means to prevent or mitigate inflammatory disorders. However, the precise mechanism underlying these effects remains unclear. Bovine colostrum (BC), the first milk from cows after calving, is a natural source of nutrients with immunomodulatory, anti-inflammatory, and gut-barrier fortifying properties. This novel study sought to investigate the transcriptome in BC-treated Zonulin transgenic mice (Ztm), characterized by dysbiotic microbiota, intestinal hyperpermeability, and mild hyperactivity, applying RNA sequencing. Seventy-five tissue samples from the duodenum, colon, and brain of Ztm and wild-type (WT) mice were dissected, processed, and RNA sequenced. The expression profiles were analyzed and integrated to identify differentially expressed genes (DEGs) and differentially expressed transcripts (DETs). These were then further examined using bioinformatics tools. RNA-seq analysis identified 1298 DEGs and 20,952 DETs in the paired (Ztm treatment vs. Ztm control) and reference (WT controls) groups. Of these, 733 DEGs and 10,476 DETs were upregulated, while 565 DEGs and 6097 DETs were downregulated. BC-treated Ztm female mice showed significant upregulation of cingulin (Cgn) and claudin 12 (Cldn12) duodenum and protein interactions, as well as molecular pathways and interactions pertaining to tight junctions, while BC-treated Ztm males displayed an upregulation of transcripts like occludin (Ocln) and Rho/Rac guanine nucleotide exchange factor 2 (Arhgf2) and cellular structures and interfaces, protein-protein interactions, and organization and response mechanisms. This comprehensive analysis reveals the influence of BC treatment on tight junctions (TJs) and Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) signaling pathway gene expressions. The present study is the first to analyze intestinal and brain samples from BC-treated Ztm mice applying high-throughput RNA sequencing. This study revealed molecular interaction in intestinal barrier function and identified hub genes and their functional pathways and biological processes in response to BC treatment in Ztm mice. Further research is needed to validate these findings and explore their implications for dietary interventions aimed at improving intestinal barrier integrity and function. The MGH Institutional Animal Care and Use Committee authorized the animal study (2013N000013).
Collapse
Affiliation(s)
- Birna Asbjornsdottir
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; (B.A.); (M.F.); (T.K.); (J.L.)
- School of Health Sciences, Faculty of Medicine, University of Iceland, 102 Reykjavik, Iceland (M.G.)
- Unit for Nutrition Research, Landspitali University Hospital, Faculty of Food Science and Nutrition, University of Iceland, 102 Reykjavik, Iceland
| | - Snaevar Sigurdsson
- School of Health Sciences, Faculty of Medicine, University of Iceland, 102 Reykjavik, Iceland (M.G.)
- Biomedical Center, University of Iceland, 102 Reykjavik, Iceland
| | - Alba Miranda-Ribera
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; (B.A.); (M.F.); (T.K.); (J.L.)
| | - Maria Fiorentino
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; (B.A.); (M.F.); (T.K.); (J.L.)
| | - Takumi Konno
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; (B.A.); (M.F.); (T.K.); (J.L.)
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Jinggang Lan
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; (B.A.); (M.F.); (T.K.); (J.L.)
| | - Larus S. Gudmundsson
- School of Health Sciences, Faculty of Pharmaceutical Sciences, University of Iceland, 102 Reykjavik, Iceland
| | - Magnus Gottfredsson
- School of Health Sciences, Faculty of Medicine, University of Iceland, 102 Reykjavik, Iceland (M.G.)
- Department of Scientific Affairs, Landspitali University Hospital, 102 Reykjavik, Iceland
- Department of Infectious Diseases, Landspitali University Hospital, 102 Reykjavik, Iceland
| | - Bertrand Lauth
- School of Health Sciences, Faculty of Medicine, University of Iceland, 102 Reykjavik, Iceland (M.G.)
- Department of Child and Adolescent Psychiatry, Landspitali University Hospital, 102 Reykjavik, Iceland
| | - Bryndis Eva Birgisdottir
- Unit for Nutrition Research, Landspitali University Hospital, Faculty of Food Science and Nutrition, University of Iceland, 102 Reykjavik, Iceland
| | - Alessio Fasano
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; (B.A.); (M.F.); (T.K.); (J.L.)
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA 02138, USA
| |
Collapse
|
37
|
Szakats S, McAtamney A, Cross H, Wilson MJ. Sex-biased gene and microRNA expression in the developing mouse brain is associated with neurodevelopmental functions and neurological phenotypes. Biol Sex Differ 2023; 14:57. [PMID: 37679839 PMCID: PMC10486049 DOI: 10.1186/s13293-023-00538-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 08/18/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Sex differences pose a challenge and an opportunity in biomedical research. Understanding how sex chromosomes and hormones affect disease-causing mechanisms will shed light on the mechanisms underlying predominantly idiopathic sex-biased neurodevelopmental disorders such as ADHD, schizophrenia, and autism. Gene expression is a crucial conduit for the influence of sex on developmental processes; therefore, this study focused on sex differences in gene expression and the regulation of gene expression. The increasing interest in microRNAs (miRNAs), small, non-coding RNAs, for their contribution to normal and pathological neurodevelopment prompted us to test how miRNA expression differs between the sexes in the developing brain. METHODS High-throughput sequencing approaches were used to identify transcripts, including miRNAs, that showed significantly different expression between male and female brains on day 15.5 of development (E15.5). RESULTS Robust sex differences were identified for some genes and miRNAs, confirming the influence of biological sex on RNA. Many miRNAs that exhibit the greatest differences between males and females have established roles in neurodevelopment, implying that sex-biased expression may drive sex differences in developmental processes. In addition to highlighting sex differences for individual miRNAs, gene ontology analysis suggested several broad categories in which sex-biased RNAs might act to establish sex differences in the embryonic mouse brain. Finally, mining publicly available SNP data indicated that some sex-biased miRNAs reside near the genomic regions associated with neurodevelopmental disorders. CONCLUSIONS Together, these findings reinforce the importance of cataloguing sex differences in molecular biology research and highlight genes, miRNAs, and pathways of interest that may be important for sexual differentiation in the mouse and possibly the human brain.
Collapse
Affiliation(s)
- Susanna Szakats
- Developmental Genomics Laboratory, Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Alice McAtamney
- Developmental Genomics Laboratory, Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Hugh Cross
- Developmental Genomics Laboratory, Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Megan J Wilson
- Developmental Genomics Laboratory, Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand.
| |
Collapse
|
38
|
Cheng JS, Chen WT, Ku HP, Chien RN, Chang ML. Characteristic geoepidemiology of primary biliary cholangitis in Taiwan: A nationwide population-based study. Hepatol Res 2023; 53:866-877. [PMID: 37060573 DOI: 10.1111/hepr.13910] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/25/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
AIM Data on the geoepidemiology and outcomes of primary biliary cholangitis (PBC) in Asia are limited; thus, we aimed to collect and assess this information for Taiwan. METHODS A nationwide population-based cohort study was undertaken using data from the Taiwan National Health Insurance Research Database. Primary biliary cholangitis was defined by the International Classification of Diseases, Ninth Revision, Clinical Modification code 571.6 based on alkaline phosphatase and antimitochondrial antibody measurements and ursodeoxycholic acid treatment. RESULTS During 2002-2015, 2737 patients (2137 female patients; mean age, 57.78 years) had PBC. The average annual age- and sex-adjusted prevalence and incidence rates of PBC were 8.092/105 and 1.148/105 , respectively. Prevalent cases peaked in patients aged 50-59 years; the female-to-male ratio was 4.21. Annual prevalence rates increased with time (average percentage change, 12.03%; p < 0.0001). The annual incidence rates decreased with time (-7.39%; p = 0.000011) in female patients (-8.94%; p = 0.000003) but remained steady in male patients. Female-to-male and northern-to-southern relative risks of PBC incidence rates ranged from 2.2675 to 4.3318 and from 1.5707 to 3.1725, respectively. The 14-year hepatocellular carcinoma (HCC) cumulative incidence was 9.11%, and the mortality rate was 32.44%; the cumulative incidences of dyslipidemia, thyroid disease, and extrahepatic cancers were 65.13%, 24.40%, and 12.79%, respectively. Higher cumulative incidences of HCC (p = 0.0064) and mortality (p < 0.0001) were noted in male than female PBC patients; southern Taiwan PBC patients had higher cumulative incidences of mortality (p < 0.0001) than their northern counterparts. CONCLUSION In Taiwan, decreasing trends in incidence rates and the female-to-male ratio of PBC patients and specific sex and geographic impacts on the incidence rates and outcomes of PBC demand further investigation.
Collapse
Affiliation(s)
- Jur-Shan Cheng
- Clinical Informatics and Medical Statistics Research Center, College of Medicine, Chang Gung University, Keelung, Taiwan
- Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Wei-Ting Chen
- Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Ping Ku
- Clinical Informatics and Medical Statistics Research Center, College of Medicine, Chang Gung University, Keelung, Taiwan
| | - Rong-Nan Chien
- Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Ling Chang
- Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
39
|
Zhao X, Zhang Y, Yu T, Cai L, Liang J, Chen Z, Pan C, Yang M. Transcriptomics-based analysis of sex-differentiated mechanisms of hepatotoxicity in zebrafish after long-term exposure to bisphenol AF. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115324. [PMID: 37556959 DOI: 10.1016/j.ecoenv.2023.115324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/23/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
Bisphenol AF (BPAF) is an emerging endocrine-disrupting chemical (EDC) prevalent in the environment as one of the main substitutes for bisphenol A. Sex-specific effects of EDCs have been commonly reported and closely linked to sexually dimorphic patterns of hormone metabolism and related gene expression during different exposure windows, but our understanding of these mechanisms is still limited. Here, following 28-day exposure of adult zebrafish to an environmentally relevant concentration of BPAF at 10 μg/L, the global transcriptional networks applying RNA sequencing (RNA-seq) and Ingenuity Pathway Analysis (IPA) were respectively investigated in the male and female fish liver, connecting the sex-dependent toxicity of the long-term exposure of BPAF to molecular responses. As a result, more differentially expressed genes (DEGs) were detected in males (811) than in females (195), and spermatogenesis was the most enriched Gene Ontology (GO) functional classification in males, while circadian regulation of gene expression was the most enriched GO term in females. The expression levels of selected DEGs were routinely verified using qRT-PCR, which showed consistent alterations with the transcriptional changes in RNA-seq data. The causal network analysis by IPA suggested that the adverse outcomes of BPAF in males including liver damage, apoptosis, inflammation of organ, and liver carcinoma, associated with the regulation of several key DEGs detected in RNA-seq, could be linked to the activation of upstream regulatory molecules ifnα, yap1, and ptger2; while, the inhibition of upstream regulators hif1α, ifng, and igf1, leading to the down-regulated expression of several key DEGs, might be involved in BPAF's effects in females. Furthermore, BPAF exposure altered hepatic histological structure and inhibited antioxidant capability in both male and female livers. Overall, this study revealed different regulation networks involved in the sex-dependent effects of BPAF on the fish liver, and these detected DEGs upon BPAF exposure might be used as potential biomarkers for further assessing sex-specific hepatotoxicity following environmental EDC exposure.
Collapse
Affiliation(s)
- Xiaoyu Zhao
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yuanyuan Zhang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Ting Yu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Ling Cai
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian 361005, China.
| | - Junlang Liang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Zhong Chen
- Department of Cardiology, Shanghai Sixth People's Hospital Fujian, Jinjiang, Fujian 362200, China
| | - Chenyuan Pan
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Ming Yang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
40
|
DeLacey PM, Sen S, Schneider-Crease IA, Chiou KL, Lemma A, Ayele F, Haile AA, Lu A, Bergman TJ, Beehner JC, Snyder-Mackler N. Vascularization underlies differences in sexually selected skin coloration in a wild primate. Mol Ecol 2023; 32:4401-4411. [PMID: 37226287 DOI: 10.1111/mec.17026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/21/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023]
Abstract
Male reproductive competition can select for condition-dependent, conspicuous traits that signal some aspect of fighting ability and facilitate assessment of potential rivals. However, the underlying mechanisms that link the signal to a male's current condition are difficult to investigate in wild populations, often requiring invasive experimental manipulation. Here, we use digital photographs and chest skin samples to investigate the mechanisms of a visual signal used in male competition in a wild primate, the red chest patch in geladas (Theropithecus gelada). We analysed photographs collected during natural (n = 144) and anaesthetized conditions (n = 38) to understand variability in male and female chest redness, and we used chest skin biopsies (n = 38) to explore sex differences in gene expression. Male and female geladas showed similar average redness, but males exhibited a wider within-individual range in redness under natural conditions. These sex differences were also reflected at the molecular level, with 10.5% of genes exhibiting significant sex differences in expression. Subadult males exhibited intermediate gene expression patterns between adult males and females, pointing to mechanisms underlying the development of the red chest patch. We found that genes more highly expressed in males were associated with blood vessel development and maintenance but not with androgen or oestrogen activity. Together, our results suggest male gelada redness variability is driven by increased blood vessel branching in the chest skin, providing a potential link between male chest redness and current condition as increased blood circulation to exposed skin could lead to heat loss in the cold, high-altitude environment of geladas.
Collapse
Affiliation(s)
- Patricia M DeLacey
- Department of Psychology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sharmi Sen
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan, USA
| | - India A Schneider-Crease
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- School of Human Evolution and Social Change, Arizona State University, Tempe, Arizona, USA
| | - Kenneth L Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | - Alemayehu Lemma
- College of Veterinary Medicine and Agriculture, Addis Ababa University, Ethiopia
| | - Ferehiwot Ayele
- College of Veterinary Medicine and Agriculture, Addis Ababa University, Ethiopia
| | | | - Amy Lu
- Department of Anthropology, Stony Brook University, New York, USA
| | - Thore J Bergman
- Department of Psychology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jacinta C Beehner
- Department of Psychology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- School of Human Evolution and Social Change, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
41
|
Moore CJ, Holstege CP, Papin JA. Metabolic modeling of sex-specific liver tissue suggests mechanism of differences in toxicological responses. PLoS Comput Biol 2023; 19:e1010927. [PMID: 37603574 PMCID: PMC10470949 DOI: 10.1371/journal.pcbi.1010927] [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: 02/02/2023] [Revised: 08/31/2023] [Accepted: 07/25/2023] [Indexed: 08/23/2023] Open
Abstract
Male subjects in animal and human studies are disproportionately used for toxicological testing. This discrepancy is evidenced in clinical medicine where females are more likely than males to experience liver-related adverse events in response to xenobiotics. While previous work has shown gene expression differences between the sexes, there is a lack of systems-level approaches to understand the direct clinical impact of these differences. Here, we integrate gene expression data with metabolic network models to characterize the impact of transcriptional changes of metabolic genes in the context of sex differences and drug treatment. We used Tasks Inferred from Differential Expression (TIDEs), a reaction-centric approach to analyzing differences in gene expression, to discover that several metabolic pathways exhibit sex differences including glycolysis, fatty acid metabolism, nucleotide metabolism, and xenobiotics metabolism. When TIDEs is used to compare expression differences in treated and untreated hepatocytes, we find several subsystems with differential expression overlap with the sex-altered pathways such as fatty acid metabolism, purine and pyrimidine metabolism, and xenobiotics metabolism. Finally, using sex-specific transcriptomic data, we create individual and averaged male and female liver models and find differences in the pentose phosphate pathway and other metabolic pathways. These results suggest potential sex differences in the contribution of the pentose phosphate pathway to oxidative stress, and we recommend further research into how these reactions respond to hepatotoxic pharmaceuticals.
Collapse
Affiliation(s)
- Connor J. Moore
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Christopher P. Holstege
- Department of Emergency Medicine, Division of Medical Toxicology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jason A. Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
42
|
Singh PP, Benayoun BA. Considerations for reproducible omics in aging research. NATURE AGING 2023; 3:921-930. [PMID: 37386258 PMCID: PMC10527412 DOI: 10.1038/s43587-023-00448-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023]
Abstract
Technical advancements over the past two decades have enabled the measurement of the panoply of molecules of cells and tissues including transcriptomes, epigenomes, metabolomes and proteomes at unprecedented resolution. Unbiased profiling of these molecular landscapes in the context of aging can reveal important details about mechanisms underlying age-related functional decline and age-related diseases. However, the high-throughput nature of these experiments creates unique analytical and design demands for robustness and reproducibility. In addition, 'omic' experiments are generally onerous, making it crucial to effectively design them to eliminate as many spurious sources of variation as possible as well as account for any biological or technical parameter that may influence such measures. In this Perspective, we provide general guidelines on best practices in the design and analysis of omic experiments in aging research from experimental design to data analysis and considerations for long-term reproducibility and validation of such studies.
Collapse
Affiliation(s)
- Param Priya Singh
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
- Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA, USA.
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
- Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, CA, USA.
- Biochemistry and Molecular Medicine Department, USC Keck School of Medicine, Los Angeles, CA, USA.
- Epigenetics and Gene Regulation, USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA.
- USC Stem Cell Initiative, Los Angeles, CA, USA.
| |
Collapse
|
43
|
Weger M, Weger BD, Gachon F. Understanding circadian dynamics: current progress and future directions for chronobiology in drug discovery. Expert Opin Drug Discov 2023. [PMID: 37300813 DOI: 10.1080/17460441.2023.2224554] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/08/2023] [Indexed: 06/12/2023]
Abstract
INTRODUCTION Most mammalian physiology is orchestrated by the circadian clock, including drug transport and metabolism. As a result, efficacy and toxicity of many drugs are influenced by the timing of their administration, which has led to the establishment of the field of chronopharmacology. AREAS COVERED In this review, the authors provide an overview of the current knowledge about the time-of-day dependent aspects of drug metabolism and the importance of chronopharmacological strategies for drug development. They also discuss the factors influencing rhythmic drug pharmacokinetic including sex, metabolic diseases, feeding rhythms, and microbiota, that are often overlooked in the context of chronopharmacology. This article summarizes the involved molecular mechanisms and functions and explains why these parameters should be considered in the process of drug discovery. EXPERT OPINION Although chronomodulated treatments have shown promising results, particularly for cancer, the practice is still underdeveloped due to the associated high cost and time investments. However, implementing this strategy at the preclinical stage could offer a new opportunity to translate preclinical discoveries into successful clinical treatments.
Collapse
Affiliation(s)
- Meltem Weger
- Institute for Molecular Bioscience, The University of Queensland, QLD, Australia
| | - Benjamin D Weger
- Institute for Molecular Bioscience, The University of Queensland, QLD, Australia
| | - Frédéric Gachon
- Institute for Molecular Bioscience, The University of Queensland, QLD, Australia
| |
Collapse
|
44
|
Darolti I, Mank JE. Sex-biased gene expression at single-cell resolution: cause and consequence of sexual dimorphism. Evol Lett 2023; 7:148-156. [PMID: 37251587 PMCID: PMC10210449 DOI: 10.1093/evlett/qrad013] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/08/2023] [Accepted: 04/06/2023] [Indexed: 05/31/2023] Open
Abstract
Gene expression differences between males and females are thought to be key for the evolution of sexual dimorphism, and sex-biased genes are often used to study the molecular footprint of sex-specific selection. However, gene expression is often measured from complex aggregations of diverse cell types, making it difficult to distinguish between sex differences in expression that are due to regulatory rewiring within similar cell types and those that are simply a consequence of developmental differences in cell-type abundance. To determine the role of regulatory versus developmental differences underlying sex-biased gene expression, we use single-cell transcriptomic data from multiple somatic and reproductive tissues of male and female guppies, a species that exhibits extensive phenotypic sexual dimorphism. Our analysis of gene expression at single-cell resolution demonstrates that nonisometric scaling between the cell populations within each tissue and heterogeneity in cell-type abundance between the sexes can influence inferred patterns of sex-biased gene expression by increasing both the false-positive and false-negative rates. Moreover, we show that, at the bulk level, the subset of sex-biased genes that are the product of sex differences in cell-type abundance can significantly confound patterns of coding-sequence evolution. Taken together, our results offer a unique insight into the effects of allometry and cellular heterogeneity on perceived patterns of sex-biased gene expression and highlight the power of single-cell RNA-sequencing in distinguishing between sex-biased genes that are the result of regulatory change and those that stem from sex differences in cell-type abundance, and hence are a consequence rather than a cause of sexual dimorphism.
Collapse
Affiliation(s)
- Iulia Darolti
- Department of Zoology and Biodiversity Research Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Ecology and Evolution, University of Lausanne, Lausanne, Switzerland
| | - Judith E Mank
- Department of Zoology and Biodiversity Research Centre, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
45
|
Allegra S, Chiara F, Di Grazia D, Gaspari M, De Francia S. Evaluation of Sex Differences in Preclinical Pharmacology Research: How Far Is Left to Go? Pharmaceuticals (Basel) 2023; 16:786. [PMID: 37375734 DOI: 10.3390/ph16060786] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/10/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Until the last quarter of the 20th century, sex was not recognized as a variable in health research, nor was it believed to be a factor that could affect health and illness. Researchers preferred studying male models for a variety of reasons, such as simplicity, lower costs, hormone confounding effects, and fear of liability from perinatal exposure in case of pregnancy. Equitable representation is imperative for determining the safety, effectiveness, and tolerance of therapeutic agents for all consumers. Decades of female models' underrepresentation in preclinical studies has resulted in inequality in the understanding, diagnosis, and treatment of disease between the sexes. Sex bias has been highlighted as one of the contributing factors to the poor translation and replicability of preclinical research. There have been multiple calls for action, and the inclusion of sex as a biological variable is increasingly supported. However, although there has been substantial progress in the efforts to include more female models in preclinical studies, disparities today remain. In the present review, we consider the current standard practice of the preclinical research setting, why the sex bias exists, why there is the need to include female models, and what risks may arise from continuing this exclusion from experimental design.
Collapse
Affiliation(s)
- Sarah Allegra
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| | - Francesco Chiara
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| | - Daniela Di Grazia
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| | - Marco Gaspari
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| | - Silvia De Francia
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| |
Collapse
|
46
|
Merchant JP, Zhu K, Henrion MYR, Zaidi SSA, Lau B, Moein S, Alamprese ML, Pearse RV, Bennett DA, Ertekin-Taner N, Young-Pearse TL, Chang R. Predictive network analysis identifies JMJD6 and other potential key drivers in Alzheimer's disease. Commun Biol 2023; 6:503. [PMID: 37188718 PMCID: PMC10185548 DOI: 10.1038/s42003-023-04791-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/31/2023] [Indexed: 05/17/2023] Open
Abstract
Despite decades of genetic studies on late-onset Alzheimer's disease, the underlying molecular mechanisms remain unclear. To better comprehend its complex etiology, we use an integrative approach to build robust predictive (causal) network models using two large human multi-omics datasets. We delineate bulk-tissue gene expression into single cell-type gene expression and integrate clinical and pathologic traits, single nucleotide variation, and deconvoluted gene expression for the construction of cell type-specific predictive network models. Here, we focus on neuron-specific network models and prioritize 19 predicted key drivers modulating Alzheimer's pathology, which we then validate by knockdown in human induced pluripotent stem cell-derived neurons. We find that neuronal knockdown of 10 of the 19 targets significantly modulates levels of amyloid-beta and/or phosphorylated tau peptides, most notably JMJD6. We also confirm our network structure by RNA sequencing in the neurons following knockdown of each of the 10 targets, which additionally predicts that they are upstream regulators of REST and VGF. Our work thus identifies robust neuronal key drivers of the Alzheimer's-associated network state which may represent therapeutic targets with relevance to both amyloid and tau pathology in Alzheimer's disease.
Collapse
Affiliation(s)
- Julie P Merchant
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kuixi Zhu
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Marc Y R Henrion
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, Pembroke Place, L3 5QA, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, PO Box 30096, Blantyre, Malawi
| | - Syed S A Zaidi
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Branden Lau
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
- Arizona Research Labs, Genetics Core, University of Arizona, Tucson, AZ, USA
| | - Sara Moein
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Melissa L Alamprese
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Richard V Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Boston, MA, USA.
| | - Rui Chang
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA.
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
- INTelico Therapeutics LLC, Tucson, AZ, USA.
- PATH Biotech LLC, Tucson, AZ, USA.
| |
Collapse
|
47
|
Ibáñez CA, Lira-León G, Reyes-Castro LA, Rodríguez-González GL, Lomas-Soria C, Hernández-Rojas A, Bravo-Flores E, Solis-Paredes JM, Estrada-Gutierrez G, Zambrano E. Programming Mechanism of Adipose Tissue Expansion in the Rat Offspring of Obese Mothers Occurs in a Sex-Specific Manner. Nutrients 2023; 15:nu15102245. [PMID: 37242132 DOI: 10.3390/nu15102245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/19/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
We investigated whether excessive retroperitoneal adipose tissue (AT) expansion programmed by maternal obesity (MO) affects adipocyte size distribution and gene expression in relation to adipocyte proliferation and differentiation in male and female offspring (F1) from control (F1C) and obese (F1MO) mothers. Female Wistar rats (F0) ate a control or high-fat diet from weaning through pregnancy and lactation. F1 were weaned onto a control diet and euthanized at 110 postnatal days. Fat depots were weighed to estimate the total AT. Serum glucose, triglyceride, leptin, insulin, and the insulin resistance index (HOMA-IR) were determined. Adipocyte size and adipogenic gene expression were examined in retroperitoneal fat. Body weight, retroperitoneal AT and adipogenesis differed between male and female F1Cs. Retroperitoneal AT, glucose, triglyceride, insulin, HOMA-IR and leptin were higher in male and female F1MO vs. F1C. Small adipocytes were reduced in F1MO females and absent in F1MO males; large adipocytes were increased in F1MO males and females vs. F1C. Wnt, PI3K-Akt, and insulin signaling pathways in F1MO males and Egr2 in F1MO females were downregulated vs. F1C. MO induced metabolic dysfunction in F1 through different sex dimorphism mechanisms, including the decreased expression of pro-adipogenic genes and reduced insulin signaling in males and lipid mobilization-related genes in females.
Collapse
Affiliation(s)
- Carlos A Ibáñez
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Gabriela Lira-León
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Luis A Reyes-Castro
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Guadalupe L Rodríguez-González
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Consuelo Lomas-Soria
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
- CONACyT-Cátedras, Investigador por México, Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City 14080, Mexico
| | - Alejandra Hernández-Rojas
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Eyerahí Bravo-Flores
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Juan Mario Solis-Paredes
- Departamento de Investigación en Salud Reproductiva y Perinatal, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Guadalupe Estrada-Gutierrez
- Dirección de Investigación, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Elena Zambrano
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| |
Collapse
|
48
|
Lalonde C, Sreetharan S, Murray A, Stoa L, Cybulski ME, Kennedy A, Landry N, Stillar A, Khurana S, Tharmalingam S, Wilson J, Khaper N, Lees SJ, Boreham D, Tai TC. Absence of Depressive and Anxious Behavior with Genetic Dysregulation in Adult C57Bl/6J Mice after Prenatal Exposure to Ionizing Radiation. Int J Mol Sci 2023; 24:ijms24108466. [PMID: 37239811 DOI: 10.3390/ijms24108466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/28/2023] Open
Abstract
The exposure of ionizing radiation during early gestation often leads to deleterious and even lethal effects; however, few extensive studies have been conducted on late gestational exposures. This research examined the behavior al effects of C57Bl/6J mouse offspring exposed to low dose ionizing gamma irradiation during the equivalent third trimester. Pregnant dams were randomly assigned to sham or exposed groups to either low dose or sublethal dose radiation (50, 300, or 1000 mGy) at gestational day 15. Adult offspring underwent a behavioral and genetic analysis after being raised under normal murine housing conditions. Our results indicate very little change in the behavioral tasks measuring general anxiety, social anxiety, and stress-management in animals exposed prenatally across the low dose radiation conditions. Quantitative real-time polymerase chain reactions were conducted on the cerebral cortex, hippocampus, and cerebellum of each animal; results indicate some dysregulation in markers of DNA damage, synaptic activity, reactive oxygen species (ROS) regulation, and methylation pathways in the offspring. Together, our results provide evidence in the C57Bl/6J strain, that exposure to sublethal dose radiation (<1000 mGy) during the last period of gestation leads to no observable changes in behaviour when assessed as adults, although some changes in gene expression were observed for specific brain regions. These results indicate that the level of oxidative stress occurring during late gestation for this mouse strain is not sufficient for a change in the assessed behavioral phenotype, but results in some modest dysregulation of the genetic profile of the brain.
Collapse
Affiliation(s)
- Christine Lalonde
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E2C6, Canada
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E2C6, Canada
- Medical Sciences Division, NOSM University, Sudbury, ON P3E2C6, Canada
| | - Shayenthiran Sreetharan
- Medical Sciences Division, NOSM University, Sudbury, ON P3E2C6, Canada
- Department of Biology, McMaster University, Hamilton, ON L8S4L8, Canada
| | - Alyssa Murray
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E2C6, Canada
- Medical Sciences Division, NOSM University, Sudbury, ON P3E2C6, Canada
| | - Lisa Stoa
- Department of Biology, McMaster University, Hamilton, ON L8S4L8, Canada
| | | | - Allison Kennedy
- Medical Sciences Division, NOSM University, Sudbury, ON P3E2C6, Canada
| | - Nicholas Landry
- Department of Psychology, Nipissing University, North Bay, ON P1B8L7, Canada
| | - Amy Stillar
- Department of Psychology, Nipissing University, North Bay, ON P1B8L7, Canada
| | - Sandhya Khurana
- Medical Sciences Division, NOSM University, Sudbury, ON P3E2C6, Canada
| | - Sujeenthar Tharmalingam
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E2C6, Canada
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E2C6, Canada
- Medical Sciences Division, NOSM University, Sudbury, ON P3E2C6, Canada
| | - Joanna Wilson
- Department of Biology, McMaster University, Hamilton, ON L8S4L8, Canada
| | - Neelam Khaper
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E2C6, Canada
- Medical Sciences Division, NOSM University, Sudbury, ON P3E2C6, Canada
| | - Simon J Lees
- Medical Sciences Division, NOSM University, Sudbury, ON P3E2C6, Canada
| | - Douglas Boreham
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E2C6, Canada
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E2C6, Canada
- Medical Sciences Division, NOSM University, Sudbury, ON P3E2C6, Canada
| | - T C Tai
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E2C6, Canada
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E2C6, Canada
- Medical Sciences Division, NOSM University, Sudbury, ON P3E2C6, Canada
| |
Collapse
|
49
|
Zhong X, Sun Y, Lu Y, Xu L. Immunomodulatory role of estrogen in ischemic stroke: neuroinflammation and effect of sex. Front Immunol 2023; 14:1164258. [PMID: 37180115 PMCID: PMC10167039 DOI: 10.3389/fimmu.2023.1164258] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/12/2023] [Indexed: 05/15/2023] Open
Abstract
Although estrogen is predominantly related to the maintenance of reproductive functioning in females, it mediates various physiological effects in nearly all tissues, especially the central nervous system. Clinical trials have revealed that estrogen, especially 17β-estradiol, can attenuate cerebral damage caused by an ischemic stroke. One mechanism underlying this effect of 17β-estradiol is by modulating the responses of immune cells, indicating its utility as a novel therapeutic strategy for ischemic stroke. The present review summarizes the effect of sex on ischemic stroke progression, the role of estrogen as an immunomodulator in immune reactions, and the potential clinical value of estrogen replacement therapy. The data presented here will help better understand the immunomodulatory function of estrogen and may provide a basis for its novel therapeutic use in ischemic stroke.
Collapse
Affiliation(s)
- Xiaojun Zhong
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yulin Sun
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yajun Lu
- Department of Internal Medicine, Sunto Women & Children’s Hospital, Jiaxing, China
| | - Lei Xu
- Department of Neurology, Zhejiang Rongjun Hospital, Jiaxing, China
| |
Collapse
|
50
|
Chen W, Zhou W, Li Q, Mao X. Sex differences in gene expression and alternative splicing in the Chinese horseshoe bat. PeerJ 2023; 11:e15231. [PMID: 37123006 PMCID: PMC10135408 DOI: 10.7717/peerj.15231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
Sexually dimorphic traits are common in sexually reproducing organisms and can be encoded by differential gene regulation between males and females. Although alternative splicing is common mechanism in generating transcriptional diversity, its role in generating sex differences relative to differential gene expression is less clear. Here, we investigate the relative roles of differential gene expression and alternative splicing between male and female the horseshoe bat species, Rhinolophus sinicus. Horseshoe bats are an excellent model to study acoustic differences between sexes. Using RNA-seq analyses of two somatic tissues (brain and liver) from males and females of the same population, we identified 3,471 and 2,208 differentially expressed genes between the sexes (DEGs) in the brain and liver, respectively. DEGs were enriched with functional categories associated with physiological difference of the sexes (e.g.,gamete generation and energy production for reproduction in females). In addition, we also detected many differentially spliced genes between the sexes (DSGs, 2,231 and 1,027 in the brain and liver, respectively) which were mainly involved in regulation of RNA splicing and mRNA metabolic process. Interestingly, we found a significant enrichment of DEGs on the X chromosome, but not for DSGs. As for the extent of overlap between the two sets of genes, more than expected overlap of DEGs and DSGs was observed in the brain but not in the liver. This suggests that more complex tissues, such as the brain, may require the intricate and simultaneous interplay of both differential gene expression and splicing of genes to govern sex-specific functions. Overall, our results support that variation in gene expression and alternative splicing are important and complementary mechanisms governing sex differences.
Collapse
Affiliation(s)
- Wenli Chen
- School of Ecological and Environmental Sciences, East China Normal University, Shanghai, China
| | - Weiwei Zhou
- School of Ecological and Environmental Sciences, East China Normal University, Shanghai, China
| | - Qianqian Li
- School of Ecological and Environmental Sciences, East China Normal University, Shanghai, China
| | - Xiuguang Mao
- School of Ecological and Environmental Sciences, East China Normal University, Shanghai, China
| |
Collapse
|