首页 > 最新文献

Biology of Sex Differences最新文献

英文 中文
Gene regulatory networks reveal sex difference in lung adenocarcinoma. 基因调控网络揭示肺腺癌的性别差异
IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-08-06 DOI: 10.1186/s13293-024-00634-y
Enakshi Saha, Marouen Ben Guebila, Viola Fanfani, Jonas Fischer, Katherine H Shutta, Panagiotis Mandros, Dawn L DeMeo, John Quackenbush, Camila M Lopes-Ramos

Background: Lung adenocarcinoma (LUAD) has been observed to have significant sex differences in incidence, prognosis, and response to therapy. However, the molecular mechanisms responsible for these disparities have not been investigated extensively.

Methods: Sample-specific gene regulatory network methods were used to analyze RNA sequencing data from non-cancerous human lung samples from The Genotype Tissue Expression Project (GTEx) and lung adenocarcinoma primary tumor samples from The Cancer Genome Atlas (TCGA); results were validated on independent data.

Results: We found that genes associated with key biological pathways including cell proliferation, immune response and drug metabolism are differentially regulated between males and females in both healthy lung tissue and tumor, and that these regulatory differences are further perturbed by tobacco smoking. We also discovered significant sex bias in transcription factor targeting patterns of clinically actionable oncogenes and tumor suppressor genes, including AKT2 and KRAS. Using differentially regulated genes between healthy and tumor samples in conjunction with a drug repurposing tool, we identified several small-molecule drugs that might have sex-biased efficacy as cancer therapeutics and further validated this observation using an independent cell line database.

Conclusions: These findings underscore the importance of including sex as a biological variable and considering gene regulatory processes in developing strategies for disease prevention and management.

背景:据观察,肺腺癌(LUAD)在发病率、预后和对治疗的反应方面存在显著的性别差异。然而,造成这些差异的分子机制尚未得到广泛研究:方法:采用样本特异性基因调控网络方法分析基因型组织表达项目(GTEx)非癌人类肺部样本和癌症基因组图谱(TCGA)肺腺癌原发肿瘤样本的RNA测序数据;结果在独立数据上得到验证:结果:我们发现,在健康肺组织和肿瘤中,与细胞增殖、免疫反应和药物代谢等关键生物通路相关的基因在男性和女性之间存在调控差异,而吸烟会进一步扰乱这些调控差异。我们还发现,临床上可发挥作用的癌基因和肿瘤抑制基因(包括 AKT2 和 KRAS)的转录因子靶向模式存在明显的性别差异。利用健康样本和肿瘤样本之间的差异调控基因,结合药物再利用工具,我们发现了几种可能具有性别偏倚疗效的小分子药物,并利用独立的细胞系数据库进一步验证了这一观察结果:这些发现强调了在制定疾病预防和管理策略时将性别作为生物变量并考虑基因调控过程的重要性。
{"title":"Gene regulatory networks reveal sex difference in lung adenocarcinoma.","authors":"Enakshi Saha, Marouen Ben Guebila, Viola Fanfani, Jonas Fischer, Katherine H Shutta, Panagiotis Mandros, Dawn L DeMeo, John Quackenbush, Camila M Lopes-Ramos","doi":"10.1186/s13293-024-00634-y","DOIUrl":"10.1186/s13293-024-00634-y","url":null,"abstract":"<p><strong>Background: </strong>Lung adenocarcinoma (LUAD) has been observed to have significant sex differences in incidence, prognosis, and response to therapy. However, the molecular mechanisms responsible for these disparities have not been investigated extensively.</p><p><strong>Methods: </strong>Sample-specific gene regulatory network methods were used to analyze RNA sequencing data from non-cancerous human lung samples from The Genotype Tissue Expression Project (GTEx) and lung adenocarcinoma primary tumor samples from The Cancer Genome Atlas (TCGA); results were validated on independent data.</p><p><strong>Results: </strong>We found that genes associated with key biological pathways including cell proliferation, immune response and drug metabolism are differentially regulated between males and females in both healthy lung tissue and tumor, and that these regulatory differences are further perturbed by tobacco smoking. We also discovered significant sex bias in transcription factor targeting patterns of clinically actionable oncogenes and tumor suppressor genes, including AKT2 and KRAS. Using differentially regulated genes between healthy and tumor samples in conjunction with a drug repurposing tool, we identified several small-molecule drugs that might have sex-biased efficacy as cancer therapeutics and further validated this observation using an independent cell line database.</p><p><strong>Conclusions: </strong>These findings underscore the importance of including sex as a biological variable and considering gene regulatory processes in developing strategies for disease prevention and management.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":"15 1","pages":"62"},"PeriodicalIF":4.9,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11302009/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141896657","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fetal endocrine axes mRNA expression levels are related to sex and intrauterine position. 胎儿内分泌轴 mRNA 表达水平与性别和宫内位置有关。
IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-08-05 DOI: 10.1186/s13293-024-00637-9
Ariel Yael, Ruth Fishman, Devorah Matas, Tirza Doniger, Yoni Vortman, Lee Koren

Background: The hypothalamic-pituitary-adrenal (HPA) and -gonadal (HPG) axes are two major pathways that connect the neural and endocrine systems in vertebrates. Factors such as prenatal stress and maternal exposure to exogenous steroids have been shown to affect these pathways during fetal development. Another less studied factor is the transfer of hormones across fetuses in multifetal pregnancies. This form of transfer has been shown to influence the morphology, anatomy, physiology, and behavior of the offspring in litter-bearing mammals, an influence termed the intrauterine position (IUP) effect. In this study, we sought to delineate how the IUP effects HPA and HPG brain receptors, peptides, and enzymes (hereafter components) in utero and how these influences may differ between males and females.

Methods: We utilized the unconventional model of culled free-ranging nutria (Myocastor coypus), with its large natural variation. We collected brain tissues from nutria fetuses and quantified the expression of key HPA and HPG components in three brain regions: prefrontal cortex, hypothalamus, and striatum.

Results: We found an interaction between sex and IUP in the mineralocorticoid receptor (MR), gonadotropin-releasing hormone receptor (GNRHR), androgen receptor (AR), and estrogen receptor alpha (ESR1). IUP was significant in both gonadotropin-releasing hormone (GnRH) and its receptor GNRHR, but in different ways. In the hypothalamus, fetuses adjacent to same-sex neighbors had higher expression of GnRH than fetuses neighboring the opposite sex. Conversely, in the cortex, GNRHR exhibited the inverse pattern, and fetuses that were neighboring the opposite sex had higher expression levels than those neighboring the same sex. Regardless of IUP, in most components that showed significant sex differences, female fetuses had higher mRNA expression levels than male fetuses. We also found that HPA and HPG components were highly related in the early stages of gestation, and that there was an interaction between sex and developmental stage. In the early stages of pregnancy, female component expression levels were more correlated than males', but in the last trimester of pregnancy, male components were more related to each other than female's.

Conclusions: This study suggests that there are sexually different mechanisms to regulate the HPA and HPG axes during fetal development. Higher mRNA expression levels of endocrine axes components may be a mechanism to help females cope with prolonged androgen exposure over a long gestational period. Additionally, these findings suggest different coordination requirements of male and female endocrine axes during stages of fetal development.

背景:下丘脑-垂体-肾上腺轴(HPA)和肾上腺-性腺轴(HPG)是连接脊椎动物神经系统和内分泌系统的两条主要通道。产前压力和母体暴露于外源性类固醇等因素已被证明会在胎儿发育过程中影响这些通路。另一个研究较少的因素是多胎妊娠中胎儿之间的激素转移。这种形式的转移已被证明会影响产仔哺乳动物后代的形态、解剖、生理和行为,这种影响被称为宫内位置效应(IUP)。在这项研究中,我们试图阐明宫内位置如何影响子宫内的 HPA 和 HPG 脑受体、肽和酶(以下简称成分),以及这些影响在雄性和雌性之间的差异:我们利用了自然变异较大的非传统散养秧鸡(Myocastor coypus)模型。我们收集了秧鸡胎儿的脑组织,并量化了前额叶皮层、下丘脑和纹状体这三个脑区中 HPA 和 HPG 关键成分的表达:结果:我们发现,在矿质皮质激素受体(MR)、促性腺激素释放激素受体(GNRHR)、雄激素受体(AR)和雌激素受体α(ESR1)中,性别与 IUP 之间存在相互作用。IUP对促性腺激素释放激素(GnRH)及其受体GNRHR都有重要作用,但作用方式不同。在下丘脑中,与同性相邻的胎儿比与异性相邻的胎儿有更高的促性腺激素释放激素表达。相反,在皮层中,GNRHR表现出相反的模式,与异性相邻的胎儿比与同性相邻的胎儿有更高的表达水平。无论 IUP 如何,在大多数表现出显著性别差异的成分中,女性胎儿的 mRNA 表达水平高于男性胎儿。我们还发现,HPA和HPG成分在妊娠早期高度相关,性别与发育阶段之间存在相互作用。在妊娠早期,女性成分表达水平的相关性高于男性,但在妊娠最后三个月,男性成分的相关性高于女性:这项研究表明,在胎儿发育过程中,HPA 和 HPG 轴的调节机制存在性别差异。内分泌轴成分的mRNA表达水平较高,这可能是帮助雌性应对妊娠期长时间雄激素暴露的一种机制。此外,这些研究结果表明,在胎儿发育阶段,男性和女性的内分泌轴需要不同的协调。
{"title":"Fetal endocrine axes mRNA expression levels are related to sex and intrauterine position.","authors":"Ariel Yael, Ruth Fishman, Devorah Matas, Tirza Doniger, Yoni Vortman, Lee Koren","doi":"10.1186/s13293-024-00637-9","DOIUrl":"10.1186/s13293-024-00637-9","url":null,"abstract":"<p><strong>Background: </strong>The hypothalamic-pituitary-adrenal (HPA) and -gonadal (HPG) axes are two major pathways that connect the neural and endocrine systems in vertebrates. Factors such as prenatal stress and maternal exposure to exogenous steroids have been shown to affect these pathways during fetal development. Another less studied factor is the transfer of hormones across fetuses in multifetal pregnancies. This form of transfer has been shown to influence the morphology, anatomy, physiology, and behavior of the offspring in litter-bearing mammals, an influence termed the intrauterine position (IUP) effect. In this study, we sought to delineate how the IUP effects HPA and HPG brain receptors, peptides, and enzymes (hereafter components) in utero and how these influences may differ between males and females.</p><p><strong>Methods: </strong>We utilized the unconventional model of culled free-ranging nutria (Myocastor coypus), with its large natural variation. We collected brain tissues from nutria fetuses and quantified the expression of key HPA and HPG components in three brain regions: prefrontal cortex, hypothalamus, and striatum.</p><p><strong>Results: </strong>We found an interaction between sex and IUP in the mineralocorticoid receptor (MR), gonadotropin-releasing hormone receptor (GNRHR), androgen receptor (AR), and estrogen receptor alpha (ESR1). IUP was significant in both gonadotropin-releasing hormone (GnRH) and its receptor GNRHR, but in different ways. In the hypothalamus, fetuses adjacent to same-sex neighbors had higher expression of GnRH than fetuses neighboring the opposite sex. Conversely, in the cortex, GNRHR exhibited the inverse pattern, and fetuses that were neighboring the opposite sex had higher expression levels than those neighboring the same sex. Regardless of IUP, in most components that showed significant sex differences, female fetuses had higher mRNA expression levels than male fetuses. We also found that HPA and HPG components were highly related in the early stages of gestation, and that there was an interaction between sex and developmental stage. In the early stages of pregnancy, female component expression levels were more correlated than males', but in the last trimester of pregnancy, male components were more related to each other than female's.</p><p><strong>Conclusions: </strong>This study suggests that there are sexually different mechanisms to regulate the HPA and HPG axes during fetal development. Higher mRNA expression levels of endocrine axes components may be a mechanism to help females cope with prolonged androgen exposure over a long gestational period. Additionally, these findings suggest different coordination requirements of male and female endocrine axes during stages of fetal development.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":"15 1","pages":"61"},"PeriodicalIF":4.9,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11301978/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141892791","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Smad4 and FoxH1 potentially interact to regulate cyp19a1a promoter in the ovary of ricefield eel (Monopterus albus). Smad4和FoxH1可能相互作用调控稻田鳗(Monopterus albus)卵巢中的cyp19a1a启动子。
IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-07-30 DOI: 10.1186/s13293-024-00636-w
Qiqi Chen, Deying Yang, Mingqiang Chen, Jinxin Xiong, Junjie Huang, Wenxiang Ding, Kuo Gao, Bolin Lai, Li Zheng, Ziting Tang, Mingwang Zhang, Taiming Yan, Zhi He

Background: Cyp19a1a is a key enzyme in the pathway that converts androgens into estrogen and is regulated by TGF-β signaling. Smad4 and FoxH1 are downstream effectors of TGF-β signaling and may play important roles in ovarian development in M. albus.

Methods: We investigated the expression pattern of the Smad4 and FoxH1 using qRT‒PCR and immunofluorescence, then tested the changes of smad4 and foxh1 by qRT‒PCR after ovary incubation with FSH in vitro, and analysed the regulation of cyp19a1a transcription by Smad4 and FoxH1 by dual-luciferase reporter assays.

Results: We found that Smad4 encoded a putative protein of 449 amino acids and harbored the three conserved domains typical of this protein family. Smad4 and foxh1 exhibited similar expression patterns during ovarian development and after FSH incubation, with Pearson's coefficients of 0.873 and 0.63-0.81, respectively. Furthermore, Smad4, FoxH1 and Cyp19a1a colocalized in the granulosa cells and theca cells of ovaries during the mid-to-late vitellogenic stage. Smad4 repressed cyp19a1a activity via SBE1 (- 1372/-1364) and SBE2 (- 415/-407) in the cyp19a1a promoter, whereas mutating SBE1 or SBE2 restored cyp19a1a promoter activity. Co-overexpression of Smad4 and FoxH1 significantly reduced cyp19a1a promoter activity.

Conclusions: This study provides new insights into the potential functions of transcription factors Smad4 and FoxH1 in ovarian development and the transcriptional regulation mechanism of cyp19a1a in M. albus, which will reveal Smad4/FoxH1-mediated TGF-β signaling in reproduction and the regulation of the cyp19a1a. Aromatase, encoded by cyp19a1a, is involved in ovarian development and plays an important role in the quality of eggs, as well the sex ratio, of the teleost fish, M. albus. The research on the transcriptional regulation of cyp19a1a has contributed to the understanding of its role in ovarian development. In previous study, it was shown that FoxH1 inhibits cyp19a1a transcription. In the present study, Smad4 was confirmed as a cyp19a1a transcriptional repressor and Smad4 may also coordinate with FoxH1 to repress cyp19a1a transcription. At present, we provide a new perspective for the transcriptional regulation of cyp19a1a by transcription factors Smad4 and FoxH1 in teleost fish ovary. In the future, the regulatory networks of Smad4 and FoxH1 will be further studied and the gene editing technology will be applied to screen specific regulatory factors of cyp191a1a gene, so as to alter the female cycle and modulate the sex ratio of the eggs production.

背景:Cyp19a1a是雄激素转化为雌激素途径中的一个关键酶,受TGF-β信号调控。Smad4和FoxH1是TGF-β信号传导的下游效应器,可能在M. albus卵巢发育过程中发挥重要作用:方法:利用qRT-PCR和免疫荧光技术研究了Smad4和FoxH1的表达模式,然后通过qRT-PCR检测了卵巢与FSH体外培养后Smad4和FoxH1的变化,并通过双荧光素酶报告实验分析了Smad4和FoxH1对cyp19a1a转录的调控作用:结果:我们发现Smad4编码一个449个氨基酸的假定蛋白,含有该蛋白家族的三个典型保守结构域。Smad4和foxh1在卵巢发育过程中和FSH孵育后表现出相似的表达模式,皮尔逊系数分别为0.873和0.63-0.81。此外,在卵黄形成的中后期,Smad4、FoxH1和Cyp19a1a共定位在卵巢的颗粒细胞和theca细胞中。Smad4通过cyp19a1a启动子中的SBE1(- 1372/-1364)和SBE2(- 415/-407)抑制cyp19a1a的活性,而突变SBE1或SBE2可恢复cyp19a1a启动子的活性。Smad4和FoxH1的共重表达显著降低了cyp19a1a启动子的活性:本研究为了解转录因子Smad4和FoxH1在卵巢发育中的潜在功能以及白僵菌cyp19a1a的转录调控机制提供了新的视角,将揭示Smad4/FoxH1介导的TGF-β信号在繁殖过程中的作用以及cyp19a1a的调控机制。由cyp19a1a编码的芳香化酶参与卵巢发育,并对远东鱼类白鲑鱼的卵子质量和性别比例起着重要作用。对cyp19a1a转录调控的研究有助于了解其在卵巢发育中的作用。之前的研究表明,FoxH1抑制cyp19a1a的转录。在本研究中,Smad4被证实是cyp19a1a的转录抑制因子,并且Smad4还可能与FoxH1协调抑制cyp19a1a的转录。目前,我们为转录因子Smad4和FoxH1在远洋鱼类卵巢中对cyp19a1a的转录调控提供了一个新的视角。未来,我们将进一步研究Smad4和FoxH1的调控网络,并应用基因编辑技术筛选cyp191a1a基因的特异性调控因子,从而改变雌性周期,调节产卵的性别比例。
{"title":"Smad4 and FoxH1 potentially interact to regulate cyp19a1a promoter in the ovary of ricefield eel (Monopterus albus).","authors":"Qiqi Chen, Deying Yang, Mingqiang Chen, Jinxin Xiong, Junjie Huang, Wenxiang Ding, Kuo Gao, Bolin Lai, Li Zheng, Ziting Tang, Mingwang Zhang, Taiming Yan, Zhi He","doi":"10.1186/s13293-024-00636-w","DOIUrl":"10.1186/s13293-024-00636-w","url":null,"abstract":"<p><strong>Background: </strong>Cyp19a1a is a key enzyme in the pathway that converts androgens into estrogen and is regulated by TGF-β signaling. Smad4 and FoxH1 are downstream effectors of TGF-β signaling and may play important roles in ovarian development in M. albus.</p><p><strong>Methods: </strong>We investigated the expression pattern of the Smad4 and FoxH1 using qRT‒PCR and immunofluorescence, then tested the changes of smad4 and foxh1 by qRT‒PCR after ovary incubation with FSH in vitro, and analysed the regulation of cyp19a1a transcription by Smad4 and FoxH1 by dual-luciferase reporter assays.</p><p><strong>Results: </strong>We found that Smad4 encoded a putative protein of 449 amino acids and harbored the three conserved domains typical of this protein family. Smad4 and foxh1 exhibited similar expression patterns during ovarian development and after FSH incubation, with Pearson's coefficients of 0.873 and 0.63-0.81, respectively. Furthermore, Smad4, FoxH1 and Cyp19a1a colocalized in the granulosa cells and theca cells of ovaries during the mid-to-late vitellogenic stage. Smad4 repressed cyp19a1a activity via SBE1 (- 1372/-1364) and SBE2 (- 415/-407) in the cyp19a1a promoter, whereas mutating SBE1 or SBE2 restored cyp19a1a promoter activity. Co-overexpression of Smad4 and FoxH1 significantly reduced cyp19a1a promoter activity.</p><p><strong>Conclusions: </strong>This study provides new insights into the potential functions of transcription factors Smad4 and FoxH1 in ovarian development and the transcriptional regulation mechanism of cyp19a1a in M. albus, which will reveal Smad4/FoxH1-mediated TGF-β signaling in reproduction and the regulation of the cyp19a1a. Aromatase, encoded by cyp19a1a, is involved in ovarian development and plays an important role in the quality of eggs, as well the sex ratio, of the teleost fish, M. albus. The research on the transcriptional regulation of cyp19a1a has contributed to the understanding of its role in ovarian development. In previous study, it was shown that FoxH1 inhibits cyp19a1a transcription. In the present study, Smad4 was confirmed as a cyp19a1a transcriptional repressor and Smad4 may also coordinate with FoxH1 to repress cyp19a1a transcription. At present, we provide a new perspective for the transcriptional regulation of cyp19a1a by transcription factors Smad4 and FoxH1 in teleost fish ovary. In the future, the regulatory networks of Smad4 and FoxH1 will be further studied and the gene editing technology will be applied to screen specific regulatory factors of cyp191a1a gene, so as to alter the female cycle and modulate the sex ratio of the eggs production.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":"15 1","pages":"60"},"PeriodicalIF":4.9,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11290265/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141854638","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The impact of sex on gene expression in the brain of schizophrenic patients: a systematic review and meta-analysis of transcriptomic studies 性别对精神分裂症患者大脑基因表达的影响:转录组研究的系统回顾和荟萃分析
IF 7.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-07-27 DOI: 10.1186/s13293-024-00635-x
Hector Carceller, Marta R. Hidalgo, María José Escartí, Juan Nacher, Maria de la Iglesia-Vayá, Francisco García-García
Schizophrenia is a severe neuropsychiatric disorder characterized by altered perception, mood, and behavior that profoundly impacts patients and society despite its relatively low prevalence. Sex-based differences have been described in schizophrenia epidemiology, symptomatology and outcomes. Different studies explored the impact of schizophrenia in the brain transcriptome, however we lack a consensus transcriptomic profile that considers sex and differentiates specific cerebral regions. We performed a systematic review on bulk RNA-sequencing studies of post-mortem brain samples. Then, we fulfilled differential expression analysis on each study and summarized their results with regions-specific meta-analyses (prefrontal cortex and hippocampus) and a global all-studies meta-analysis. Finally, we used the consensus transcriptomic profiles to functionally characterize the impact of schizophrenia in males and females by protein-protein interaction networks, enriched biological processes and dysregulated transcription factors. We discovered the sex-based dysregulation of 265 genes in the prefrontal cortex, 1.414 genes in the hippocampus and 66 genes in the all-studies meta-analyses. The functional characterization of these gene sets unveiled increased processes related to immune response functions in the prefrontal cortex in male and the hippocampus in female schizophrenia patients and the overexpression of genes related to neurotransmission and synapses in the prefrontal cortex of female schizophrenia patients. Considering a meta-analysis of all brain regions available, we encountered the relative overexpression of genes related to synaptic plasticity and transmission in females and the overexpression of genes involved in organizing genetic information and protein folding in male schizophrenia patients. The protein-protein interaction networks and transcription factors activity analyses supported these sex-based profiles. Our results report multiple sex-based transcriptomic alterations in specific brain regions of schizophrenia patients, which provides new insight into the role of sex in schizophrenia. Moreover, we unveil a partial overlapping of inflammatory processes in the prefrontal cortex of males and the hippocampus of females. Schizophrenia is a serious illness characterised by changes in perception, mood and behaviour that profoundly affect patients and society. The frequency, symptoms and progression of schizophrenia are different in women and men, but the biological reason for this is not understood. The identification of disease mechanisms specific in men and women, is relevant because it would allow a better understanding of this pathology, as well as improving the personalisation of diagnoses and treatments for patients. To achieve this goal, in this work we reviewed all available RNA sequencing studies of post-mortem brain samples from women and men affected by schizophrenia. Then, we compared gene expression in each study by sex, and i
精神分裂症是一种以知觉、情绪和行为改变为特征的严重神经精神疾病,尽管发病率相对较低,但却对患者和社会产生了深远的影响。在精神分裂症的流行病学、症状学和预后方面,已经出现了基于性别的差异。不同的研究探讨了精神分裂症对大脑转录组的影响,但我们缺乏一个考虑性别并区分特定脑区的共识转录组图谱。我们对死后大脑样本的大量 RNA 序列研究进行了系统回顾。然后,我们对每项研究进行了差异表达分析,并通过特定区域荟萃分析(前额叶皮层和海马)和全球所有研究荟萃分析总结了研究结果。最后,我们利用共识转录组图谱,通过蛋白质-蛋白质相互作用网络、富集的生物过程和失调的转录因子,从功能上描述了精神分裂症对男性和女性的影响。我们发现前额叶皮层中有 265 个基因、海马中有 1.414 个基因以及所有研究的荟萃分析中有 66 个基因存在基于性别的失调。这些基因组的功能特征揭示了男性精神分裂症患者前额叶皮层和女性精神分裂症患者海马体中与免疫反应功能相关的过程增多,以及女性精神分裂症患者前额叶皮层中与神经传递和突触相关的基因过度表达。通过对所有脑区进行荟萃分析,我们发现女性精神分裂症患者中与突触可塑性和传递相关的基因相对过度表达,而男性精神分裂症患者中与遗传信息组织和蛋白质折叠相关的基因过度表达。蛋白质-蛋白质相互作用网络和转录因子活性分析支持这些基于性别的特征。我们的研究结果表明,在精神分裂症患者的特定脑区存在多种基于性别的转录组改变,这为我们了解性别在精神分裂症中的作用提供了新的视角。此外,我们还揭示了男性前额叶皮层和女性海马体中炎症过程的部分重叠。精神分裂症是一种以知觉、情绪和行为改变为特征的严重疾病,对患者和社会造成了深远影响。女性和男性患精神分裂症的频率、症状和病情发展都有所不同,但其生物学原因尚不清楚。确定男性和女性特有的疾病机制具有重要意义,因为这将有助于更好地了解这种病理现象,并改进对患者的个性化诊断和治疗。为了实现这一目标,在这项工作中,我们回顾了所有现有的关于受精神分裂症影响的女性和男性死后大脑样本的 RNA 测序研究。然后,我们按性别比较了每项研究中的基因表达,并整合了不同脑区的所有研究结果:前额叶皮层、海马和所有研究。我们发现了男女之间的重大变化:前额叶皮层有 265 个基因表达不同,海马体有 1414 个基因表达不同,所有研究的元分析中有 66 个基因表达不同。对这些基因的研究显示,男性精神分裂症患者前额叶皮层和女性精神分裂症患者海马体中的免疫反应功能增强,女性精神分裂症患者前额叶皮层中的神经传递和突触增加。我们的研究结果表明,精神分裂症患者特定脑区的多种基因表达发生了变化,这为我们了解性别在精神分裂症中的作用提供了新的视角。
{"title":"The impact of sex on gene expression in the brain of schizophrenic patients: a systematic review and meta-analysis of transcriptomic studies","authors":"Hector Carceller, Marta R. Hidalgo, María José Escartí, Juan Nacher, Maria de la Iglesia-Vayá, Francisco García-García","doi":"10.1186/s13293-024-00635-x","DOIUrl":"https://doi.org/10.1186/s13293-024-00635-x","url":null,"abstract":"Schizophrenia is a severe neuropsychiatric disorder characterized by altered perception, mood, and behavior that profoundly impacts patients and society despite its relatively low prevalence. Sex-based differences have been described in schizophrenia epidemiology, symptomatology and outcomes. Different studies explored the impact of schizophrenia in the brain transcriptome, however we lack a consensus transcriptomic profile that considers sex and differentiates specific cerebral regions. We performed a systematic review on bulk RNA-sequencing studies of post-mortem brain samples. Then, we fulfilled differential expression analysis on each study and summarized their results with regions-specific meta-analyses (prefrontal cortex and hippocampus) and a global all-studies meta-analysis. Finally, we used the consensus transcriptomic profiles to functionally characterize the impact of schizophrenia in males and females by protein-protein interaction networks, enriched biological processes and dysregulated transcription factors. We discovered the sex-based dysregulation of 265 genes in the prefrontal cortex, 1.414 genes in the hippocampus and 66 genes in the all-studies meta-analyses. The functional characterization of these gene sets unveiled increased processes related to immune response functions in the prefrontal cortex in male and the hippocampus in female schizophrenia patients and the overexpression of genes related to neurotransmission and synapses in the prefrontal cortex of female schizophrenia patients. Considering a meta-analysis of all brain regions available, we encountered the relative overexpression of genes related to synaptic plasticity and transmission in females and the overexpression of genes involved in organizing genetic information and protein folding in male schizophrenia patients. The protein-protein interaction networks and transcription factors activity analyses supported these sex-based profiles. Our results report multiple sex-based transcriptomic alterations in specific brain regions of schizophrenia patients, which provides new insight into the role of sex in schizophrenia. Moreover, we unveil a partial overlapping of inflammatory processes in the prefrontal cortex of males and the hippocampus of females. Schizophrenia is a serious illness characterised by changes in perception, mood and behaviour that profoundly affect patients and society. The frequency, symptoms and progression of schizophrenia are different in women and men, but the biological reason for this is not understood. The identification of disease mechanisms specific in men and women, is relevant because it would allow a better understanding of this pathology, as well as improving the personalisation of diagnoses and treatments for patients. To achieve this goal, in this work we reviewed all available RNA sequencing studies of post-mortem brain samples from women and men affected by schizophrenia. Then, we compared gene expression in each study by sex, and i","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":"43 1","pages":""},"PeriodicalIF":7.9,"publicationDate":"2024-07-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141771828","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Adult sex change leads to extensive forebrain reorganization in clownfish. 成年变性导致小丑鱼前脑广泛重组
IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-07-23 DOI: 10.1186/s13293-024-00632-0
Coltan G Parker, George W Gruenhagen, Brianna E Hegarty, Abigail R Histed, Jeffrey T Streelman, Justin S Rhodes, Zachary V Johnson

Background: Sexual differentiation of the brain occurs in all major vertebrate lineages but is not well understood at a molecular and cellular level. Unlike most vertebrates, sex-changing fishes have the remarkable ability to change reproductive sex during adulthood in response to social stimuli, offering a unique opportunity to understand mechanisms by which the nervous system can initiate and coordinate sexual differentiation.

Methods: This study explores sexual differentiation of the forebrain using single nucleus RNA-sequencing in the anemonefish Amphiprion ocellaris, producing the first cellular atlas of a sex-changing brain.

Results: We uncover extensive sex differences in cell type-specific gene expression, relative proportions of cells, baseline neuronal excitation, and predicted inter-neuronal communication. Additionally, we identify the cholecystokinin, galanin, and estrogen systems as central molecular axes of sexual differentiation. Supported by these findings, we propose a model of sexual differentiation in the conserved vertebrate social decision-making network spanning multiple subtypes of neurons and glia, including neuronal subpopulations within the preoptic area that are positioned to regulate gonadal differentiation.

Conclusions: This work deepens our understanding of sexual differentiation in the vertebrate brain and defines a rich suite of molecular and cellular pathways that differentiate during adult sex change in anemonefish.

背景:大脑的性别分化发生在所有主要的脊椎动物谱系中,但在分子和细胞水平上却没有得到很好的理解。与大多数脊椎动物不同,变性鱼类具有在成年期因社会刺激而改变生殖性别的非凡能力,这为了解神经系统启动和协调性分化的机制提供了一个独特的机会:本研究利用单核 RNA 测序技术探讨了鳗鲡前脑的性分化,首次绘制了变性大脑的细胞图谱:结果:我们发现了在细胞类型特异性基因表达、细胞相对比例、基线神经元兴奋和预测的神经元间通讯方面存在的广泛性别差异。此外,我们还发现胆囊收缩素、加拉宁和雌激素系统是性分化的核心分子轴。在这些发现的支持下,我们提出了一个在保守的脊椎动物社会决策网络中进行性分化的模型,该网络跨越多种亚型神经元和神经胶质细胞,包括视前区的神经元亚群,它们被定位为调节性腺分化:这项研究加深了我们对脊椎动物大脑中性分化的理解,并定义了一套丰富的分子和细胞通路,这些通路在鲶鱼成年后的性变化过程中发生分化。
{"title":"Adult sex change leads to extensive forebrain reorganization in clownfish.","authors":"Coltan G Parker, George W Gruenhagen, Brianna E Hegarty, Abigail R Histed, Jeffrey T Streelman, Justin S Rhodes, Zachary V Johnson","doi":"10.1186/s13293-024-00632-0","DOIUrl":"10.1186/s13293-024-00632-0","url":null,"abstract":"<p><strong>Background: </strong>Sexual differentiation of the brain occurs in all major vertebrate lineages but is not well understood at a molecular and cellular level. Unlike most vertebrates, sex-changing fishes have the remarkable ability to change reproductive sex during adulthood in response to social stimuli, offering a unique opportunity to understand mechanisms by which the nervous system can initiate and coordinate sexual differentiation.</p><p><strong>Methods: </strong>This study explores sexual differentiation of the forebrain using single nucleus RNA-sequencing in the anemonefish Amphiprion ocellaris, producing the first cellular atlas of a sex-changing brain.</p><p><strong>Results: </strong>We uncover extensive sex differences in cell type-specific gene expression, relative proportions of cells, baseline neuronal excitation, and predicted inter-neuronal communication. Additionally, we identify the cholecystokinin, galanin, and estrogen systems as central molecular axes of sexual differentiation. Supported by these findings, we propose a model of sexual differentiation in the conserved vertebrate social decision-making network spanning multiple subtypes of neurons and glia, including neuronal subpopulations within the preoptic area that are positioned to regulate gonadal differentiation.</p><p><strong>Conclusions: </strong>This work deepens our understanding of sexual differentiation in the vertebrate brain and defines a rich suite of molecular and cellular pathways that differentiate during adult sex change in anemonefish.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":"15 1","pages":"58"},"PeriodicalIF":4.9,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11267845/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141750951","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Sex differences in the social motivation of rats: Insights from social operant conditioning, behavioural economics, and video tracking. 大鼠社交动机的性别差异:从社会操作性条件反射、行为经济学和视频追踪中获得的启示。
IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-07-19 DOI: 10.1186/s13293-024-00612-4
Joel S Raymond, Simone Rehn, Morgan H James, Nicholas A Everett, Michael T Bowen
<p><strong>Background: </strong>Social behaviour plays a key role in mental health and wellbeing, and developing greater understanding of mechanisms underlying social interaction-particularly social motivation-holds substantial transdiagnostic impact. Common rodent behavioural assays used to assess social behaviour are limited in their assessment of social motivation, whereas the social operant conditioning model can provide unique and valuable insights into social motivation. Further characterisation of common experimental parameters that may influence social motivation within the social operant model, as well as complementary methodological and analytical approaches, are warranted.</p><p><strong>Methods: </strong>This study investigated the effects of biological sex, housing condition, and time-of-day, on social motivation using the social operant model. This involved training rats to lever press (FR1) for 60-s access to a social reward (same-sex conspecific stimulus). Subjects were male and female Wistar rats, housed under individual or paired conditions, and sessions were conducted either in the mid-late light phase (ZT6-10) or early-mid dark phase (ZT13-17). A behavioural economics approach was implemented to measure social demand and the influence of stimulus partner sex (same- vs. opposite-sex stimulus) on social operant responding. Additionally, video tracking analyses were conducted to assess the degree of convergence between social appetitive and consummatory behaviours.</p><p><strong>Results: </strong>Biological sex, housing conditions, the interaction between sex and housing, and stimulus partner sex potently influenced social motivation, whereas time-of-day did not. Behavioural economics demonstrated that sex, housing, and their interaction influence both the hedonic set-point and elasticity of social demand. Video analysis of social interaction during social operant sessions revealed that social appetitive and consummatory behaviours are not necessarily convergent, and indicate potential social satiety. Lastly, oestrus phase of female experimental and stimulus rats did not impact social motivation within the model.</p><p><strong>Conclusions: </strong>Social isolation-dependent sex differences exist in social motivation for rats, as assessed by social operant conditioning. The social operant model represents an optimal preclinical assay that comprehensively evaluates social motivation and offers a platform for future investigations of neurobiological mechanisms underlying sex differences in social motivation. These findings highlight the importance of continued consideration and inclusion of sex as a biological variable in future social operant conditioning studies. Humans are social creatures-our everyday interactions with others and the support this provides play a key role in our wellbeing. For those experiencing mental health conditions, people's motivation to engage with others can wane, which can lead them to withdraw from thos
背景:社会行为在心理健康和幸福中起着关键作用,进一步了解社会互动的基本机制,尤其是社会动机,对跨诊断具有重大影响。用于评估社交行为的常见啮齿动物行为测定方法在评估社交动机方面存在局限性,而社交操作性条件反射模型则能为社交动机提供独特而有价值的见解。有必要进一步确定社会操作模型中可能影响社会动机的常见实验参数的特征,以及补充方法学和分析方法:本研究使用社会操作模型调查了生理性别、饲养条件和时间对社会动机的影响。这包括训练大鼠按下杠杆(FR1)以获得 60 天的社会奖赏(同性同种刺激)。受试者为雄性和雌性 Wistar 大鼠,饲养条件为单独饲养或配对饲养,训练在光照中后期(ZT6-10)或光照早中期(ZT13-17)进行。实验采用行为经济学方法测量社会需求以及刺激伙伴性别(同性刺激与异性刺激)对社会操作反应的影响。此外,还进行了视频追踪分析,以评估社会性食欲和消耗行为的趋同程度:结果:生物性别、饲养条件、性别与饲养条件之间的交互作用以及刺激伙伴的性别对社会动机有很大影响,而时间则没有影响。行为经济学证明,性别、住房条件及其相互作用会影响享乐设定点和社会需求弹性。对社交操作过程中的社交互动进行的视频分析表明,社交食欲和消费行为并不一定趋同,它们表明潜在的社交饱腹感。最后,雌性实验鼠和刺激鼠的发情期不会影响模型中的社交动机:结论:通过社会操作性条件反射评估,大鼠的社会动机存在依赖于社会隔离的性别差异。社会操作模型是一种能全面评估社会动机的最佳临床前检测方法,并为今后研究社会动机性别差异的神经生物学机制提供了一个平台。这些发现强调了在未来的社会操作性条件反射研究中继续考虑并将性别作为生物变量的重要性。人类是社会性动物--我们与他人的日常互动以及这种互动所提供的支持对我们的身心健康起着关键作用。对于那些患有精神疾病的人来说,与他人交往的动力可能会减弱,这可能会导致他们从支持他们的人那里退缩。因此,为了针对这些症状制定更好的治疗策略,我们需要更深入地了解社交动机。在动物身上研究社交行为可以促进对社交动机的研究,因为它可以让我们了解潜在神经生物学的因果关系,而这在人体实验中是不可能实现的。研究动物社会动机的最佳方法是使用社会操作性条件反射模型,即让大鼠学会按下杠杆,从而打开一扇门,并允许它们与另一只大鼠进行短时间的互动。本研究通过测试性别、饲养条件、时间和刺激伙伴的性别是否会影响大鼠寻求与另一只大鼠互动的动机,来描述社会操作模式的特点。我们发现,雌性大鼠比雄性大鼠更有社会动机,而单独生活的大鼠比与另一只大鼠生活的大鼠更有社会动机;有趣的是,居住条件对雌性大鼠的影响比对雄性大鼠的影响更大。无论性别如何,大鼠都更愿意与异性大鼠交往。这些发现提供了关于大鼠社会动机性别差异的见解,以及对社会操作模型的新认识,这将有助于指导未来对社会动机和其他心理健康问题的研究。
{"title":"Sex differences in the social motivation of rats: Insights from social operant conditioning, behavioural economics, and video tracking.","authors":"Joel S Raymond, Simone Rehn, Morgan H James, Nicholas A Everett, Michael T Bowen","doi":"10.1186/s13293-024-00612-4","DOIUrl":"10.1186/s13293-024-00612-4","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Social behaviour plays a key role in mental health and wellbeing, and developing greater understanding of mechanisms underlying social interaction-particularly social motivation-holds substantial transdiagnostic impact. Common rodent behavioural assays used to assess social behaviour are limited in their assessment of social motivation, whereas the social operant conditioning model can provide unique and valuable insights into social motivation. Further characterisation of common experimental parameters that may influence social motivation within the social operant model, as well as complementary methodological and analytical approaches, are warranted.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;This study investigated the effects of biological sex, housing condition, and time-of-day, on social motivation using the social operant model. This involved training rats to lever press (FR1) for 60-s access to a social reward (same-sex conspecific stimulus). Subjects were male and female Wistar rats, housed under individual or paired conditions, and sessions were conducted either in the mid-late light phase (ZT6-10) or early-mid dark phase (ZT13-17). A behavioural economics approach was implemented to measure social demand and the influence of stimulus partner sex (same- vs. opposite-sex stimulus) on social operant responding. Additionally, video tracking analyses were conducted to assess the degree of convergence between social appetitive and consummatory behaviours.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;Biological sex, housing conditions, the interaction between sex and housing, and stimulus partner sex potently influenced social motivation, whereas time-of-day did not. Behavioural economics demonstrated that sex, housing, and their interaction influence both the hedonic set-point and elasticity of social demand. Video analysis of social interaction during social operant sessions revealed that social appetitive and consummatory behaviours are not necessarily convergent, and indicate potential social satiety. Lastly, oestrus phase of female experimental and stimulus rats did not impact social motivation within the model.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusions: &lt;/strong&gt;Social isolation-dependent sex differences exist in social motivation for rats, as assessed by social operant conditioning. The social operant model represents an optimal preclinical assay that comprehensively evaluates social motivation and offers a platform for future investigations of neurobiological mechanisms underlying sex differences in social motivation. These findings highlight the importance of continued consideration and inclusion of sex as a biological variable in future social operant conditioning studies. Humans are social creatures-our everyday interactions with others and the support this provides play a key role in our wellbeing. For those experiencing mental health conditions, people's motivation to engage with others can wane, which can lead them to withdraw from thos","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":"15 1","pages":"57"},"PeriodicalIF":4.9,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11264584/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141726824","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Sex differences in airway disease: estrogen and airway surface liquid dynamics. 气道疾病的性别差异:雌激素与气道表面液体动力学。
IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-07-18 DOI: 10.1186/s13293-024-00633-z
Brian J Harvey, Noel G McElvaney

Biological sex differences exist for many airway diseases in which females have either worse or better health outcomes. Inflammatory airway diseases such as cystic fibrosis (CF) and asthma display a clear male advantage in post-puberty while a female benefit is observed in asthma during the pre-puberty years. The influence of menstrual cycle stage and pregnancy on the frequency and severity of pulmonary exacerbations in CF and asthma point to a role for sex steroid hormones, particularly estrogen, in underpinning biological sex differences in these diseases. There are many ways by which estrogen may aggravate asthma and CF involving disturbances in airway surface liquid (ASL) dynamics, inappropriate hyper-immune and allergenic responses, as well as exacerbation of pathogen virulence. The deleterious effect of estrogen on pulmonary function in CF and asthma contrasts with the female advantage observed in airway diseases characterised by pulmonary edema such as pneumonia, acute respiratory distress syndrome (ARDS) and COVID-19. Airway surface liquid hypersecretion and alveolar flooding are hallmarks of ARDS and COVID-19, and contribute to the morbidity and mortality of severe forms of these diseases. ASL dynamics encompasses the intrinsic features of the thin lining of fluid covering the airway epithelium which regulate mucociliary clearance (ciliary beat, ASL height, volume, pH, viscosity, mucins, and channel activating proteases) in addition to innate defence mechanisms (pathogen virulence, cytokines, defensins, specialised pro-resolution lipid mediators, and metabolism). Estrogen regulation of ASL dynamics contributing to biological sex differences in CF, asthma and COVID-19 is a major focus of this review.

许多气道疾病都存在生物学性别差异,女性的健康状况要么更差,要么更好。囊性纤维化(CF)和哮喘等气道炎症性疾病在青春期后显示出明显的男性优势,而哮喘在青春期前则显示出女性优势。月经周期阶段和怀孕对 CF 和哮喘肺部恶化的频率和严重程度的影响表明,性类固醇激素(尤其是雌激素)在这些疾病的生物性别差异中扮演着重要角色。雌激素可通过多种途径加重哮喘和 CF 的病情,包括气道表面液体(ASL)动力学紊乱、不适当的高免疫和过敏原反应,以及病原体毒力的增强。雌激素对 CF 和哮喘患者肺功能的有害影响,与在肺炎、急性呼吸窘迫综合征(ARDS)和 COVID-19 等以肺水肿为特征的气道疾病中观察到的女性优势形成鲜明对比。气道表面液体分泌过多和肺泡充血是 ARDS 和 COVID-19 的特征,也是导致这些疾病的发病率和死亡率的重要原因。ASL 动态包括覆盖气道上皮的液体薄层的固有特征,除了先天防御机制(病原体毒力、细胞因子、防御素、特殊的促溶解脂质介质和新陈代谢)外,它还能调节粘液纤毛清除(纤毛节拍、ASL 高度、体积、pH 值、粘度、粘蛋白和通道激活蛋白酶)。雌激素对 ASL 动态的调节导致了 CF、哮喘和 COVID-19 的生物性别差异,这是本综述的重点。
{"title":"Sex differences in airway disease: estrogen and airway surface liquid dynamics.","authors":"Brian J Harvey, Noel G McElvaney","doi":"10.1186/s13293-024-00633-z","DOIUrl":"10.1186/s13293-024-00633-z","url":null,"abstract":"<p><p>Biological sex differences exist for many airway diseases in which females have either worse or better health outcomes. Inflammatory airway diseases such as cystic fibrosis (CF) and asthma display a clear male advantage in post-puberty while a female benefit is observed in asthma during the pre-puberty years. The influence of menstrual cycle stage and pregnancy on the frequency and severity of pulmonary exacerbations in CF and asthma point to a role for sex steroid hormones, particularly estrogen, in underpinning biological sex differences in these diseases. There are many ways by which estrogen may aggravate asthma and CF involving disturbances in airway surface liquid (ASL) dynamics, inappropriate hyper-immune and allergenic responses, as well as exacerbation of pathogen virulence. The deleterious effect of estrogen on pulmonary function in CF and asthma contrasts with the female advantage observed in airway diseases characterised by pulmonary edema such as pneumonia, acute respiratory distress syndrome (ARDS) and COVID-19. Airway surface liquid hypersecretion and alveolar flooding are hallmarks of ARDS and COVID-19, and contribute to the morbidity and mortality of severe forms of these diseases. ASL dynamics encompasses the intrinsic features of the thin lining of fluid covering the airway epithelium which regulate mucociliary clearance (ciliary beat, ASL height, volume, pH, viscosity, mucins, and channel activating proteases) in addition to innate defence mechanisms (pathogen virulence, cytokines, defensins, specialised pro-resolution lipid mediators, and metabolism). Estrogen regulation of ASL dynamics contributing to biological sex differences in CF, asthma and COVID-19 is a major focus of this review.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":"15 1","pages":"56"},"PeriodicalIF":4.9,"publicationDate":"2024-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11264786/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141722986","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Sex-specific role of high-fat diet and stress on behavior, energy metabolism, and the ventromedial hypothalamus. 高脂饮食和压力对行为、能量代谢和腹内侧下丘脑的作用具有性别特异性。
IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-07-15 DOI: 10.1186/s13293-024-00628-w
Sanutha Shetty, Samuel J Duesman, Sanil Patel, Pacific Huynh, Pamela Toh, Sanjana Shroff, Anika Das, Disha Chowhan, Benjamin Keller, Johana Alvarez, Rachel Fisher-Foye, Robert Sebra, Kristin Beaumont, Cameron S McAlpine, Prashant Rajbhandari, Abha K Rajbhandari

Background: Scientific evidence highlights the influence of biological sex on the relationship between stress and metabolic dysfunctions. However, there is limited understanding of how diet and stress concurrently contribute to metabolic dysregulation in both males and females. Our study aimed to investigate the combined effects of high-fat diet (HFD) induced obesity and repeated stress on fear-related behaviors, metabolic, immune, and hypothalamic outcomes in male and female mice.

Methods: To investigate this, we used a highly reliable rodent behavioral model that faithfully recapitulates key aspects of post-traumatic stress disorder (PTSD)-like fear. We subjected mice to footshock stressor followed by a weekly singular footshock stressor or no stressor for 14 weeks while on either an HFD or chow diet. At weeks 10 and 14 we conducted glucose tolerance and insulin sensitivity measurements. Additionally, we placed the mice in metabolic chambers to perform indirect calorimetric measurements. Finally, we collected brain and peripheral tissues for cellular analysis.

Results: We observed that HFD-induced obesity disrupted fear memory extinction, increased glucose intolerance, and affected energy expenditure specifically in male mice. Conversely, female mice on HFD exhibited reduced respiratory exchange ratio (RER), and a significant defect in glucose tolerance only when subjected to repeated stress. Furthermore, the combination of repeated stress and HFD led to sex-specific alterations in proinflammatory markers and hematopoietic stem cells across various peripheral metabolic tissues. Single-nuclei RNA sequencing (snRNAseq) analysis of the ventromedial hypothalamus (VMH) revealed microglial activation in female mice on HFD, while male mice on HFD exhibited astrocytic activation under repeated stress.

Conclusions: Overall, our findings provide insights into complex interplay between repeated stress, high-fat diet regimen, and their cumulative effects on health, including their potential contribution to the development of PTSD-like stress and metabolic dysfunctions, emphasizing the need for further research to fully understand these interconnected pathways and their implications for health.

背景:科学证据强调了生理性别对压力和代谢功能障碍之间关系的影响。然而,人们对饮食和压力如何同时导致男性和女性代谢失调的了解还很有限。我们的研究旨在调查高脂饮食(HFD)诱导肥胖和反复应激对雌雄小鼠恐惧相关行为、代谢、免疫和下丘脑结果的综合影响:为了研究这个问题,我们使用了一种高度可靠的啮齿动物行为模型,它能忠实地再现创伤后应激障碍(PTSD)样恐惧的关键方面。我们对小鼠进行了为期 14 周的脚震应激,随后每周进行一次单次脚震应激或不进行应激,同时让小鼠食用高脂低糖食物或饲料。在第 10 周和第 14 周,我们进行了葡萄糖耐量和胰岛素敏感性测量。此外,我们还将小鼠放入代谢室进行间接热量测量。最后,我们收集了脑组织和外周组织进行细胞分析:结果:我们观察到,高密度脂蛋白胆固醇诱导的肥胖破坏了恐惧记忆的消退,增加了葡萄糖不耐受性,并影响了雄性小鼠的能量消耗。相反,雌性高氟日粮小鼠只有在反复应激时才会表现出呼吸交换比(RER)降低和葡萄糖耐量的显著缺陷。此外,重复应激和高氟酸膳食的组合导致了不同性别小鼠外周代谢组织中促炎标志物和造血干细胞的改变。腹内侧下丘脑(VMH)的单核糖核酸测序(snRNAseq)分析显示,高频分解雌性小鼠的小胶质细胞活化,而高频分解雄性小鼠在反复应激下表现出星形胶质细胞活化:总之,我们的研究结果提供了有关反复应激、高脂饮食方案及其对健康的累积影响之间复杂相互作用的见解,包括它们对创伤后应激障碍样应激和代谢功能障碍的潜在贡献,强调了进一步研究的必要性,以充分了解这些相互关联的途径及其对健康的影响。
{"title":"Sex-specific role of high-fat diet and stress on behavior, energy metabolism, and the ventromedial hypothalamus.","authors":"Sanutha Shetty, Samuel J Duesman, Sanil Patel, Pacific Huynh, Pamela Toh, Sanjana Shroff, Anika Das, Disha Chowhan, Benjamin Keller, Johana Alvarez, Rachel Fisher-Foye, Robert Sebra, Kristin Beaumont, Cameron S McAlpine, Prashant Rajbhandari, Abha K Rajbhandari","doi":"10.1186/s13293-024-00628-w","DOIUrl":"10.1186/s13293-024-00628-w","url":null,"abstract":"<p><strong>Background: </strong>Scientific evidence highlights the influence of biological sex on the relationship between stress and metabolic dysfunctions. However, there is limited understanding of how diet and stress concurrently contribute to metabolic dysregulation in both males and females. Our study aimed to investigate the combined effects of high-fat diet (HFD) induced obesity and repeated stress on fear-related behaviors, metabolic, immune, and hypothalamic outcomes in male and female mice.</p><p><strong>Methods: </strong>To investigate this, we used a highly reliable rodent behavioral model that faithfully recapitulates key aspects of post-traumatic stress disorder (PTSD)-like fear. We subjected mice to footshock stressor followed by a weekly singular footshock stressor or no stressor for 14 weeks while on either an HFD or chow diet. At weeks 10 and 14 we conducted glucose tolerance and insulin sensitivity measurements. Additionally, we placed the mice in metabolic chambers to perform indirect calorimetric measurements. Finally, we collected brain and peripheral tissues for cellular analysis.</p><p><strong>Results: </strong>We observed that HFD-induced obesity disrupted fear memory extinction, increased glucose intolerance, and affected energy expenditure specifically in male mice. Conversely, female mice on HFD exhibited reduced respiratory exchange ratio (RER), and a significant defect in glucose tolerance only when subjected to repeated stress. Furthermore, the combination of repeated stress and HFD led to sex-specific alterations in proinflammatory markers and hematopoietic stem cells across various peripheral metabolic tissues. Single-nuclei RNA sequencing (snRNAseq) analysis of the ventromedial hypothalamus (VMH) revealed microglial activation in female mice on HFD, while male mice on HFD exhibited astrocytic activation under repeated stress.</p><p><strong>Conclusions: </strong>Overall, our findings provide insights into complex interplay between repeated stress, high-fat diet regimen, and their cumulative effects on health, including their potential contribution to the development of PTSD-like stress and metabolic dysfunctions, emphasizing the need for further research to fully understand these interconnected pathways and their implications for health.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":"15 1","pages":"55"},"PeriodicalIF":4.9,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11247790/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619216","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Sex differences in membrane properties and cellular excitability of dopamine D1 receptor-expressing neurons within the shell of the nucleus accumbens of pre- and mid-adolescent mice. 青春期前期和中期小鼠大脑核外壳内多巴胺 D1 受体表达神经元的膜特性和细胞兴奋性的性别差异。
IF 5.4 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-07-13 DOI: 10.1186/s13293-024-00631-1
Heather C Aziz, Regina A Mangieri

Background: The transition from childhood to adulthood, or adolescence, a developmental stage, is characterized by psychosocial and biological changes. The nucleus accumbens (NAc), a striatal brain region composed of the core (NAcC) and shell (NAcSh), has been linked to risk-taking behavior and implicated in reward seeking and evaluation. Most neurons in the NAc are medium spiny neurons (MSNs) that express dopamine D1 receptors (D1R +) and/or dopamine D2 receptors (D2R +). Changes in dopaminergic and glutamatergic systems occur during adolescence and converge in the NAc. While there are previous investigations into sex differences in membrane excitability and synaptic glutamate transmission in both subdivisions of the NAc, to our knowledge, none have specified NAcSh D1R + MSNs from mice during pre- and mid-adolescence.

Methods: Sagittal brain slices containing the NAc were prepared from B6.Cg-Tg(Drd1a-tdTomato)6Calak/J mice of both sexes from postnatal days 21-25 and 35-47, representing pre- and mid-adolescence, respectively. Whole-cell electrophysiology recordings were collected from NAcSh D1R + MSNs in the form of membrane-voltage responses to current injections, to assess membrane properties and action potential waveform characteristics, and spontaneous excitatory postsynaptic currents (sEPSCs) to assess glutamatergic synaptic activity.

Results: Relative to pre-adolescent males, pre-adolescent female NAcSh D1R + MSNs exhibited a less hyperpolarized resting membrane potential, increased input resistance, and smaller action potential afterhyperpolarization amplitudes. During mid-adolescence, decreased input resistance and a shorter action potential duration in females were the only sex differences observed.

Conclusions: Taken together, our results indicate that NAcSh D1R + MSNs in mice exhibit sex differences in membrane properties and AP waveform during pre-adolescence that are overall indicative of increased cellular excitability in females and are suggestive of possible sex differences in glycine receptors, inwardly-rectifying potassium channels, and large conductance voltage-gated potassium channels. These differences do not appear to persist into mid-adolescence, when sex was observed to affect input resistance oppositely to that of pre-adolescence and AP waveform in a manner suggestive of differences in voltage-gated potassium channels.

背景:从童年到成年(即青春期)这一发育阶段的过渡,以社会心理和生理变化为特征。大脑纹状体核(NAc)由核心区(NAcC)和外壳区(NAcSh)组成,与冒险行为有关,并与奖赏寻求和评估有关。NAc 中的大多数神经元是中刺神经元(MSN),它们表达多巴胺 D1 受体(D1R +)和/或多巴胺 D2 受体(D2R +)。多巴胺能和谷氨酸能系统的变化发生在青春期,并在 NAc 中汇聚。虽然以前曾对 NAc 两个分支的膜兴奋性和突触谷氨酸传递的性别差异进行过研究,但据我们所知,还没有研究对青春期前中期小鼠的 NAcSh D1R + MSNs 进行过专门研究:方法:从B6.Cg-Tg(Drd1a-tdTomato)6Calak/J雌雄小鼠出生后第21-25天和第35-47天(分别代表青春期前期和中期)制备含有NAc的矢状脑切片。采集了NAcSh D1R + MSNs的全细胞电生理记录,记录形式包括对电流注入的膜电压反应,以评估膜特性和动作电位波形特征,以及自发兴奋性突触后电流(sEPSCs),以评估谷氨酸能突触活动:与青春期前的男性相比,青春期前的女性NAcSh D1R + MSN表现出较低的超极化静息膜电位、较高的输入阻抗和较小的动作电位超极化后振幅。在青春期中期,雌性的输入阻力降低和动作电位持续时间缩短是观察到的唯一性别差异:综上所述,我们的研究结果表明,小鼠的 NAcSh D1R + MSNs 在青春前期的膜特性和 AP 波形上表现出性别差异,总体上表明雌性细胞的兴奋性增加,并提示甘氨酸受体、内向整流钾通道和大电导电压门控钾通道可能存在性别差异。这些差异似乎不会持续到青春期中期,因为在中期观察到性别对输入电阻的影响与青春期前期相反,对 AP 波形的影响也与电压门控钾通道的差异有关。
{"title":"Sex differences in membrane properties and cellular excitability of dopamine D1 receptor-expressing neurons within the shell of the nucleus accumbens of pre- and mid-adolescent mice.","authors":"Heather C Aziz, Regina A Mangieri","doi":"10.1186/s13293-024-00631-1","DOIUrl":"10.1186/s13293-024-00631-1","url":null,"abstract":"<p><strong>Background: </strong>The transition from childhood to adulthood, or adolescence, a developmental stage, is characterized by psychosocial and biological changes. The nucleus accumbens (NAc), a striatal brain region composed of the core (NAcC) and shell (NAcSh), has been linked to risk-taking behavior and implicated in reward seeking and evaluation. Most neurons in the NAc are medium spiny neurons (MSNs) that express dopamine D1 receptors (D1R +) and/or dopamine D2 receptors (D2R +). Changes in dopaminergic and glutamatergic systems occur during adolescence and converge in the NAc. While there are previous investigations into sex differences in membrane excitability and synaptic glutamate transmission in both subdivisions of the NAc, to our knowledge, none have specified NAcSh D1R + MSNs from mice during pre- and mid-adolescence.</p><p><strong>Methods: </strong>Sagittal brain slices containing the NAc were prepared from B6.Cg-Tg(Drd1a-tdTomato)6Calak/J mice of both sexes from postnatal days 21-25 and 35-47, representing pre- and mid-adolescence, respectively. Whole-cell electrophysiology recordings were collected from NAcSh D1R + MSNs in the form of membrane-voltage responses to current injections, to assess membrane properties and action potential waveform characteristics, and spontaneous excitatory postsynaptic currents (sEPSCs) to assess glutamatergic synaptic activity.</p><p><strong>Results: </strong>Relative to pre-adolescent males, pre-adolescent female NAcSh D1R + MSNs exhibited a less hyperpolarized resting membrane potential, increased input resistance, and smaller action potential afterhyperpolarization amplitudes. During mid-adolescence, decreased input resistance and a shorter action potential duration in females were the only sex differences observed.</p><p><strong>Conclusions: </strong>Taken together, our results indicate that NAcSh D1R + MSNs in mice exhibit sex differences in membrane properties and AP waveform during pre-adolescence that are overall indicative of increased cellular excitability in females and are suggestive of possible sex differences in glycine receptors, inwardly-rectifying potassium channels, and large conductance voltage-gated potassium channels. These differences do not appear to persist into mid-adolescence, when sex was observed to affect input resistance oppositely to that of pre-adolescence and AP waveform in a manner suggestive of differences in voltage-gated potassium channels.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":"15 1","pages":"54"},"PeriodicalIF":5.4,"publicationDate":"2024-07-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11245857/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141603265","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MicroRNA-21 modulates brown adipose tissue adipogenesis and thermogenesis in a mouse model of polycystic ovary syndrome. MicroRNA-21 可调节多囊卵巢综合征小鼠模型中棕色脂肪组织的脂肪生成和产热。
IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-07-10 DOI: 10.1186/s13293-024-00630-2
Samar Rezq, Alexandra M Huffman, Jelina Basnet, Amira E Alsemeh, Jussara M do Carmo, Licy L Yanes Cardozo, Damian G Romero

Background: Polycystic ovary syndrome (PCOS), the most common endocrine disorder in premenopausal women, is associated with increased obesity, hyperandrogenism, and altered brown adipose tissue (BAT) thermogenesis. MicroRNAs play critical functions in brown adipocyte differentiation and maintenance. We aim to study the role of microRNA-21 (miR-21) in altered energy homeostasis and BAT thermogenesis in a PCOS mouse model of peripubertal androgen exposure.

Methods: Three-week-old miR-21 knockout (miR21KO) or wild-type (WT) female mice were treated with dihydrotestosterone (DHT) or vehicle for 90 days. Body composition was determined by EchoMRI. Energy expenditure (EE), oxygen consumption (VO2), carbon dioxide production (VCO2), and respiratory exchange ratio (RER) were measured by indirect calorimetry. Androgen receptor (AR), and markers of adipogenesis, de novo lipogenesis, angiogenesis, extracellular matrix remodeling, and thermogenesis were quantified by RT-qPCR and/or Western-blot.

Results: MiR-21 ablation attenuated DHT-mediated increase in body weight while having no effect on fat or BAT mass. MiR-21 ablation attenuated DHT-mediated BAT AR upregulation. MiR-21 ablation did not alter EE; however, miR21KO DHT-treated mice have reduced VO2, VCO2, and RER. MiR-21 ablation reversed DHT-mediated decrease in food intake and increase in sleep time. MiR-21 ablation decreased some adipogenesis (Adipoq, Pparγ, and Cebpβ) and extracellular matrix remodeling (Mmp-9 and Timp-1) markers expression in DHT-treated mice. MiR-21 ablation abolished DHT-mediated increases in thermogenesis markers Cpt1a and Cpt1b, while decreasing CIDE-A expression.

Conclusions: Our findings suggest that BAT miR-21 may play a role in regulating DHT-mediated thermogenic dysfunction in PCOS. Modulation of BAT miR-21 levels could be a novel therapeutic approach for the treatment of PCOS-associated metabolic derangements.

背景:多囊卵巢综合征(PCOS)是绝经前妇女最常见的内分泌疾病,与肥胖增加、高雄激素和棕色脂肪组织(BAT)产热改变有关。微RNA在棕色脂肪细胞的分化和维持中发挥着关键作用。我们旨在研究在围青春期雄激素暴露的多囊卵巢综合征小鼠模型中,microRNA-21(miR-21)在改变能量平衡和棕色脂肪组织产热中的作用:方法:用双氢睾酮(DHT)或药物治疗三周大的 miR-21 基因敲除(miR21KO)或野生型(WT)雌性小鼠 90 天。身体成分通过回波磁共振成像测定。能量消耗(EE)、耗氧量(VO2)、二氧化碳产生量(VCO2)和呼吸交换比(RER)由间接量热法测量。雄激素受体(AR)以及脂肪生成、新脂肪生成、血管生成、细胞外基质重塑和产热的标记物通过 RT-qPCR 和/或 Western-blot 进行量化:结果:MiR-21消减减轻了DHT介导的体重增加,但对脂肪或BAT质量没有影响。消融 MiR-21 可减轻 DHT 介导的 BAT AR 上调。消减 MiR-21 并未改变 EE;然而,miR21KO DHT 处理的小鼠的 VO2、VCO2 和 RER 均降低。MiR-21 消融逆转了 DHT 介导的食物摄入量减少和睡眠时间增加。消减 MiR-21 可减少 DHT 治疗小鼠的一些脂肪生成(Adipoq、Pparγ 和 Cebpβ)和细胞外基质重塑(Mmp-9 和 Timp-1)标记物的表达。MiR-21的消减抑制了DHT介导的产热标志物Cpt1a和Cpt1b的增加,同时降低了CIDE-A的表达:我们的研究结果表明,在多囊卵巢综合征中,BAT miR-21 可能在调节 DHT 介导的生热功能障碍中发挥作用。调节BAT miR-21水平可能是治疗多囊卵巢综合征相关代谢紊乱的一种新型治疗方法。
{"title":"MicroRNA-21 modulates brown adipose tissue adipogenesis and thermogenesis in a mouse model of polycystic ovary syndrome.","authors":"Samar Rezq, Alexandra M Huffman, Jelina Basnet, Amira E Alsemeh, Jussara M do Carmo, Licy L Yanes Cardozo, Damian G Romero","doi":"10.1186/s13293-024-00630-2","DOIUrl":"10.1186/s13293-024-00630-2","url":null,"abstract":"<p><strong>Background: </strong>Polycystic ovary syndrome (PCOS), the most common endocrine disorder in premenopausal women, is associated with increased obesity, hyperandrogenism, and altered brown adipose tissue (BAT) thermogenesis. MicroRNAs play critical functions in brown adipocyte differentiation and maintenance. We aim to study the role of microRNA-21 (miR-21) in altered energy homeostasis and BAT thermogenesis in a PCOS mouse model of peripubertal androgen exposure.</p><p><strong>Methods: </strong>Three-week-old miR-21 knockout (miR21KO) or wild-type (WT) female mice were treated with dihydrotestosterone (DHT) or vehicle for 90 days. Body composition was determined by EchoMRI. Energy expenditure (EE), oxygen consumption (VO2), carbon dioxide production (VCO2), and respiratory exchange ratio (RER) were measured by indirect calorimetry. Androgen receptor (AR), and markers of adipogenesis, de novo lipogenesis, angiogenesis, extracellular matrix remodeling, and thermogenesis were quantified by RT-qPCR and/or Western-blot.</p><p><strong>Results: </strong>MiR-21 ablation attenuated DHT-mediated increase in body weight while having no effect on fat or BAT mass. MiR-21 ablation attenuated DHT-mediated BAT AR upregulation. MiR-21 ablation did not alter EE; however, miR21KO DHT-treated mice have reduced VO2, VCO2, and RER. MiR-21 ablation reversed DHT-mediated decrease in food intake and increase in sleep time. MiR-21 ablation decreased some adipogenesis (Adipoq, Pparγ, and Cebpβ) and extracellular matrix remodeling (Mmp-9 and Timp-1) markers expression in DHT-treated mice. MiR-21 ablation abolished DHT-mediated increases in thermogenesis markers Cpt1a and Cpt1b, while decreasing CIDE-A expression.</p><p><strong>Conclusions: </strong>Our findings suggest that BAT miR-21 may play a role in regulating DHT-mediated thermogenic dysfunction in PCOS. Modulation of BAT miR-21 levels could be a novel therapeutic approach for the treatment of PCOS-associated metabolic derangements.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":"15 1","pages":"53"},"PeriodicalIF":4.9,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11238487/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141578916","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Biology of Sex Differences
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1