首页 > 最新文献

Biology of Sex Differences最新文献

英文 中文
XX sex chromosome complement modulates immune responses to heat-killed Streptococcus pneumoniae immunization in a microbiome-dependent manner XX性染色体互补以微生物依赖的方式调节对热杀灭肺炎链球菌免疫的免疫反应
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-03-14 DOI: 10.1186/s13293-024-00597-0
Carly J. Amato-Menker, Quinn Hopen, Andrea Pettit, Jasleen Gandhi, Gangqing Hu, Rosana Schafer, Jennifer Franko
Differences in male vs. female immune responses are well-documented and have significant clinical implications. While the immunomodulatory effects of sex hormones are well established, the contributions of sex chromosome complement (XX vs. XY) and gut microbiome diversity on immune sexual dimorphisms have only recently become appreciated. Here we investigate the individual and collaborative influences of sex chromosome complements and gut microbiota on humoral immune activation. Male and female Four Core Genotype (FCG) mice were immunized with heat-killed Streptococcus pneumoniae (HKSP). Humoral immune responses were assessed, and X-linked immune-related gene expression was evaluated to explain the identified XX-dependent phenotype. The functional role of Kdm6a, an X-linked epigenetic regulatory gene of interest, was evaluated ex vivo using mitogen stimulation of B cells. Additional influences of the gut microbiome on sex chromosome-dependent B cell activation was also evaluated by antibiotically depleting gut microbiota prior to HKSP immunization. Reconstitution of the depleted microbiome with short-chain fatty acid (SCFA)-producing bacteria tested the impact of SCFAs on XX-dependent immune activation. XX mice exhibited higher HKSP-specific IgM-secreting B cells and plasma cell frequencies than XY mice, regardless of gonadal sex. Although Kdm6a was identified as an X-linked gene overexpressed in XX B cells, inhibition of its enzymatic activity did not affect mitogen-induced plasma cell differentiation or antibody production in a sex chromosome-dependent manner ex vivo. Enhanced humoral responses in XX vs. XY immunized FCG mice were eliminated after microbiome depletion, indicating that the microbiome contributes to the identified XX-dependent immune enhancement. Reconstituting microbiota-depleted mice with select SCFA-producing bacteria enhanced fecal SCFA concentrations and increased humoral responses in XX, but not XY, FCG mice. However, exposure to the SCFA propionate alone did not enhance mitogenic B cell stimulation in ex vivo studies. FCG mice have been used to assess sex hormone and sex chromosome complement influences on various sexually dimorphic traits. The current study indicates that the gut microbiome impacts humoral responses in an XX-dependent manner, suggesting that the collaborative influence of gut bacteria and other sex-specific factors should be considered when interpreting data aimed at delineating the mechanisms that promote sexual dimorphism. Male and female immune systems differ in their ability to respond to infectious challenge. While males tend to be more susceptible to infection and produce lower amounts of antibodies in response to vaccination, females are more prone to develop autoimmune and inflammatory diseases. Key contributors to these differences include sex hormones, sex chromosome complement (XX in females vs. XY in males), and distinct gut microbial communities capable of regulating immune activation. While
男性与女性免疫反应的差异是有据可查的,并具有重要的临床意义。虽然性激素的免疫调节作用已得到公认,但性染色体互补(XX 与 XY)和肠道微生物群多样性对免疫性双态性的贡献最近才得到重视。在此,我们研究了性染色体互补和肠道微生物群对体液免疫激活的个体和协同影响。雄性和雌性四核心基因型(FCG)小鼠用热灭活肺炎链球菌(HKSP)进行免疫。评估了体液免疫反应,并评估了 X 连锁免疫相关基因的表达,以解释已确定的 XX 依赖性表型。利用有丝分裂原刺激 B 细胞,对 Kdm6a(一种 X 连锁表观遗传调控基因)的功能作用进行了体内外评估。在进行 HKSP 免疫之前,还通过抗生素消耗肠道微生物群来评估肠道微生物群对性染色体依赖性 B 细胞活化的其他影响。用产生短链脂肪酸(SCFA)的细菌重建被耗竭的微生物群,测试了SCFA对XX依赖性免疫激活的影响。XX小鼠比XY小鼠表现出更高的HKSP特异性IgM分泌B细胞和浆细胞频率,与性腺性别无关。虽然 Kdm6a 被鉴定为在 XX B 细胞中过度表达的 X 连锁基因,但抑制其酶活性并不会影响有丝分裂原诱导的浆细胞分化,也不会以性染色体依赖的方式影响体内抗体的产生。微生物群耗竭后,XX 与 XY 免疫 FCG 小鼠增强的体液反应消失了,这表明微生物群有助于已确定的 XX 依赖性免疫增强。用精选的产生 SCFA 的细菌重组微生物群耗竭的小鼠可提高粪便中 SCFA 的浓度,并增强 XX 而非 XY FCG 小鼠的体液反应。然而,在体外研究中,单独接触 SCFA 丙酸盐并不会增强有丝分裂原 B 细胞刺激。FCG 小鼠已被用于评估性激素和性染色体互补对各种性二态性状的影响。目前的研究表明,肠道微生物组以依赖 XX 的方式影响体液反应,这表明在解释旨在阐明促进性双态性机制的数据时,应考虑肠道细菌和其他性别特异性因素的共同影响。男性和女性的免疫系统在应对传染性挑战的能力上有所不同。男性往往更容易受到感染,接种疫苗后产生的抗体量也较低,而女性则更容易患上自身免疫性和炎症性疾病。造成这些差异的主要因素包括性激素、性染色体互补(女性为 XX,男性为 XY)以及能够调节免疫激活的独特肠道微生物群落。虽然对每个因素都进行了单独研究,但这项研究强调了这些因素共同影响免疫激活的潜力。在本研究中,XX 与 XY 性染色体互补被证明能增强对热杀灭肺炎链球菌疫苗接种的抗体反应。在试图确定这种免疫增强的根本原因时,发现肠道微生物组起着至关重要的作用。在缺乏完整肠道微生物群的情况下,XX 免疫激活降低到了与拥有 XY 性染色体补体的小鼠相似的水平。用精选的可产生 SCFA 的细菌物种替代被耗竭的肠道微生物群,可提高抗生素处理的小鼠体内的 SCFA 水平,并挽救 XX 依赖性免疫增强,这表明 SCFA 起了介导作用。要确定这些选择性细菌如何以性染色体互补依赖的方式影响免疫激活,还需要进一步的研究。我们的研究结果突出表明,在试图了解免疫性别偏差时,需要考虑单个性别特异性因素的协同作用,因为更好地了解这些相互作用很可能会为改进针对两性的疗法和疫苗铺平道路。
{"title":"XX sex chromosome complement modulates immune responses to heat-killed Streptococcus pneumoniae immunization in a microbiome-dependent manner","authors":"Carly J. Amato-Menker, Quinn Hopen, Andrea Pettit, Jasleen Gandhi, Gangqing Hu, Rosana Schafer, Jennifer Franko","doi":"10.1186/s13293-024-00597-0","DOIUrl":"https://doi.org/10.1186/s13293-024-00597-0","url":null,"abstract":"Differences in male vs. female immune responses are well-documented and have significant clinical implications. While the immunomodulatory effects of sex hormones are well established, the contributions of sex chromosome complement (XX vs. XY) and gut microbiome diversity on immune sexual dimorphisms have only recently become appreciated. Here we investigate the individual and collaborative influences of sex chromosome complements and gut microbiota on humoral immune activation. Male and female Four Core Genotype (FCG) mice were immunized with heat-killed Streptococcus pneumoniae (HKSP). Humoral immune responses were assessed, and X-linked immune-related gene expression was evaluated to explain the identified XX-dependent phenotype. The functional role of Kdm6a, an X-linked epigenetic regulatory gene of interest, was evaluated ex vivo using mitogen stimulation of B cells. Additional influences of the gut microbiome on sex chromosome-dependent B cell activation was also evaluated by antibiotically depleting gut microbiota prior to HKSP immunization. Reconstitution of the depleted microbiome with short-chain fatty acid (SCFA)-producing bacteria tested the impact of SCFAs on XX-dependent immune activation. XX mice exhibited higher HKSP-specific IgM-secreting B cells and plasma cell frequencies than XY mice, regardless of gonadal sex. Although Kdm6a was identified as an X-linked gene overexpressed in XX B cells, inhibition of its enzymatic activity did not affect mitogen-induced plasma cell differentiation or antibody production in a sex chromosome-dependent manner ex vivo. Enhanced humoral responses in XX vs. XY immunized FCG mice were eliminated after microbiome depletion, indicating that the microbiome contributes to the identified XX-dependent immune enhancement. Reconstituting microbiota-depleted mice with select SCFA-producing bacteria enhanced fecal SCFA concentrations and increased humoral responses in XX, but not XY, FCG mice. However, exposure to the SCFA propionate alone did not enhance mitogenic B cell stimulation in ex vivo studies. FCG mice have been used to assess sex hormone and sex chromosome complement influences on various sexually dimorphic traits. The current study indicates that the gut microbiome impacts humoral responses in an XX-dependent manner, suggesting that the collaborative influence of gut bacteria and other sex-specific factors should be considered when interpreting data aimed at delineating the mechanisms that promote sexual dimorphism. Male and female immune systems differ in their ability to respond to infectious challenge. While males tend to be more susceptible to infection and produce lower amounts of antibodies in response to vaccination, females are more prone to develop autoimmune and inflammatory diseases. Key contributors to these differences include sex hormones, sex chromosome complement (XX in females vs. XY in males), and distinct gut microbial communities capable of regulating immune activation. While ","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140125841","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
Microbial composition, functionality, and stress resilience or susceptibility: unraveling sex-specific patterns. 微生物组成、功能和应激复原力或易感性:揭示性别特异性模式。
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-02-26 DOI: 10.1186/s13293-024-00590-7
Arax Tanelian, Bistra Nankova, Mariam Miari, Esther L Sabban

Background: Following exposure to traumatic stress, women are twice as likely as men to develop mood disorders. Yet, individual responses to such stress vary, with some people developing stress-induced psychopathologies while others exhibit resilience. The factors influencing sex-related disparities in affective disorders as well as variations in resilience remain unclear; however, emerging evidence suggests differences in the gut microbiota play a role. In this study, using the single prolonged stress (SPS) model of post-traumatic stress disorder, we investigated pre- and post-existing differences in microbial composition, functionality, and metabolites that affect stress susceptibility or resilience in each sex.

Methods: Male and female Sprague-Dawley rats were randomly assigned to control or SPS groups. Two weeks following SPS, the animals were exposed to a battery of behavioral tests and decapitated a day later. Based on their anxiety index, they were further categorized as SPS-resilient (SPS-R) or SPS-susceptible (SPS-S). On the day of dissection, cecum, and selected brain tissues were isolated. Stool samples were collected before and after SPS, whereas urine samples were taken before and 30 min into the SPS.

Results: Before SPS exposure, the sympathoadrenal axis exhibited alterations within male subgroups only. Expression of tight junction protein claudin-5 was lower in brain of SPS-S males, but higher in SPS-R females following SPS. Across the study, alpha diversity remained consistently lower in males compared to females. Beta diversity revealed distinct separations between male and female susceptible groups before SPS, with this separation becoming evident in the resilient groups following SPS. At the genus level, Lactobacillus, Lachnospiraceae_Incertae_Sedis, and Barnesiella exhibited sex-specific alterations, displaying opposing abundances in each sex. Additionally, sex-specific changes were observed in microbial predictive functionality and targeted functional modules both before and after SPS. Alterations in the microbial short-chain fatty acids (SCFAs), were also observed, with major and minor SCFAs being lower in SPS-susceptible males whereas branched-chain SCFAs being higher in SPS-susceptible females.

Conclusion: This study highlights distinct pre- and post-trauma differences in microbial composition, functionality, and metabolites, associated with stress resilience in male and female rats. The findings underscore the importance of developing sex-specific therapeutic strategies to effectively address stress-related disorders. Highlights SPS model induces divergent anxiety and social behavioral responses to traumatic stress in both male and female rodents. SPS-resilient females displayed less anxiety-like behavior and initiated more interactions towards a juvenile rat than SPS-resilient males. Sex-specific pre-existing and SPS-induced differen

背景:遭受创伤性压力后,女性患情绪障碍的几率是男性的两倍。然而,个体对这种压力的反应各不相同,有些人会出现由压力引起的精神病态,而有些人则表现出抗压能力。影响情感障碍中与性别有关的差异以及复原力差异的因素仍不清楚;不过,新出现的证据表明,肠道微生物群的差异在其中发挥了作用。在这项研究中,我们利用创伤后应激障碍的单次长期应激(SPS)模型,研究了影响男女应激易感性或复原力的微生物组成、功能和代谢物的前后差异:雄性和雌性 Sprague-Dawley 大鼠被随机分配到对照组或 SPS 组。SPS两周后,对动物进行一系列行为测试,一天后将其斩首。根据大鼠的焦虑指数,将它们进一步分为 SPS 抗性组(SPS-R)和 SPS 易感组(SPS-S)。解剖当天,分离盲肠和部分脑组织。在接触 SPS 之前和之后采集粪便样本,在接触 SPS 之前和 30 分钟后采集尿液样本:结果:在接触 SPS 之前,交感肾上腺轴仅在男性亚组中出现了变化。SPS-S男性大脑中紧密连接蛋白claudin-5的表达量较低,而SPS-R女性大脑中紧密连接蛋白claudin-5的表达量较高。在整个研究过程中,与女性相比,男性的α多样性一直较低。Beta 多样性显示,在 SPS 之前,男性和女性易感群体之间存在明显的分离,而在 SPS 之后,这种分离在抗病群体中变得更加明显。在属一级,乳酸杆菌、Lachnospiraceae_Incertae_Sedis 和 Barnesiella 表现出性别特异性变化,在每种性别中的丰度相反。此外,在 SPS 前后,微生物的预测功能和目标功能模块都发生了性别特异性变化。还观察到微生物短链脂肪酸(SCFAs)的变化,易受 SPS 影响的雄性微生物的主要和次要 SCFAs 较低,而易受 SPS 影响的雌性微生物的支链 SCFAs 较高:本研究强调了与雌雄大鼠应激复原力相关的微生物组成、功能和代谢物在创伤前后的明显差异。这些发现强调了开发针对不同性别的治疗策略以有效解决应激相关疾病的重要性。亮点 SPS 模型诱导雄性和雌性啮齿动物对创伤性应激做出不同的焦虑和社会行为反应。与具有 SPS 抵抗能力的雄性啮齿动物相比,具有 SPS 抵抗能力的雌性啮齿动物表现出的焦虑行为更少,与幼鼠的互动也更多。在易感大鼠和抗应激大鼠中,观察到了肠道微生物组成和预测功能的性别差异。对 SPS 有抵抗力的雄性大鼠的盲肠乙酸盐含量升高,而对 SPS 易感的雌性大鼠的支链 SCFAs 含量升高。
{"title":"Microbial composition, functionality, and stress resilience or susceptibility: unraveling sex-specific patterns.","authors":"Arax Tanelian, Bistra Nankova, Mariam Miari, Esther L Sabban","doi":"10.1186/s13293-024-00590-7","DOIUrl":"10.1186/s13293-024-00590-7","url":null,"abstract":"<p><strong>Background: </strong>Following exposure to traumatic stress, women are twice as likely as men to develop mood disorders. Yet, individual responses to such stress vary, with some people developing stress-induced psychopathologies while others exhibit resilience. The factors influencing sex-related disparities in affective disorders as well as variations in resilience remain unclear; however, emerging evidence suggests differences in the gut microbiota play a role. In this study, using the single prolonged stress (SPS) model of post-traumatic stress disorder, we investigated pre- and post-existing differences in microbial composition, functionality, and metabolites that affect stress susceptibility or resilience in each sex.</p><p><strong>Methods: </strong>Male and female Sprague-Dawley rats were randomly assigned to control or SPS groups. Two weeks following SPS, the animals were exposed to a battery of behavioral tests and decapitated a day later. Based on their anxiety index, they were further categorized as SPS-resilient (SPS-R) or SPS-susceptible (SPS-S). On the day of dissection, cecum, and selected brain tissues were isolated. Stool samples were collected before and after SPS, whereas urine samples were taken before and 30 min into the SPS.</p><p><strong>Results: </strong>Before SPS exposure, the sympathoadrenal axis exhibited alterations within male subgroups only. Expression of tight junction protein claudin-5 was lower in brain of SPS-S males, but higher in SPS-R females following SPS. Across the study, alpha diversity remained consistently lower in males compared to females. Beta diversity revealed distinct separations between male and female susceptible groups before SPS, with this separation becoming evident in the resilient groups following SPS. At the genus level, Lactobacillus, Lachnospiraceae_Incertae_Sedis, and Barnesiella exhibited sex-specific alterations, displaying opposing abundances in each sex. Additionally, sex-specific changes were observed in microbial predictive functionality and targeted functional modules both before and after SPS. Alterations in the microbial short-chain fatty acids (SCFAs), were also observed, with major and minor SCFAs being lower in SPS-susceptible males whereas branched-chain SCFAs being higher in SPS-susceptible females.</p><p><strong>Conclusion: </strong>This study highlights distinct pre- and post-trauma differences in microbial composition, functionality, and metabolites, associated with stress resilience in male and female rats. The findings underscore the importance of developing sex-specific therapeutic strategies to effectively address stress-related disorders. Highlights SPS model induces divergent anxiety and social behavioral responses to traumatic stress in both male and female rodents. SPS-resilient females displayed less anxiety-like behavior and initiated more interactions towards a juvenile rat than SPS-resilient males. Sex-specific pre-existing and SPS-induced differen","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-02-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10898170/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139970870","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
Consideration of sex as a biological variable in diabetes research across twenty years 二十年来,在糖尿病研究中将性别视为一个生物变量
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-02-26 DOI: 10.1186/s13293-024-00595-2
Celena M. Cherian, Hayley R. Reeves, Duneesha De Silva, Serena Tsao, Katie E. Marshall, Elizabeth J. Rideout
Sex differences exist in the risk of developing type 1 and type 2 diabetes, and in the risk of developing diabetes-associated complications. Sex differences in glucose homeostasis, islet and β cell biology, and peripheral insulin sensitivity have also been reported. Yet, we lack detailed information on the mechanisms underlying these differences, preventing the development of sex-informed therapeutic strategies for persons living with diabetes. To chart a path toward greater inclusion of biological sex as a variable in diabetes research, we first need a detailed assessment of common practices in the field. We developed a scoring system to evaluate the inclusion of biological sex in manuscripts published in Diabetes, a journal published by the American Diabetes Association. We chose Diabetes as this journal focuses solely on diabetes and diabetes-related research, and includes manuscripts that use both clinical and biomedical approaches. We scored papers published across 3 years within a 20-year period (1999, 2009, 2019), a timeframe that spans the introduction of funding agency and journal policies designed to improve the consideration of biological sex as a variable. Our analysis showed fewer than 15% of papers used sex-based analysis in even one figure across all study years, a trend that was reproduced across journal-defined categories of diabetes research (e.g., islet studies, signal transduction). Single-sex studies accounted for approximately 40% of all manuscripts, of which > 87% used male subjects only. While we observed a modest increase in the overall inclusion of sex as a biological variable during our study period, our data highlight significant opportunities for improvement in diabetes research practices. We also present data supporting a positive role for journal policies in promoting better consideration of biological sex in diabetes research. Our analysis provides significant insight into common practices in diabetes research related to the consideration of biological sex as a variable. Based on our analysis we recommend ways that diabetes researchers can improve inclusion of biological sex as a variable. In the long term, improved practices will reveal sex-specific mechanisms underlying diabetes risk and complications, generating knowledge to enable the development of sex-informed prevention and treatment strategies. Men and women have a different risk of developing type 1 and type 2 diabetes. Men and women also live with different complications of diabetes and show different treatment benefits. One reason for these differences is that biological sex affects diabetes risk, complications, and treatment efficacy. Unfortunately, a lot of diabetes research does not consider whether biological sex might affect the study results. As a result, we do not have enough information to match an individual’s sex with the best diabetes prevention and treatment strategies. We are tackling this problem by learning how diabetes researchers conside
1 型和 2 型糖尿病的发病风险以及糖尿病相关并发症的发病风险存在性别差异。葡萄糖稳态、胰岛和β细胞生物学以及外周胰岛素敏感性方面的性别差异也有报道。然而,我们缺乏有关这些差异的详细机制信息,因此无法为糖尿病患者制定基于性别的治疗策略。要想在糖尿病研究中更多地考虑生理性别这一变量,我们首先需要对该领域的常见做法进行详细评估。我们开发了一套评分系统,用于评估在《糖尿病》(美国糖尿病协会出版的期刊)上发表的稿件中纳入生物性别的情况。我们之所以选择《糖尿病》,是因为该杂志只关注糖尿病和糖尿病相关研究,并收录了采用临床和生物医学方法的稿件。我们对 20 年内(1999 年、2009 年、2019 年)发表的 3 年论文进行了评分,这一时间段跨越了资助机构和期刊政策出台的时间,这些政策旨在改善对生理性别这一变量的考虑。我们的分析表明,在所有研究年份中,只有不到15%的论文在哪怕一个图表中使用了基于性别的分析,这一趋势在期刊定义的糖尿病研究类别(如胰岛研究、信号转导)中也有体现。单一性别研究约占所有稿件的 40%,其中大于 87% 的研究仅使用男性受试者。在我们的研究期间,虽然我们观察到将性别作为生物变量纳入研究的总体情况略有增加,但我们的数据凸显了改进糖尿病研究实践的重要机会。我们还提供数据支持期刊政策在促进糖尿病研究更好地考虑生物性别方面的积极作用。我们的分析提供了有关糖尿病研究中将生理性别作为变量的常见做法的重要见解。根据我们的分析,我们建议糖尿病研究人员如何改进将生理性别作为变量的做法。从长远来看,改进后的研究方法将揭示糖尿病风险和并发症的性别特异性机制,为制定以性别为基础的预防和治疗策略提供知识。男性和女性患 1 型和 2 型糖尿病的风险不同。男女患糖尿病并发症的情况也不同,治疗效果也不同。造成这些差异的原因之一是生理性别会影响糖尿病风险、并发症和治疗效果。遗憾的是,很多糖尿病研究并没有考虑生理性别是否会影响研究结果。因此,我们没有足够的信息来根据个人的性别制定最佳的糖尿病预防和治疗策略。我们正在通过了解糖尿病研究人员如何在研究中考虑生理性别来解决这个问题。我们阅读了 800 多篇糖尿病研究论文并进行了评分,以了解他们在研究中是否考虑了生理性别以及考虑得如何。根据我们的结果,我们推荐了几种简便的方法,让糖尿病研究人员在工作中更好地考虑生理性别。随着越来越多的研究人员考虑生理性别,他们将更多地了解个人性别对糖尿病风险、并发症和治疗效果的影响。这些信息将使整个糖尿病界受益,因为它代表了将个人性别与最佳预防和治疗策略相匹配的第一步。
{"title":"Consideration of sex as a biological variable in diabetes research across twenty years","authors":"Celena M. Cherian, Hayley R. Reeves, Duneesha De Silva, Serena Tsao, Katie E. Marshall, Elizabeth J. Rideout","doi":"10.1186/s13293-024-00595-2","DOIUrl":"https://doi.org/10.1186/s13293-024-00595-2","url":null,"abstract":"Sex differences exist in the risk of developing type 1 and type 2 diabetes, and in the risk of developing diabetes-associated complications. Sex differences in glucose homeostasis, islet and β cell biology, and peripheral insulin sensitivity have also been reported. Yet, we lack detailed information on the mechanisms underlying these differences, preventing the development of sex-informed therapeutic strategies for persons living with diabetes. To chart a path toward greater inclusion of biological sex as a variable in diabetes research, we first need a detailed assessment of common practices in the field. We developed a scoring system to evaluate the inclusion of biological sex in manuscripts published in Diabetes, a journal published by the American Diabetes Association. We chose Diabetes as this journal focuses solely on diabetes and diabetes-related research, and includes manuscripts that use both clinical and biomedical approaches. We scored papers published across 3 years within a 20-year period (1999, 2009, 2019), a timeframe that spans the introduction of funding agency and journal policies designed to improve the consideration of biological sex as a variable. Our analysis showed fewer than 15% of papers used sex-based analysis in even one figure across all study years, a trend that was reproduced across journal-defined categories of diabetes research (e.g., islet studies, signal transduction). Single-sex studies accounted for approximately 40% of all manuscripts, of which > 87% used male subjects only. While we observed a modest increase in the overall inclusion of sex as a biological variable during our study period, our data highlight significant opportunities for improvement in diabetes research practices. We also present data supporting a positive role for journal policies in promoting better consideration of biological sex in diabetes research. Our analysis provides significant insight into common practices in diabetes research related to the consideration of biological sex as a variable. Based on our analysis we recommend ways that diabetes researchers can improve inclusion of biological sex as a variable. In the long term, improved practices will reveal sex-specific mechanisms underlying diabetes risk and complications, generating knowledge to enable the development of sex-informed prevention and treatment strategies. Men and women have a different risk of developing type 1 and type 2 diabetes. Men and women also live with different complications of diabetes and show different treatment benefits. One reason for these differences is that biological sex affects diabetes risk, complications, and treatment efficacy. Unfortunately, a lot of diabetes research does not consider whether biological sex might affect the study results. As a result, we do not have enough information to match an individual’s sex with the best diabetes prevention and treatment strategies. We are tackling this problem by learning how diabetes researchers conside","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-02-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139968621","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
Sex-specific modulation of early life vocalization and cognition by Fmr1 gene dosage in a mouse model of Fragile X Syndrome 在脆性 X 综合征小鼠模型中,Fmr1 基因剂量对生命早期发声和认知的性别特异性调控
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-02-21 DOI: 10.1186/s13293-024-00594-3
Gabriele Giua, Daniela Iezzi, Alba Caceres-Rodriguez, Benjamin Strauss, Pascale Chavis, Olivier J. Manzoni
Pup-dam ultrasonic vocalizations (USVs) are essential to cognitive and socio-emotional development. In autism and Fragile X Syndrome (FXS), disruptions in pup-dam USV communication hint at a possible connection between abnormal early developmental USV communication and the later emergence of communication and social deficits. Here, we gathered USVs from PND 10 FXS pups during a short period of separation from their mothers, encompassing animals of all possible genotypes and both sexes (i.e., Fmr1-/y vs. Fmr1+/y males and Fmr1+/+, +/-, and -/- females). This allowed comparing the influence of sex and gene dosage on pups’ communication capabilities. Leveraging DeepSqueak and analyzing vocal patterns, intricate vocal behaviors such as call structure, duration, frequency modulation, and temporal patterns were examined. Furthermore, homing behavior was assessed as a sensitive indicator of early cognitive development and social discrimination. This behavior relies on the use of olfactory and thermal cues to navigate and search for the maternal or nest odor in the surrounding space. The results show that FMRP-deficient pups of both sexes display an increased inclination to vocalize when separated from their mothers, and this behavior is accompanied by significant sex-specific changes in the main features of their USVs as well as in body weight. Analysis of the vocal repertoire and syntactic usage revealed that Fmr1 gene silencing primarily alters the USVs’ qualitative composition in males. Moreover, sex-specific effects of Fmr1 silencing on locomotor activity and homing behavior were observed. FMRP deficiency in females increased activity, reduced nest-reaching time, and extended nest time. In males, it prolonged nest-reaching time and reduced nest time without affecting locomotion. These findings highlight the interplay between Fmr1 gene dosage and sex in influencing communicative and cognitive skills during infancy. In this study, we investigated ultrasonic vocalizations (USVs) and homing behavior in a mouse model of Fragile X Syndrome (FXS), a leading genetic cause of autism spectrum disorder (ASD) caused by a mutation of the X-chromosome linked Fmr1 gene. Disruptions in pup-dam USV communication and cognitive skills may be linked to the later emergence of communication and social deficits in ASD. USVs were collected from 10-day-old FXS pups of all possible genotypes and both sexes during a short period of separation from their mothers. We utilized DeepSqueak, an advanced deep learning system, to examine vocal patterns and intricate vocal behaviors, including call structure, duration, frequency modulation, and their temporal patterns. Homing, a sensitive indicator of early cognitive development and social discrimination was assessed at P13. The results showed that FXS pups of both sexes displayed an increased inclination to vocalize when separated from their mothers. Examination of the vocal repertoire and its syntactic usage revealed that the silenc
幼犬的超声波发声(USV)对认知和社会情感的发展至关重要。在自闭症和脆性 X 综合征(FXS)中,幼犬与母犬之间的 USV 交流中断暗示着早期发育异常的 USV 交流与后来出现的交流和社交障碍之间可能存在联系。在此,我们收集了 PND 10 FXS 幼崽在与母亲短暂分离期间的 USV,其中包括所有可能的基因型和性别(即 Fmr1-/y 与 Fmr1+/y 雄性和 Fmr1+/+ 、+/- 和 -/- 雌性)。这样就可以比较性别和基因剂量对幼鼠交流能力的影响。通过利用 DeepSqueak 和分析发声模式,研究了复杂的发声行为,如叫声结构、持续时间、频率调制和时间模式。此外,还对归巢行为进行了评估,将其作为早期认知发展和社会辨别能力的敏感指标。这种行为依赖于利用嗅觉和热暗示来导航和寻找周围空间中的母体或巢穴气味。结果表明,FMRP缺失的雌雄幼鼠在与母亲分离时都会表现出更强烈的发声倾向,而且伴随着这种行为,其USV的主要特征和体重也会发生显著的性别差异。对发声曲目和句法用法的分析表明,Fmr1基因沉默主要改变了雄性USV的质量组成。此外,还观察到Fmr1基因沉默对运动活动和归巢行为的性别特异性影响。在雌性中,FMRP的缺失会增加活动量、减少筑巢时间并延长筑巢时间。在雄性中,它延长了筑巢时间并缩短了筑巢时间,但不影响运动。这些发现突显了Fmr1基因剂量和性别在影响婴儿期交流和认知能力方面的相互作用。脆性X综合征(FXS)是自闭症谱系障碍(ASD)的主要遗传病因,由与X染色体相连的Fmr1基因突变引起。幼鼠与母鼠之间的USV交流和认知技能障碍可能与ASD后来出现的交流和社交障碍有关。在与母亲短暂分离期间,我们从 10 天大的 FXS 幼崽(包括所有可能的基因型和男女)身上收集了 USV。我们利用先进的深度学习系统 DeepSqueak 来研究发声模式和复杂的发声行为,包括叫声结构、持续时间、频率调制及其时间模式。归巢是早期认知发展和社会辨别能力的敏感指标,我们在幼鼠13岁时对其进行了评估。结果表明,FXS幼鼠在与母亲分离时,雌雄均表现出更强的发声倾向。对发声曲目及其句法用法的研究表明,Fmr1基因的沉默主要改变了雄性幼鼠超声波交流的质量组成。在USV中观察到的性别特异性变化伴随着体重的改变。在归巢行为方面,FMRP的缺失会导致活动、到达巢穴的时间和筑巢时间出现相反的缺陷,这取决于性别。综上所述,这些发现凸显了Fmr1基因剂量和性别在塑造婴儿期交流和认知能力方面的相互作用。我们研究了脆性X综合征(FXS)小鼠模型的早期沟通和认知能力,脆性X综合征是自闭症谱系障碍的主要遗传原因,由X染色体上的Fmr1基因突变引起。在与母亲短暂分离期间,分别从10天和13天大的FXS幼崽(包括所有可能的基因型和雌雄幼崽)身上收集了超声波发声(USV)和归家行为。雄性和雌性FXS幼鼠在与母亲分离时都表现出更强的发声倾向,同时其USV、归家行为和体重的主要特征也发生了显著的性别特异性改变。Fmr1基因的沉默主要影响雄性动物超声波通讯的质量组成。在归巢行为方面,FMRP基因的缺失会导致活动量、到达巢穴的时间和筑巢时间出现相反的缺陷,这取决于性别。Fmr1基因剂量与性别之间的相互作用影响了婴儿早期的交流和认知。
{"title":"Sex-specific modulation of early life vocalization and cognition by Fmr1 gene dosage in a mouse model of Fragile X Syndrome","authors":"Gabriele Giua, Daniela Iezzi, Alba Caceres-Rodriguez, Benjamin Strauss, Pascale Chavis, Olivier J. Manzoni","doi":"10.1186/s13293-024-00594-3","DOIUrl":"https://doi.org/10.1186/s13293-024-00594-3","url":null,"abstract":"Pup-dam ultrasonic vocalizations (USVs) are essential to cognitive and socio-emotional development. In autism and Fragile X Syndrome (FXS), disruptions in pup-dam USV communication hint at a possible connection between abnormal early developmental USV communication and the later emergence of communication and social deficits. Here, we gathered USVs from PND 10 FXS pups during a short period of separation from their mothers, encompassing animals of all possible genotypes and both sexes (i.e., Fmr1-/y vs. Fmr1+/y males and Fmr1+/+, +/-, and -/- females). This allowed comparing the influence of sex and gene dosage on pups’ communication capabilities. Leveraging DeepSqueak and analyzing vocal patterns, intricate vocal behaviors such as call structure, duration, frequency modulation, and temporal patterns were examined. Furthermore, homing behavior was assessed as a sensitive indicator of early cognitive development and social discrimination. This behavior relies on the use of olfactory and thermal cues to navigate and search for the maternal or nest odor in the surrounding space. The results show that FMRP-deficient pups of both sexes display an increased inclination to vocalize when separated from their mothers, and this behavior is accompanied by significant sex-specific changes in the main features of their USVs as well as in body weight. Analysis of the vocal repertoire and syntactic usage revealed that Fmr1 gene silencing primarily alters the USVs’ qualitative composition in males. Moreover, sex-specific effects of Fmr1 silencing on locomotor activity and homing behavior were observed. FMRP deficiency in females increased activity, reduced nest-reaching time, and extended nest time. In males, it prolonged nest-reaching time and reduced nest time without affecting locomotion. These findings highlight the interplay between Fmr1 gene dosage and sex in influencing communicative and cognitive skills during infancy. In this study, we investigated ultrasonic vocalizations (USVs) and homing behavior in a mouse model of Fragile X Syndrome (FXS), a leading genetic cause of autism spectrum disorder (ASD) caused by a mutation of the X-chromosome linked Fmr1 gene. Disruptions in pup-dam USV communication and cognitive skills may be linked to the later emergence of communication and social deficits in ASD. USVs were collected from 10-day-old FXS pups of all possible genotypes and both sexes during a short period of separation from their mothers. We utilized DeepSqueak, an advanced deep learning system, to examine vocal patterns and intricate vocal behaviors, including call structure, duration, frequency modulation, and their temporal patterns. Homing, a sensitive indicator of early cognitive development and social discrimination was assessed at P13. The results showed that FXS pups of both sexes displayed an increased inclination to vocalize when separated from their mothers. Examination of the vocal repertoire and its syntactic usage revealed that the silenc","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-02-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139923472","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
Different mechanisms underlie compulsive alcohol self-administration in male and female rats 雌雄大鼠强迫性酒精自我摄取的机制不同
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-02-17 DOI: 10.1186/s13293-024-00592-5
Sanne Toivainen, Li Xu, Francesco Gobbo, Andrea Della Valle, Andrea Coppola, Markus Heilig, Esi Domi
Sex is an important factor in the progression and treatment of alcohol addiction, and therapeutic approaches may have to be tailored to potential sex differences. This highlights the importance of understanding sex differences in behaviors that reflect key elements of clinical alcohol addiction, such as continued use despite negative consequences (“compulsive use”). Studies in experimental animals can help provide an understanding of the role sex plays to influence these behaviors. Large populations of genetically heterogeneous male and female Wistar rats were tested in an established model of compulsive alcohol self-administration, operationalized as alcohol responding despite contingent foot shock punishment. We also tested baseline (fixed ratio, unpunished) operant alcohol self-administration, motivation to self-administer alcohol (progressive ratio), and temporal discounting for alcohol reward. In search of predictors of compulsivity, animals were screened for novelty-induced place preference, anxiety-like behavior, pain sensitivity and corticosterone levels. The estrous cycle was monitored throughout the study. Unpunished self-administration of alcohol did not differ between males and females when alcohol intake was corrected for body weight. Overall, females showed higher levels of compulsive responding for alcohol. Compulsive response rates showed bimodal distributions in male but not in female rats when intermediate shock intensities were used (0.2 and 0.25 mA); at higher shock intensities, responding was uniformly suppressed in both males and females. We also found less steep discounting in females when alcohol was devalued by delaying its delivery. Males exhibited a stronger motivation to obtain alcohol under unpunished conditions, while females showed higher corticosterone levels at baseline. Factor analysis showed that an underlying dimension related to stress and pain predicted compulsivity in females, while compulsivity in males was predicted by a reward factor. We did not find differences in alcohol-related behaviors throughout the various stages of the estrous cycle. Our results suggest that mechanisms promoting compulsivity, a key feature of alcohol addiction, likely differ between males and females. This underscores the importance of considering sex as a biological variable in both preclinical and clinical research, and has potential treatment implications in alcohol addiction. Sex plays an important role in the progression and treatment of alcohol addiction. While men show a higher prevalence of alcohol addiction, women are more susceptible to the adverse effects of excessive alcohol consumption. Additionally, women often rely on heavy drinking as a maladaptive coping mechanism to alleviate stress and anxiety, driven by negative affect. On the other hand, men are more likely to report heavy drinking and relapse in response to positive emotions and social influences. These sex-based differences underline the importance of unders
在酒精成瘾的发展和治疗过程中,性别是一个重要因素,治疗方法可能必须针对潜在的性别差异进行调整。这凸显了了解行为性别差异的重要性,这些行为反映了临床酒精成瘾的关键因素,如不顾不良后果继续使用("强迫性使用")。对实验动物的研究有助于了解性别对这些行为的影响。我们对大量基因不同的雌雄 Wistar 大鼠进行了强迫性酒精自我给药模型的测试,该模型被定义为在受到脚震惩罚的情况下仍对酒精做出反应。我们还测试了基线(固定比率、未受惩罚)操作性酒精自我管理、自我管理酒精的动机(渐进比率)以及酒精奖励的时间折扣。为了寻找强迫性的预测因素,对动物进行了新奇诱发的场所偏好、焦虑样行为、疼痛敏感性和皮质酮水平的筛查。在整个研究过程中,对动物的发情周期进行了监测。在对酒精摄入量进行体重校正后,雌雄动物在不受惩罚地自我饮酒方面没有差异。总体而言,雌性对酒精的强迫反应水平更高。在使用中间电击强度(0.2 和 0.25 mA)时,雄性大鼠的强迫反应率呈双峰分布,而雌性大鼠则没有;在使用较高电击强度时,雄性和雌性大鼠的反应均受到抑制。我们还发现,当酒精通过延迟交付而贬值时,雌性大鼠的陡峭折扣较少。在不受惩罚的条件下,男性表现出更强的获取酒精的动机,而女性则表现出更高的皮质酮基线水平。因子分析显示,与压力和痛苦相关的潜在维度预测了女性的强迫性,而男性的强迫性则是由奖励因子预测的。在发情周期的不同阶段,我们没有发现酒精相关行为的差异。我们的研究结果表明,酒精成瘾的一个关键特征--强迫性的促进机制可能在男性和女性之间存在差异。这强调了在临床前和临床研究中将性别作为生物变量的重要性,并对酒精成瘾的治疗具有潜在的影响。性别在酒精成瘾的发展和治疗中起着重要作用。男性酒精成瘾的发病率更高,而女性则更容易受到过量饮酒的不良影响。此外,在负面情绪的驱使下,女性往往依赖大量饮酒作为一种不适应的应对机制,以缓解压力和焦虑。另一方面,男性则更有可能在积极情绪和社会影响的作用下大量饮酒并复发。这些性别差异凸显了了解雌雄两性对酒精成瘾的易感性和治疗方法有何不同的重要性。我们利用基因异质性大鼠来探索导致强迫性的行为特征,强迫性是酒精成瘾的一个关键临床特征。我们发现,雄性自我饮酒的动机更高,而雌性则表现出更高的强迫性自我饮酒。在男性中,自我饮酒的动机与强迫性有显著的相关性,而在女性中,基础皮质酮水平较高可预测强迫性。这些发现揭示了性别特异性因素在强迫性酒精自我管理中的重要性,对预防和治疗酒精成瘾具有潜在的意义。
{"title":"Different mechanisms underlie compulsive alcohol self-administration in male and female rats","authors":"Sanne Toivainen, Li Xu, Francesco Gobbo, Andrea Della Valle, Andrea Coppola, Markus Heilig, Esi Domi","doi":"10.1186/s13293-024-00592-5","DOIUrl":"https://doi.org/10.1186/s13293-024-00592-5","url":null,"abstract":"Sex is an important factor in the progression and treatment of alcohol addiction, and therapeutic approaches may have to be tailored to potential sex differences. This highlights the importance of understanding sex differences in behaviors that reflect key elements of clinical alcohol addiction, such as continued use despite negative consequences (“compulsive use”). Studies in experimental animals can help provide an understanding of the role sex plays to influence these behaviors. Large populations of genetically heterogeneous male and female Wistar rats were tested in an established model of compulsive alcohol self-administration, operationalized as alcohol responding despite contingent foot shock punishment. We also tested baseline (fixed ratio, unpunished) operant alcohol self-administration, motivation to self-administer alcohol (progressive ratio), and temporal discounting for alcohol reward. In search of predictors of compulsivity, animals were screened for novelty-induced place preference, anxiety-like behavior, pain sensitivity and corticosterone levels. The estrous cycle was monitored throughout the study. Unpunished self-administration of alcohol did not differ between males and females when alcohol intake was corrected for body weight. Overall, females showed higher levels of compulsive responding for alcohol. Compulsive response rates showed bimodal distributions in male but not in female rats when intermediate shock intensities were used (0.2 and 0.25 mA); at higher shock intensities, responding was uniformly suppressed in both males and females. We also found less steep discounting in females when alcohol was devalued by delaying its delivery. Males exhibited a stronger motivation to obtain alcohol under unpunished conditions, while females showed higher corticosterone levels at baseline. Factor analysis showed that an underlying dimension related to stress and pain predicted compulsivity in females, while compulsivity in males was predicted by a reward factor. We did not find differences in alcohol-related behaviors throughout the various stages of the estrous cycle. Our results suggest that mechanisms promoting compulsivity, a key feature of alcohol addiction, likely differ between males and females. This underscores the importance of considering sex as a biological variable in both preclinical and clinical research, and has potential treatment implications in alcohol addiction. Sex plays an important role in the progression and treatment of alcohol addiction. While men show a higher prevalence of alcohol addiction, women are more susceptible to the adverse effects of excessive alcohol consumption. Additionally, women often rely on heavy drinking as a maladaptive coping mechanism to alleviate stress and anxiety, driven by negative affect. On the other hand, men are more likely to report heavy drinking and relapse in response to positive emotions and social influences. These sex-based differences underline the importance of unders","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139751655","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
Nrf2 activation rescues stress-induced depression-like behaviour and inflammatory responses in male but not female rats. 激活 Nrf2 可挽救雄性大鼠(而非雌性大鼠)由压力诱发的抑郁样行为和炎症反应。
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-02-13 DOI: 10.1186/s13293-024-00589-0
Ryan T McCallum, Rachel-Karson Thériault, Joshua D Manduca, Isaac S B Russell, Angel M Culmer, Janan Shoja Doost, Tami A Martino, Melissa L Perreault

Background: Major depressive disorder (MDD) is a recurring affective disorder that is two times more prevalent in females than males. Evidence supports immune system dysfunction as a major contributing factor to MDD, notably in a sexually dimorphic manner. Nuclear factor erythroid 2-related factor 2 (Nrf2), a regulator of antioxidant signalling during inflammation, is dysregulated in many chronic inflammatory disorders; however, its role in depression and the associated sex differences have yet to be explored. Here, we investigated the sex-specific antidepressant and immunomodulatory effects of the potent Nrf2 activator dimethyl fumarate (DMF), as well as the associated gene expression profiles.

Methods: Male and female rats were treated with vehicle or DMF (25 mg/kg) whilst subjected to 8 weeks of chronic unpredictable stress. The effect of DMF treatment on stress-induced depression- and anxiety-like behaviours, as well as deficits in recognition and spatial learning and memory were then assessed. Sex differences in hippocampal (HIP) microglial activation and gene expression response were also evaluated.

Results: DMF treatment during stress exposure had antidepressant effects in male but not female rats, with no anxiolytic effects in either sex. Recognition learning and memory and spatial learning and memory were impaired in chronically stressed males and females, respectively, and DMF treatment rescued these deficits. DMF treatment also prevented stress-induced HIP microglial activation in males. Conversely, females displayed no HIP microglial activation associated with stress exposure. Last, chronic stress elicited sex-specific alterations in HIP gene expression, many of which were normalized in animals treated with DMF. Of note, most of the differentially expressed genes in males normalized by DMF were related to antioxidant, inflammatory or immune responses.

Conclusions: Collectively, these findings support a greater role of immune processes in males than females in a rodent model of depression. This suggests that pharmacotherapies that target Nrf2 have the potential to be an effective sex-specific treatment for depression.

背景:重度抑郁障碍(MDD)是一种反复发作的情感障碍,女性发病率是男性的两倍。有证据表明,免疫系统功能障碍是导致重度抑郁症的一个主要因素,尤其是以性别二态的方式存在。核因子红细胞2相关因子2(Nrf2)是炎症过程中抗氧化信号的调节因子,在许多慢性炎症性疾病中都会出现失调;然而,它在抑郁症中的作用以及相关的性别差异还有待探索。在这里,我们研究了强效Nrf2激活剂富马酸二甲酯(DMF)的性别特异性抗抑郁和免疫调节作用,以及相关的基因表达谱:雄性和雌性大鼠在接受为期8周的慢性不可预测应激的同时,还接受了药物或DMF(25 mg/kg)的治疗。然后评估DMF处理对应激诱发的抑郁和焦虑样行为以及识别和空间学习记忆缺陷的影响。此外,还评估了海马(HIP)小胶质细胞活化和基因表达反应的性别差异:结果:应激暴露期间的 DMF 处理对雄性大鼠有抗抑郁作用,但对雌性大鼠没有抗焦虑作用。长期应激的雄性大鼠和雌性大鼠的识别学习和记忆以及空间学习和记忆分别受到损害,而DMF治疗可缓解这些缺陷。DMF还能防止应激诱导的雄性HIP小胶质细胞活化。相反,女性的 HIP 小胶质细胞活化与压力暴露无关。最后,慢性应激引起了 HIP 基因表达的性别特异性改变,其中许多改变在接受 DMF 治疗的动物体内恢复了正常。值得注意的是,DMF 使雄性动物中的大部分差异表达基因恢复正常,这些基因与抗氧化、炎症或免疫反应有关:总之,这些研究结果表明,在抑郁症的啮齿类动物模型中,雄性动物的免疫过程比雌性动物发挥着更大的作用。这表明以 Nrf2 为靶点的药物疗法有可能成为治疗抑郁症的一种有效的性别特异性疗法。
{"title":"Nrf2 activation rescues stress-induced depression-like behaviour and inflammatory responses in male but not female rats.","authors":"Ryan T McCallum, Rachel-Karson Thériault, Joshua D Manduca, Isaac S B Russell, Angel M Culmer, Janan Shoja Doost, Tami A Martino, Melissa L Perreault","doi":"10.1186/s13293-024-00589-0","DOIUrl":"10.1186/s13293-024-00589-0","url":null,"abstract":"<p><strong>Background: </strong>Major depressive disorder (MDD) is a recurring affective disorder that is two times more prevalent in females than males. Evidence supports immune system dysfunction as a major contributing factor to MDD, notably in a sexually dimorphic manner. Nuclear factor erythroid 2-related factor 2 (Nrf2), a regulator of antioxidant signalling during inflammation, is dysregulated in many chronic inflammatory disorders; however, its role in depression and the associated sex differences have yet to be explored. Here, we investigated the sex-specific antidepressant and immunomodulatory effects of the potent Nrf2 activator dimethyl fumarate (DMF), as well as the associated gene expression profiles.</p><p><strong>Methods: </strong>Male and female rats were treated with vehicle or DMF (25 mg/kg) whilst subjected to 8 weeks of chronic unpredictable stress. The effect of DMF treatment on stress-induced depression- and anxiety-like behaviours, as well as deficits in recognition and spatial learning and memory were then assessed. Sex differences in hippocampal (HIP) microglial activation and gene expression response were also evaluated.</p><p><strong>Results: </strong>DMF treatment during stress exposure had antidepressant effects in male but not female rats, with no anxiolytic effects in either sex. Recognition learning and memory and spatial learning and memory were impaired in chronically stressed males and females, respectively, and DMF treatment rescued these deficits. DMF treatment also prevented stress-induced HIP microglial activation in males. Conversely, females displayed no HIP microglial activation associated with stress exposure. Last, chronic stress elicited sex-specific alterations in HIP gene expression, many of which were normalized in animals treated with DMF. Of note, most of the differentially expressed genes in males normalized by DMF were related to antioxidant, inflammatory or immune responses.</p><p><strong>Conclusions: </strong>Collectively, these findings support a greater role of immune processes in males than females in a rodent model of depression. This suggests that pharmacotherapies that target Nrf2 have the potential to be an effective sex-specific treatment for depression.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10863247/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139728882","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 effects of individual anxiety state on regional responses to negative emotional scenes. 个体焦虑状态对负面情绪场景区域反应的影响存在性别差异。
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-02-13 DOI: 10.1186/s13293-024-00591-6
Shefali Chaudhary, Hak Kei Wong, Yu Chen, Sheng Zhang, Chiang-Shan R Li

Background: Men and women are known to show differences in the incidence and clinical manifestations of mood and anxiety disorders. Many imaging studies have investigated the neural correlates of sex differences in emotion processing. However, it remains unclear how anxiety might impact emotion processing differently in men and women.

Method: We recruited 119 healthy adults and assessed their levels of anxiety using State-Trait Anxiety Inventory (STAI) State score. With functional magnetic resonance imaging (fMRI), we examined regional responses to negative vs. neutral (Neg-Neu) picture matching in the Hariri task. Behavioral data were analyzed using regression and repeated-measures analysis of covariance with age as a covariate, and fMRI data were analyzed using a full-factorial model with sex as a factor and age as a covariate.

Results: Men and women did not differ in STAI score, or accuracy rate or reaction time (RT) (Neg-Neu). However, STAI scores correlated positively with RT (Neg-Neu) in women but not in men. Additionally, in women, STAI score correlated positively with lingual gyrus (LG) and negatively with medial prefrontal cortex (mPFC) and superior frontal gyrus (SFG) activity during Neg vs. Neu trials. The parameter estimates (βs) of mPFC also correlated with RT (Neg-Neu) in women but not in men. Generalized psychophysiological interaction (gPPI) analysis in women revealed mPFC connectivity with the right inferior frontal gyrus, right SFG, and left parahippocampal gyrus during Neg vs. Neu trials in positive correlation with both STAI score and RT (Neg-Neu). In a mediation analysis, mPFC gPPI but not mPFC activity fully mediated the association between STAI scores and RT (Neg-Neu).

Conclusion: With anxiety affecting the behavioral and neural responses to negative emotions in women but not in men and considering the known roles of the mPFC in emotion regulation, we discussed heightened sensitivity and regulatory demands during negative emotion processing as neurobehavioral markers of anxiety in women.

背景:众所周知,男性和女性在情绪和焦虑症的发病率和临床表现方面存在差异。许多影像学研究都对情绪处理中的性别差异的神经相关性进行了调查。然而,目前仍不清楚焦虑会如何对男性和女性的情绪处理产生不同的影响:方法:我们招募了 119 名健康成年人,并使用状态-特质焦虑量表(STAI)状态评分评估了他们的焦虑水平。通过功能磁共振成像(fMRI),我们研究了哈里里任务中负性与中性(Neg-Neu)图片匹配的区域反应。行为数据采用回归和重复测量协方差分析法进行分析,年龄为协方差;fMRI 数据采用全因子模型进行分析,性别为因子,年龄为协方差:男性和女性在STAI得分、准确率或反应时间(RT)(Neg-Neu)方面没有差异。但是,女性的 STAI 分数与 RT(Neg-Neu)呈正相关,而男性则不然。此外,在Neg与Neu试验中,女性的STAI得分与舌回(LG)呈正相关,而与内侧前额叶皮层(mPFC)和额上回(SFG)的活动呈负相关。在女性中,mPFC 的参数估计(βs)也与 RT(Neg-Neu)相关,但在男性中则不相关。对女性进行的广义心理生理学交互作用(gPPI)分析显示,在 "否定 "与 "新 "的试验中,mPFC与右侧额叶下回、右侧SFG和左侧海马旁回的连接与STAI得分和RT("否定 "与 "新")呈正相关。在中介分析中,mPFC gPPI而非mPFC活动完全中介了STAI得分和RT(Neg-Neu)之间的关联:结论:焦虑会影响女性而非男性对负面情绪的行为和神经反应,考虑到 mPFC 在情绪调节中的已知作用,我们讨论了女性在负面情绪处理过程中作为焦虑神经行为标记的高度敏感性和调节需求。
{"title":"Sex differences in the effects of individual anxiety state on regional responses to negative emotional scenes.","authors":"Shefali Chaudhary, Hak Kei Wong, Yu Chen, Sheng Zhang, Chiang-Shan R Li","doi":"10.1186/s13293-024-00591-6","DOIUrl":"10.1186/s13293-024-00591-6","url":null,"abstract":"<p><strong>Background: </strong>Men and women are known to show differences in the incidence and clinical manifestations of mood and anxiety disorders. Many imaging studies have investigated the neural correlates of sex differences in emotion processing. However, it remains unclear how anxiety might impact emotion processing differently in men and women.</p><p><strong>Method: </strong>We recruited 119 healthy adults and assessed their levels of anxiety using State-Trait Anxiety Inventory (STAI) State score. With functional magnetic resonance imaging (fMRI), we examined regional responses to negative vs. neutral (Neg-Neu) picture matching in the Hariri task. Behavioral data were analyzed using regression and repeated-measures analysis of covariance with age as a covariate, and fMRI data were analyzed using a full-factorial model with sex as a factor and age as a covariate.</p><p><strong>Results: </strong>Men and women did not differ in STAI score, or accuracy rate or reaction time (RT) (Neg-Neu). However, STAI scores correlated positively with RT (Neg-Neu) in women but not in men. Additionally, in women, STAI score correlated positively with lingual gyrus (LG) and negatively with medial prefrontal cortex (mPFC) and superior frontal gyrus (SFG) activity during Neg vs. Neu trials. The parameter estimates (βs) of mPFC also correlated with RT (Neg-Neu) in women but not in men. Generalized psychophysiological interaction (gPPI) analysis in women revealed mPFC connectivity with the right inferior frontal gyrus, right SFG, and left parahippocampal gyrus during Neg vs. Neu trials in positive correlation with both STAI score and RT (Neg-Neu). In a mediation analysis, mPFC gPPI but not mPFC activity fully mediated the association between STAI scores and RT (Neg-Neu).</p><p><strong>Conclusion: </strong>With anxiety affecting the behavioral and neural responses to negative emotions in women but not in men and considering the known roles of the mPFC in emotion regulation, we discussed heightened sensitivity and regulatory demands during negative emotion processing as neurobehavioral markers of anxiety in women.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10863151/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139728883","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
Remembering the null hypothesis when searching for brain sex differences. 寻找大脑性别差异时记住无效假设
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-02-09 DOI: 10.1186/s13293-024-00585-4
Lise Eliot

Human brain sex differences have fascinated scholars for centuries and become a key focus of neuroscientists since the dawn of MRI. We recently published a major review in Neuroscience and Biobehavioral Reviews showing that most male-female brain differences in humans are small and few have been reliably replicated. Although widely cited, this work was the target of a critical Commentary by DeCasien et al. (Biol Sex Differ 13:43, 2022). In this response, I update our findings and confirm the small effect sizes and pronounced scatter across recent large neuroimaging studies of human sex/gender difference. Based on the sum of data, neuroscientists would be well-advised to take the null hypothesis seriously: that men and women's brains are fundamentally similar, or "monomorphic". This perspective has important implications for how we study the genesis of behavioral and neuropsychiatric gender disparities.

几个世纪以来,人类大脑的性别差异一直令学者们着迷,自核磁共振成像问世以来,它已成为神经科学家关注的焦点。最近,我们在《神经科学与生物行为评论》(Neuroscience and Biobehavioral Reviews)上发表了一篇重要的评论文章,指出人类大脑中的大多数男女差异都很小,而且很少被可靠地复制。尽管这项研究成果被广泛引用,但它还是成为了 DeCasien 等人的批评性评论(Biol Sex Differ 13:43, 2022)的目标。在这篇回应中,我将更新我们的研究结果,并证实最近关于人类性别差异的大型神经影像学研究的效应量较小且明显分散。根据这些数据,神经科学家最好认真对待零假设:即男女大脑从根本上是相似的,或者说是 "单形的"。这一观点对于我们如何研究行为和神经精神性别差异的起源具有重要意义。
{"title":"Remembering the null hypothesis when searching for brain sex differences.","authors":"Lise Eliot","doi":"10.1186/s13293-024-00585-4","DOIUrl":"10.1186/s13293-024-00585-4","url":null,"abstract":"<p><p>Human brain sex differences have fascinated scholars for centuries and become a key focus of neuroscientists since the dawn of MRI. We recently published a major review in Neuroscience and Biobehavioral Reviews showing that most male-female brain differences in humans are small and few have been reliably replicated. Although widely cited, this work was the target of a critical Commentary by DeCasien et al. (Biol Sex Differ 13:43, 2022). In this response, I update our findings and confirm the small effect sizes and pronounced scatter across recent large neuroimaging studies of human sex/gender difference. Based on the sum of data, neuroscientists would be well-advised to take the null hypothesis seriously: that men and women's brains are fundamentally similar, or \"monomorphic\". This perspective has important implications for how we study the genesis of behavioral and neuropsychiatric gender disparities.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-02-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10854110/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139711293","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 role of microRNAs in understanding sex-based differences in Alzheimer's disease. 微RNA在理解阿尔茨海默病性别差异中的作用。
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-01-31 DOI: 10.1186/s13293-024-00588-1
Jaime Llera-Oyola, Héctor Carceller, Zoraida Andreu, Marta R Hidalgo, Irene Soler-Sáez, Fernando Gordillo, Borja Gómez-Cabañes, Beatriz Roson, Maria de la Iglesia-Vayá, Roberta Mancuso, Franca R Guerini, Akiko Mizokami, Francisco García-García

Background: The incidence of Alzheimer's disease (AD)-the most frequent cause of dementia-is expected to increase as life expectancies rise across the globe. While sex-based differences in AD have previously been described, there remain uncertainties regarding any association between sex and disease-associated molecular mechanisms. Studying sex-specific expression profiles of regulatory factors such as microRNAs (miRNAs) could contribute to more accurate disease diagnosis and treatment.

Methods: A systematic review identified six studies of microRNA expression in AD patients that incorporated information regarding the biological sex of samples in the Gene Expression Omnibus repository. A differential microRNA expression analysis was performed, considering disease status and patient sex. Subsequently, results were integrated within a meta-analysis methodology, with a functional enrichment of meta-analysis results establishing an association between altered miRNA expression and relevant Gene Ontology terms.

Results: Meta-analyses of miRNA expression profiles in blood samples revealed the alteration of sixteen miRNAs in female and 22 miRNAs in male AD patients. We discovered nine miRNAs commonly overexpressed in both sexes, suggesting a shared miRNA dysregulation profile. Functional enrichment results based on miRNA profiles revealed sex-based differences in biological processes; most affected processes related to ubiquitination, regulation of different kinase activities, and apoptotic processes in males, but RNA splicing and translation in females. Meta-analyses of miRNA expression profiles in brain samples revealed the alteration of six miRNAs in female and four miRNAs in male AD patients. We observed a single underexpressed miRNA in female and male AD patients (hsa-miR-767-5p); however, the functional enrichment analysis for brain samples did not reveal any specifically affected biological process.

Conclusions: Sex-specific meta-analyses supported the detection of differentially expressed miRNAs in female and male AD patients, highlighting the relevance of sex-based information in biomedical data. Further studies on miRNA regulation in AD patients should meet the criteria for comparability and standardization of information.

背景:阿尔茨海默病(AD)是导致痴呆症的最常见原因,随着全球人口寿命的延长,预计其发病率还会上升。虽然以前曾描述过阿尔茨海默病的性别差异,但性别与疾病相关分子机制之间的关系仍不确定。研究微小RNA(miRNA)等调控因子的性别特异性表达谱有助于更准确地诊断和治疗疾病:方法:通过系统性回顾,在基因表达总库(Gene Expression Omnibus)中找到了六项关于AD患者microRNA表达的研究,这些研究都包含了样本的生物学性别信息。在考虑疾病状态和患者性别的基础上,进行了微RNA表达差异分析。随后,在荟萃分析方法中整合了分析结果,并对荟萃分析结果进行了功能富集,确定了miRNA表达改变与相关基因本体术语之间的关联:血液样本中 miRNA 表达谱的元分析显示,女性 AD 患者和男性 AD 患者分别有 16 个和 22 个 miRNA 发生变化。我们发现有九种miRNA在男女患者中普遍表达过高,这表明男女患者存在共同的miRNA失调特征。基于miRNA图谱的功能富集结果显示了生物过程中的性别差异;男性受影响最大的过程与泛素化、不同激酶活性的调节和细胞凋亡过程有关,而女性则与RNA剪接和翻译有关。对大脑样本中 miRNA 表达谱的元分析表明,女性和男性 AD 患者分别有六种和四种 miRNA 发生了改变。我们在女性和男性AD患者中观察到了一种表达不足的miRNA(hsa-miR-767-5p);然而,脑样本的功能富集分析并未发现任何具体受影响的生物过程:结论:性别特异性荟萃分析支持在女性和男性AD患者中检测到不同表达的miRNA,突出了生物医学数据中基于性别的信息的相关性。有关AD患者miRNA调控的进一步研究应符合信息的可比性和标准化标准。
{"title":"The role of microRNAs in understanding sex-based differences in Alzheimer's disease.","authors":"Jaime Llera-Oyola, Héctor Carceller, Zoraida Andreu, Marta R Hidalgo, Irene Soler-Sáez, Fernando Gordillo, Borja Gómez-Cabañes, Beatriz Roson, Maria de la Iglesia-Vayá, Roberta Mancuso, Franca R Guerini, Akiko Mizokami, Francisco García-García","doi":"10.1186/s13293-024-00588-1","DOIUrl":"10.1186/s13293-024-00588-1","url":null,"abstract":"<p><strong>Background: </strong>The incidence of Alzheimer's disease (AD)-the most frequent cause of dementia-is expected to increase as life expectancies rise across the globe. While sex-based differences in AD have previously been described, there remain uncertainties regarding any association between sex and disease-associated molecular mechanisms. Studying sex-specific expression profiles of regulatory factors such as microRNAs (miRNAs) could contribute to more accurate disease diagnosis and treatment.</p><p><strong>Methods: </strong>A systematic review identified six studies of microRNA expression in AD patients that incorporated information regarding the biological sex of samples in the Gene Expression Omnibus repository. A differential microRNA expression analysis was performed, considering disease status and patient sex. Subsequently, results were integrated within a meta-analysis methodology, with a functional enrichment of meta-analysis results establishing an association between altered miRNA expression and relevant Gene Ontology terms.</p><p><strong>Results: </strong>Meta-analyses of miRNA expression profiles in blood samples revealed the alteration of sixteen miRNAs in female and 22 miRNAs in male AD patients. We discovered nine miRNAs commonly overexpressed in both sexes, suggesting a shared miRNA dysregulation profile. Functional enrichment results based on miRNA profiles revealed sex-based differences in biological processes; most affected processes related to ubiquitination, regulation of different kinase activities, and apoptotic processes in males, but RNA splicing and translation in females. Meta-analyses of miRNA expression profiles in brain samples revealed the alteration of six miRNAs in female and four miRNAs in male AD patients. We observed a single underexpressed miRNA in female and male AD patients (hsa-miR-767-5p); however, the functional enrichment analysis for brain samples did not reveal any specifically affected biological process.</p><p><strong>Conclusions: </strong>Sex-specific meta-analyses supported the detection of differentially expressed miRNAs in female and male AD patients, highlighting the relevance of sex-based information in biomedical data. Further studies on miRNA regulation in AD patients should meet the criteria for comparability and standardization of information.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10832236/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139650236","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 effects of repeated ketamine infusions on bone markers in patients with unipolar and bipolar depression. 重复注射氯胺酮对单相抑郁症和双相抑郁症患者骨标记物影响的性别差异。
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-01-29 DOI: 10.1186/s13293-024-00587-2
Xiaofeng Lan, Haiyan Liu, Chengyu Wang, Weicheng Li, Fan Zhang, Zhibo Hu, Xiaoyu Chen, Zerui You, Yuping Ning, Yanling Zhou

Background: Patients with depression, especially women, are associated with low bone mineral density (BMD). Traditional antidepressants are associated with negative effects on BMD. Few studies have examined the effect of ketamine on BMD, and it remains unclear whether there are sex differences in the effects of ketamine on BMD in patients with depression.

Methods: A total of 102 patients with unipolar and bipolar depression were administered six infusions of intravenous ketamine over a 12-day period. Plasma levels of eight bone markers were examined at baseline, 24 h after the sixth infusion and again 2 weeks (Days 13 and 26).

Results: Linear mixed models showed all bone markers had significant time main effect (all p < 0.05). Compared with baseline, the whole sample showed increased levels of leptin and osteoprotegerin at Days 13 and 26, as well as Dickkopf-related protein 1 at Day 13, and decreased levels of osteocalcin, sclerostin, osteopontin, parathyroid hormone and fibroblast growth factor 23 at Days 13 and 26 (all p < 0.05). Females had a higher level of leptin at Days 13 and 26, and lower levels of osteocalcin and sclerostin at Day 13 than males (all p < 0.05). Increases of leptin were associated with depressive symptom improvements at Day 13 and Day 26 in females (both p < 0.05). In males, higher baseline osteocalcin levels were associated with greater depressive symptom improvement at Day 26 (β = 0.414, p = 0.009).

Conclusions: Our results suggest that repeated ketamine infusions may be associated with modulation of bone markers in patients with depression and present sex differences. Baseline osteocalcin level may be served as a predictor for the antidepressant effects of ketamine in males. Trial registration Data were derived from an open label clinical trial, which was registered at Chinese Clinical Trial Registry (ChiCTR-OOC-17012239). Registered 26 May 2017. http://www.chictr.org.cn.

背景:抑郁症患者(尤其是女性)的骨质密度(BMD)较低。传统的抗抑郁药对骨密度有负面影响。目前还不清楚氯胺酮对抑郁症患者骨密度的影响是否存在性别差异:共有 102 名单相和双相抑郁症患者在 12 天内接受了六次氯胺酮静脉注射。在基线、第六次输液后 24 小时以及两周后(第 13 天和第 26 天)对八种骨标志物的血浆水平进行了检测:线性混合模型显示,所有骨标记物都有显著的时间主效应(均为 p我们的研究结果表明,重复输注氯胺酮可能与抑郁症患者骨标记物的调节有关,并存在性别差异。基线骨钙素水平可作为氯胺酮对男性抗抑郁作用的预测因子。试验注册 数据来自一项开放标签临床试验,该试验已在中国临床试验注册中心注册(ChiCTR-OOC-17012239)。注册时间为2017年5月26日。http://www.chictr.org.cn。
{"title":"Sex differences in the effects of repeated ketamine infusions on bone markers in patients with unipolar and bipolar depression.","authors":"Xiaofeng Lan, Haiyan Liu, Chengyu Wang, Weicheng Li, Fan Zhang, Zhibo Hu, Xiaoyu Chen, Zerui You, Yuping Ning, Yanling Zhou","doi":"10.1186/s13293-024-00587-2","DOIUrl":"10.1186/s13293-024-00587-2","url":null,"abstract":"<p><strong>Background: </strong>Patients with depression, especially women, are associated with low bone mineral density (BMD). Traditional antidepressants are associated with negative effects on BMD. Few studies have examined the effect of ketamine on BMD, and it remains unclear whether there are sex differences in the effects of ketamine on BMD in patients with depression.</p><p><strong>Methods: </strong>A total of 102 patients with unipolar and bipolar depression were administered six infusions of intravenous ketamine over a 12-day period. Plasma levels of eight bone markers were examined at baseline, 24 h after the sixth infusion and again 2 weeks (Days 13 and 26).</p><p><strong>Results: </strong>Linear mixed models showed all bone markers had significant time main effect (all p < 0.05). Compared with baseline, the whole sample showed increased levels of leptin and osteoprotegerin at Days 13 and 26, as well as Dickkopf-related protein 1 at Day 13, and decreased levels of osteocalcin, sclerostin, osteopontin, parathyroid hormone and fibroblast growth factor 23 at Days 13 and 26 (all p < 0.05). Females had a higher level of leptin at Days 13 and 26, and lower levels of osteocalcin and sclerostin at Day 13 than males (all p < 0.05). Increases of leptin were associated with depressive symptom improvements at Day 13 and Day 26 in females (both p < 0.05). In males, higher baseline osteocalcin levels were associated with greater depressive symptom improvement at Day 26 (β = 0.414, p = 0.009).</p><p><strong>Conclusions: </strong>Our results suggest that repeated ketamine infusions may be associated with modulation of bone markers in patients with depression and present sex differences. Baseline osteocalcin level may be served as a predictor for the antidepressant effects of ketamine in males. Trial registration Data were derived from an open label clinical trial, which was registered at Chinese Clinical Trial Registry (ChiCTR-OOC-17012239). Registered 26 May 2017. http://www.chictr.org.cn.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10826032/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139575224","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