首页 > 最新文献

Biology of Sex Differences最新文献

英文 中文
Sexually dimorphic effects of prenatal alcohol exposure on the murine skeleton. 产前酒精暴露对小鼠骨骼的性别双态影响
IF 4.9 2区 医学 Q1 Social Sciences Pub Date : 2024-06-18 DOI: 10.1186/s13293-024-00626-y
Lucie E Bourne, Soher N Jayash, Lysanne V Michels, Mark Hopkinson, Fergus M Guppy, Claire E Clarkin, Paul Gard, Nigel Brissett, Katherine A Staines

Background: Prenatal alcohol exposure (PAE) can result in lifelong disabilities known as foetal alcohol spectrum disorder (FASD) and is associated with childhood growth deficiencies and increased bone fracture risk. However, the effects of PAE on the adult skeleton remain unclear and any potential sexual dimorphism is undetermined. Therefore, we utilised a murine model to examine sex differences with PAE on in vitro bone formation, and in the juvenile and adult skeleton.

Methods: Pregnant C57BL/6J female mice received 5% ethanol in their drinking water during gestation. Primary calvarial osteoblasts were isolated from neonatal offspring and mineralised bone nodule formation and gene expression assessed. Skeletal phenotyping of 4- and 12-week-old male and female offspring was conducted by micro-computed tomography (µCT), 3-point bending, growth plate analyses, and histology.

Results: Osteoblasts from male and female PAE mice displayed reduced bone formation, compared to control (≤ 30%). Vegfa, Vegfb, Bmp6, Tgfbr1, Flt1 and Ahsg were downregulated in PAE male osteoblasts only, whilst Ahsg was upregulated in PAE females. In 12-week-old mice, µCT analysis revealed a sex and exposure interaction across several trabecular bone parameters. PAE was detrimental to the trabecular compartment in male mice compared to control, yet PAE females were unaffected. Both male and female mice had significant reductions in cortical parameters with PAE. Whilst male mice were negatively affected along the tibial length, females were only distally affected. Posterior cortical porosity was increased in PAE females only. Mechanical testing revealed PAE males had significantly reduced bone stiffness compared to controls; maximum load and yield were reduced in both sexes. PAE had no effect on total body weight or tibial bone length in either sex. However, total growth plate width in male PAE mice compared to control was reduced, whilst female PAE mice were unaffected. 4-week-old mice did not display the altered skeletal phenotype with PAE observed in 12-week-old animals.

Conclusions: Evidence herein suggests, for the first time, that PAE exerts divergent sex effects on the skeleton, possibly influenced by underlying sex-specific transcriptional mechanisms of osteoblasts. Establishing these sex differences will support future policies and clinical management of FASD.

背景:产前酒精暴露(PAE)可导致终身残疾,即胎儿酒精谱系障碍(FASD),并与儿童生长缺陷和骨折风险增加有关。然而,PAE 对成人骨骼的影响仍不清楚,任何潜在的性别二形性也未确定。因此,我们利用小鼠模型来研究 PAE 对体外骨形成以及幼年和成年骨骼的性别差异:方法:妊娠期的 C57BL/6J 雌性小鼠在饮用水中加入 5% 的乙醇。方法:妊娠期的 C57BL/6J 雌性小鼠在饮用水中加入 5% 的乙醇,从新生仔鼠体内分离出原始犊骨成骨细胞,并对矿化骨结节的形成和基因表达进行评估。通过微型计算机断层扫描(μCT)、三点弯曲、生长板分析和组织学,对 4 周大和 12 周大的雄性和雌性后代进行骨骼表型分析:结果:与对照组相比,雄性和雌性 PAE 小鼠的成骨细胞显示骨形成减少(≤ 30%)。仅 PAE 雄性成骨细胞中 Vegfa、Vegfb、Bmp6、Tgfbr1、Flt1 和 Ahsg 下调,而 PAE 雌性成骨细胞中 Ahsg 上调。在 12 周大的小鼠中,µCT 分析表明性别与暴露在多个骨小梁参数上存在交互作用。与对照组相比,PAE 对雄性小鼠的骨小梁有害,而 PAE 雌性小鼠则不受影响。在 PAE 的作用下,雄性和雌性小鼠的皮质参数都显著降低。雄性小鼠的胫骨长度受到负面影响,而雌性小鼠仅在远端受到影响。只有 PAE 雌鼠后部皮质孔隙率增加。机械测试表明,与对照组相比,PAE 雄性小鼠的骨刚度明显降低;雌雄小鼠的最大负荷和屈服均降低。PAE 对雄性和雌性的总重量或胫骨长度均无影响。然而,与对照组相比,雄性 PAE 小鼠的生长板总宽度减少,而雌性 PAE 小鼠则不受影响。4周龄的小鼠没有表现出12周龄小鼠所观察到的PAE骨骼表型的改变:本文的证据首次表明 PAE 对骨骼产生了不同的性别影响,这可能受到成骨细胞潜在的性别特异性转录机制的影响。这些性别差异的确定将有助于未来针对 FASD 的政策和临床管理。
{"title":"Sexually dimorphic effects of prenatal alcohol exposure on the murine skeleton.","authors":"Lucie E Bourne, Soher N Jayash, Lysanne V Michels, Mark Hopkinson, Fergus M Guppy, Claire E Clarkin, Paul Gard, Nigel Brissett, Katherine A Staines","doi":"10.1186/s13293-024-00626-y","DOIUrl":"10.1186/s13293-024-00626-y","url":null,"abstract":"<p><strong>Background: </strong>Prenatal alcohol exposure (PAE) can result in lifelong disabilities known as foetal alcohol spectrum disorder (FASD) and is associated with childhood growth deficiencies and increased bone fracture risk. However, the effects of PAE on the adult skeleton remain unclear and any potential sexual dimorphism is undetermined. Therefore, we utilised a murine model to examine sex differences with PAE on in vitro bone formation, and in the juvenile and adult skeleton.</p><p><strong>Methods: </strong>Pregnant C57BL/6J female mice received 5% ethanol in their drinking water during gestation. Primary calvarial osteoblasts were isolated from neonatal offspring and mineralised bone nodule formation and gene expression assessed. Skeletal phenotyping of 4- and 12-week-old male and female offspring was conducted by micro-computed tomography (µCT), 3-point bending, growth plate analyses, and histology.</p><p><strong>Results: </strong>Osteoblasts from male and female PAE mice displayed reduced bone formation, compared to control (≤ 30%). Vegfa, Vegfb, Bmp6, Tgfbr1, Flt1 and Ahsg were downregulated in PAE male osteoblasts only, whilst Ahsg was upregulated in PAE females. In 12-week-old mice, µCT analysis revealed a sex and exposure interaction across several trabecular bone parameters. PAE was detrimental to the trabecular compartment in male mice compared to control, yet PAE females were unaffected. Both male and female mice had significant reductions in cortical parameters with PAE. Whilst male mice were negatively affected along the tibial length, females were only distally affected. Posterior cortical porosity was increased in PAE females only. Mechanical testing revealed PAE males had significantly reduced bone stiffness compared to controls; maximum load and yield were reduced in both sexes. PAE had no effect on total body weight or tibial bone length in either sex. However, total growth plate width in male PAE mice compared to control was reduced, whilst female PAE mice were unaffected. 4-week-old mice did not display the altered skeletal phenotype with PAE observed in 12-week-old animals.</p><p><strong>Conclusions: </strong>Evidence herein suggests, for the first time, that PAE exerts divergent sex effects on the skeleton, possibly influenced by underlying sex-specific transcriptional mechanisms of osteoblasts. Establishing these sex differences will support future policies and clinical management of FASD.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11186175/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141417547","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 and gender differences in adverse events following influenza and COVID-19 vaccination. 接种流感疫苗和 COVID-19 疫苗后不良事件的性别差异。
IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-06-18 DOI: 10.1186/s13293-024-00625-z
Anna Yin, Nadia Wang, Patrick J Shea, Erica N Rosser, Helen Kuo, Janna R Shapiro, Katherine Z J Fenstermacher, Andrew Pekosz, Richard E Rothman, Sabra L Klein, Rosemary Morgan

Introduction: Active and passive surveillance studies have found that a greater proportion of females report adverse events (AE) following receipt of either the COVID-19 or seasonal influenza vaccine compared to males. In a predominately young adult female population of healthcare workers, we sought to determine the intersection of biological sex and sociocultural gender differences in prospective active reporting of vaccine outcomes, which remains poorly characterized.

Methods: This cohort study enrolled Johns Hopkins Health System healthcare workers (HCWs) who were recruited from the mandatory annual fall 2019-2022 influenza vaccine and the fall 2022 COVID-19 bivalent vaccine campaigns. Vaccine recipients were enrolled the day of vaccination and AE surveys were administered two days post-vaccination for bivalent COVID-19 and influenza vaccine recipients. Data were collected regarding the presence of a series of solicited local and systemic AEs. Open-ended answers about participants' experiences with AEs also were collected for the COVID-19 vaccine recipients.

Results: Females were more likely to report local AEs after either influenza (OR = 2.28, p = 0.001) or COVID-19 (OR = 2.57, p = 0.008) vaccination compared to males, regardless of age or race. Males and females had comparable probabilities of reporting systemic AEs after either influenza (OR = 1.18, p = 0.552) or COVID-19 (OR = 0.96, p = 0.907) vaccination. Hormonal birth control use did not impact the rates of reported AEs following influenza vaccination among reproductive-aged female HCWs. Women reported more interruptions in their daily routine following COVID-19 vaccination than men and were more likely to seek out self-treatment. More women than men scheduled their COVID-19 vaccination before their days off in anticipation of AEs.

Conclusions: Our findings highlight the need for sex- and gender-inclusive policies to inform more effective mandatory occupational health vaccination strategies. Further research is needed to evaluate the potential disruption of AEs on occupational responsibilities following mandated vaccination for healthcare workers, a predominately female population, and to more fully characterize the post-vaccination behavioral differences between men and women.

导言:主动和被动监测研究发现,与男性相比,女性在接种 COVID-19 或季节性流感疫苗后报告不良事件(AE)的比例更高。在以年轻女性为主的医护人员群体中,我们试图确定生物性别和社会文化性别差异在疫苗结果前瞻性主动报告中的交叉点,而这一交叉点的特征还很不明显:这项队列研究招募了约翰霍普金斯卫生系统的医护人员(HCWs),他们是从 2019-2022 年秋季流感疫苗和 2022 年秋季 COVID-19 二价疫苗强制接种活动中招募的。疫苗接种者在接种当天进行登记,接种两天后对 COVID-19 二价疫苗和流感疫苗接种者进行 AE 调查。调查收集了有关一系列局部和全身性AE的数据。此外,还收集了 COVID-19 疫苗接种者对AEs经历的开放式回答:结果:与男性相比,无论年龄或种族如何,女性在接种流感疫苗(OR = 2.28,p = 0.001)或 COVID-19 疫苗(OR = 2.57,p = 0.008)后更有可能出现局部不良反应。男性和女性在接种流感疫苗(OR = 1.18,p = 0.552)或 COVID-19 疫苗(OR = 0.96,p = 0.907)后报告全身性 AE 的概率相当。使用荷尔蒙避孕药不会影响育龄女性医护人员接种流感疫苗后报告的不良反应发生率。与男性相比,女性在接种 COVID-19 疫苗后会中断更多的日常工作,并且更有可能寻求自我治疗。与男性相比,更多的女性将COVID-19疫苗接种安排在休息日之前,以防发生AEs:我们的研究结果强调了制定性别包容政策的必要性,以便为更有效的强制性职业健康疫苗接种策略提供依据。还需要进一步研究,以评估医疗保健工作者(女性占绝大多数)在强制接种疫苗后可能出现的AE对职业责任的干扰,并更全面地描述男性和女性在接种疫苗后的行为差异。
{"title":"Sex and gender differences in adverse events following influenza and COVID-19 vaccination.","authors":"Anna Yin, Nadia Wang, Patrick J Shea, Erica N Rosser, Helen Kuo, Janna R Shapiro, Katherine Z J Fenstermacher, Andrew Pekosz, Richard E Rothman, Sabra L Klein, Rosemary Morgan","doi":"10.1186/s13293-024-00625-z","DOIUrl":"10.1186/s13293-024-00625-z","url":null,"abstract":"<p><strong>Introduction: </strong>Active and passive surveillance studies have found that a greater proportion of females report adverse events (AE) following receipt of either the COVID-19 or seasonal influenza vaccine compared to males. In a predominately young adult female population of healthcare workers, we sought to determine the intersection of biological sex and sociocultural gender differences in prospective active reporting of vaccine outcomes, which remains poorly characterized.</p><p><strong>Methods: </strong>This cohort study enrolled Johns Hopkins Health System healthcare workers (HCWs) who were recruited from the mandatory annual fall 2019-2022 influenza vaccine and the fall 2022 COVID-19 bivalent vaccine campaigns. Vaccine recipients were enrolled the day of vaccination and AE surveys were administered two days post-vaccination for bivalent COVID-19 and influenza vaccine recipients. Data were collected regarding the presence of a series of solicited local and systemic AEs. Open-ended answers about participants' experiences with AEs also were collected for the COVID-19 vaccine recipients.</p><p><strong>Results: </strong>Females were more likely to report local AEs after either influenza (OR = 2.28, p = 0.001) or COVID-19 (OR = 2.57, p = 0.008) vaccination compared to males, regardless of age or race. Males and females had comparable probabilities of reporting systemic AEs after either influenza (OR = 1.18, p = 0.552) or COVID-19 (OR = 0.96, p = 0.907) vaccination. Hormonal birth control use did not impact the rates of reported AEs following influenza vaccination among reproductive-aged female HCWs. Women reported more interruptions in their daily routine following COVID-19 vaccination than men and were more likely to seek out self-treatment. More women than men scheduled their COVID-19 vaccination before their days off in anticipation of AEs.</p><p><strong>Conclusions: </strong>Our findings highlight the need for sex- and gender-inclusive policies to inform more effective mandatory occupational health vaccination strategies. Further research is needed to evaluate the potential disruption of AEs on occupational responsibilities following mandated vaccination for healthcare workers, a predominately female population, and to more fully characterize the post-vaccination behavioral differences between men and women.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11184791/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141417546","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
Differently different?: A commentary on the emerging social cognitive neuroscience of female autism. 与众不同?:关于女性自闭症的新兴社会认知神经科学的评论。
IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-06-13 DOI: 10.1186/s13293-024-00621-3
Gina Rippon

Autism is a neurodevelopmental condition, behaviourally identified, which is generally characterised by social communication differences, and restrictive and repetitive patterns of behaviour and interests. It has long been claimed that it is more common in males. This observed preponderance of males in autistic populations has served as a focussing framework in all spheres of autism-related issues, from recognition and diagnosis through to theoretical models and research agendas. One related issue is the near total absence of females in key research areas. For example, this paper reports a review of over 120 brain-imaging studies of social brain processes in autism that reveals that nearly 70% only included male participants or minimal numbers (just one or two) of females. Authors of such studies very rarely report that their cohorts are virtually female-free and discuss their findings as though applicable to all autistic individuals. The absence of females can be linked to exclusionary consequences of autism diagnostic procedures, which have mainly been developed on male-only cohorts. There is clear evidence that disproportionately large numbers of females do not meet diagnostic criteria and are then excluded from ongoing autism research. Another issue is a long-standing assumption that the female autism phenotype is broadly equivalent to that of the male autism phenotype. Thus, models derived from male-based studies could be applicable to females. However, it is now emerging that certain patterns of social behaviour may be very different in females. This includes a specific type of social behaviour called camouflaging or masking, linked to attempts to disguise autistic characteristics. With respect to research in the field of sex/gender cognitive neuroscience, there is emerging evidence of female differences in patterns of connectivity and/or activation in the social brain that are at odds with those reported in previous, male-only studies. Decades of research have excluded or overlooked females on the autistic spectrum, resulting in the construction of inaccurate and misleading cognitive neuroscience models, and missed opportunities to explore the brain bases of this highly complex condition. A note of warning needs to be sounded about inferences drawn from past research, but if future research addresses this problem of male bias, then a deeper understanding of autism as a whole, as well as in previously overlooked females, will start to emerge.

自闭症是一种神经发育性疾病,从行为学上可以确定,一般表现为社会交流差异、行为和兴趣模式的限制性和重复性。一直以来,自闭症在男性中更为常见。自闭症患者中男性居多的现象已成为自闭症相关问题(从识别和诊断到理论模型和研究议程)各个领域的焦点框架。与此相关的一个问题是,在关键研究领域几乎完全没有女性的身影。例如,本文对 120 多项有关自闭症患者大脑社交过程的脑成像研究进行了回顾,结果显示,近 70% 的研究仅包括男性参与者或极少数(仅一或两名)女性参与者。此类研究的作者很少报告他们的研究对象中几乎没有女性,并且在讨论他们的研究结果时,好像他们的研究结果适用于所有自闭症患者。自闭症患者中没有女性可能与自闭症诊断程序的排斥性后果有关,这些程序主要是针对男性群体开发的。有明显证据表明,不符合诊断标准的女性人数过多,因此被排除在正在进行的自闭症研究之外。另一个问题是长期以来存在的一种假设,即女性自闭症表型与男性自闭症表型大致相同。因此,从基于男性的研究中得出的模型可以适用于女性。然而,现在人们发现,女性的某些社会行为模式可能与男性有很大不同。这包括一种被称为伪装或掩饰的特殊社交行为,与试图掩饰自闭症特征有关。在性/性别认知神经科学领域的研究方面,新出现的证据表明,女性在社交大脑的连接和/或激活模式上存在差异,这与以往仅针对男性的研究报告中的内容不符。数十年来的研究排除或忽视了自闭症谱系中的女性,导致构建了不准确和误导性的认知神经科学模型,错失了探索这一高度复杂病症的大脑基础的机会。对于从过去的研究中得出的推论,我们需要敲响警钟,但如果未来的研究能够解决男性偏见的问题,那么对自闭症整体以及之前被忽视的女性的更深入了解就会开始出现。
{"title":"Differently different?: A commentary on the emerging social cognitive neuroscience of female autism.","authors":"Gina Rippon","doi":"10.1186/s13293-024-00621-3","DOIUrl":"10.1186/s13293-024-00621-3","url":null,"abstract":"<p><p>Autism is a neurodevelopmental condition, behaviourally identified, which is generally characterised by social communication differences, and restrictive and repetitive patterns of behaviour and interests. It has long been claimed that it is more common in males. This observed preponderance of males in autistic populations has served as a focussing framework in all spheres of autism-related issues, from recognition and diagnosis through to theoretical models and research agendas. One related issue is the near total absence of females in key research areas. For example, this paper reports a review of over 120 brain-imaging studies of social brain processes in autism that reveals that nearly 70% only included male participants or minimal numbers (just one or two) of females. Authors of such studies very rarely report that their cohorts are virtually female-free and discuss their findings as though applicable to all autistic individuals. The absence of females can be linked to exclusionary consequences of autism diagnostic procedures, which have mainly been developed on male-only cohorts. There is clear evidence that disproportionately large numbers of females do not meet diagnostic criteria and are then excluded from ongoing autism research. Another issue is a long-standing assumption that the female autism phenotype is broadly equivalent to that of the male autism phenotype. Thus, models derived from male-based studies could be applicable to females. However, it is now emerging that certain patterns of social behaviour may be very different in females. This includes a specific type of social behaviour called camouflaging or masking, linked to attempts to disguise autistic characteristics. With respect to research in the field of sex/gender cognitive neuroscience, there is emerging evidence of female differences in patterns of connectivity and/or activation in the social brain that are at odds with those reported in previous, male-only studies. Decades of research have excluded or overlooked females on the autistic spectrum, resulting in the construction of inaccurate and misleading cognitive neuroscience models, and missed opportunities to explore the brain bases of this highly complex condition. A note of warning needs to be sounded about inferences drawn from past research, but if future research addresses this problem of male bias, then a deeper understanding of autism as a whole, as well as in previously overlooked females, will start to emerge.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11177439/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141316689","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
Sexual dimorphism in colorectal cancer: molecular mechanisms and treatment strategies. 结直肠癌的性别双态性:分子机制和治疗策略。
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-06-12 DOI: 10.1186/s13293-024-00623-1
Yair Rodríguez-Santiago, Claudia Angelica Garay-Canales, Karen Elizabeth Nava-Castro, Jorge Morales-Montor

Introduction: Sexual dimorphism significantly influences cancer incidence and prognosis. Notably, females exhibit a lower risk and favorable prognosis for non-reproductive cancers compared to males, a pattern observable beyond the scope of risk behaviors such as alcohol consumption and smoking. Colorectal cancer, ranking third in global prevalence and second in mortality, disproportionately affects men. Sex steroid hormones, particularly estrogens and androgens, play crucial roles in cancer progression, considering epidemiological in vivo and in vitro, in general estrogens imparting a protective effect in females and androgens correlating with an increasing risk of colorectal cancer development.

Main body: The hormonal impact on immune response is mediated by receptor interactions, resulting in heightened inflammation, modulation of NF-kB, and fostering an environment conducive to cancer progression and metastasis. These molecules also influence the enteric nervous system, that is a pivotal in neuromodulator release and intestinal neuron stimulation, also contributes to cancer development, as evidenced by nerve infiltration into tumors. Microbiota diversity further intersects with immune, hormonal, and neural mechanisms, influencing colorectal cancer dynamics. A comprehensive understanding of hormonal influences on colorectal cancer progression, coupled with the complex interplay between immune responses, microbiota diversity and neurotransmitter imbalances, underpins the development of more targeted and effective therapies.

Conclusions: Estrogens mitigate colorectal cancer risk by modulating anti-tumor immune responses, enhancing microbial diversity, and curbing the pro-tumor actions of the sympathetic and enteric nervous systems. Conversely, androgens escalate tumor growth by dampening anti-tumor immune activity, reducing microbial diversity, and facilitating the release of tumor-promoting factors by the nervous system. These findings hold significant potential for the strategic purposing of drugs to fine-tune the extensive impacts of sex hormones within the tumor microenvironment, promising advancements in colorectal cancer therapies.

简介性别二形性对癌症发病率和预后有重大影响。值得注意的是,与男性相比,女性罹患非生殖系统癌症的风险较低,预后较好,这种模式超出了饮酒和吸烟等危险行为的范围。结肠直肠癌在全球发病率中排名第三,在死亡率中排名第二,对男性的影响尤为严重。考虑到体内和体外的流行病学研究,性类固醇激素,尤其是雌激素和雄激素,在癌症的发展过程中起着至关重要的作用,一般来说,雌激素对女性有保护作用,而雄激素则与结直肠癌发病风险的增加有关:激素对免疫反应的影响是通过受体相互作用介导的,从而导致炎症加剧、NF-kB 调节,并形成有利于癌症进展和转移的环境。这些分子还影响肠道神经系统,该系统是神经调节剂释放和肠道神经元刺激的关键,也有助于癌症的发展,神经浸润肿瘤就是证明。微生物群多样性与免疫、激素和神经机制进一步交叉,影响结直肠癌的动态变化。全面了解激素对结直肠癌进展的影响,以及免疫反应、微生物群多样性和神经递质失衡之间复杂的相互作用,是开发更有针对性的有效疗法的基础:结论:雌激素通过调节抗肿瘤免疫反应、增强微生物多样性以及抑制交感神经和肠道神经系统的促肿瘤作用来降低结直肠癌风险。相反,雄激素则通过抑制抗肿瘤免疫活动、降低微生物多样性以及促进神经系统释放促肿瘤因子来加剧肿瘤生长。这些发现为药物的战略目的提供了巨大的潜力,以微调性激素在肿瘤微环境中的广泛影响,从而有望推动结直肠癌疗法的发展。
{"title":"Sexual dimorphism in colorectal cancer: molecular mechanisms and treatment strategies.","authors":"Yair Rodríguez-Santiago, Claudia Angelica Garay-Canales, Karen Elizabeth Nava-Castro, Jorge Morales-Montor","doi":"10.1186/s13293-024-00623-1","DOIUrl":"10.1186/s13293-024-00623-1","url":null,"abstract":"<p><strong>Introduction: </strong>Sexual dimorphism significantly influences cancer incidence and prognosis. Notably, females exhibit a lower risk and favorable prognosis for non-reproductive cancers compared to males, a pattern observable beyond the scope of risk behaviors such as alcohol consumption and smoking. Colorectal cancer, ranking third in global prevalence and second in mortality, disproportionately affects men. Sex steroid hormones, particularly estrogens and androgens, play crucial roles in cancer progression, considering epidemiological in vivo and in vitro, in general estrogens imparting a protective effect in females and androgens correlating with an increasing risk of colorectal cancer development.</p><p><strong>Main body: </strong>The hormonal impact on immune response is mediated by receptor interactions, resulting in heightened inflammation, modulation of NF-kB, and fostering an environment conducive to cancer progression and metastasis. These molecules also influence the enteric nervous system, that is a pivotal in neuromodulator release and intestinal neuron stimulation, also contributes to cancer development, as evidenced by nerve infiltration into tumors. Microbiota diversity further intersects with immune, hormonal, and neural mechanisms, influencing colorectal cancer dynamics. A comprehensive understanding of hormonal influences on colorectal cancer progression, coupled with the complex interplay between immune responses, microbiota diversity and neurotransmitter imbalances, underpins the development of more targeted and effective therapies.</p><p><strong>Conclusions: </strong>Estrogens mitigate colorectal cancer risk by modulating anti-tumor immune responses, enhancing microbial diversity, and curbing the pro-tumor actions of the sympathetic and enteric nervous systems. Conversely, androgens escalate tumor growth by dampening anti-tumor immune activity, reducing microbial diversity, and facilitating the release of tumor-promoting factors by the nervous system. These findings hold significant potential for the strategic purposing of drugs to fine-tune the extensive impacts of sex hormones within the tumor microenvironment, promising advancements in colorectal cancer therapies.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-06-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11170921/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141309854","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
Relationship between sex biases in gene expression and sex biases in autism and Alzheimer's disease. 基因表达的性别偏差与自闭症和阿尔茨海默病的性别偏差之间的关系。
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-06-07 DOI: 10.1186/s13293-024-00622-2
Stuart B Fass, Bernard Mulvey, Rebecca Chase, Wei Yang, Din Selmanovic, Sneha M Chaturvedi, Eric Tycksen, Lauren A Weiss, Joseph D Dougherty

Background: Sex differences in the brain may play an important role in sex-differential prevalence of neuropsychiatric conditions.

Methods: In order to understand the transcriptional basis of sex differences, we analyzed multiple, large-scale, human postmortem brain RNA-Seq datasets using both within-region and pan-regional frameworks.

Results: We find evidence of sex-biased transcription in many autosomal genes, some of which provide evidence for pathways and cell population differences between chromosomally male and female individuals. These analyses also highlight regional differences in the extent of sex-differential gene expression. We observe an increase in specific neuronal transcripts in male brains and an increase in immune and glial function-related transcripts in female brains. Integration with single-nucleus data suggests this corresponds to sex differences in cellular states rather than cell abundance. Integration with case-control gene expression studies suggests a female molecular predisposition towards Alzheimer's disease, a female-biased disease. Autism, a male-biased diagnosis, does not exhibit a male predisposition pattern in our analysis.

Conclusion: Overall, these analyses highlight mechanisms by which sex differences may interact with sex-biased conditions in the brain. Furthermore, we provide region-specific analyses of sex differences in brain gene expression to enable additional studies at the interface of gene expression and diagnostic differences.

背景:大脑中的性别差异可能在神经精神疾病的性别差异患病率中扮演重要角色:大脑中的性别差异可能在神经精神疾病的性别差异患病率中扮演重要角色:为了了解性别差异的转录基础,我们利用区域内和泛区域框架分析了多个大规模人类死后大脑RNA-Seq数据集:结果:我们发现了许多常染色体基因存在性别偏向转录的证据,其中一些基因为染色体上男性和女性个体之间的通路和细胞群差异提供了证据。这些分析还凸显了性别差异基因表达程度的地区差异。我们观察到男性大脑中特定神经元转录本增加,而女性大脑中免疫和神经胶质功能相关转录本增加。与单核数据的整合表明,这与细胞状态的性别差异而非细胞丰度相对应。与病例对照基因表达研究相结合表明,女性对阿尔茨海默病这种女性偏好的疾病有分子倾向。自闭症是一种偏向男性的诊断,但在我们的分析中并没有表现出男性易感模式:总之,这些分析凸显了性别差异可能与大脑中的性别偏向性疾病相互作用的机制。此外,我们还提供了大脑基因表达中性别差异的特定区域分析,以便在基因表达和诊断差异的界面上开展更多研究。
{"title":"Relationship between sex biases in gene expression and sex biases in autism and Alzheimer's disease.","authors":"Stuart B Fass, Bernard Mulvey, Rebecca Chase, Wei Yang, Din Selmanovic, Sneha M Chaturvedi, Eric Tycksen, Lauren A Weiss, Joseph D Dougherty","doi":"10.1186/s13293-024-00622-2","DOIUrl":"10.1186/s13293-024-00622-2","url":null,"abstract":"<p><strong>Background: </strong>Sex differences in the brain may play an important role in sex-differential prevalence of neuropsychiatric conditions.</p><p><strong>Methods: </strong>In order to understand the transcriptional basis of sex differences, we analyzed multiple, large-scale, human postmortem brain RNA-Seq datasets using both within-region and pan-regional frameworks.</p><p><strong>Results: </strong>We find evidence of sex-biased transcription in many autosomal genes, some of which provide evidence for pathways and cell population differences between chromosomally male and female individuals. These analyses also highlight regional differences in the extent of sex-differential gene expression. We observe an increase in specific neuronal transcripts in male brains and an increase in immune and glial function-related transcripts in female brains. Integration with single-nucleus data suggests this corresponds to sex differences in cellular states rather than cell abundance. Integration with case-control gene expression studies suggests a female molecular predisposition towards Alzheimer's disease, a female-biased disease. Autism, a male-biased diagnosis, does not exhibit a male predisposition pattern in our analysis.</p><p><strong>Conclusion: </strong>Overall, these analyses highlight mechanisms by which sex differences may interact with sex-biased conditions in the brain. Furthermore, we provide region-specific analyses of sex differences in brain gene expression to enable additional studies at the interface of gene expression and diagnostic differences.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11157820/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141282887","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
Testosterone deficiency promotes arterial stiffening independent of sex chromosome complement. 睾酮缺乏会促进动脉僵化,与性染色体互补无关。
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-06-06 DOI: 10.1186/s13293-024-00624-0
Anil Sakamuri, Bruna Visniauskas, Isabella Kilanowski-Doroh, Alexandra B McNally, Ariane Imulinde, Anne Kamau, Divya Sengottaian, John McLachlan, Montserrat Anguera, Franck Mauvais-Jarvis, Sarah H Lindsey, Benard O Ogola

Background: Sex hormones and sex chromosomes play a vital role in cardiovascular disease. Testosterone plays a crucial role in men's health. Lower testosterone level is associated with cardiovascular and cardiometabolic diseases, including inflammation, atherosclerosis, and type 2 diabetes. Testosterone replacement is beneficial or neutral to men's cardiovascular health. Testosterone deficiency is associated with cardiovascular events. Testosterone supplementation to hypogonadal men improves libido, increases muscle strength, and enhances mood. We hypothesized that sex chromosomes (XX and XY) interaction with testosterone plays a role in arterial stiffening.

Methods: We used four core genotype male mice to understand the inherent contribution of sex hormones and sex chromosome complement in arterial stiffening. Age-matched mice were either gonadal intact or castrated at eight weeks plus an additional eight weeks to clear endogenous sex hormones. This was followed by assessing blood pressure, pulse wave velocity, echocardiography, and ex vivo passive vascular mechanics.

Results: Arterial stiffening but not blood pressure was more significant in castrated than testes-intact mice independent of sex chromosome complement. Castrated mice showed a leftward shift in stress-strain curves and carotid wall thinning. Sex chromosome complement (XX) in the absence of testosterone increased collagen deposition in the aorta and Kdm6a gene expression.

Conclusion: Testosterone deprivation increases arterial stiffening and vascular wall remodeling. Castration increases Col1α1 in male mice with XX sex chromosome complement. Our study shows decreased aortic contractile genes in castrated mice with XX than XY sex chromosomes.

背景:性激素和性染色体在心血管疾病中起着至关重要的作用。睾酮对男性健康起着至关重要的作用。睾酮水平降低与心血管和心脏代谢疾病有关,包括炎症、动脉粥样硬化和 2 型糖尿病。睾酮替代对男性心血管健康有益或无益。睾酮缺乏与心血管事件有关。性腺功能低下的男性补充睾酮可提高性欲、增强肌肉力量并改善情绪。我们假设性染色体(XX 和 XY)与睾酮的相互作用在动脉僵化中发挥作用:我们使用四种核心基因型雄性小鼠来了解性激素和性染色体互补在动脉僵化中的内在作用。年龄匹配的小鼠要么性腺完好无损,要么在八周时被阉割,再经过八周清除内源性性激素。随后对血压、脉搏波速度、超声心动图和体内外被动血管力学进行评估:结果:与性染色体互补无关,阉割小鼠的动脉僵化比睾丸未受损的小鼠更明显,但血压却不明显。阉割小鼠的应力-应变曲线左移,颈动脉壁变薄。在没有睾酮的情况下,性染色体互补(XX)增加了主动脉中胶原蛋白的沉积和Kdm6a基因的表达:结论:睾酮剥夺会增加动脉僵化和血管壁重塑。阉割会增加具有 XX 性染色体互补的雄性小鼠的 Col1α1。我们的研究显示,性染色体为 XX 的阉割小鼠的主动脉收缩基因比 XY 小鼠减少。
{"title":"Testosterone deficiency promotes arterial stiffening independent of sex chromosome complement.","authors":"Anil Sakamuri, Bruna Visniauskas, Isabella Kilanowski-Doroh, Alexandra B McNally, Ariane Imulinde, Anne Kamau, Divya Sengottaian, John McLachlan, Montserrat Anguera, Franck Mauvais-Jarvis, Sarah H Lindsey, Benard O Ogola","doi":"10.1186/s13293-024-00624-0","DOIUrl":"10.1186/s13293-024-00624-0","url":null,"abstract":"<p><strong>Background: </strong>Sex hormones and sex chromosomes play a vital role in cardiovascular disease. Testosterone plays a crucial role in men's health. Lower testosterone level is associated with cardiovascular and cardiometabolic diseases, including inflammation, atherosclerosis, and type 2 diabetes. Testosterone replacement is beneficial or neutral to men's cardiovascular health. Testosterone deficiency is associated with cardiovascular events. Testosterone supplementation to hypogonadal men improves libido, increases muscle strength, and enhances mood. We hypothesized that sex chromosomes (XX and XY) interaction with testosterone plays a role in arterial stiffening.</p><p><strong>Methods: </strong>We used four core genotype male mice to understand the inherent contribution of sex hormones and sex chromosome complement in arterial stiffening. Age-matched mice were either gonadal intact or castrated at eight weeks plus an additional eight weeks to clear endogenous sex hormones. This was followed by assessing blood pressure, pulse wave velocity, echocardiography, and ex vivo passive vascular mechanics.</p><p><strong>Results: </strong>Arterial stiffening but not blood pressure was more significant in castrated than testes-intact mice independent of sex chromosome complement. Castrated mice showed a leftward shift in stress-strain curves and carotid wall thinning. Sex chromosome complement (XX) in the absence of testosterone increased collagen deposition in the aorta and Kdm6a gene expression.</p><p><strong>Conclusion: </strong>Testosterone deprivation increases arterial stiffening and vascular wall remodeling. Castration increases Col1α1 in male mice with XX sex chromosome complement. Our study shows decreased aortic contractile genes in castrated mice with XX than XY sex chromosomes.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-06-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11155160/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141282888","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
Low testosterone levels relate to poorer cognitive function in women in an APOE-ε4-dependant manner. 低睾酮水平与女性较差的认知功能有关,其方式取决于 APOE-ε4。
IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-06-05 DOI: 10.1186/s13293-024-00620-4
Melanie A Dratva, Sarah J Banks, Matthew S Panizzon, Douglas Galasko, Erin E Sundermann

Background: Past research suggests that low testosterone levels relate to poorer cognitive function and higher Alzheimer's disease (AD) risk; however, these findings are inconsistent and are mostly derived from male samples, despite similar age-related testosterone decline in females. Both animal and human studies demonstrate that testosterone's effects on brain health may be moderated by apolipoprotein E ε4 allele (APOE-ε4) carrier status, which may explain some previous inconsistencies. We examined how testosterone relates to cognitive function in older women versus men across healthy aging and the AD continuum and the moderating role of APOE-ε4 genotype.

Methods: Five hundred and sixty one participants aged 55-90 (155 cognitively normal (CN), 294 mild cognitive impairment (MCI), 112 AD dementia) from the Alzheimer's Disease Neuroimaging Initiative (ADNI), who had baseline cognitive and plasma testosterone data, as measured by the Rules Based Medicine Human DiscoveryMAP Panel were included. There were 213 females and 348 males (self-reported sex assigned at birth), and 52% of the overall sample were APOE-ε4 carriers. We tested the relationship of plasma testosterone levels and its interaction with APOE-ε4 status on clinical diagnostic group (CN vs. MCI vs. AD), global, and domain-specific cognitive performance using ANOVAs and linear regression models in sex-stratified samples. Cognitive domains included verbal memory, executive function, processing speed, and language.

Results: We did not observe a significant difference in testosterone levels between clinical diagnostic groups in either sex, regrardless of APOE-ε4 status. Across clinical diagnostic group, we found a significant testosterone by APOE-ε4 interaction in females, such that lower testosterone levels related to worse global cognition, processing speed, and verbal memory in APOE-ε4 carriers only. We did not find that testosterone, nor its interaction with APOE-ε4, related to cognitive outcomes in males.

Conclusions: Findings suggest that low testosterone levels in older female APOE-ε4 carriers across the aging-MCI-AD continuum may have deleterious, domain-specific effects on cognitive performance. Although future studies including additional sex hormones and longitudinal cognitive trajectories are needed, our results highlight the importance of including both sexes and considering APOE-ε4 carrier status when examining testosterone's role in cognitive health.

背景:过去的研究表明,睾酮水平低与认知功能较差和阿尔茨海默病(AD)风险较高有关;然而,这些研究结果并不一致,而且大多来自男性样本,尽管女性也存在与年龄相关的睾酮下降。动物和人体研究都表明,睾酮对大脑健康的影响可能会受到载脂蛋白 E ε4等位基因(APOE-ε4)携带者身份的影响,这可能解释了之前的一些不一致之处。我们研究了睾酮与老年女性认知功能的关系,以及APOE-ε4基因型的调节作用:研究人员纳入了来自阿尔茨海默病神经影像学倡议(ADNI)的 561 名 55-90 岁参与者(155 名认知正常者(CN)、294 名轻度认知障碍者(MCI)、112 名 AD 痴呆者),这些参与者都有基线认知和血浆睾酮数据,这些数据是由基于规则的医学人类发现 MAP 小组测量的。其中有 213 名女性和 348 名男性(出生时自我报告的性别),总体样本中有 52% 是 APOE-ε4 携带者。我们在性别分层样本中使用方差分析和线性回归模型检验了血浆睾酮水平及其与 APOE-ε4 状态的交互作用与临床诊断组(CN vs. MCI vs. AD)、整体和特定领域认知能力的关系。认知领域包括言语记忆、执行功能、处理速度和语言:我们没有观察到临床诊断组之间男女睾酮水平的显著差异,与 APOE-ε4 状态无关。在不同的临床诊断组中,我们发现女性的睾酮与APOE-ε4之间存在显著的交互作用,即睾酮水平越低,APOE-ε4携带者的整体认知能力、处理速度和语言记忆力越差。我们没有发现睾酮或其与APOE-ε4的相互作用与男性的认知结果有关:研究结果表明,老年女性APOE-ε4携带者在整个衰老-MCI-AD过程中的低睾酮水平可能会对认知能力产生有害的、特定领域的影响。尽管未来的研究还需要包括更多的性激素和纵向认知轨迹,但我们的研究结果强调了在研究睾酮在认知健康中的作用时,将两性都包括在内并考虑 APOE-ε4 携带者状态的重要性。
{"title":"Low testosterone levels relate to poorer cognitive function in women in an APOE-ε4-dependant manner.","authors":"Melanie A Dratva, Sarah J Banks, Matthew S Panizzon, Douglas Galasko, Erin E Sundermann","doi":"10.1186/s13293-024-00620-4","DOIUrl":"10.1186/s13293-024-00620-4","url":null,"abstract":"<p><strong>Background: </strong>Past research suggests that low testosterone levels relate to poorer cognitive function and higher Alzheimer's disease (AD) risk; however, these findings are inconsistent and are mostly derived from male samples, despite similar age-related testosterone decline in females. Both animal and human studies demonstrate that testosterone's effects on brain health may be moderated by apolipoprotein E ε4 allele (APOE-ε4) carrier status, which may explain some previous inconsistencies. We examined how testosterone relates to cognitive function in older women versus men across healthy aging and the AD continuum and the moderating role of APOE-ε4 genotype.</p><p><strong>Methods: </strong>Five hundred and sixty one participants aged 55-90 (155 cognitively normal (CN), 294 mild cognitive impairment (MCI), 112 AD dementia) from the Alzheimer's Disease Neuroimaging Initiative (ADNI), who had baseline cognitive and plasma testosterone data, as measured by the Rules Based Medicine Human DiscoveryMAP Panel were included. There were 213 females and 348 males (self-reported sex assigned at birth), and 52% of the overall sample were APOE-ε4 carriers. We tested the relationship of plasma testosterone levels and its interaction with APOE-ε4 status on clinical diagnostic group (CN vs. MCI vs. AD), global, and domain-specific cognitive performance using ANOVAs and linear regression models in sex-stratified samples. Cognitive domains included verbal memory, executive function, processing speed, and language.</p><p><strong>Results: </strong>We did not observe a significant difference in testosterone levels between clinical diagnostic groups in either sex, regrardless of APOE-ε4 status. Across clinical diagnostic group, we found a significant testosterone by APOE-ε4 interaction in females, such that lower testosterone levels related to worse global cognition, processing speed, and verbal memory in APOE-ε4 carriers only. We did not find that testosterone, nor its interaction with APOE-ε4, related to cognitive outcomes in males.</p><p><strong>Conclusions: </strong>Findings suggest that low testosterone levels in older female APOE-ε4 carriers across the aging-MCI-AD continuum may have deleterious, domain-specific effects on cognitive performance. Although future studies including additional sex hormones and longitudinal cognitive trajectories are needed, our results highlight the importance of including both sexes and considering APOE-ε4 carrier status when examining testosterone's role in cognitive health.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11151480/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141247264","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
CGRP as a potential mediator for the sexually dimorphic responses to traumatic brain injury. CGRP 作为创伤性脑损伤性双态反应的潜在介质。
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-05-30 DOI: 10.1186/s13293-024-00619-x
Chunyan Li, Erum Ajmal, Khaled Alok, Keren Powell, Steven Wadolowski, Willians Tambo, Justin Turpin, Ernest Barthélemy, Yousef Al-Abed, David LeDoux

Background: The outcomes of traumatic brain injury (TBI) exhibit variance contingent upon biological sex. Although female sex hormones exert neuroprotective effects, the administration of estrogen and progesterone has not yielded conclusive results. Hence, it is conceivable that additional mediators, distinct from female sex hormones, merit consideration due to their potential differential impact on TBI outcomes. Calcitonin gene-related peptide (CGRP) exhibits sexually dimorphic expression and demonstrates neuroprotective effects in acute brain injuries. In this study, we aimed to examine sex-based variations in TBI structural and functional outcomes with respect to CGRP expression.

Methods: Male and female Sprague Dawley rats were exposed to controlled cortical impact to induce severe TBI, followed by interventions with and without CGRP inhibition. In the acute phase of TBI, the study centered on elucidating the influence of CGRP on oxidative stress, nuclear factor erythroid 2-related factor 2 (Nrf2) and endothelial nitric oxide synthase (eNOS) signaling in the peri-impact tissue. Subsequently, during the chronic phase of TBI, the investigation expanded to evaluate CGRP expression in relation to lesion volume, microvascular dysfunction, and white matter injury, as well as working and spatial memory, anxiety-like, and depression-like behaviors in subjects of both sexes.

Results: Female rats exhibited elevated levels of CGRP in the peri-impact brain tissue during both baseline conditions and in the acute and chronic phases of TBI, in comparison to age-matched male counterparts. Enhanced CGRP levels in specific brain sub-regions among female rats correlated with superior structural and functional outcomes following TBI compared to their male counterparts. CGRP inhibition induced heightened oxidative stress and a reduction in the expression of Nrf2 and eNOS in both male and female rats, with the observed alteration being more pronounced in females than in males.

Conclusions: This study marks the inaugural identification of CGRP as a downstream mediator contributing to the sexually dimorphic response observed in TBI outcomes.

背景:创伤性脑损伤(TBI)的结果因性别而异。虽然女性性激素具有神经保护作用,但雌激素和孕酮的应用尚未产生结论性结果。因此,除了女性性激素之外,其他介质对创伤性脑损伤结果的潜在影响也值得考虑。降钙素基因相关肽(CGRP)在急性脑损伤中表现出性别双态性,并具有神经保护作用。在这项研究中,我们旨在研究 TBI 结构和功能结果中 CGRP 表达的性别差异:方法:将雌雄 Sprague Dawley 大鼠暴露于可控的皮质冲击下,诱发严重的 TBI,然后进行抑制 CGRP 和不抑制 CGRP 的干预。在创伤性脑损伤的急性期,研究重点是阐明 CGRP 对冲击周围组织中氧化应激、核因子红细胞 2 相关因子 2(Nrf2)和内皮一氧化氮合酶(eNOS)信号传导的影响。随后,在创伤性脑损伤的慢性阶段,研究扩大到评估 CGRP 表达与病变体积、微血管功能障碍和白质损伤的关系,以及男女受试者的工作记忆和空间记忆、焦虑样和抑郁样行为:结果:与年龄相匹配的雄性大鼠相比,雌性大鼠在创伤性脑损伤的基线状态、急性和慢性阶段表现出冲击周围脑组织中 CGRP 水平的升高。与雄性大鼠相比,雌性大鼠特定脑亚区域中 CGRP 水平的升高与创伤性脑损伤后的结构和功能结果相关。在雄性和雌性大鼠中,CGRP抑制诱导氧化应激增加,Nrf2和eNOS的表达减少,观察到的变化在雌性大鼠中比在雄性大鼠中更明显:本研究首次发现 CGRP 是导致创伤性脑损伤结果出现性别双态反应的下游介质。
{"title":"CGRP as a potential mediator for the sexually dimorphic responses to traumatic brain injury.","authors":"Chunyan Li, Erum Ajmal, Khaled Alok, Keren Powell, Steven Wadolowski, Willians Tambo, Justin Turpin, Ernest Barthélemy, Yousef Al-Abed, David LeDoux","doi":"10.1186/s13293-024-00619-x","DOIUrl":"10.1186/s13293-024-00619-x","url":null,"abstract":"<p><strong>Background: </strong>The outcomes of traumatic brain injury (TBI) exhibit variance contingent upon biological sex. Although female sex hormones exert neuroprotective effects, the administration of estrogen and progesterone has not yielded conclusive results. Hence, it is conceivable that additional mediators, distinct from female sex hormones, merit consideration due to their potential differential impact on TBI outcomes. Calcitonin gene-related peptide (CGRP) exhibits sexually dimorphic expression and demonstrates neuroprotective effects in acute brain injuries. In this study, we aimed to examine sex-based variations in TBI structural and functional outcomes with respect to CGRP expression.</p><p><strong>Methods: </strong>Male and female Sprague Dawley rats were exposed to controlled cortical impact to induce severe TBI, followed by interventions with and without CGRP inhibition. In the acute phase of TBI, the study centered on elucidating the influence of CGRP on oxidative stress, nuclear factor erythroid 2-related factor 2 (Nrf2) and endothelial nitric oxide synthase (eNOS) signaling in the peri-impact tissue. Subsequently, during the chronic phase of TBI, the investigation expanded to evaluate CGRP expression in relation to lesion volume, microvascular dysfunction, and white matter injury, as well as working and spatial memory, anxiety-like, and depression-like behaviors in subjects of both sexes.</p><p><strong>Results: </strong>Female rats exhibited elevated levels of CGRP in the peri-impact brain tissue during both baseline conditions and in the acute and chronic phases of TBI, in comparison to age-matched male counterparts. Enhanced CGRP levels in specific brain sub-regions among female rats correlated with superior structural and functional outcomes following TBI compared to their male counterparts. CGRP inhibition induced heightened oxidative stress and a reduction in the expression of Nrf2 and eNOS in both male and female rats, with the observed alteration being more pronounced in females than in males.</p><p><strong>Conclusions: </strong>This study marks the inaugural identification of CGRP as a downstream mediator contributing to the sexually dimorphic response observed in TBI outcomes.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-05-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11138127/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141178532","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
Fatty Liver Index (FLI) is the best score to predict MASLD with 50% lower cut-off value in women than in men. 脂肪肝指数(FLI)是预测 MASLD 的最佳指标,女性的临界值比男性低 50%。
IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-05-17 DOI: 10.1186/s13293-024-00617-z
Lucilla Crudele, Carlo De Matteis, Fabio Novielli, Ersilia Di Buduo, Stefano Petruzzelli, Alessia De Giorgi, Gianfranco Antonica, Elsa Berardi, Antonio Moschetta

Background: Metabolic dysfunction-associated steatotic liver disease (MASLD) is defined by the presence of hepatic steatosis, detected on ultrasonography (US) imaging or histology, and at least one of criteria for Metabolic Syndrome diagnosis. Simple non-invasive tests (NITs) have been proposed as an acceptable alternative when US and biopsy are not available or feasible but have not been validated for MASLD. In this observational study, we investigated the reliability of NITs for MASLD detection and whether sex-differences in screening methods should be considered.

Methods: We included 1069 individuals (48% males and 52% females) who underwent their first clinical examination for Metabolic Syndrome in the period between January 2015 and December 2022. Liver steatosis was detected through US and anthropometric and clinical parameters were recorded.

Results: Liver steatosis was detected in 648 patients and MASLD was diagnosed in 630 subjects (355 males; 275 females). Women with MASLD showed better metabolic profile and lower prevalence of Metabolic Syndrome criteria than men. Among NITs, Fatty Liver Index (FLI) showed the best ability for detection of MASLD, with a cut-off value of 44 (AUC = 0.82). When considering the two sexes for MASLD detection via FLI, despite no substantial differences regarding FLI correlations with metabolic biomarkers except for age, women showed marked lower FLI cut-off value (32; AUC = 0.80) than men (60; AUC = 0.80).

Conclusions: In this study, we found that FLI is the best non-invasive predictor of both liver steatosis and MASLD. The finding that in women FLI cut-off value for MASLD detection is 50% lower than in men suggests the need of a sex-specific personalized program of screening and prevention of dysmetabolism-related liver diseases, despite outwardly healthy biomarkers profile.

背景:代谢功能障碍相关性脂肪性肝病(MASLD)的定义是:超声波成像(US)或组织学检查发现肝脏脂肪变性,且至少有一项代谢综合征诊断标准。有人提出,在无法使用或无法进行 US 和活组织检查时,可将简单的无创检查(NIT)作为一种可接受的替代方法,但这种方法尚未针对 MASLD 进行验证。在这项观察性研究中,我们调查了 NIT 检测 MASLD 的可靠性,以及是否应考虑筛查方法的性别差异:我们纳入了 2015 年 1 月至 2022 年 12 月期间首次接受代谢综合征临床检查的 1069 人(男性占 48%,女性占 52%)。通过 US 检测肝脏脂肪变性,并记录人体测量和临床参数:结果:648 名患者检测出肝脏脂肪变性,630 名受试者(355 名男性;275 名女性)确诊为 MASLD。与男性相比,女性脂肪肝患者的代谢状况更好,代谢综合征标准的发病率更低。在 NITs 中,脂肪肝指数(FLI)检测 MASLD 的能力最强,临界值为 44(AUC = 0.82)。在通过 FLI 检测 MASLD 时,尽管除年龄外 FLI 与代谢生物标志物的相关性没有实质性差异,但女性的 FLI 临界值(32;AUC = 0.80)明显低于男性(60;AUC = 0.80):在这项研究中,我们发现 FLI 是肝脏脂肪变性和 MASLD 的最佳无创预测指标。发现女性的 FLI 临界值比男性低 50%,这表明,尽管女性的生物标志物外表健康,但仍需要针对不同性别的个性化方案来筛查和预防代谢异常相关的肝病。
{"title":"Fatty Liver Index (FLI) is the best score to predict MASLD with 50% lower cut-off value in women than in men.","authors":"Lucilla Crudele, Carlo De Matteis, Fabio Novielli, Ersilia Di Buduo, Stefano Petruzzelli, Alessia De Giorgi, Gianfranco Antonica, Elsa Berardi, Antonio Moschetta","doi":"10.1186/s13293-024-00617-z","DOIUrl":"10.1186/s13293-024-00617-z","url":null,"abstract":"<p><strong>Background: </strong>Metabolic dysfunction-associated steatotic liver disease (MASLD) is defined by the presence of hepatic steatosis, detected on ultrasonography (US) imaging or histology, and at least one of criteria for Metabolic Syndrome diagnosis. Simple non-invasive tests (NITs) have been proposed as an acceptable alternative when US and biopsy are not available or feasible but have not been validated for MASLD. In this observational study, we investigated the reliability of NITs for MASLD detection and whether sex-differences in screening methods should be considered.</p><p><strong>Methods: </strong>We included 1069 individuals (48% males and 52% females) who underwent their first clinical examination for Metabolic Syndrome in the period between January 2015 and December 2022. Liver steatosis was detected through US and anthropometric and clinical parameters were recorded.</p><p><strong>Results: </strong>Liver steatosis was detected in 648 patients and MASLD was diagnosed in 630 subjects (355 males; 275 females). Women with MASLD showed better metabolic profile and lower prevalence of Metabolic Syndrome criteria than men. Among NITs, Fatty Liver Index (FLI) showed the best ability for detection of MASLD, with a cut-off value of 44 (AUC = 0.82). When considering the two sexes for MASLD detection via FLI, despite no substantial differences regarding FLI correlations with metabolic biomarkers except for age, women showed marked lower FLI cut-off value (32; AUC = 0.80) than men (60; AUC = 0.80).</p><p><strong>Conclusions: </strong>In this study, we found that FLI is the best non-invasive predictor of both liver steatosis and MASLD. The finding that in women FLI cut-off value for MASLD detection is 50% lower than in men suggests the need of a sex-specific personalized program of screening and prevention of dysmetabolism-related liver diseases, despite outwardly healthy biomarkers profile.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-05-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11100212/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140955638","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 impacts of prenatal bisphenol A exposure on genes associated with cortical development, social behaviors, and autism in the offspring's prefrontal cortex. 产前双酚 A 暴露对后代前额叶皮层中与皮层发育、社交行为和自闭症相关基因的性别特异性影响。
IF 7.9 2区 医学 Q1 Social Sciences Pub Date : 2024-05-15 DOI: 10.1186/s13293-024-00614-2
Songphon Kanlayaprasit, Thanit Saeliw, Surangrat Thongkorn, Pawinee Panjabud, Kasidit Kasitipradit, Pattanachat Lertpeerapan, Kwanjira Songsritaya, Wasana Yuwattana, Thanawin Jantheang, Depicha Jindatip, Valerie W Hu, Takako Kikkawa, Noriko Osumi, Tewarit Sarachana

Background: Recent studies have shown that prenatal BPA exposure altered the transcriptome profiles of autism-related genes in the offspring's hippocampus, disrupting hippocampal neuritogenesis and causing male-specific deficits in learning. However, the sex differences in the effects of prenatal BPA exposure on the developing prefrontal cortex, which is another brain region highly implicated in autism spectrum disorder (ASD), have not been investigated.

Methods: We obtained transcriptome data from RNA sequencing analysis of the prefrontal cortex of male and female rat pups prenatally exposed to BPA or control and reanalyzed. BPA-responsive genes associated with cortical development and social behaviors were selected for confirmation by qRT-PCR analysis. Neuritogenesis of primary cells from the prefrontal cortex of pups prenatally exposed to BPA or control was examined. The social behaviors of the pups were assessed using the two-trial and three-chamber tests. The male-specific impact of the downregulation of a selected BPA-responsive gene (i.e., Sema5a) on cortical development in vivo was interrogated using siRNA-mediated knockdown by an in utero electroporation technique.

Results: Genes disrupted by prenatal BPA exposure were associated with ASD and showed sex-specific dysregulation. Sema5a and Slc9a9, which were involved in neuritogenesis and social behaviors, were downregulated only in males, while Anxa2 and Junb, which were also linked to neuritogenesis and social behaviors, were suppressed only in females. Neuritogenesis was increased in males and showed a strong inverse correlation with Sema5a and Slc9a9 expression levels, whereas, in the females, neuritogenesis was decreased and correlated with Anxa2 and Junb levels. The siRNA-mediated knockdown of Sema5a in males also impaired cortical development in utero. Consistent with Anxa2 and Junb downregulations, deficits in social novelty were observed only in female offspring but not in males.

Conclusion: This is the first study to show that prenatal BPA exposure dysregulated the expression of ASD-related genes and functions, including cortical neuritogenesis and development and social behaviors, in a sex-dependent manner. Our findings suggest that, besides the hippocampus, BPA could also exert its adverse effects through sex-specific molecular mechanisms in the offspring's prefrontal cortex, which in turn would lead to sex differences in ASD-related neuropathology and clinical manifestations, which deserves further investigation.

背景:最近的研究表明,产前暴露于双酚A会改变后代海马中自闭症相关基因的转录组图谱,破坏海马神经元的生成,导致男性特有的学习障碍。然而,产前暴露于双酚 A 对发育中的前额叶皮层(另一个与自闭症谱系障碍(ASD)高度相关的脑区)的影响的性别差异尚未得到研究:我们从RNA测序分析中获得了产前暴露于双酚A或对照组的雌雄幼鼠前额叶皮层的转录组数据,并对其进行了重新分析。通过 qRT-PCR 分析,筛选出与大脑皮层发育和社会行为相关的 BPA 反应基因进行确认。检测了产前暴露于双酚 A 或对照组的幼鼠前额叶皮层原代细胞的神经发生。使用两试验和三腔试验评估了幼鼠的社会行为。通过宫内电穿孔技术,使用 siRNA 介导的基因敲除技术,研究了选定的双酚 A 反应基因(即 Sema5a)下调对体内大脑皮层发育的雄性特异性影响:结果:因产前暴露于双酚 A 而被破坏的基因与 ASD 有关,并表现出性别特异性失调。参与神经元发生和社会行为的 Sema5a 和 Slc9a9 仅在男性中被下调,而同样与神经元发生和社会行为有关的 Anxa2 和 Junb 仅在女性中被抑制。雄性的神经发生增加,并与Sema5a和Slc9a9的表达水平呈强烈的反相关,而雌性的神经发生减少,并与Anexa2和Junb的表达水平相关。siRNA 介导的男性 Sema5a 基因敲除也会影响子宫内的大脑皮层发育。与 Anxa2 和 Junb 的下调相一致的是,只在雌性后代中观察到社交新奇性缺陷,而在雄性后代中没有观察到:这是首次研究表明,产前暴露于双酚 A 会导致 ASD 相关基因和功能的表达失调,包括大脑皮层神经元的发生和发育以及社会行为。我们的研究结果表明,除了海马体外,双酚A还可能通过后代前额叶皮层的性别特异性分子机制产生不良影响,进而导致ASD相关神经病理学和临床表现的性别差异,这值得进一步研究。
{"title":"Sex-specific impacts of prenatal bisphenol A exposure on genes associated with cortical development, social behaviors, and autism in the offspring's prefrontal cortex.","authors":"Songphon Kanlayaprasit, Thanit Saeliw, Surangrat Thongkorn, Pawinee Panjabud, Kasidit Kasitipradit, Pattanachat Lertpeerapan, Kwanjira Songsritaya, Wasana Yuwattana, Thanawin Jantheang, Depicha Jindatip, Valerie W Hu, Takako Kikkawa, Noriko Osumi, Tewarit Sarachana","doi":"10.1186/s13293-024-00614-2","DOIUrl":"10.1186/s13293-024-00614-2","url":null,"abstract":"<p><strong>Background: </strong>Recent studies have shown that prenatal BPA exposure altered the transcriptome profiles of autism-related genes in the offspring's hippocampus, disrupting hippocampal neuritogenesis and causing male-specific deficits in learning. However, the sex differences in the effects of prenatal BPA exposure on the developing prefrontal cortex, which is another brain region highly implicated in autism spectrum disorder (ASD), have not been investigated.</p><p><strong>Methods: </strong>We obtained transcriptome data from RNA sequencing analysis of the prefrontal cortex of male and female rat pups prenatally exposed to BPA or control and reanalyzed. BPA-responsive genes associated with cortical development and social behaviors were selected for confirmation by qRT-PCR analysis. Neuritogenesis of primary cells from the prefrontal cortex of pups prenatally exposed to BPA or control was examined. The social behaviors of the pups were assessed using the two-trial and three-chamber tests. The male-specific impact of the downregulation of a selected BPA-responsive gene (i.e., Sema5a) on cortical development in vivo was interrogated using siRNA-mediated knockdown by an in utero electroporation technique.</p><p><strong>Results: </strong>Genes disrupted by prenatal BPA exposure were associated with ASD and showed sex-specific dysregulation. Sema5a and Slc9a9, which were involved in neuritogenesis and social behaviors, were downregulated only in males, while Anxa2 and Junb, which were also linked to neuritogenesis and social behaviors, were suppressed only in females. Neuritogenesis was increased in males and showed a strong inverse correlation with Sema5a and Slc9a9 expression levels, whereas, in the females, neuritogenesis was decreased and correlated with Anxa2 and Junb levels. The siRNA-mediated knockdown of Sema5a in males also impaired cortical development in utero. Consistent with Anxa2 and Junb downregulations, deficits in social novelty were observed only in female offspring but not in males.</p><p><strong>Conclusion: </strong>This is the first study to show that prenatal BPA exposure dysregulated the expression of ASD-related genes and functions, including cortical neuritogenesis and development and social behaviors, in a sex-dependent manner. Our findings suggest that, besides the hippocampus, BPA could also exert its adverse effects through sex-specific molecular mechanisms in the offspring's prefrontal cortex, which in turn would lead to sex differences in ASD-related neuropathology and clinical manifestations, which deserves further investigation.</p>","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11094985/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140943904","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