首页 > 最新文献

Nature aging最新文献

英文 中文
Targeting dysregulated phago-/auto-lysosomes in Sertoli cells to ameliorate late-onset hypogonadism 靶向调节失常的 Sertoli 细胞吞噬体/自动溶酶体,改善晚发性性腺功能减退症
Pub Date : 2024-04-22 DOI: 10.1038/s43587-024-00614-2
Zhiwen Deng, Liangyu Zhao, Sha Li, Xiaoyang Chen, Xiaohan Ling, Jiajun Zheng, Kunkun Yu, Jing Xu, Chencheng Yao, Sha Han, Jiayi Liang, Huimin Feng, Lanlan Wu, Peng Li, Ruhui Tian, Tao Jing, Yuxin Tang, Yingbo Dai, Minbo Yan, Chenchen Wang, Zheng Li, Zhi Zhou
Age-related changes in testicular function can impact health and well-being. The mechanisms underlying age-related testicular dysfunction, such as late-onset hypogonadism (LOH), remain incompletely understood. Using single-cell RNA sequencing on human testes with LOH, we delineated Sertoli cells (SCs) as pivotal metabolic coordinators within the testicular microenvironment. In particular, lysosomal acidity probing revealed compromised degradative capacity in aged SCs, hindering autophagy and phagocytic flux. Consequently, SCs accumulated metabolites, including cholesterol, and have increased inflammatory gene expression; thus, we termed these cells as phago-/auto-lysosomal deregulated SCs. Exposure to a high-fat diet-induced phago-/auto-lysosomal dysregulated-like SCs, recapitulating LOH features in mice. Notably, efferent ductular injection and systemic TRPML1 agonist administration restored lysosomal function, normalizing testosterone deficiency and associated abnormalities in high-fat diet-induced LOH mice. Our findings underscore the central role of SCs in testis aging, presenting a promising therapeutic avenue for LOH. Late-onset hypogonadism (LOH) can occur with male reproductive aging and is characterized by declining testosterone levels as well as other clinical symptoms. Here the authors show that dysregulated phago-/auto-lysosomes in Sertoli cells are a key feature of LOH, linking metabolism and aging, and that pharmaceutical targeting of lysosome dysfunction can alleviate LOH in mice.
与年龄有关的睾丸功能变化会影响健康和幸福。人们对与年龄相关的睾丸功能障碍(如晚发性腺功能减退症(LOH))的机制仍不完全了解。通过对患有 LOH 的人类睾丸进行单细胞 RNA 测序,我们发现 Sertoli 细胞(SC)是睾丸微环境中关键的代谢协调者。特别是,溶酶体酸度探测显示,老化的SCs降解能力受损,阻碍了自噬和吞噬通量。因此,SCs积累了包括胆固醇在内的代谢物,并增加了炎症基因的表达;因此,我们将这些细胞称为吞噬/自溶酶体失调的SCs。暴露于高脂肪饮食会诱发吞噬/自动溶酶体失调样SC,再现小鼠的LOH特征。值得注意的是,传出导管注射和全身服用TRPML1激动剂可恢复溶酶体功能,使高脂饮食诱导的LOH小鼠的睾酮缺乏症和相关异常恢复正常。我们的研究结果强调了SC在睾丸衰老中的核心作用,为LOH的治疗提供了一个前景广阔的途径。
{"title":"Targeting dysregulated phago-/auto-lysosomes in Sertoli cells to ameliorate late-onset hypogonadism","authors":"Zhiwen Deng, Liangyu Zhao, Sha Li, Xiaoyang Chen, Xiaohan Ling, Jiajun Zheng, Kunkun Yu, Jing Xu, Chencheng Yao, Sha Han, Jiayi Liang, Huimin Feng, Lanlan Wu, Peng Li, Ruhui Tian, Tao Jing, Yuxin Tang, Yingbo Dai, Minbo Yan, Chenchen Wang, Zheng Li, Zhi Zhou","doi":"10.1038/s43587-024-00614-2","DOIUrl":"10.1038/s43587-024-00614-2","url":null,"abstract":"Age-related changes in testicular function can impact health and well-being. The mechanisms underlying age-related testicular dysfunction, such as late-onset hypogonadism (LOH), remain incompletely understood. Using single-cell RNA sequencing on human testes with LOH, we delineated Sertoli cells (SCs) as pivotal metabolic coordinators within the testicular microenvironment. In particular, lysosomal acidity probing revealed compromised degradative capacity in aged SCs, hindering autophagy and phagocytic flux. Consequently, SCs accumulated metabolites, including cholesterol, and have increased inflammatory gene expression; thus, we termed these cells as phago-/auto-lysosomal deregulated SCs. Exposure to a high-fat diet-induced phago-/auto-lysosomal dysregulated-like SCs, recapitulating LOH features in mice. Notably, efferent ductular injection and systemic TRPML1 agonist administration restored lysosomal function, normalizing testosterone deficiency and associated abnormalities in high-fat diet-induced LOH mice. Our findings underscore the central role of SCs in testis aging, presenting a promising therapeutic avenue for LOH. Late-onset hypogonadism (LOH) can occur with male reproductive aging and is characterized by declining testosterone levels as well as other clinical symptoms. Here the authors show that dysregulated phago-/auto-lysosomes in Sertoli cells are a key feature of LOH, linking metabolism and aging, and that pharmaceutical targeting of lysosome dysfunction can alleviate LOH in mice.","PeriodicalId":94150,"journal":{"name":"Nature aging","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140637065","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The advent of Alzheimer treatments will change the trajectory of human aging 老年痴呆症治疗方法的出现将改变人类衰老的轨迹
Pub Date : 2024-04-19 DOI: 10.1038/s43587-024-00611-5
Dennis J. Selkoe
Slowing neurodegenerative disorders of late life has lagged behind progress on other chronic diseases. But advances in two areas, biochemical pathology and human genetics, have now identified early pathogenic events, enabling molecular hypotheses and disease-modifying treatments. A salient example is the discovery that antibodies to amyloid ß-protein, long debated as a causative factor in Alzheimer’s disease (AD), clear amyloid plaques, decrease levels of abnormal tau proteins and slow cognitive decline. Approval of amyloid antibodies as the first disease-modifying treatments means a gradually rising fraction of the world’s estimated 60 million people with symptomatic disease may decline less or even stabilize. Society is entering an era in which the unchecked devastation of AD is no longer inevitable. This Perspective considers the impact of slowing AD and other neurodegenerative disorders on the trajectory of aging, allowing people to survive into late life with less functional decline. The implications of this moment for medicine and society are profound. The advent of plaque-clearing antibodies to the amyloid-β as the first disease-modifying treatment for Alzheimer’s disease will change the course of this disease, the most common type of dementia. Related progress will gradually alter the trajectory of human aging.
减缓晚年神经退行性疾病的进展一直落后于其他慢性疾病。但是,生化病理学和人类遗传学这两个领域的进步现已发现了早期致病事件,从而能够提出分子假说和改变疾病的治疗方法。一个突出的例子是,人们发现淀粉样ß蛋白抗体(长期以来一直被认为是阿尔茨海默病(AD)的致病因素)可以清除淀粉样斑块,降低异常tau蛋白的水平,减缓认知能力的衰退。淀粉样蛋白抗体作为第一种改变病情的治疗方法获得批准,意味着在全球约 6000 万有症状的患者中,有一部分人的病情可能会逐渐减轻,甚至趋于稳定。社会正在进入这样一个时代:注意力缺失症的肆意破坏不再不可避免。本视角探讨了减缓注意力缺失症和其他神经退行性疾病对衰老轨迹的影响,使人们能够以较少的功能衰退存活到晚年。这一时刻对医学和社会的影响是深远的。淀粉样蛋白-β斑块清除抗体作为阿尔茨海默病的第一种疾病调节疗法的出现,将改变这种最常见的痴呆症的病程。相关进展将逐步改变人类衰老的轨迹。
{"title":"The advent of Alzheimer treatments will change the trajectory of human aging","authors":"Dennis J. Selkoe","doi":"10.1038/s43587-024-00611-5","DOIUrl":"10.1038/s43587-024-00611-5","url":null,"abstract":"Slowing neurodegenerative disorders of late life has lagged behind progress on other chronic diseases. But advances in two areas, biochemical pathology and human genetics, have now identified early pathogenic events, enabling molecular hypotheses and disease-modifying treatments. A salient example is the discovery that antibodies to amyloid ß-protein, long debated as a causative factor in Alzheimer’s disease (AD), clear amyloid plaques, decrease levels of abnormal tau proteins and slow cognitive decline. Approval of amyloid antibodies as the first disease-modifying treatments means a gradually rising fraction of the world’s estimated 60 million people with symptomatic disease may decline less or even stabilize. Society is entering an era in which the unchecked devastation of AD is no longer inevitable. This Perspective considers the impact of slowing AD and other neurodegenerative disorders on the trajectory of aging, allowing people to survive into late life with less functional decline. The implications of this moment for medicine and society are profound. The advent of plaque-clearing antibodies to the amyloid-β as the first disease-modifying treatment for Alzheimer’s disease will change the course of this disease, the most common type of dementia. Related progress will gradually alter the trajectory of human aging.","PeriodicalId":94150,"journal":{"name":"Nature aging","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140621395","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Aging-induced MCPH1 translocation activates necroptosis and impairs hematopoietic stem cell function 衰老诱导的 MCPH1 易位可激活坏死并损害造血干细胞功能
Pub Date : 2024-04-17 DOI: 10.1038/s43587-024-00609-z
Hanqing He, Yuqian Wang, Baixue Tang, Qiongye Dong, Chou Wu, Wanling Sun, Jianwei Wang
DNA damage contributes to the aging of hematopoietic stem cells (HSCs), yet the underlying molecular mechanisms are not fully understood. In this study, we identified a heterogeneous functional role of microcephalin (MCPH1) in the nucleus and cytoplasm of mouse HSCs. In the nucleus, MCPH1 maintains genomic stability, whereas in the cytoplasm, it prevents necroptosis by binding with p-RIPK3. Aging triggers MCPH1 translocation from cytosol to nucleus, reducing its cytoplasmic retention and leading to the activation of necroptosis and deterioration of HSC function. Mechanistically, we found that KAT7-mediated lysine acetylation within the NLS motif of MCPH1 in response to DNA damage facilitates its nuclear translocation. Targeted mutation of these lysines inhibits MCPH1 translocation and, consequently, compromises necroptosis. The dysfunction of necroptosis signaling, in turn, improves the function of aged HSCs. In summary, our findings demonstrate that DNA damage-induced redistribution of MCPH1 promotes HSC aging and could have broader implications for aging and aging-related diseases. He et al. characterizes a role of microcephalin (MCPH1), a known regulator of DNA damage response, in hematopoietic stem cell (HSC) aging demonstrating nuclear MCPH1 translocation that leads to activation of necroptosis and deterioration of HSC function with age.
DNA损伤导致造血干细胞(HSCs)衰老,但其潜在的分子机制尚未完全明了。在这项研究中,我们发现了小鼠造血干细胞的细胞核和细胞质中微头孢素(MCPH1)的异质性功能作用。在细胞核中,MCPH1维持基因组的稳定,而在细胞质中,它通过与p-RIPK3结合防止坏死。衰老会引发 MCPH1 从细胞质转位到细胞核,减少其在细胞质中的保留,导致坏死的激活和造血干细胞功能的退化。从机理上讲,我们发现在 DNA 损伤时,KAT7 介导的 MCPH1 NLS 矩阵内的赖氨酸乙酰化促进了其核转位。这些赖氨酸的靶向突变会抑制 MCPH1 的转位,从而影响坏死。坏死突变信号传导功能的失调反过来又会改善衰老造血干细胞的功能。总之,我们的研究结果表明,DNA损伤诱导的MCPH1重新分布促进了造血干细胞的衰老,并可能对衰老和衰老相关疾病产生更广泛的影响。
{"title":"Aging-induced MCPH1 translocation activates necroptosis and impairs hematopoietic stem cell function","authors":"Hanqing He, Yuqian Wang, Baixue Tang, Qiongye Dong, Chou Wu, Wanling Sun, Jianwei Wang","doi":"10.1038/s43587-024-00609-z","DOIUrl":"10.1038/s43587-024-00609-z","url":null,"abstract":"DNA damage contributes to the aging of hematopoietic stem cells (HSCs), yet the underlying molecular mechanisms are not fully understood. In this study, we identified a heterogeneous functional role of microcephalin (MCPH1) in the nucleus and cytoplasm of mouse HSCs. In the nucleus, MCPH1 maintains genomic stability, whereas in the cytoplasm, it prevents necroptosis by binding with p-RIPK3. Aging triggers MCPH1 translocation from cytosol to nucleus, reducing its cytoplasmic retention and leading to the activation of necroptosis and deterioration of HSC function. Mechanistically, we found that KAT7-mediated lysine acetylation within the NLS motif of MCPH1 in response to DNA damage facilitates its nuclear translocation. Targeted mutation of these lysines inhibits MCPH1 translocation and, consequently, compromises necroptosis. The dysfunction of necroptosis signaling, in turn, improves the function of aged HSCs. In summary, our findings demonstrate that DNA damage-induced redistribution of MCPH1 promotes HSC aging and could have broader implications for aging and aging-related diseases. He et al. characterizes a role of microcephalin (MCPH1), a known regulator of DNA damage response, in hematopoietic stem cell (HSC) aging demonstrating nuclear MCPH1 translocation that leads to activation of necroptosis and deterioration of HSC function with age.","PeriodicalId":94150,"journal":{"name":"Nature aging","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140614165","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Small extracellular vesicles from young plasma reverse age-related functional declines by improving mitochondrial energy metabolism 年轻血浆中的细胞外小泡可通过改善线粒体能量代谢逆转与年龄有关的功能衰退
IF 17 Pub Date : 2024-04-16 DOI: 10.1038/s43587-024-00612-4
Xiaorui Chen, Yang Luo, Qing Zhu, Jingzi Zhang, Huan Huang, Yansheng Kan, Dian Li, Ming Xu, Shuohan Liu, Jianxiao Li, Jinmeng Pan, Li Zhang, Yan Guo, Binghao Wang, Guantong Qi, Zhen Zhou, Chen-Yu Zhang, Lei Fang, Yanbo Wang, Xi Chen
Recent investigations into heterochronic parabiosis have unveiled robust rejuvenating effects of young blood on aged tissues. However, the specific rejuvenating mechanisms remain incompletely elucidated. Here we demonstrate that small extracellular vesicles (sEVs) from the plasma of young mice counteract pre-existing aging at molecular, mitochondrial, cellular and physiological levels. Intravenous injection of young sEVs into aged mice extends their lifespan, mitigates senescent phenotypes and ameliorates age-associated functional declines in multiple tissues. Quantitative proteomic analyses identified substantial alterations in the proteomes of aged tissues after young sEV treatment, and these changes are closely associated with metabolic processes. Mechanistic investigations reveal that young sEVs stimulate PGC-1α expression in vitro and in vivo through their miRNA cargoes, thereby improving mitochondrial functions and mitigating mitochondrial deficits in aged tissues. Overall, this study demonstrates that young sEVs reverse degenerative changes and age-related dysfunction, at least in part, by stimulating PGC-1α expression and enhancing mitochondrial energy metabolism. Circulating factors have an important role in aging. Here the authors show that small extracellular vesicles derived from young plasma rejuvenate whole-body physiology in aged mice, at least in part, by stimulating PGC-1α expression and improving mitochondrial energy metabolism.
最近对异种同种异体移植的研究揭示了年轻血液对衰老组织的强大恢复活力作用。然而,具体的年轻化机制仍未完全阐明。在这里,我们证明了年轻小鼠血浆中的小细胞外囊泡(sEVs)能在分子、线粒体、细胞和生理水平上抵消先前存在的衰老。向衰老小鼠静脉注射年轻的 sEVs 可延长其寿命、减轻衰老表型并改善多种组织中与年龄相关的功能衰退。定量蛋白质组分析发现,年轻 sEV 处理后,衰老组织的蛋白质组发生了重大变化,这些变化与新陈代谢过程密切相关。机理研究发现,年轻 sEV 通过其 miRNA 载体在体外和体内刺激 PGC-1α 的表达,从而改善线粒体功能并减轻衰老组织中线粒体的缺陷。总之,这项研究表明,年轻的 sEV 至少部分是通过刺激 PGC-1α 的表达和增强线粒体的能量代谢来逆转退行性变化和与年龄相关的功能障碍的。
{"title":"Small extracellular vesicles from young plasma reverse age-related functional declines by improving mitochondrial energy metabolism","authors":"Xiaorui Chen, Yang Luo, Qing Zhu, Jingzi Zhang, Huan Huang, Yansheng Kan, Dian Li, Ming Xu, Shuohan Liu, Jianxiao Li, Jinmeng Pan, Li Zhang, Yan Guo, Binghao Wang, Guantong Qi, Zhen Zhou, Chen-Yu Zhang, Lei Fang, Yanbo Wang, Xi Chen","doi":"10.1038/s43587-024-00612-4","DOIUrl":"10.1038/s43587-024-00612-4","url":null,"abstract":"Recent investigations into heterochronic parabiosis have unveiled robust rejuvenating effects of young blood on aged tissues. However, the specific rejuvenating mechanisms remain incompletely elucidated. Here we demonstrate that small extracellular vesicles (sEVs) from the plasma of young mice counteract pre-existing aging at molecular, mitochondrial, cellular and physiological levels. Intravenous injection of young sEVs into aged mice extends their lifespan, mitigates senescent phenotypes and ameliorates age-associated functional declines in multiple tissues. Quantitative proteomic analyses identified substantial alterations in the proteomes of aged tissues after young sEV treatment, and these changes are closely associated with metabolic processes. Mechanistic investigations reveal that young sEVs stimulate PGC-1α expression in vitro and in vivo through their miRNA cargoes, thereby improving mitochondrial functions and mitigating mitochondrial deficits in aged tissues. Overall, this study demonstrates that young sEVs reverse degenerative changes and age-related dysfunction, at least in part, by stimulating PGC-1α expression and enhancing mitochondrial energy metabolism. Circulating factors have an important role in aging. Here the authors show that small extracellular vesicles derived from young plasma rejuvenate whole-body physiology in aged mice, at least in part, by stimulating PGC-1α expression and improving mitochondrial energy metabolism.","PeriodicalId":94150,"journal":{"name":"Nature aging","volume":null,"pages":null},"PeriodicalIF":17.0,"publicationDate":"2024-04-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s43587-024-00612-4.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140579174","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Human skeletal muscle aging atlas 人类骨骼肌老化图谱
Pub Date : 2024-04-15 DOI: 10.1038/s43587-024-00613-3
Veronika R. Kedlian, Yaning Wang, Tianliang Liu, Xiaoping Chen, Liam Bolt, Catherine Tudor, Zhuojian Shen, Eirini S. Fasouli, Elena Prigmore, Vitalii Kleshchevnikov, Jan Patrick Pett, Tong Li, John E. G. Lawrence, Shani Perera, Martin Prete, Ni Huang, Qin Guo, Xinrui Zeng, Lu Yang, Krzysztof Polański, Nana-Jane Chipampe, Monika Dabrowska, Xiaobo Li, Omer Ali Bayraktar, Minal Patel, Natsuhiko Kumasaka, Krishnaa T. Mahbubani, Andy Peng Xiang, Kerstin B. Meyer, Kourosh Saeb-Parsy, Sarah A. Teichmann, Hongbo Zhang
Skeletal muscle aging is a key contributor to age-related frailty and sarcopenia with substantial implications for global health. Here we profiled 90,902 single cells and 92,259 single nuclei from 17 donors to map the aging process in the adult human intercostal muscle, identifying cellular changes in each muscle compartment. We found that distinct subsets of muscle stem cells exhibit decreased ribosome biogenesis genes and increased CCL2 expression, causing different aging phenotypes. Our atlas also highlights an expansion of nuclei associated with the neuromuscular junction, which may reflect re-innervation, and outlines how the loss of fast-twitch myofibers is mitigated through regeneration and upregulation of fast-type markers in slow-twitch myofibers with age. Furthermore, we document the function of aging muscle microenvironment in immune cell attraction. Overall, we present a comprehensive human skeletal muscle aging resource ( https://www.muscleageingcellatlas.org/ ) together with an in-house mouse muscle atlas to study common features of muscle aging across species. The Muscle Aging Cell Atlas presents approximately 200,000 single-cell and single-nuclei transcriptomes from 17 human donors across different ages, uncovering mechanisms of aging in muscle stem cells, myofibers and microenvironment cells, and demonstrates parallels in mouse muscle aging.
骨骼肌衰老是造成与年龄相关的虚弱和肌肉疏松症的关键因素,对全球健康具有重大影响。在这里,我们对来自 17 位供体的 90,902 个单细胞和 92,259 个单个细胞核进行了分析,以绘制成年人类肋间肌的衰老过程图,确定每个肌肉区的细胞变化。我们发现,不同的肌肉干细胞亚群表现出核糖体生物生成基因减少和CCL2表达增加,从而导致不同的衰老表型。我们的图谱还突出显示了与神经肌肉接头相关的细胞核的扩大,这可能反映了神经支配的再支配,并概述了随着年龄的增长,快肌肌纤维的损失是如何通过慢肌肌纤维的再生和快型标志物的上调而得到缓解的。此外,我们还记录了衰老肌肉微环境在吸引免疫细胞方面的功能。总之,我们提供了一个全面的人类骨骼肌衰老资源(https://www.muscleageingcellatlas.org/)和一个内部小鼠肌肉图谱,用于研究不同物种肌肉衰老的共同特征。
{"title":"Human skeletal muscle aging atlas","authors":"Veronika R. Kedlian, Yaning Wang, Tianliang Liu, Xiaoping Chen, Liam Bolt, Catherine Tudor, Zhuojian Shen, Eirini S. Fasouli, Elena Prigmore, Vitalii Kleshchevnikov, Jan Patrick Pett, Tong Li, John E. G. Lawrence, Shani Perera, Martin Prete, Ni Huang, Qin Guo, Xinrui Zeng, Lu Yang, Krzysztof Polański, Nana-Jane Chipampe, Monika Dabrowska, Xiaobo Li, Omer Ali Bayraktar, Minal Patel, Natsuhiko Kumasaka, Krishnaa T. Mahbubani, Andy Peng Xiang, Kerstin B. Meyer, Kourosh Saeb-Parsy, Sarah A. Teichmann, Hongbo Zhang","doi":"10.1038/s43587-024-00613-3","DOIUrl":"10.1038/s43587-024-00613-3","url":null,"abstract":"Skeletal muscle aging is a key contributor to age-related frailty and sarcopenia with substantial implications for global health. Here we profiled 90,902 single cells and 92,259 single nuclei from 17 donors to map the aging process in the adult human intercostal muscle, identifying cellular changes in each muscle compartment. We found that distinct subsets of muscle stem cells exhibit decreased ribosome biogenesis genes and increased CCL2 expression, causing different aging phenotypes. Our atlas also highlights an expansion of nuclei associated with the neuromuscular junction, which may reflect re-innervation, and outlines how the loss of fast-twitch myofibers is mitigated through regeneration and upregulation of fast-type markers in slow-twitch myofibers with age. Furthermore, we document the function of aging muscle microenvironment in immune cell attraction. Overall, we present a comprehensive human skeletal muscle aging resource ( https://www.muscleageingcellatlas.org/ ) together with an in-house mouse muscle atlas to study common features of muscle aging across species. The Muscle Aging Cell Atlas presents approximately 200,000 single-cell and single-nuclei transcriptomes from 17 human donors across different ages, uncovering mechanisms of aging in muscle stem cells, myofibers and microenvironment cells, and demonstrates parallels in mouse muscle aging.","PeriodicalId":94150,"journal":{"name":"Nature aging","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s43587-024-00613-3.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140579201","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
FOXP1 is a gatekeeper of cellular senescence with ovarian aging FOXP1 是卵巢衰老过程中细胞衰老的看门人
Pub Date : 2024-04-15 DOI: 10.1038/s43587-024-00608-0
Our analysis of the spatiotemporal transcriptional features of human ovarian aging at the single-cell level identified the DNA damage response as a fundamental attribute in oocyte senescence. FOXP1, a gatekeeper both in granulosa and in theca and stroma cellular senescence, can be activated by quercetin treatment to delay ovarian aging.
我们在单细胞水平上分析了人类卵巢衰老的时空转录特征,发现DNA损伤反应是卵母细胞衰老的基本属性。FOXP1是颗粒细胞和卵巢及基质细胞衰老的看门人,它可被槲皮素治疗激活,从而延缓卵巢衰老。
{"title":"FOXP1 is a gatekeeper of cellular senescence with ovarian aging","authors":"","doi":"10.1038/s43587-024-00608-0","DOIUrl":"10.1038/s43587-024-00608-0","url":null,"abstract":"Our analysis of the spatiotemporal transcriptional features of human ovarian aging at the single-cell level identified the DNA damage response as a fundamental attribute in oocyte senescence. FOXP1, a gatekeeper both in granulosa and in theca and stroma cellular senescence, can be activated by quercetin treatment to delay ovarian aging.","PeriodicalId":94150,"journal":{"name":"Nature aging","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140579175","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting aging and age-related diseases with vaccines 用疫苗防治衰老和老年相关疾病
Pub Date : 2024-04-15 DOI: 10.1038/s43587-024-00597-0
Ruochen Wu, Fei Sun, Weiqi Zhang, Jie Ren, Guang-Hui Liu
Aging is a major risk factor for numerous chronic diseases. Vaccination offers a promising strategy to combat these age-related diseases by targeting specific antigens and inducing immune responses. Here, we provide a comprehensive overview of recent advances in vaccine-based interventions targeting these diseases, including Alzheimer’s disease, type II diabetes, hypertension, abdominal aortic aneurysm, atherosclerosis, osteoarthritis, fibrosis and cancer, summarizing current approaches for identifying disease-associated antigens and inducing immune responses against these targets. Further, we reflect on the recent development of vaccines targeting senescent cells, as a strategy for more broadly targeting underlying causes of aging and associated pathologies. In addition to highlighting recent progress in these areas, we discuss important next steps to advance the therapeutic potential of these vaccines, including improving and robustly demonstrating efficacy in human clinical trials, as well as rigorously evaluating the safety and long-term effects of these vaccine strategies. Wu et al. explore vaccine strategies targeting age-related diseases, as well as senescent cells specifically, as potential underlying drivers of aging itself. They discuss challenges faced in clinical trials, as well as further optimizations required to increase therapeutic efficacy.
衰老是许多慢性疾病的主要风险因素。疫苗接种通过靶向特定抗原和诱导免疫反应,为防治这些与年龄有关的疾病提供了一种前景广阔的策略。在此,我们全面概述了针对这些疾病(包括阿尔茨海默病、II 型糖尿病、高血压、腹主动脉瘤、动脉粥样硬化、骨关节炎、纤维化和癌症)的疫苗干预的最新进展,总结了目前识别疾病相关抗原和诱导针对这些靶点的免疫反应的方法。此外,我们还反思了针对衰老细胞的疫苗的最新发展,这是一种更广泛地针对衰老和相关病症的根本原因的策略。除了重点介绍这些领域的最新进展外,我们还讨论了推进这些疫苗治疗潜力的下一步重要措施,包括在人体临床试验中改进和有力地证明疗效,以及严格评估这些疫苗策略的安全性和长期效果。
{"title":"Targeting aging and age-related diseases with vaccines","authors":"Ruochen Wu, Fei Sun, Weiqi Zhang, Jie Ren, Guang-Hui Liu","doi":"10.1038/s43587-024-00597-0","DOIUrl":"10.1038/s43587-024-00597-0","url":null,"abstract":"Aging is a major risk factor for numerous chronic diseases. Vaccination offers a promising strategy to combat these age-related diseases by targeting specific antigens and inducing immune responses. Here, we provide a comprehensive overview of recent advances in vaccine-based interventions targeting these diseases, including Alzheimer’s disease, type II diabetes, hypertension, abdominal aortic aneurysm, atherosclerosis, osteoarthritis, fibrosis and cancer, summarizing current approaches for identifying disease-associated antigens and inducing immune responses against these targets. Further, we reflect on the recent development of vaccines targeting senescent cells, as a strategy for more broadly targeting underlying causes of aging and associated pathologies. In addition to highlighting recent progress in these areas, we discuss important next steps to advance the therapeutic potential of these vaccines, including improving and robustly demonstrating efficacy in human clinical trials, as well as rigorously evaluating the safety and long-term effects of these vaccine strategies. Wu et al. explore vaccine strategies targeting age-related diseases, as well as senescent cells specifically, as potential underlying drivers of aging itself. They discuss challenges faced in clinical trials, as well as further optimizations required to increase therapeutic efficacy.","PeriodicalId":94150,"journal":{"name":"Nature aging","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140579178","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A lipidome landscape of aging in mice 小鼠衰老的脂质体图谱
Pub Date : 2024-04-12 DOI: 10.1038/s43587-024-00610-6
Hiroshi Tsugawa, Tomoaki Ishihara, Kota Ogasa, Seigo Iwanami, Aya Hori, Mikiko Takahashi, Yutaka Yamada, Naoko Satoh-Takayama, Hiroshi Ohno, Aki Minoda, Makoto Arita
Understanding the molecular mechanisms of aging is crucial for enhancing healthy longevity. We conducted untargeted lipidomics across 13 biological samples from mice at various life stages (2, 12, 19 and 24 months) to explore the potential link between aging and lipid metabolism, considering sex (male or female) and microbiome (specific pathogen-free or germ-free) dependencies. By analyzing 2,704 molecules from 109 lipid subclasses, we characterized common and tissue-specific lipidome alterations associated with aging. For example, the levels of bis(monoacylglycero)phosphate containing polyunsaturated fatty acids increased in various organs during aging, whereas the levels of other phospholipids containing saturated and monounsaturated fatty acids decreased. In addition, we discovered age-dependent sulfonolipid accumulation, absent in germ-free mice, correlating with Alistipes abundance determined by 16S ribosomal RNA gene amplicon sequencing. In the male kidney, glycolipids such as galactosylceramides, galabiosylceramides (Gal2Cer), trihexosylceramides (Hex3Cer), and mono- and digalactosyldiacylglycerols were detected, with two lipid classes—Gal2Cer and Hex3Cer—being significantly enriched in aged mice. Integrated analysis of the kidney transcriptome revealed uridine diphosphate galactosyltransferase 8A (UGT8a), alkylglycerone phosphate synthase and fatty acyl-coenzyme A reductase 1 as potential enzymes responsible for the male-specific glycolipid biosynthesis in vivo, which would be relevant to sex dependency in kidney diseases. Inhibiting UGT8 reduced the levels of these glycolipids and the expression of inflammatory cytokines in the kidney. Our study provides a valuable resource for clarifying potential links between lipid metabolism and aging. Lipid changes across the lifespan and their role in health and longevity are incompletely understood. Here, Tsugawa and colleagues conduct untargeted lipidomics across 13 sample types and four ages in mice, considering sex and microbiome dependencies. This study provides a comprehensive resource of lipid changes with aging and highlights regulatory metabolic components, such as the enzyme UGT8, as potentially responsible for male-specific glycolipid biosynthesis in the kidney.
了解衰老的分子机制对于提高健康长寿至关重要。我们对小鼠不同生命阶段(2、12、19 和 24 个月)的 13 份生物样本进行了非靶向脂质组学研究,以探索衰老与脂质代谢之间的潜在联系,同时考虑到性别(雄性或雌性)和微生物组(特定无病原体或无病菌)的依赖性。通过分析 109 种脂质亚类中的 2,704 个分子,我们确定了与衰老相关的常见和组织特异性脂质体改变的特征。例如,在衰老过程中,各器官中含有多不饱和脂肪酸的磷酸二(单酰基甘油)酯的含量增加,而含有饱和脂肪酸和单不饱和脂肪酸的其他磷脂的含量则下降。此外,我们还发现了无菌小鼠体内不存在的年龄依赖性磺脂积累,这与 16S 核糖体 RNA 基因扩增片段测序确定的 Alistipes 丰度相关。在雄性小鼠的肾脏中,检测到了半乳糖基甘油酰胺、半生物基甘油酰胺(Gal2Cer)、三己基甘油酰胺(Hex3Cer)、单半乳糖基和双半乳糖基二乙酰甘油等糖脂,其中两类脂质--Gal2Cer和Hex3Cer--在老龄小鼠中显著富集。肾脏转录组的综合分析表明,二磷酸尿苷半乳糖基转移酶8A(UGT8a)、磷酸烷基甘油酯合成酶和脂肪酰辅酶A还原酶1是体内负责雄性特异性糖脂生物合成的潜在酶,这与肾脏疾病中的性别依赖有关。抑制 UGT8 可降低肾脏中这些糖脂的水平和炎症细胞因子的表达。我们的研究为阐明脂质代谢与衰老之间的潜在联系提供了宝贵的资料。
{"title":"A lipidome landscape of aging in mice","authors":"Hiroshi Tsugawa, Tomoaki Ishihara, Kota Ogasa, Seigo Iwanami, Aya Hori, Mikiko Takahashi, Yutaka Yamada, Naoko Satoh-Takayama, Hiroshi Ohno, Aki Minoda, Makoto Arita","doi":"10.1038/s43587-024-00610-6","DOIUrl":"10.1038/s43587-024-00610-6","url":null,"abstract":"Understanding the molecular mechanisms of aging is crucial for enhancing healthy longevity. We conducted untargeted lipidomics across 13 biological samples from mice at various life stages (2, 12, 19 and 24 months) to explore the potential link between aging and lipid metabolism, considering sex (male or female) and microbiome (specific pathogen-free or germ-free) dependencies. By analyzing 2,704 molecules from 109 lipid subclasses, we characterized common and tissue-specific lipidome alterations associated with aging. For example, the levels of bis(monoacylglycero)phosphate containing polyunsaturated fatty acids increased in various organs during aging, whereas the levels of other phospholipids containing saturated and monounsaturated fatty acids decreased. In addition, we discovered age-dependent sulfonolipid accumulation, absent in germ-free mice, correlating with Alistipes abundance determined by 16S ribosomal RNA gene amplicon sequencing. In the male kidney, glycolipids such as galactosylceramides, galabiosylceramides (Gal2Cer), trihexosylceramides (Hex3Cer), and mono- and digalactosyldiacylglycerols were detected, with two lipid classes—Gal2Cer and Hex3Cer—being significantly enriched in aged mice. Integrated analysis of the kidney transcriptome revealed uridine diphosphate galactosyltransferase 8A (UGT8a), alkylglycerone phosphate synthase and fatty acyl-coenzyme A reductase 1 as potential enzymes responsible for the male-specific glycolipid biosynthesis in vivo, which would be relevant to sex dependency in kidney diseases. Inhibiting UGT8 reduced the levels of these glycolipids and the expression of inflammatory cytokines in the kidney. Our study provides a valuable resource for clarifying potential links between lipid metabolism and aging. Lipid changes across the lifespan and their role in health and longevity are incompletely understood. Here, Tsugawa and colleagues conduct untargeted lipidomics across 13 sample types and four ages in mice, considering sex and microbiome dependencies. This study provides a comprehensive resource of lipid changes with aging and highlights regulatory metabolic components, such as the enzyme UGT8, as potentially responsible for male-specific glycolipid biosynthesis in the kidney.","PeriodicalId":94150,"journal":{"name":"Nature aging","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140579268","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A conserved complex lipid signature marks human muscle aging and responds to short-term exercise 一种保守的复合脂质特征标志着人类肌肉老化并对短期运动做出反应
Pub Date : 2024-04-12 DOI: 10.1038/s43587-024-00595-2
Georges E. Janssens, Marte Molenaars, Katharina Herzog, Lotte Grevendonk, Carlijn M. E. Remie, Martin A. T. Vervaart, Hyung L. Elfrink, Eric J. M. Wever, Bauke V. Schomakers, Simone W. Denis, Hans R. Waterham, Mia L. Pras-Raves, Michel van Weeghel, Antoine H. C. van Kampen, Alessandra Tammaro, Loes M. Butter, Sanne van der Rijt, Sandrine Florquin, Aldo Jongejan, Perry D. Moerland, Joris Hoeks, Patrick Schrauwen, Frédéric M. Vaz, Riekelt H. Houtkooper
Studies in preclinical models suggest that complex lipids, such as phospholipids, play a role in the regulation of longevity. However, identification of universally conserved complex lipid changes that occur during aging, and how these respond to interventions, is lacking. Here, to comprehensively map how complex lipids change during aging, we profiled ten tissues in young versus aged mice using a lipidomics platform. Strikingly, from >1,200 unique lipids, we found a tissue-wide accumulation of bis(monoacylglycero)phosphate (BMP) during mouse aging. To investigate translational value, we assessed muscle tissue of young and older people, and found a similar marked BMP accumulation in the human aging lipidome. Furthermore, we found that a healthy-aging intervention consisting of moderate-to-vigorous exercise was able to lower BMP levels in postmenopausal female research participants. Our work implicates complex lipid biology as central to aging, identifying a conserved aging lipid signature of BMP accumulation that is modifiable upon a short-term healthy-aging intervention. Aging dynamics of complex lipids are incompletely understood. Here Janssens and colleagues describe lipids that change with age across ten tissues in mice. Notably, bis(monoacylglycerol)phosphate accumulated with age. This lipid also accumulated in muscle of older humans, and reduced upon a short bout of exercise.
对临床前模型的研究表明,磷脂等复合脂质在调节长寿方面发挥着作用。然而,目前还缺乏对衰老过程中发生的普遍一致的复合脂质变化以及这些变化如何对干预措施做出反应的鉴定。在这里,为了全面描绘复杂脂质在衰老过程中的变化,我们利用脂质组学平台分析了年轻小鼠和衰老小鼠的十种组织。令人震惊的是,我们从1200种独特的脂质中发现,在小鼠衰老过程中,双(单酰基甘油)磷酸酯(BMP)会在整个组织中积累。为了研究其转化价值,我们对年轻人和老年人的肌肉组织进行了评估,发现在人类衰老脂质组中也有类似的明显 BMP 累积。此外,我们还发现,由中度到剧烈运动组成的健康老龄化干预措施能够降低绝经后女性研究参与者体内的 BMP 水平。我们的工作表明,复杂的脂质生物学是衰老的核心,我们发现了一种保守的 BMP 积累的衰老脂质特征,这种特征在短期的健康衰老干预措施下是可以改变的。
{"title":"A conserved complex lipid signature marks human muscle aging and responds to short-term exercise","authors":"Georges E. Janssens, Marte Molenaars, Katharina Herzog, Lotte Grevendonk, Carlijn M. E. Remie, Martin A. T. Vervaart, Hyung L. Elfrink, Eric J. M. Wever, Bauke V. Schomakers, Simone W. Denis, Hans R. Waterham, Mia L. Pras-Raves, Michel van Weeghel, Antoine H. C. van Kampen, Alessandra Tammaro, Loes M. Butter, Sanne van der Rijt, Sandrine Florquin, Aldo Jongejan, Perry D. Moerland, Joris Hoeks, Patrick Schrauwen, Frédéric M. Vaz, Riekelt H. Houtkooper","doi":"10.1038/s43587-024-00595-2","DOIUrl":"10.1038/s43587-024-00595-2","url":null,"abstract":"Studies in preclinical models suggest that complex lipids, such as phospholipids, play a role in the regulation of longevity. However, identification of universally conserved complex lipid changes that occur during aging, and how these respond to interventions, is lacking. Here, to comprehensively map how complex lipids change during aging, we profiled ten tissues in young versus aged mice using a lipidomics platform. Strikingly, from >1,200 unique lipids, we found a tissue-wide accumulation of bis(monoacylglycero)phosphate (BMP) during mouse aging. To investigate translational value, we assessed muscle tissue of young and older people, and found a similar marked BMP accumulation in the human aging lipidome. Furthermore, we found that a healthy-aging intervention consisting of moderate-to-vigorous exercise was able to lower BMP levels in postmenopausal female research participants. Our work implicates complex lipid biology as central to aging, identifying a conserved aging lipid signature of BMP accumulation that is modifiable upon a short-term healthy-aging intervention. Aging dynamics of complex lipids are incompletely understood. Here Janssens and colleagues describe lipids that change with age across ten tissues in mice. Notably, bis(monoacylglycerol)phosphate accumulated with age. This lipid also accumulated in muscle of older humans, and reduced upon a short bout of exercise.","PeriodicalId":94150,"journal":{"name":"Nature aging","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140579198","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Spatiotemporal transcriptomic changes of human ovarian aging and the regulatory role of FOXP1 人类卵巢衰老的时空转录组变化及 FOXP1 的调控作用
Pub Date : 2024-04-09 DOI: 10.1038/s43587-024-00607-1
Meng Wu, Weicheng Tang, Ying Chen, Liru Xue, Jun Dai, Yan Li, Xiaoran Zhu, Chuqing Wu, Jiaqiang Xiong, Jinjin Zhang, Tong Wu, Su Zhou, Dan Chen, Chaoyang Sun, Jing Yu, Hongyi Li, Yican Guo, Yibao Huang, Qingqing Zhu, Simin Wei, Ziliang Zhou, Mingfu Wu, Ya Li, Tao Xiang, Huiying Qiao, Shixuan Wang
Limited understanding exists regarding how aging impacts the cellular and molecular aspects of the human ovary. This study combines single-cell RNA sequencing and spatial transcriptomics to systematically characterize human ovarian aging. Spatiotemporal molecular signatures of the eight types of ovarian cells during aging are observed. An analysis of age-associated changes in gene expression reveals that DNA damage response may be a key biological pathway in oocyte aging. Three granulosa cells subtypes and five theca and stromal cells subtypes, as well as their spatiotemporal transcriptomics changes during aging, are identified. FOXP1 emerges as a regulator of ovarian aging, declining with age and inhibiting CDKN1A transcription. Silencing FOXP1 results in premature ovarian insufficiency in mice. These findings offer a comprehensive understanding of spatiotemporal variability in human ovarian aging, aiding the prioritization of potential diagnostic biomarkers and therapeutic strategies. Ovarian aging has an important role in health and fertility; however, the molecular mechanisms underlying it remain incompletely understood. Here the authors use single-cell and spatial transcriptomics in reproductively young, middle-aged and older human ovarian tissue to elucidate ovarian aging. They describe spatiotemporal changes in ovarian cells and highlight the important regulatory role of FOXP1.
人们对衰老如何影响人类卵巢的细胞和分子方面的了解有限。这项研究结合了单细胞 RNA 测序和空间转录组学,系统地描述了人类卵巢衰老的特征。研究观察了衰老过程中八种卵巢细胞的时空分子特征。对与年龄相关的基因表达变化的分析表明,DNA损伤反应可能是卵母细胞衰老的一个关键生物学途径。确定了三种颗粒细胞亚型和五种透明带及基质细胞亚型,以及它们在衰老过程中的时空转录组学变化。FOXP1是卵巢衰老的调节因子,随着年龄的增长而下降,并抑制CDKN1A的转录。沉默 FOXP1 会导致小鼠卵巢早衰。这些发现使人们对人类卵巢衰老的时空变异有了全面的了解,有助于确定潜在诊断生物标志物和治疗策略的优先次序。
{"title":"Spatiotemporal transcriptomic changes of human ovarian aging and the regulatory role of FOXP1","authors":"Meng Wu, Weicheng Tang, Ying Chen, Liru Xue, Jun Dai, Yan Li, Xiaoran Zhu, Chuqing Wu, Jiaqiang Xiong, Jinjin Zhang, Tong Wu, Su Zhou, Dan Chen, Chaoyang Sun, Jing Yu, Hongyi Li, Yican Guo, Yibao Huang, Qingqing Zhu, Simin Wei, Ziliang Zhou, Mingfu Wu, Ya Li, Tao Xiang, Huiying Qiao, Shixuan Wang","doi":"10.1038/s43587-024-00607-1","DOIUrl":"10.1038/s43587-024-00607-1","url":null,"abstract":"Limited understanding exists regarding how aging impacts the cellular and molecular aspects of the human ovary. This study combines single-cell RNA sequencing and spatial transcriptomics to systematically characterize human ovarian aging. Spatiotemporal molecular signatures of the eight types of ovarian cells during aging are observed. An analysis of age-associated changes in gene expression reveals that DNA damage response may be a key biological pathway in oocyte aging. Three granulosa cells subtypes and five theca and stromal cells subtypes, as well as their spatiotemporal transcriptomics changes during aging, are identified. FOXP1 emerges as a regulator of ovarian aging, declining with age and inhibiting CDKN1A transcription. Silencing FOXP1 results in premature ovarian insufficiency in mice. These findings offer a comprehensive understanding of spatiotemporal variability in human ovarian aging, aiding the prioritization of potential diagnostic biomarkers and therapeutic strategies. Ovarian aging has an important role in health and fertility; however, the molecular mechanisms underlying it remain incompletely understood. Here the authors use single-cell and spatial transcriptomics in reproductively young, middle-aged and older human ovarian tissue to elucidate ovarian aging. They describe spatiotemporal changes in ovarian cells and highlight the important regulatory role of FOXP1.","PeriodicalId":94150,"journal":{"name":"Nature aging","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s43587-024-00607-1.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140579195","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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
Nature aging
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