%A Li,Yinchuan %A Mi,Panpan %A Wu,Jiabao %A Tang,Yunge %A Liu,Xiaohua %A Cheng,Jinmei %A Huang,Yingying %A Qin,Weibing %A Cheng,C. Yan %A Sun,Fei %D 2022 %J Frontiers in Immunology %C %F %G English %K ScRNA-seq,Leydig Cells,senescence,Androgen synthesis,Cytokines,experimental autoimmune orchitis (EAO),Glutathione metabolism,Fibrosis,complement %Q %R 10.3389/fimmu.2021.771373 %W %L %M %P %7 %8 2022-January-17 %9 Original Research %+ Weibing Qin,NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital),China,sunfei@ntu.edu.cn %+ C. Yan Cheng,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council,United States,sunfei@ntu.edu.cn %+ Fei Sun,Institute of Reproductive Medicine, Medical School of Nantong University,China,sunfei@ntu.edu.cn %# %! Leydig cell senescence and EAO %* %< %T High Throughput scRNA-Seq Provides Insights Into Leydig Cell Senescence Induced by Experimental Autoimmune Orchitis: A Prominent Role of Interstitial Fibrosis and Complement Activation %U https://www.frontiersin.org/articles/10.3389/fimmu.2021.771373 %V 12 %0 JOURNAL ARTICLE %@ 1664-3224 %X Leydig cells (Lc), located in the interstitial space of the testis between seminiferous tubules, produce 95% of testosterone in male individuals, which is pivotal for male sexual differentiation, spermatogenesis, and maintenance of the male secondary sex characteristics. Lc are prone to senescence in aging testes, resulting in compromised androgen synthesis capability upon aging. However, little is known about whether Lc undergo senescence in a chronic inflammatory environment. To investigate this question, mouse models of experimental autoimmune orchitis (EAO) were used, and Lc were analyzed by high throughput scRNA-Seq. Data were screened and analyzed by correlating signaling pathways with senescence, apoptosis, androgen synthesis, and cytokine/chemokine signaling pathways. EAO did induce Lc senescence, and Lc senescence in turn antagonized androgen synthesis. Based on the correlation screening of pathways inducing Lc senescence, a plethora of pathways were found to play potential roles in triggering Lc senescence during EAO, among which the Arf6 and angiopoietin receptor pathways were highly correlated with senescence signature. Notably, complement and interstitial fibrosis activated by EAO worsened Lc senescence and strongly antagonized androgen synthesis. Furthermore, most proinflammatory cytokines enhanced both senescence and apoptosis in Lc and spermatogonia (Sg) during EAO, and proinflammatory cytokine antagonism of the glutathione metabolism pathway may be key in inducing cellular senescence during EAO.