Volume 13 Issue 8
Aug.  2023
Turn off MathJax
Article Contents
Wei Zhang, Siyu Xia, Jinhuan Ou, Min Cao, Guangqing Cheng, Zhijie Li, Jigang Wang, Chuanbin Yang. A single-cell landscape of triptolide-associated testicular toxicity in mice[J]. Journal of Pharmaceutical Analysis, 2023, 13(8): 880-893. doi: 10.1016/j.jpha.2023.04.006
Citation: Wei Zhang, Siyu Xia, Jinhuan Ou, Min Cao, Guangqing Cheng, Zhijie Li, Jigang Wang, Chuanbin Yang. A single-cell landscape of triptolide-associated testicular toxicity in mice[J]. Journal of Pharmaceutical Analysis, 2023, 13(8): 880-893. doi: 10.1016/j.jpha.2023.04.006

A single-cell landscape of triptolide-associated testicular toxicity in mice

doi: 10.1016/j.jpha.2023.04.006
Funds:

The work was supported by grants from the National Key Research and Development Program of China (Grant Nos.: 2020YFA0908000 and 2022YFC2303600), the Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine (Grant No.: ZYYCXTD-C-202002), the National Natural Science Foundation of China (Grant Nos.: 82201786, 82141001, 82274182, 82074098 and 82173914), the CACMS Innovation Fund (Grant Nos.: CI2021A05101 and CI2021A05104), the Scientific and Technological Innovation Project of China Academy of Chinese Medical Sciences (Grant No.: CI2021B014), the Science and Technology Foundation of Shenzhen (Grant Nos.: JCYJ20220818102613029, JCYJ20210324114014039, and JCYJ20210324115800001), Guangdong Basic and Applied Basic Research Foundation (Grant Nos.: 2020A1515110549, 2021A1515110646), the Science and Technology Foundation of Shenzhen (Shenzhen Clinical Medical Research Center for Geriatric Diseases), the National Key R&D Program of China Key projects for international cooperation on science, technology and innovation (Grant No.: 2020YFE0205100), and the Fundamental Research Funds for the Central Public Welfare Research Institutes (Grant Nos.: ZZ14-YQ-050, ZZ14-YQ-051, ZZ14-YQ-052, ZZ14-FL-002, ZZ14-ND-010 and ZZ15-ND-10).

  • Received Date: Dec. 28, 2022
  • Rev Recd Date: Apr. 09, 2023
  • Available Online: Aug. 29, 2023
  • Triptolide is a key active component of the widely used traditional Chinese herb medicine Tripterygium wilfordii Hook. F. Although triptolide exerts multiple biological activities and shows promising efficacy in treating inflammatory-related diseases, its well-known safety issues, especially reproductive toxicity has aroused concerns. However, a comprehensive dissection of triptolide-associated testicular toxicity at single cell resolution is still lacking. Here, we observed testicular toxicity after 14 days of triptolide exposure, and then constructed a single-cell transcriptome map of 59,127 cells in mouse testes upon triptolide-treatment. We identified triptolide-associated shared and cell-type specific differentially expressed genes, enriched pathways, and ligand-receptor pairs in different cell types of mouse testes. In addition to the loss of germ cells, our results revealed increased macrophages and the inflammatory response in triptolide-treated mouse testes, suggesting a critical role of inflammation in triptolide-induced testicular injury. We also found increased reactive oxygen species (ROS) signaling and downregulated pathways associated with spermatid development in somatic cells, especially Leydig and Sertoli cells, in triptolide-treated mice, indicating that dysregulation of these signaling pathways may contribute to triptolide-induced testicular toxicity. Overall, our high-resolution single-cell landscape offers comprehensive information regarding triptolide-associated gene expression profiles in major cell types of mouse testes at single cell resolution, providing an invaluable resource for understanding the underlying mechanism of triptolide-associated testicular injury and additional discoveries of therapeutic targets of triptolide-induced male reproductive toxicity.
  • loading
  • M. Vander Borght, C. Wyns. Fertility and infertility: Definition and epidemiology, Clin. Biochem. 62 (2018) 2-10.
    A. Agarwal, S. Baskaran, N. Parekh, et al., Male infertility, Lancet. 397 (2021) 319-333.
    N. E. Skakkebaek, E. Rajpert-De Meyts, K. M. Main. Testicular dysgenesis syndrome: an increasingly common developmental disorder with environmental aspects, Hum. Reprod. 16 (2001) 972-978.
    M. Semet, M. Paci, J. Saias-Magnan, et al., The impact of drugs on male fertility: a review, Andrology. 5 (2017) 640-663.
    Y. Liu, X. He, Y. Wang, et al., Aristolochic acid I induces impairment in spermatogonial stem cell in rodents. Toxicol. Res. 10 (2021) 436-445.
    Y. Zhang, X. Mao, W. Li, et al., Tripterygium wilfordii: An inspiring resource for rheumatoid arthritis treatment, Med. Res. Rev. 41 (2021) 1337-1374.
    G. Xun, Y. Tian, Y. Gao, et al., Identification and comparison of compounds in commercial Tripterygium wilfordii genus preparations with HPLC-QTOF/MS based on molecular networking and multivariate statistical analysis, J. Pharm. Biomed. Anal. 216 (2022), 114811.
    J. Gao, Y. Zhang, X. Liu, et al., Triptolide: pharmacological spectrum, biosynthesis, chemical synthesis and derivatives, Theranostics. 11 (2021) 7199-7221.
    Y. Liu, F. Song, W. K. Wu, et al., Triptolide inhibits colon cancer cell proliferation and induces cleavage and translocation of 14-3-3 epsilon, Cell. Biochem. Funct. 30 (2012) 271-278.
    M. Lai, L. Liu, L. Zhu, et al., Triptolide reverses epithelial-mesenchymal transition in glioma cells via inducing autophagy, Ann. Transl. Med. 9 (2021), 1304.
    K. Li, P. Gao, P. Xiang, et al., Molecular mechanisms of PFOA-induced toxicity in animals and humans: Implications for health risks, Environ. Int. 99 (2017) 43-54.
    Y. Wang, S. H. Guo, X. J. Shang, et al., Triptolide induces Sertoli cell apoptosis in mice via ROS/JNK-dependent activation of the mitochondrial pathway and inhibition of Nrf2-mediated antioxidant response, Acta. Pharmacol. Sin. 39 (2018) 311-327.
    A. N. Shami, X. Zheng, S. K. Munyoki, et al., Single-Cell RNA Sequencing of Human, Macaque, and Mouse Testes Uncovers Conserved and Divergent Features of Mammalian Spermatogenesis, Dev. Cell. 54 (2020) 529-547.e512.
    W. Zhang, S. Xia, W. Xiao, et al., A Single-Cell Transcriptomic Landscape of Mouse Testicular Aging, J. Adv. Res. 2022. https://doi.org/10.1016/j.jare.2022.12.007.
    S. Xia, W. Zhang, J. Yang, et al., A single-cell atlas of bisphenol A (BPA)-induced testicular injury in mice, Clin. Transl. Med. 12 (2022), e789.
    Q. Zhao, J. F. Huang, Y. Cheng, et al., Polyamine metabolism links gut microbiota and testicular dysfunction, Microbiome. 9 (2021), 224.
    J. Chen, P. Luo, C. Wang, et al., Integrated single-cell transcriptomics and proteomics reveal cellular-specific responses and microenvironment remodeling in aristolochic acid nephropathy, JCI. Insight. 16 (2022), e157360.
    R. D. Cardiff, C. H. Miller, R. J. Munn. Manual hematoxylin and eosin staining of mouse tissue sections, Cold. Spring. Harb. Protoc. 2014 (2014) 655-658.
    M. S. Balzer, T. Doke, Y. W. Yang, et al., Single-cell analysis highlights differences in druggable pathways underlying adaptive or fibrotic kidney regeneration, Nat. Commun. 13 (2022), 4018.
    W. Zhang, S. Xia, X. Zhong, et al., Characterization of 2,2',4,4'-tetrabromodiphenyl ether (BDE47)-induced testicular toxicity via single-cell RNA-sequencing, Precis. Clin. Med. 5 (2022), pbac016.
    A. Butler, P. Hoffman, P. Smibert, et al., Integrating single-cell transcriptomic data across different conditions, technologies, and species, Nat. Biotechnol. 36 (2018) 411-420.
    C. S. Mcginnis, L. M. Murrow, Z. J. Gartner. DoubletFinder: Doublet Detection in Single-Cell RNA Sequencing Data Using Artificial Nearest Neighbors, Cell. Syst. 8 (2019) 329-337.e324.
    M. Jung, D. Wells, J. Rusch, et al., Unified single-cell analysis of testis gene regulation and pathology in five mouse strains, Elife. 8 (2019), e43966.
    T. Wu, E. Hu, S. Xu, et al., clusterProfiler 4.0: A universal enrichment tool for interpreting omics data, Innovation. 2 (2021), 100141.
    S. Aibar, C. B. Gonzalez-Blas, T. Moerman, et al., SCENIC: single-cell regulatory network inference and clustering, Nat. Methods. 14 (2017) 1083-1086.
    V. A. Huynh-Thu, A. Irrthum, L. Wehenkel, et al., Inferring regulatory networks from expression data using tree-based methods, PLoS One. 5 (2010), e12776.
    P. Shannon, A. Markiel, O. Ozier, et al., Cytoscape: a software environment for integrated models of biomolecular interaction networks, Genome. Res. 13 (2003) 2498-2504.
    A. Liberzon, C. Birger, H. Thorvaldsdottir, et al., The Molecular Signatures Database (MSigDB) hallmark gene set collection, Cell. Syst. 1 (2015) 417-425.
    S. Jin, C. F. Guerrero-Juarez, L. Zhang, et al., Inference and analysis of cell-cell communication using CellChat, Nat. Commun. 12 (2021), 1088.
    J. Yang, W. Zhang, L. Jia, et al., A novel autophagy activator ginsenoside Rh2 enhances the efficacy of immunogenic chemotherapy, Clin. Transl. Med. 13 (2023), e1109.
    C. Yang, Z. Zhu, B. C. Tong, et al., A stress response p38 MAP kinase inhibitor SB202190 promoted TFEB/TFE3-dependent autophagy and lysosomal biogenesis independent of p38, Redox. Biol. 32 (2020), 101445.
    S. Wang, G. Wang, W. Wu, et al., Autophagy activation by dietary piceatannol enhances the efficacy of immunogenic chemotherapy, Front. Immunol. 13 (2022), 968686.
    C. D. Green, Q. Ma, G. L. Manske, et al., A Comprehensive Roadmap of Murine Spermatogenesis Defined by Single-Cell RNA-Seq, Dev. Cell. 46 (2018) 651-667.e610.
    M. Daigle, P. Roumaud, L. J. Martin. Expressions of Sox9, Sox5, and Sox13 transcription factors in mice testis during postnatal development, Mol. Cell. Biochem. 407 (2015) 209-221.
    N. Mossadegh-Keller, M. H. Sieweke. Testicular macrophages: Guardians of fertility, Cell. Immunol. 330 (2018) 120-125.
    S. Ren, X. Chen, X. Tian, et al., The expression, function, and utilization of Protamine1: a literature review, Transl. Cancer. Res. 10 (2021) 4947-4957.
    A. R. Moschen, T. E. Adolph, R. R. Gerner, et al., Lipocalin-2: A Master Mediator of Intestinal and Metabolic Inflammation, Trends. Endocrinol. Metab. 28 (2017) 388-397.
    J. A. Ramilowski, T. Goldberg, J. Harshbarger, et al., A draft network of ligand-receptor-mediated multicellular signalling in human, Nat. Commun. 6 (2015), 7866.
    J. H. Van Der Meer, T. Van Der Poll, C. Van'T Veer. TAM receptors, Gas6, and protein S: roles in inflammation and hemostasis, Blood. 123 (2014) 2460-2469.
    C. Yang, C. Su, A. Iyaswamy, et al., Celastrol enhances transcription factor EB (TFEB)-mediated autophagy and mitigates Tau pathology: Implications for Alzheimer's disease therapy, Acta. Pharm. Sin B. 12 (2022) 1707-1722.
    W. Zhang, C. Xu, J. Sun, et al., Impairment of the autophagy-lysosomal pathway in Alzheimer's diseases: Pathogenic mechanisms and therapeutic potential, Acta. Pharm. Sin B. 12 (2022) 1019-1040.
    M. Wang, L. Zeng, P. Su, et al., Autophagy: a multifaceted player in the fate of sperm, Hum. Reprod. Update. 28 (2022) 200-231.
    Y. Cheng, Y. Zhao, Y. Zheng. Therapeutic potential of triptolide in autoimmune diseases and strategies to reduce its toxicity, Chin. Med. 16 (2021), 114.
    K. Yuan, X. Li, Q. Lu, et al., Application and Mechanisms of Triptolide in the Treatment of Inflammatory Diseases-A Review, Front. Pharmacol. 10 (2019), 1469.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Article Metrics

    Article views (189) PDF downloads(20) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return