101
|
Cai Z, Chen J, Yu Z, Li H, Liu Z, Deng D, Liu J, Chen C, Zhang C, Ou Z, Chen M, Hu J, Zu X. BCAT2 Shapes a Noninflamed Tumor Microenvironment and Induces Resistance to Anti-PD-1/PD-L1 Immunotherapy by Negatively Regulating Proinflammatory Chemokines and Anticancer Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207155. [PMID: 36642843 PMCID: PMC10015882 DOI: 10.1002/advs.202207155] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Indexed: 06/17/2023]
Abstract
To improve response rate of monotherapy of immune checkpoint blockade (ICB), it is necessary to find an emerging target in combination therapy. Through analyzing tumor microenvironment (TME)-related indicators, it is validated that BCAT2 shapes a noninflamed TME in bladder cancer. The outcomes of multiomics indicate that BCAT2 has an inhibitory effect on cytotoxic lymphocyte recruitment by restraining activities of proinflammatory cytokine/chemokine-related pathways and T-cell-chemotaxis pathway. Immunoassays reveal that secretion of CD8+ T-cell-related chemokines keeps a robust negative correlation with BCAT2, generating a decreasing tendency of CD8+ T cells around BCAT2+ tumor cells from far to near. Cotreatment of BCAT2 deficiency and anti-PD-1 antibody has a synergistic effect in vivo, implying the potential of BCAT2 in combination therapy. Moreover, the value of BCAT2 in predicting efficacy of immunotherapy is validated in multiple immunotherapy cohorts. Together, as a key molecule in TME, BCAT2 is an emerging target in combination with ICB and a biomarker of guiding precision therapy.
Collapse
Affiliation(s)
- Zhiyong Cai
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jinbo Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zhengzheng Yu
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- Research Center of Carcinogenesis and Targeted TherapyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Huihuang Li
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zhi Liu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Dingshan Deng
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jinhui Liu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Chunliang Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Chunyu Zhang
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zhenyu Ou
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Minfeng Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jiao Hu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Xiongbing Zu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| |
Collapse
|
102
|
Singh A, Hermann BP. Conserved Transcriptome Features Define Prepubertal Primate Spermatogonial Stem Cells as A dark Spermatogonia and Identify Unique Regulators. Int J Mol Sci 2023; 24:4755. [PMID: 36902187 PMCID: PMC10002546 DOI: 10.3390/ijms24054755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/25/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Antineoplastic treatments for cancer and other non-malignant disorders can result in long-term or permanent male infertility by ablating spermatogonial stem cells (SSCs). SSC transplantation using testicular tissue harvested before a sterilizing treatment is a promising approach for restoring male fertility in these cases, but a lack of exclusive biomarkers to unequivocally identify prepubertal SSCs limits their therapeutic potential. To address this, we performed single-cell RNA-seq on testis cells from immature baboons and macaques and compared these cells with published data from prepubertal human testis cells and functionally-defined mouse SSCs. While we found discrete groups of human spermatogonia, baboon and rhesus spermatogonia appeared less heterogenous. A cross-species analysis revealed cell types analogous to human SSCs in baboon and rhesus germ cells, but a comparison with mouse SSCs revealed significant differences with primate SSCs. Primate-specific SSC genes were enriched for components and regulators of the actin cytoskeleton and participate in cell-adhesion, which may explain why the culture conditions for rodent SSCs are not appropriate for primate SSCs. Furthermore, correlating the molecular definitions of human SSC, progenitor and differentiating spermatogonia with the histological definitions of Adark/Apale spermatogonia indicates that both SSCs and progenitor spermatogonia are Adark, while Apale spermatogonia appear biased towards differentiation. These results resolve the molecular identity of prepubertal human SSCs, define novel pathways that could be leveraged for advancing their selection and propagation in vitro, and confirm that the human SSC pool resides entirely within Adark spermatogonia.
Collapse
Affiliation(s)
| | - Brian P. Hermann
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| |
Collapse
|
103
|
Voigt AL, Dardari R, Lara NLM, He T, Steele H, Dufour A, Orwig KE, Dobrinski I. Multiomics approach to profiling Sertoli cell maturation during development of the spermatogonial stem cell niche. Mol Hum Reprod 2023; 29:gaad004. [PMID: 36688722 PMCID: PMC9976880 DOI: 10.1093/molehr/gaad004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/10/2022] [Indexed: 01/24/2023] Open
Abstract
Spermatogonial stem cells (SSCs) are the basis of spermatogenesis, a complex process supported by a specialized microenvironment, called the SSC niche. Postnatal development of SSCs is characterized by distinct metabolic transitions from prepubertal to adult stages. An understanding of the niche factors that regulate these maturational events is critical for the clinical application of SSCs in fertility preservation. To investigate the niche maturation events that take place during SSC maturation, we combined different '-omics' technologies. Serial single cell RNA sequencing analysis revealed changes in the transcriptomes indicative of niche maturation that was initiated at 11 years of age in humans and at 8 weeks of age in pigs, as evident by Monocle analysis of Sertoli cells and peritubular myoid cell (PMC) development in humans and Sertoli cell analysis in pigs. Morphological niche maturation was associated with lipid droplet accumulation, a characteristic that was conserved between species. Lipidomic profiling revealed an increase in triglycerides and a decrease in sphingolipids with Sertoli cell maturation in the pig model. Quantitative (phospho-) proteomics analysis detected the activation of distinct pathways with porcine Sertoli cell maturation. We show here that the main aspects of niche maturation coincide with the morphological maturation of SSCs, which is followed by their metabolic maturation. The main aspects are also conserved between the species and can be predicted by changes in the niche lipidome. Overall, this knowledge is pivotal to establishing cell/tissue-based biomarkers that could gauge stem cell maturation to facilitate laboratory techniques that allow for SSC transplantation for restoration of fertility.
Collapse
Affiliation(s)
- A L Voigt
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - R Dardari
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - N L M Lara
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - T He
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - H Steele
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - A Dufour
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Canada
| | - K E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Women’s Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - I Dobrinski
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
104
|
Diabetes-Induced Autophagy Dysregulation Engenders Testicular Impairment via Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:4365895. [PMID: 36778206 PMCID: PMC9918358 DOI: 10.1155/2023/4365895] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/01/2022] [Accepted: 11/24/2022] [Indexed: 02/05/2023]
Abstract
Testes produce sperms, and gamete generation relies on a proper niche environment. The disruption of hierarchical regulatory homeostasis in Leydig or Sertoli cells may evoke a sterile phenotype in humans. In this study, we recapitulated type 2 diabetes mellitus by using a high-fat diet- (HFD-) fed mouse model to identify the phenotype and potential mechanism of diabetes-induced testicular impairment. At the end of the study, blood glucose levels, testosterone structure, testicular antioxidant capacity, and testosterone level and the expression of hypoxia-inducible factor- (HIF-) 1α, apoptosis-related protein cleaved-caspase3, and autophagy-related proteins such as LC3I/II, p62, and Beclin1 were evaluated. We found that long-term HFD treatment causes the development of diabetes mellitus, implicating increased serum glucose level, cell apoptosis, and testicular atrophy (P < 0.05 vs. Ctrl). Mechanistically, the results showed enhanced expression of HIF-1α in both Sertoli and Leydig cells (P < 0.05 vs. Ctrl). Advanced glycation end products (AGEs) were demonstrated to be a potential factor leading to HIF-1α upregulation in both cell types. In Sertoli cells, high glucose treatment had minor effects on Sertoli cell autophagy. However, AGE treatment stagnated the autophagy flux and escalated cell apoptosis (P < 0.05 vs. Ctrl+Ctrl). In Leydig cells, high glucose treatment was adequate to encumber autophagy induction and enhance oxidative stress. Similarly, AGE treatment facilitated HIF-1α expression and hampered testosterone production (P < 0.05 vs. Ctrl+Ctrl). Overall, these findings highlight the dual effects of diabetes on autophagy regulation in Sertoli and Leydig cells while imposing oxidative stress in both cell types. Furthermore, the upregulation of HIF-1α, which could be triggered by AGE treatment, may negatively affect both cell types. Together, these findings will help us further understand the molecular mechanism of diabetes-induced autophagy dysregulation and testicular impairment, enriching the content of male reproductive biology in diabetic patients.
Collapse
|
105
|
Luo X, Zheng H, Nai Z, Li M, Li Y, Lin N, Li Y, Wu Z. Identification of biomarkers associated with macrophage infiltration in non-obstructive azoospermia using single-cell transcriptomic and microarray data. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:55. [PMID: 36819497 PMCID: PMC9929779 DOI: 10.21037/atm-22-5601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/29/2022] [Indexed: 01/19/2023]
Abstract
Background Non-obstructive azoospermia (NOA) is a common clinical cause of male infertility. Research suggests that macrophages are linked to testicular function; however, their involvement in NOA remains unknown. Methods To evaluate the importance of macrophages infiltration in NOA and identify the macrophage-related biomarkers, the gene-expression microarray data GSE45885 and the single-cell transcriptomic data GSE149512 were utilized from the Gene Expression Omnibus (GEO). A single-sample gene set enrichment analysis (ssGSEA) was conducted to investigate immune cell proliferation. The Seurat package was used for the single-cell data analysis, and the limma package was used to identify the differentially expressed genes between the NOA and normal samples. Moreover, we conducted a weighted gene co-expression network analysis (WGCNA) to identify the macrophage-related key modules and genes, and conducted Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses for the functional exploration. To identify the macrophage-related biomarkers, we conducted least absolute shrinkage and selection operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE) analyses. Real-time quantitative polymerase chain reaction (RT-qPCR) was used to verify the marker genes present in NOA. Results We confirmed that open reading frame 72 gene on chromosome 9 (C9orf72) [area under the curve (AUC) =0.861] and cartilage-associated protein (CRTAP) (AUC =0.917) were the hub genes of NOA, and the RT-qPCR analysis revealed the critical expression of both genes in NOA. Conclusions Through the combination of tissue transcriptomic and single-cell RNA-sequencing analyses, we concluded that macrophage infiltration is significant in different subtypes of NOA, and we hypothesized that C9orf72 and CRTAP play critical roles in NOA due to their high expression in macrophages.
Collapse
Affiliation(s)
- Xi Luo
- Department of Reproductive Medicine, The First People’s Hospital of Yunnan Province, Kunming, China;,Reproductive Medical Center of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China;,NHC Key Laboratory of Periconception Health Birth in Western China, Kunming, China;,Faculty of Life science and Technology, Kunming University of Science and Technology, Kunming, China;,Medical school, Kunming University of Science and Technology, Kunming, China
| | - Haishan Zheng
- Department of Reproductive Medicine, The First People’s Hospital of Yunnan Province, Kunming, China;,Reproductive Medical Center of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China;,NHC Key Laboratory of Periconception Health Birth in Western China, Kunming, China
| | - Zhen Nai
- Department of Reproductive Medicine, The First People’s Hospital of Yunnan Province, Kunming, China;,Reproductive Medical Center of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China;,NHC Key Laboratory of Periconception Health Birth in Western China, Kunming, China
| | - Mingying Li
- Department of Reproductive Medicine, The First People’s Hospital of Yunnan Province, Kunming, China;,Reproductive Medical Center of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China;,NHC Key Laboratory of Periconception Health Birth in Western China, Kunming, China
| | - Yonggang Li
- Department of Reproductive Medicine, The First People’s Hospital of Yunnan Province, Kunming, China;,Reproductive Medical Center of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China;,NHC Key Laboratory of Periconception Health Birth in Western China, Kunming, China
| | - Na Lin
- Department of Reproductive Medicine, The First People’s Hospital of Yunnan Province, Kunming, China;,Reproductive Medical Center of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China;,NHC Key Laboratory of Periconception Health Birth in Western China, Kunming, China
| | - Yunxiu Li
- Department of Reproductive Medicine, The First People’s Hospital of Yunnan Province, Kunming, China;,Reproductive Medical Center of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China;,NHC Key Laboratory of Periconception Health Birth in Western China, Kunming, China
| | - Ze Wu
- Department of Reproductive Medicine, The First People’s Hospital of Yunnan Province, Kunming, China;,Reproductive Medical Center of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China;,NHC Key Laboratory of Periconception Health Birth in Western China, Kunming, China
| |
Collapse
|
106
|
Di Persio S, Neuhaus N. Human spermatogonial stem cells and their niche in male (in)fertility: novel concepts from single-cell RNA-sequencing. Hum Reprod 2023; 38:1-13. [PMID: 36409992 PMCID: PMC9825264 DOI: 10.1093/humrep/deac245] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
The amount of single-cell RNA-sequencing (scRNA-seq) data produced in the field of human male reproduction has steadily increased. Transcriptional profiles of thousands of testicular cells have been generated covering the human neonatal, prepubertal, pubertal and adult period as well as different types of male infertility; the latter include non-obstructive azoospermia, cryptozoospermia, Klinefelter syndrome and azoospermia factor deletions. In this review, we provide an overview of transcriptional changes in different testicular subpopulations during postnatal development and in cases of male infertility. Moreover, we review novel concepts regarding the existence of spermatogonial and somatic cell subtypes as well as their crosstalk and provide corresponding marker genes to facilitate their identification. We discuss the potential clinical implications of scRNA-seq findings, the need for spatial information and the necessity to corroborate findings by exploring other levels of regulation, including at the epigenetic or protein level.
Collapse
Affiliation(s)
- Sara Di Persio
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Münster, Germany
| | - Nina Neuhaus
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Münster, Germany
| |
Collapse
|
107
|
Chen Y, Sun T, Gu L, Ouyang S, Liu K, Yuan P, Liu C. Identification of hub genes and biological mechanisms underlying the pathogenesis of asthenozoospermia and chronic epididymitis. Front Genet 2023; 14:1110218. [PMID: 37152990 PMCID: PMC10160426 DOI: 10.3389/fgene.2023.1110218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Objective: Asthenozoospermia (AZS) is one of the most common causes of male fertility, affecting family wellbeing and population growth. Chronic epididymitis (CE) is a common and lingering inflammatory disease in the scrotum. Inflammation in the epididymis has a severe impact on sperm motility. This study aimed to explore the genetic profile and critical pathways involved in the pathological mechanisms of AZS and CE, and discover potential biomarkers. Methods: Genomic datasets of AZS and CE were obtained from the Gene Expression Omnibus (GEO) database, and relevant differentially expressed genes (DEGs) were identified. GO and pathway enrichment analyses, construction of a protein-protein interaction network, and receiver operator characteristic curve analysis were conducted. The expression profile of hub genes was validated in immunohistochemical data and testicular cell data. Immune infiltration, miRNA-hub gene interactions, and gene-disease interactions were explored. The mRNA levels of hub genes were further measured by qRT-PCR. Results: A total of 109 DEGs were identified between the AZS/CE and healthy control groups. Pathways of the immune system, neutrophil degranulation, and interleukin-4 and interleukin-13 signaling were enriched in AZS and CE. Five hub genes (CD300LB, CMKLR1, CCR4, B3GALT5, and CTSK) were selected, and their diagnostic values were validated in AZS, CE, and independent validation sets (area under the curve >0.7). Furthermore, the five-hub gene signature was well characterized in testicular immunohistochemical staining and testicular cells from healthy controls. Immune infiltration analysis showed that infiltration of CD8+ cells and T helper cells was significantly related to the expression level of five hub genes. In addition, a miRNA-hub gene network and interaction of other diseases were displayed. The mRNA levels of hub genes (CD300LB, CMKLR1, CCR4, and B3GALT5) were significantly elevated in the patient group. The mRNA level of CTSK also showed a similar trend. Conclusion: Our study uncovered the genetic profile involved in AZS and CE, and elucidated enriched pathways and molecular associations between hub genes and immune infiltration. This finding provides novel insight into the common pathogenesis of both diseases as well as the potential biomarkers for CE-associated AZS.
Collapse
Affiliation(s)
- Yinwei Chen
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Taotao Sun
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Longjie Gu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Song Ouyang
- Department of Urology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Kang Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Penghui Yuan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Penghui Yuan, ; Chang Liu,
| | - Chang Liu
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
- *Correspondence: Penghui Yuan, ; Chang Liu,
| |
Collapse
|
108
|
Yu C, Zhou C, Lin F, Zhang W, Wang X, Hu L, Lu R. Analysis of the Growth and Development of Children Born with ICSI of Epididymal and Testicular Spermatozoa: A Propensity Matching Study. Curr Pharm Des 2023; 29:2668-2678. [PMID: 37929741 DOI: 10.2174/0113816128264448231022201641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/10/2023] [Accepted: 09/21/2023] [Indexed: 11/07/2023]
Abstract
OBJECTIVE The study aimed to evaluate whether singleton live births (at 0, 1, 6, 12, and 24 months) following intracytoplasmic sperm injection (ICSI) using sperm of different origins (ejaculated or non-ejaculated sperm) are associated with the growth and development of children born. METHODS This was a retrospective cohort study conducted at a single center from January 2016 to December 2019. Follow-up data of the children were obtained from the Jiangsu Province Maternal and Child database. A total of 350 singleton live births after fresh embryo transfer (ET) with ICSI were included. Based on the origin of the sperm, the patients were divided into two groups: the ejaculated group (n = 310) and the non-ejaculated group (n = 40). Propensity score matching was used to control for multiple baseline covariates, resulting in 80 singleton live births (ejaculated sperm) matched to 40 singleton live births (non-ejaculated). The non-ejaculated group was further divided into two subgroups: the PESA group (n = 23) and the TESA group (n = 17). The primary outcome of the study was the growth and development of children. Secondary outcomes included the 2PN rate, high-cleavage embryo rate, blastocyst formation rate, and others. RESULTS After matching parental age, BMI, occupation, and maternal serum AMH level, there was no significant difference found in the growth and development of children between the non-ejaculated and ejaculated group or the PESA group and TESA group, respectively. However, the 2PN rate and the blastocyst formation rate were higher in the ejaculated group compared to the non-ejaculated group (91.02 and 85.45, P = 0.002) and (67.37 and 56.06, P = 0.019), respectively. The high-quality cleavage embryo rate was also higher in the TESA group compared to the PESA group (85.06 and 65.63, P = 0.001). CONCLUSION This study suggests that there are no significant differences in the growth and development of children born following ICSI using sperm of different origins (ejaculated or non-ejaculated). For nonobstructive azoospermia (OA) patients, sperm derived from the testis may be more effective than derived from the epididymis. However, due to the limited sample size of the non-ejaculated group in this study, further investigations with larger sample sizes are needed to validate these findings.
Collapse
Affiliation(s)
- Chunmei Yu
- Changzhou Medical Center, Changzhou Maternal and Child Health Care Hospital, Nanjing Medical University, Jiangsu, Changzhou, China
| | - Chao Zhou
- Reproduction Medical Center, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Feng Lin
- Department of Wuqia County People Hospital, The Xinjiang Uygur Autonomous Region, China
| | - Wanchao Zhang
- Department of Wuqia County People Hospital, The Xinjiang Uygur Autonomous Region, China
| | - Xiaoyu Wang
- Changzhou Medical Center, Changzhou Maternal and Child Health Care Hospital, Nanjing Medical University, Jiangsu, Changzhou, China
| | - Lingmin Hu
- Changzhou Medical Center, Changzhou Maternal and Child Health Care Hospital, Nanjing Medical University, Jiangsu, Changzhou, China
| | - Renjie Lu
- Changzhou Medical Center, Changzhou Third People's Hospital, Nanjing Medical University, Jiangsu, China
| |
Collapse
|
109
|
Transcriptomic differences between fibrotic and non-fibrotic testicular tissue reveal possible key players in Klinefelter syndrome-related testicular fibrosis. Sci Rep 2022; 12:21518. [PMID: 36513788 PMCID: PMC9748020 DOI: 10.1038/s41598-022-26011-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Klinefelter syndrome (KS; 47,XXY) affects 1-2 in 1000 males. Most men with KS suffer from an early germ cell loss and testicular fibrosis from puberty onwards. Mechanisms responsible for these processes remain unknown. Previous genomics studies on testis tissue from men with KS focused on germ cell loss, while a transcriptomic analysis focused on testicular fibrosis has not yet been performed. This study aimed to identify factors involved in the fibrotic remodelling of KS testes by analysing the transcriptome of fibrotic and non-fibrotic testicular tissue. RNA sequencing was performed to compare the genes expressed in testicular samples with (KS and testis atrophy) and without (Sertoli cell-only syndrome and fertile controls) fibrosis (n = 5, each). Additionally, differentially expressed genes (DEGs) between KS and testis atrophy samples were studied to reveal KS-specific fibrotic genes. DEGs were considered significant when p < 0.01 and log2FC > 2. Next, downstream analyses (GO and KEGG) were performed. Lastly, RNA in situ hybridization was performed to validate the results. The first analysis (fibrotic vs non-fibrotic) resulted in 734 significant DEGs (167 up- and 567 down-regulated). Genes involved in the extracellular structure organization (e.g. VCAM1) were found up-regulated. KEGG analysis showed an up-regulation of genes involved in the TGF-β pathway. The KS vs testis atrophy analysis resulted in 539 significant DEGs (59 up- and 480 down-regulated). Chronic inflammatory response genes were found up-regulated. The overlap of X-linked DEGs from the two analyses revealed three genes: matrix-remodelling associated 5 (MXRA5), doublecortin (DCX) and variable charge X-Linked 3B (VCX3B). RNA in situ hybridization showed an overexpression of VCAM1, MXRA5 and DCX within the fibrotic group compared with the non-fibrotic group. To summarize, this study revealed DEGs between fibrotic and non-fibrotic testis tissue, including VCAM1. In addition, X-linked fibrotic genes were revealed, e.g. MXRA5, DCX and VCX3B. Their potential role in KS-related testicular fibrosis needs further study.
Collapse
|
110
|
Huang L, Zhang J, Zhang P, Huang X, Yang W, Liu R, Sun Q, Lu Y, Zhang M, Fu Q. Single-cell RNA sequencing uncovers dynamic roadmap and cell-cell communication during buffalo spermatogenesis. iScience 2022; 26:105733. [PMID: 36582818 PMCID: PMC9793287 DOI: 10.1016/j.isci.2022.105733] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/24/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Spermatogenesis carries the task of precise intergenerational transmission of genetic information from the paternal genome and involves complex developmental processes regulated by the testicular microenvironment. Studies performed mainly in mouse models have established the theoretical basis for spermatogenesis, yet the wide interspecies differences preclude direct translation of the findings, and farm animal studies are progressing slowly. More than 32,000 cells from prepubertal (3-month-old) and pubertal (24-month-old) buffalo testes were analyzed by using single-cell RNA sequencing (scRNA-seq), and dynamic gene expression roadmaps of germ and somatic cell development were generated. In addition to identifying the dynamic processes of sequential cell fate transitions, the global cell-cell communication essential to maintain regular spermatogenesis in the buffalo testicular microenvironment was uncovered. The findings provide the theoretical basis for establishing buffalo germline stem cells in vitro or culturing organoids and facilitating the expansion of superior livestock breeding.
Collapse
Affiliation(s)
- Liangfeng Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Junjun Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Pengfei Zhang
- Institute of Medical and Health, Guangxi Academy of Sciences, Nanning 530007, China
| | - Xingchen Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Weihan Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Runfeng Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Qinqiang Sun
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Yangqing Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China,Corresponding author
| | - Ming Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China,Corresponding author
| | - Qiang Fu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China,Corresponding author
| |
Collapse
|
111
|
Rabbani M, Zheng X, Manske GL, Vargo A, Shami AN, Li JZ, Hammoud SS. Decoding the Spermatogenesis Program: New Insights from Transcriptomic Analyses. Annu Rev Genet 2022; 56:339-368. [PMID: 36070560 PMCID: PMC10722372 DOI: 10.1146/annurev-genet-080320-040045] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Spermatogenesis is a complex differentiation process coordinated spatiotemporally across and along seminiferous tubules. Cellular heterogeneity has made it challenging to obtain stage-specific molecular profiles of germ and somatic cells using bulk transcriptomic analyses. This has limited our ability to understand regulation of spermatogenesis and to integrate knowledge from model organisms to humans. The recent advancement of single-cell RNA-sequencing (scRNA-seq) technologies provides insights into the cell type diversity and molecular signatures in the testis. Fine-grained cell atlases of the testis contain both known and novel cell types and define the functional states along the germ cell developmental trajectory in many species. These atlases provide a reference system for integrated interspecies comparisons to discover mechanistic parallels and to enable future studies. Despite recent advances, we currently lack high-resolution data to probe germ cell-somatic cell interactions in the tissue environment, but the use of highly multiplexed spatial analysis technologies has begun to resolve this problem. Taken together, recent single-cell studies provide an improvedunderstanding of gametogenesis to examine underlying causes of infertility and enable the development of new therapeutic interventions.
Collapse
Affiliation(s)
- Mashiat Rabbani
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA;
| | - Xianing Zheng
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA;
| | - Gabe L Manske
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Alexander Vargo
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA;
| | - Adrienne N Shami
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA;
| | - Jun Z Li
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA;
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Saher Sue Hammoud
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA;
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Urology, University of Michigan, Ann Arbor, Michigan, USA
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
112
|
Low-Intensity Pulsed Ultrasound Counteracts Advanced Glycation End Products-Induced Corpus Cavernosal Endothelial Cell Dysfunction via Activating Mitophagy. Int J Mol Sci 2022; 23:ijms232314887. [PMID: 36499213 PMCID: PMC9740783 DOI: 10.3390/ijms232314887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/13/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Injury to corpus cavernosal endothelial cells (CCECs) is an important pathological basis of diabetes mellitus-induced erectile dysfunction (DMED), while low-intensity pulsed ultrasound (LIPUS) has been shown to improve erectile function in DMED. To further understand its therapeutic mechanism of action, in this study, we first demonstrated increased apoptosis and shedding in the CCECs of DMED patients, accompanied by significant mitochondrial injury by immunohistochemistry and electron microscopy of corpus cavernosum tissue. Next, we used advanced glycation end products (AGEs) to simulate the diabetic environment in vitro and found that AGES damaged mitochondria and inhibited angiogenesis in CCECs in a dose-dependent manner, while LIPUS treatment significantly reversed its effects. Mechanistic studies based on transcriptome sequencing showed that LIPUS significantly up-regulated LC3 and PARKIN protein levels in mitochondria, promoted mitophagy, and affected mitochondrial dynamics and reactive oxygen species (ROS) production. In addition, the protective effects of LIPUS were abrogated when mitophagy was inhibited by 3-methyladenine. In summary, LIPUS exerted potent inhibitory effects on AGES-induced CCEC failure via mitophagy, providing a theoretical basis for DMED treatment that encompasses the protection of endothelial structure and function.
Collapse
|
113
|
Zhong Y, Chen X, Zhao J, Deng H, Li X, Xie Z, Zhou B, Xian Z, Li X, Luo G, Li H. Integrative analyses of potential biomarkers and pathways for non-obstructive azoospermia. Front Genet 2022; 13:988047. [PMID: 36506310 PMCID: PMC9730279 DOI: 10.3389/fgene.2022.988047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/01/2022] [Indexed: 11/26/2022] Open
Abstract
Background: Non-obstructive azoospermia (NOA) is the most severe form of male infertility. Currently, the molecular mechanisms underlying NOA pathology have not yet been elucidated. Hence, elucidation of the mechanisms of NOA and exploration of potential biomarkers are essential for accurate diagnosis and treatment of this disease. In the present study, we aimed to screen for biomarkers and pathways involved in NOA and reveal their potential molecular mechanisms using integrated bioinformatics. Methods: We downloaded two gene expression datasets from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) in NOA and matched the control group tissues were identified using the limma package in R software. Subsequently, Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), gene set enrichment analysis (GSEA), protein-protein interaction (PPI) network, gene-microRNAs network, and transcription factor (TF)-hub genes regulatory network analyses were performed to identify hub genes and associated pathways. Finally, we conducted immune infiltration analysis using CIBERSORT to evaluate the relationship between the hub genes and the NOA immune infiltration levels. Results: We identified 698 common DEGs, including 87 commonly upregulated and 611 commonly downregulated genes in the two datasets. GO analysis indicated that the most significantly enriched gene was protein polyglycylation, and KEGG pathway analysis revealed that the DEGs were most significantly enriched in taste transduction and pancreatic secretion signaling pathways. GSEA showed that DEGs affected the biological functions of the ribosome, focaladhesion, and protein_expor. We further identified the top 31 hub genes from the PPI network, and friends analysis of hub genes in the PPI network showed that NR4A2 had the highest score. In addition, immune infiltration analysis found that CD8+ T cells and plasma cells were significantly correlated with ODF3 expression, whereas naive B cells, plasma cells, monocytes, M2 macrophages, and resting mast cells showed significant variation in the NR4A2 gene expression group, and there were differences in T cell regulatory immune cell infiltration in the FOS gene expression groups. Conclusion: The present study successfully constructed a regulatory network of DEGs between NOA and normal controls and screened three hub genes using integrative bioinformatics analysis. In addition, our results suggest that functional changes in several immune cells in the immune microenvironment may play an important role in spermatogenesis. Our results provide a novel understanding of the molecular mechanisms of NOA and offer potential biomarkers for its diagnosis and treatment.
Collapse
Affiliation(s)
- Yucheng Zhong
- Assisted Reproductive Technology Center, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Xiaoqing Chen
- Department of Breast Surgical Oncology, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Jun Zhao
- Assisted Reproductive Technology Center, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Hao Deng
- Assisted Reproductive Technology Center, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Xiaohang Li
- Assisted Reproductive Technology Center, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Zhongju Xie
- Assisted Reproductive Technology Center, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Bingyu Zhou
- Assisted Reproductive Technology Center, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Zhuojie Xian
- Assisted Reproductive Technology Center, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Xiaoqin Li
- Assisted Reproductive Technology Center, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Guoqun Luo
- Assisted Reproductive Technology Center, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China,*Correspondence: Guoqun Luo, ; Huan Li,
| | - Huan Li
- Assisted Reproductive Technology Center, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China,*Correspondence: Guoqun Luo, ; Huan Li,
| |
Collapse
|
114
|
Wang HY, Liu X, Chen JY, Huang Y, Lu Y, Tan F, Liu Q, Yang M, Li S, Zhang X, Qin Y, Ma W, Yang Y, Meng L, Liu K, Wang Q, Fan G, Nóbrega RH, Liu S, Piferrer F, Shao C. Single-cell-resolution transcriptome map revealed novel genes involved in testicular germ cell progression and somatic cells specification in Chinese tongue sole with sex reversal. SCIENCE CHINA LIFE SCIENCES 2022; 66:1151-1169. [PMID: 36437386 DOI: 10.1007/s11427-021-2236-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/28/2022] [Indexed: 11/29/2022]
Abstract
Female-to-male sex reversals (pseudomales) are common in lower vertebrates and have been found in natural populations, which is a concern under rapid changes in environmental conditions. Pseudomales can exhibit altered spermatogenesis. However, the regulatory mechanisms underlying pseudomale spermatogenesis remain unclear. Here, we characterized spermatogenesis in Chinese tongue sole (Cynoglossus semilaevis), a species with genetic and environmental sex determination, based on a high-resolution single-cell RNA-seq atlas of cells derived from the testes of genotypic males and pseudomales. We identified five germ cell types and six somatic cell types and obtained a single-cell atlas of dynamic changes in gene expression during spermatogenesis in Chinese tongue sole, including alterations in pseudomales. We detected decreased levels of Ca2+ signaling pathway-related genes in spermatogonia, insufficient meiotic initiation in spermatocytes, and a malfunction of somatic niche cells in pseudomales. However, a cluster of CaSR genes and MAPK signaling factors were upregulated in undifferentiated spermatogonia of pseudomales. Additionally, we revealed that Z chromosome-specific genes, such as piwil2, dhx37, and ehmt1, were important for spermatogenesis. These results improve our understanding of reproduction after female-to-male sex-reversal and provide new insights into the adaptability of reproductive strategies in lower vertebrates.
Collapse
|
115
|
Wu X, Yun D, Sang M, Liu J, Zhou L, Shi J, Wang L, Bu T, Li L, Huang Y, Lin D, Sun F, Cheng CY. Defects of microtubule cytoskeletal organization in NOA human testes. Reprod Biol Endocrinol 2022; 20:154. [PMID: 36329464 PMCID: PMC9632130 DOI: 10.1186/s12958-022-01026-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022] Open
Abstract
The importance of actin and microtubule (MT) cytoskeletons in testis function in rodents is known to some extent, but its role in the etiology of azoospermia in humans remains unexplored. Here, we examined if MT cytoskeleton was defective in NOA (non-obstructive azoospermia) testes versus normal human testes based on histopathological, immunofluorescence (IF), and scRNA-Seq transcriptome profiling. Testis biopsy samples from n = 6 normal men versus n = 3 Sertoli cell only (SCO) and n = 3 MA (meiotic arrest) of NOA patients were used for histopathological analysis. IF analysis was also used to examine MT organization across the seminiferous epithelium, investigating the likely involvement of microtubule-associated proteins (MAPs). scRNA-Seq transcriptome profiling datasets from testes of 3 SCO patients versus 3 normal men in public domain in Gene Expression Omnibus (GEO) Sample (GSM) with identifiers were analyzed to examine relevant genes that regulate MT dynamics. NOA testes of MA and SCO patients displayed notable defects in MT organization across the epithelium with extensive truncation, mis-alignments and appeared as collapsed structures near the base of the tubules. These changes are in contrast to MTs in testes of normal men. scRNA-Seq analyses revealed considerable loss of spermatogenesis capacity in SCO testes of NOA patients versus normal men. An array of genes that support MT dynamics displayed considerable changes in expression and in spatial distribution. In summary, defects in MT cytoskeleton were noted in testes of NOA (SCO) patients, possibly mediated by defective spatial expression and/or distribution of MAPs. These changes, in turn, may impede spermatogenesis in SCO testes of NOA patients.
Collapse
Affiliation(s)
- Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Damin Yun
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Mengmeng Sang
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Jianpeng Liu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Liwei Zhou
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Jie Shi
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Lingling Wang
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Tiao Bu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Linxi Li
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - YingYing Huang
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Dengfeng Lin
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Fei Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
| | - C Yan Cheng
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China.
- Center for Biomedical Research, The Mary M. Wohlford Laboratory for Male Contraceptive Research, Population Council, 1230 York Ave, New York, NY, 10065, USA.
| |
Collapse
|
116
|
Zhang L, Guo M, Liu Z, Liu R, Zheng Y, Yu T, Lv Y, Lu H, Zeng W, Zhang T, Pan C. Single-cell RNA-seq analysis of testicular somatic cell development in pigs. J Genet Genomics 2022; 49:1016-1028. [PMID: 35436608 DOI: 10.1016/j.jgg.2022.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 12/29/2022]
Abstract
Spermatogenesis is the process by which diploid male germ cells propagate and differentiate into haploid flagellated spermatozoa. This highly complicated process is dependent on testicular somatic cells maturation. While the role of these somatic cells in spermatogenesis is relatively well established, knowledge about their development and maturation, particularly at the molecular level, is limited. In this study, we profiled the testicular single-cell transcriptomes of Guanzhong black pigs at the age of 7, 30, 60, 90, and 150 days. Five types of Sertoli cells, five types of Leydig cells, and four types of peritubular myoid cells were identified. Histological analysis revealed the changes in proliferation levels and marker gene expressions, and the prion-like protein gene (PRND) was identified as a novel marker for Sertoli cells. Additionally, integrated analyses of porcine and human datasets revealed similarities between human and pig testicular somatic cells. Overall, the data obtained in this study contribute to the understanding of testicular development in pigs as a model species.
Collapse
Affiliation(s)
- Lingkai Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ming Guo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zidong Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ruifang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yi Zheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Taiyong Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yinghua Lv
- College of Chemistry and Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hongzhao Lu
- College of Chemistry and Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wenxian Zeng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Tao Zhang
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, Shaanxi 723001, China.
| | - Chuanying Pan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
117
|
Wu Y, Zhang R, Shen C, Xu J, Wu T, Huang X, Liu M, Li H, Xu D, Zheng B. Testis-enriched Asb15 is not required for spermatogenesis and male fertility in mice. Am J Transl Res 2022; 14:6978-6990. [PMID: 36398235 PMCID: PMC9641487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/06/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The function of Asb15, which encodes an ASB protein with ankyrin (ANK) repeats and a C-terminal suppressor of cytokine signaling (SOCS) box motif, in male germ cells is poorly understood. Because expression of Asb15 is enriched in mouse testis, it may have a role in spermatogenesis. METHODS AND RESULTS We used a computer-assisted sperm analysis (CASA) system to analyze sperm from Asb15 gene knockout (KO) mice that we generated using the clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) technique. Histological staining and immunostaining were used to evaluate spermatogenesis in Asb15-KO mice. Asb15-KO and wild-type mice showed no differences in histology or in semen quality, fertility, or sperm apoptosis. Asb15- and Asb17-double KO (dKO) mice were generated to determine whether Asb17 compensated for the loss of Asb15. However, Asb15/17-dKO mice also showed normal fertility, except for an increase in giant cells in testicular tubules, suggesting a minor functional compensation between the two genes during spermatogenesis. CONCLUSIONS Our study suggests that Asb15 was individually not required for spermatogenesis or for fertility in mice. However, further investigation might be needed to reach a firm conclusion. These findings can prevent redundant research by other scientists and provides new information for further studies on the genetics of fertility in humans.
Collapse
Affiliation(s)
- Yibo Wu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan UniversityWuxi 214122, Jiangsu, China
| | - Ranran Zhang
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan UniversityWuxi 214122, Jiangsu, China
| | - Cong Shen
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical UniversitySuzhou 215002, Jiangsu, China
| | - Jinfu Xu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical UniversityNanjing 211166, Jiangsu, China
| | - Tiantian Wu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical UniversityNanjing 211166, Jiangsu, China
| | - Xiaoyan Huang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical UniversityNanjing 211166, Jiangsu, China
| | - Mingxi Liu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical UniversityNanjing 211166, Jiangsu, China
| | - Hong Li
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical UniversitySuzhou 215002, Jiangsu, China
| | - Dewu Xu
- Teaching Affairs Department, Affiliated Hospital of Jiangnan UniversityWuxi 214122, Jiangsu, China
| | - Bo Zheng
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical UniversitySuzhou 215002, Jiangsu, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University)Hefei 230032, Anhui, China
| |
Collapse
|
118
|
Deng CC, Zhang JP, Huo YN, Xue HY, Wang W, Zhang JJ, Wang XZ. Melatonin alleviates the heat stress-induced impairment of Sertoli cells by reprogramming glucose metabolism. J Pineal Res 2022; 73:e12819. [PMID: 35906194 DOI: 10.1111/jpi.12819] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/20/2022] [Accepted: 07/28/2022] [Indexed: 11/28/2022]
Abstract
Sertoli cells (SCs) provide structural and nutritional support for developing germ cells. Normal glucose metabolism of SCs is necessary for spermatogenesis. Melatonin could alleviate the effects of heat stress on spermatogenesis. However, the influences of heat stress on glucose metabolism in SCs remain unclear, and the potential protective mechanisms of melatonin on SCs need more exploration. In this study, boar SCs were treated at 43°C for 30 min, and different concentrations of melatonin were added to protect SCs from heat stress-induced impairment. These results showed that heat stress-induced oxidative stress caused cell apoptosis, inhibited the pentose phosphate pathway, and decreased the ATP content. Furthermore, heat stress increased the expressions of glucose intake- and glycolytic-related enzymes, which enhanced the glycolysis activity to compensate for the energy deficit. Melatonin relieved heat stress-induced oxidative stress and apoptosis by activating the Kelch-like ECH-associated protein 1 (KEAP1)/NF-E2-related factor 2 signaling pathway to increase the capacity of antioxidants. In addition, melatonin enhanced heat-shock protein 90 (HSP90) expression through melatonin receptor 1B (MTNR1B), thereby stabilizing hypoxia-inducible factor-1α (HIF-1α). Activation of the HIF-1α signaling pathway enhanced glycolysis, promoted the pentose phosphate pathway, and increased cell viability. Our results suggest that melatonin reprograms glucose metabolism in SCs through the MTNR1B-HSP90-HIF-1α axis and provides a theoretical basis for preventing heat stress injury.
Collapse
Affiliation(s)
- Cheng-Chen Deng
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Chongqing, Beibei, People's Republic of China
| | - Ji-Pan Zhang
- College of Animal Science and Technology, Southwest University, Chongqing, People's Republic of China
| | - Yuan-Nan Huo
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Chongqing, Beibei, People's Republic of China
| | - Hong-Yan Xue
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Chongqing, Beibei, People's Republic of China
| | - Wenxiu Wang
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, Shandong, People's Republic of China
| | - Jiao-Jiao Zhang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Chongqing, Beibei, People's Republic of China
| | - Xian-Zhong Wang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Chongqing, Beibei, People's Republic of China
| |
Collapse
|
119
|
Qian B, Li Y, Yan R, Han S, Bu Z, Gong J, Zheng B, Yuan Z, Ren S, He Q, Zhang J, Xu C, Wang R, Sun Z, Lin M, Zhou J, Ye L. RNA binding protein RBM46 regulates mitotic-to-meiotic transition in spermatogenesis. SCIENCE ADVANCES 2022; 8:eabq2945. [PMID: 36001654 PMCID: PMC9401620 DOI: 10.1126/sciadv.abq2945] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/08/2022] [Indexed: 06/15/2023]
Abstract
Meiosis entry during spermatogenesis requires reprogramming from mitotic to meiotic gene expression profiles. Transcriptional regulation has been extensively studied in meiosis entry, but gain of function for master transcription factors is insufficient to down-regulate mitotic genes. RNA helicase YTHDC2 and its partner MEIOC emerge as essential posttranscriptional regulators of meiotic entry. However, it is unclear what governs the RNA binding specificity of YTHDC2/MEIOC. Here, we identified RNA binding protein RBM46 as a component of the YTHDC2/MEIOC complex. Testis-specific Rbm46 knockout in mice causes infertility with defective mitotic-to-meiotic transition, phenocopying global Ythdc2 or Meioc knockout. RBM46 binds to 3' UTR of mitotic transcripts within 100 nucleotides from YTHDC2 U-rich motifs and targets these transcripts for degradation. Dysregulated RBM46 expression is associated with human male fertility disorders. These findings establish the RBM46/YTHDC2/MEIOC complex as the major posttranscriptional regulator responsible for down-regulating mitotic transcripts during meiosis entry in mammalian spermatogenesis, with implications for understanding meiosis-related fertility disorders.
Collapse
Affiliation(s)
- Baomei Qian
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yang Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Ruoyu Yan
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Shenglin Han
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Zhiwen Bu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Jie Gong
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Bangjin Zheng
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Zihan Yuan
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Sen Ren
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Qing He
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Jinwen Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Chen Xu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Ruilin Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Zheng Sun
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mingyan Lin
- Department of Neurobiology, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, People’s Republic of China
| | - Jian Zhou
- Department of Pediatric Laboratory, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Lan Ye
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
120
|
Santacroce L, Imbimbo C, Ballini A, Crocetto F, Scacco S, Cantore S, Di Zazzo E, Colella M, Jirillo E. Testicular Immunity and Its Connection with the Microbiota. Physiological and Clinical Implications in the Light of Personalized Medicine. J Pers Med 2022; 12:1335. [PMID: 36013286 PMCID: PMC9409709 DOI: 10.3390/jpm12081335] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 12/03/2022] Open
Abstract
Reproduction is a complex process, which is based on the cooperation between the endocrine-immune system and the microbiota. Testicular immunity is characterized by the so-called immune privilege, a mechanism that avoids autoimmune attacks against proteins expressed by spermatozoa. Testicular microbiota is connected with the gut microbiota, the most prevalent site of commensals inthe body. Both microbiotas take part inthe development of the immune system and protection againstpathogen invasion. Dysbiosis is caused by concurrent pathologies, such as obesity, diabetes, infections and trauma. The substitution of beneficial bacteria with pathogens may lead to destruction of spermatozoa directly or indirectly and, ultimately, to male infertility. Novel therapeutic interventions, i.e., nutritional interventions and supplementation of natural products, such as, probiotics, prebiotics, antioxidants and polyphenols, may lead to the restoration of the otherwise-impaired male reproductive potential, even if experimental and clinical results are not always concordant. In this review, the structure and immune function of the testis will be described with special reference to the blood-testisbarrier. The regulatory role of both the gut and testicular microbiota will be illustrated in health and disease, also emphasizing therapeutic attempts with natural products for the correction of male infertility, in the era of personalized medicine.
Collapse
Affiliation(s)
- Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Ciro Imbimbo
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples “Federico II”, 80131Naples, Italy
| | - Andrea Ballini
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Felice Crocetto
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples “Federico II”, 80131Naples, Italy
| | - Salvatore Scacco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Stefania Cantore
- Independent Researcher, Sorriso & Benessere—Ricerca e Clinica, 70129 Bari, Italy
| | - Erika Di Zazzo
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| | - Marica Colella
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Emilio Jirillo
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| |
Collapse
|
121
|
Luo SW, Tang L, Zhou D, Bo H, Fan LQ. FOXP4 promotes proliferation of human spermatogonial stem cells. Asian J Androl 2022; 25:322-330. [PMID: 36018067 DOI: 10.4103/aja202254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Continuous self-renewal and differentiation of spermatogonial stem cells (SSCs) is vital for maintenance of adult spermatogenesis. Although several spermatogonial stem cell regulators have been extensively investigated in rodents, regulatory mechanisms of human SSC self-renewal and differentiation have not been fully established. We analyzed single-cell sequencing data from the human testis and found that forkhead box P4 (FOXP4) expression gradually increased with development of SSCs. Further analysis of its expression patterns in human testicular tissues revealed that FOXP4 specifically marks a subset of spermatogonia with stem cell potential. Conditional inactivation of FOXP4 in human SSC lines suppressed SSC proliferation and significantly activated apoptosis. FOXP4 expressions were markedly suppressed in tissues with dysregulated spermatogenesis. These findings imply that FOXP4 is involved in human SSC proliferation, which will help elucidate on the mechanisms controlling the fate decisions in human SSCs.
Collapse
Affiliation(s)
- Shu-Wei Luo
- Reproductive Medicine Center, Hunan Provincial Maternal and Child Health Care Hospital, Changsha 410000, China.,NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha 410000, China
| | - Le Tang
- Reproductive Medicine Center, Hunan Provincial Maternal and Child Health Care Hospital, Changsha 410000, China.,NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha 410000, China
| | - Dai Zhou
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha 410000, China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410000, China
| | - Hao Bo
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha 410000, China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410000, China
| | - Li-Qing Fan
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha 410000, China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410000, China
| |
Collapse
|
122
|
Sagaradze G, Monakova A, Basalova N, Popov V, Balabanyan V, Efimenko A. Regenerative medicine for male infertility: A focus on stem cell niche injury models. Biomed J 2022; 45:607-614. [PMID: 35123107 PMCID: PMC9486244 DOI: 10.1016/j.bj.2022.01.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/19/2021] [Accepted: 01/27/2022] [Indexed: 02/08/2023] Open
Abstract
Stem and progenitor cells located within stem cell niches maintain the renewal and regeneration of tissues and organs throughout the life of an adult organism. Stem cell niche component dysfunction might alter the activity of stem cells and ultimately lead to the development of difficult-to-treat chronic or acute disorders. Of note, some cases of idiopathic male infertility, a highly prevalent diagnosis with no specific treatment options, might be associated with a spermatogonial stem cell(SSC) niche disturbance. To overcome this disease entity, approaches aiming at launching the regeneration of an altered stem cell niche are worth considering. Particularly, mesenchymal stromal cells (MSCs) or their secretome might fulfill this task due to their promising contribution in recovering injured stem cell niches. However, the successful application of MSC-based treatment is limited by the uncovered mechanisms of action of MSCs and their secretome. Specific animal models should be developed or adapted to reveal the role of MSCs and their secretome in a stem cell niche recovery. In this review, in a bid to consider MSCs and their secretome as a therapeutic regenerative approach for idiopathic male infertility we focus on the rationale of SSC niche injury modeling.
Collapse
Affiliation(s)
- Georgy Sagaradze
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Anna Monakova
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Nataliya Basalova
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia; Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir Popov
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia; Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Vadim Balabanyan
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Anastasia Efimenko
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia; Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
123
|
Davalieva K, Rusevski A, Velkov M, Noveski P, Kubelka-Sabit K, Filipovski V, Plaseski T, Dimovski A, Plaseska-Karanfilska D. Comparative proteomics analysis of human FFPE testicular tissues reveals new candidate biomarkers for distinction among azoospermia types and subtypes. J Proteomics 2022; 267:104686. [PMID: 35914715 DOI: 10.1016/j.jprot.2022.104686] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 10/16/2022]
Abstract
Understanding molecular mechanisms that underpin azoospermia and discovery of biomarkers that could enable reliable, non-invasive diagnosis is highly needed. Using label-free data-independent LC-MS/MS acquisition coupled with ion mobility, we compared the FFPE testicular proteome of patients with obstructive (OA) and non-obstructive azoospermia (NOA) subtypes hypospermatogenesis (Hyp) and Sertoli cell-only syndrome (SCO). Out of 2044 proteins identified based on ≥2 peptides, 61 proteins had the power to quantitatively discriminate OA from NOA and 30 to quantitatively discriminate SCO from Hyp and OA. Among these, H1-6, RANBP1 and TKTL2 showed superior potential for quantitative discrimination among OA, Hyp and SCO. Integrin signaling pathway, adherens junction, planar cell polarity/convergent extension pathway and Dectin-1 mediated noncanonical NF-kB signaling were significantly associated with the proteins that could discriminate OA from NOA. Comparison with 2 transcriptome datasets revealed 278 and 55 co-differentially expressed proteins/genes with statistically significant positive correlation. Gene expression analysis by qPCR of 6 genes (H1-6, RANBP1, TKTL2, TKTL1, H2BC1, and ACTL7B) with the highest discriminatory power on protein level and the same regulation trend with transcriptomic datasets, confirmed proteomics results. In summary, our results suggest some underlying pathways in azoospermia and broaden the range of potential novel candidates for diagnosis. SIGNIFICANCE: Using a comparative proteomics approach on testicular tissue we have identified several pathways associated with azoospermia and a number of testis-specific and germ cell-specific proteins that have the potential to pinpoint the type of spermatogenesis failure. Furthermore, comparison with transcriptomics datasets based on genome-wide gene expression analyses of human testis specimens from azoospermia patients identified proteins that could discriminate between obstructive and non-obstructive azoospermia subtypes on both protein and mRNA levels. Up to our knowledge, this is the first integrated comparative analysis of proteomics and transcriptomics data from testicular tissues. We believe that the data from our study contributes significantly to increase the knowledge of molecular mechanisms of azoospermia and pave the way for new investigations in regards to non-invasive diagnosis.
Collapse
Affiliation(s)
- Katarina Davalieva
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia, Macedonia.
| | - Aleksandar Rusevski
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia, Macedonia
| | - Milan Velkov
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia, Macedonia
| | - Predrag Noveski
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia, Macedonia
| | - Katerina Kubelka-Sabit
- Laboratory for Histopathology, Clinical Hospital "Sistina", 1000 Skopje, North Macedonia, Macedonia
| | - Vanja Filipovski
- Laboratory for Histopathology, Clinical Hospital "Sistina", 1000 Skopje, North Macedonia, Macedonia
| | - Toso Plaseski
- Faculty of Medicine, Endocrinology and Metabolic Disorders Clinic, 1000 Skopje, North Macedonia, Macedonia
| | - Aleksandar Dimovski
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia, Macedonia; Faculty of Pharmacy, University "St. Cyril and Methodius", 1000 Skopje, North Macedonia, Macedonia
| | - Dijana Plaseska-Karanfilska
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia, Macedonia.
| |
Collapse
|
124
|
Single-cell transcriptome atlas of the human corpus cavernosum. Nat Commun 2022; 13:4302. [PMID: 35879305 PMCID: PMC9314400 DOI: 10.1038/s41467-022-31950-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 07/11/2022] [Indexed: 11/30/2022] Open
Abstract
The corpus cavernosum is the most important structure for penile erection, and its dysfunction causes many physiological and psychological problems. However, its cellular heterogeneity and signalling networks at the molecular level are poorly understood because of limited access to samples. Here, we profile 64,993 human cavernosal single-cell transcriptomes from three males with normal erection and five organic erectile dysfunction patients. Cell communication analysis reveals that cavernosal fibroblasts are central to the paracrine signalling network and regulate microenvironmental homeostasis. Combining with immunohistochemical staining, we reveal the cellular heterogeneity and describe a detailed spatial distribution map for each fibroblast, smooth muscle and endothelial subcluster in the corpus cavernosum. Furthermore, comparative analysis and related functional experiments identify candidate regulatory signalling pathways in the pathological process. Our study provides an insight into the human corpus cavernosum microenvironment and a reference for potential erectile dysfunction therapies. The corpus cavernosum is the most important structure for penile erection, and its dysfunction causes physiological and psychological problems. Here the authors perform single-cell RNA-sequencing on corpus cavernosum samples from males with normal erection and erectile dysfunction patients, providing insights into this pathology.
Collapse
|
125
|
Liu Q, Chen X, Qiao J. Advances in studying human gametogenesis and embryonic development in China. Biol Reprod 2022; 107:12-26. [PMID: 35788258 DOI: 10.1093/biolre/ioac134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/21/2022] [Accepted: 06/20/2022] [Indexed: 11/12/2022] Open
Abstract
Reproductive medicine in China has developed rapidly since 1988 due to the support from the government and scientific exploration. However, the success rate of assisted reproduction technology (ART) is around 30-40% and many unknown "black boxes" in gametogenesis and embryo development are still present. With the development of single-cell and low-input sequencing technologies, the network of transcriptome and epigenetic regulation (DNA methylation, chromatin accessibility, and histone modifications) during the development of human primordial germ cells (PGCs), gametes and embryos has been investigated in depth. Furthermore, pre-implantation genetic testing (PGT) has also rapidly developed. In this review, we summarize and analyze China's outstanding progress in these fields.
Collapse
Affiliation(s)
- Qiang Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Xi Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.,Beijing Advanced Innovation Center for Genomics, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.,Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
126
|
Chen KQ, Wei BH, Hao SL, Yang WX. The PI3K/AKT signaling pathway: How does it regulate development of Sertoli cells and spermatogenic cells? Histol Histopathol 2022; 37:621-636. [PMID: 35388905 DOI: 10.14670/hh-18-457] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The PI3K/AKT signaling pathway is one of the most crucial regulatory mechanisms in animal cells, which can mainly regulate proliferation, survival and anti-apoptosis in cell lines. In the seminiferous epithelium, most studies were concentrated on the role of PI3K/AKT signaling in immature Sertoli cells (SCs) and spermatogonia stem cells (SSCs). PI3K/AKT signaling can facilitate the proliferation and anti-apoptosis of immature Sertoli cells and spermatogenic cells. Besides, in mature Sertoli cells, this pathway can disintegrate the structure of the blood-testis barrier (BTB) via regulatory protein synthesis and the cytoskeleton of Sertoli cells. All of these effects can directly and indirectly maintain and promote spermatogenesis in male testis.
Collapse
Affiliation(s)
- Kuang-Qi Chen
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bang-Hong Wei
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shuang-Li Hao
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
127
|
Wu D, Zhang K, Khan FA, Pandupuspitasari NS, Liang W, Huang C, Sun F. Smtnl2 regulates apoptotic germ cell clearance and lactate metabolism in mouse Sertoli cells. Mol Cell Endocrinol 2022; 551:111664. [PMID: 35551947 DOI: 10.1016/j.mce.2022.111664] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 12/25/2022]
Abstract
Smtnl2 is an epithelial Smoothelin that binds to actin filaments and is crucial for epithelial morphogenesis. We examined the role of Smtnl2 in Sertoli cells, which undergo dynamic cytoskeleton reorganization to phagocytose apoptotic germ cells, a process known as efferocytosis. We observed Smtnl2 expression in primary mouse Sertoli cells and the 15P1 Sertoli cell line. Smtnl2 expression increased in 15P1 cells committing efferocytosis. Smtnl2-deficient Sertoli cells exhibited defective ability to engulf apoptotic germ cells and importantly, the phenomenon occurred in the setting of an unaffected maturation of phagosome. We demonstrated that Smtnl2 regulates the engulfment process through the function of branched actin nucleation protein ARP3, an actin assembly dictator. Intriguingly, a shift in glucose metabolism that restricts lactate production in Sertoli cells was induced upon Smtnl2 depletion, leading to the activation of downstream AMPK and AKT signaling. Using an in vivo RNAi approach, we found that silencing of Smtnl2 in testis triggers an obvious disruption in cytoskeleton architecture and blood-testis barrier integrity across seminiferous epithelium, causing the detachment of massive germ cells from their nest, as evidenced by their exfoliation into the lumen. Overall, our study identifies Smtnl2 as a determinant for Sertoli cells' functioning in supporting spermatogenesis.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Faheem Ahmed Khan
- Department of Zoology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54782, Pakistan; Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | | | - Wangzhang Liang
- Department of Pathology, Second Hospital of Shanxi Medical University, Shanxi, 030001, China
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| |
Collapse
|
128
|
Yang C, Lin X, Ji Z, Huang Y, Zhang L, Luo J, Chen H, Li P, Tian R, Zhi E, Hong Y, Zhou Z, Zhang F, Li Z, Yao C. Novel bi-allelic variants in KASH5 are associated with meiotic arrest and non-obstructive azoospermia. Mol Hum Reprod 2022; 28:gaac021. [PMID: 35674372 DOI: 10.1093/molehr/gaac021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/11/2022] [Indexed: 11/12/2022] Open
Abstract
KASH5 is an essential component of the LINC (linker of the nucleoskeleton and cytoskeleton) complex that regulates chromosome movements and nuclear envelope (NE) remodeling in mouse spermatocytes during meiosis prophase I, but its expression and function in human cells, as well as its association with male infertility are largely unknown. In this study, a novel heterozygous copy number variation (CNV) (seq [GRCh37] del(19) (19q13.33) chr19: g.49894043-49903011del) and a heterozygous loss of function variant (NM_144688: c.979_980del: p.R327Sfs*21) in human KASH5 were identified in a non-obstructive azoospermia (NOA)-affected patient and in his infertile sister by whole-exome sequencing and CNV array. Spermatogenesis in the proband was arrested at zygotene-like stage with a deficiency in homolog pairing and synapsis. KASH5 protein expression in human spermatocytes was evaluated and reported first in this study. Single-cell RNA sequencing demonstrated that the LINC complex and associated genes in human and mouse shared a similar expression pattern, indicating a conserved mechanism in the regulation of chromosome movements and NE remodeling. Kash5 knockout mouse displayed similar phenotypes, including a meiotic arrest at a zygotene-like stage and impaired pairing and synapsis. Collectively, we have identified novel rare variants within human KASH5 in patients with NOA and meiosis arrest. Our study expands the knowledge of KASH5 and associated proteins in regulating human meiosis prophase I progress and provides new insight into the genetic etiology of NOA.
Collapse
Affiliation(s)
- Chao Yang
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiaoqi Lin
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), State Key Laboratory of Genetic Engineering, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Zhiyong Ji
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhua Huang
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Zhang
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Jiaqiang Luo
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huixing Chen
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Li
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruhui Tian
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Erlei Zhi
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Hong
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi Zhou
- School of Life Sciences and Technology, Shanghai Tech University, Shanghai, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), State Key Laboratory of Genetic Engineering, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Zheng Li
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chencheng Yao
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
129
|
Tan XH, Gu SJ, Tian WJ, Song WP, Gu YY, Yuan YM, Li XS, Xin ZC, Kim SW, Guan RL, Bae WJ. Extracellular vesicles derived from human Sertoli cells: characterizations, proteomic analysis, and miRNA profiling. Mol Biol Rep 2022; 49:4673-4681. [PMID: 35366759 DOI: 10.1007/s11033-022-07316-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 03/02/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Extracellular vesicles (EVs) contain thousands of proteins and nucleic acids, playing an important role in cell-cell communications. Sertoli cells have been essential in the testis as a "nurse cell". However, EVs derived from human Sertoli cells (HSerCs) have not been well investigated. METHODS EVs were isolated from HSerCs via ultracentrifugation and characterized by transmission electron microscopy, tunable resistive pulse sensing, and Western blotting. The cargo carried by HSerCs-EVs was measured via liquid chromatography-mass spectrometry and GeneChip miRNA Arrays. Bioinformatic analysis was performed to reveal potential functions of HSerCs-EVs. RESULTS A total of 860 proteins with no less than 2 unique peptides and 88 microRNAs with high signal values were identified in HSerCs-EVs. Biological processes related to molecular binding, enzyme activity, and regulation of cell cycle were significantly enriched. Specifically, many proteins in HSerCs-EVs were associated with spermatogenesis and regulation of immune system, including Septins, Large proline-rich protein BAG6, Clusterin, and Galectin-1. Moreover, abundant microRNAs within HSerCs-EVs (miR-638, miR-149-3p, miR-1246, etc.) had a possible impact on male reproductive disorders such as asthenozoospermia and oligozoospermia. CONCLUSIONS Our study has shown that HSerCs-EVs contain diverse components such as proteins and microRNAs. Further research is required to evaluate HSerCs-EVs in spermatogenesis, which are underutilized but highly potent resources with particular promise for male infertility.
Collapse
Affiliation(s)
- Xiao-Hui Tan
- Department of Urology, Peking University First Hospital, Beijing, 100034, People's Republic of China
- Institute of Urology, Peking University, Beijing, 100034, People's Republic of China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, People's Republic of China
| | - Sheng-Ji Gu
- Department of Urology, Peking University First Hospital, Beijing, 100034, People's Republic of China
- Institute of Urology, Peking University, Beijing, 100034, People's Republic of China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, People's Republic of China
| | - Wen-Jie Tian
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Catholic Integrative Medicine Research Institute, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Wen-Peng Song
- Institute of Urology, Peking University, Beijing, 100034, People's Republic of China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, People's Republic of China
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Yang-Yang Gu
- Institute of Urology, Peking University, Beijing, 100034, People's Republic of China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, People's Republic of China
- Department of Radiation Medicine, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, People's Republic of China
| | - Yi-Ming Yuan
- Department of Urology, Peking University First Hospital, Beijing, 100034, People's Republic of China
- Institute of Urology, Peking University, Beijing, 100034, People's Republic of China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, People's Republic of China
| | - Xue-Song Li
- Department of Urology, Peking University First Hospital, Beijing, 100034, People's Republic of China
- Institute of Urology, Peking University, Beijing, 100034, People's Republic of China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, People's Republic of China
| | - Zhong-Cheng Xin
- Department of Urology, Peking University First Hospital, Beijing, 100034, People's Republic of China
- Institute of Urology, Peking University, Beijing, 100034, People's Republic of China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, People's Republic of China
| | - Sae Woong Kim
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Catholic Integrative Medicine Research Institute, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Rui-Li Guan
- Department of Urology, Peking University First Hospital, Beijing, 100034, People's Republic of China.
- Institute of Urology, Peking University, Beijing, 100034, People's Republic of China.
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, People's Republic of China.
| | - Woong Jin Bae
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Catholic Integrative Medicine Research Institute, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
130
|
Liu H, Xue J, Li L, Mo H. Shenjing Guben Wan promotes sperm development by increasing the activity of seminiferous epithelium Sertoli cells. Transl Androl Urol 2022; 11:867-876. [PMID: 35812203 PMCID: PMC9262746 DOI: 10.21037/tau-22-381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/20/2022] [Indexed: 11/15/2022] Open
Abstract
Background Infertility is an important social problem. Asthenozoospermia (AZS) is a common pathological cause of male infertility, but its pathogenesis is unclear. Shenjing Guben Wan (SJGBW), a traditional Chinese medicine, has shown remarkable effects during the clinical treatment of oligozoospermia or AZS. Methods In this study, clinical evaluations were carried out on 184 AZS patients receiving SJGBW treatment, including sperm count, sperm quality, and pregnancy rate. Also, ornidazole was used to build an AZS mouse model, and SJGBW treatment was administered. The sperm quantity and fertility of mice in different groups were evaluated; a cholecystokinin octapeptide-8 (CCK-8) experiment was carried out to test the activity of seminiferous epithelium Sertoli cells, and immunohistochemistry and the Terminal Deoxynucleotidyl Transferase-mediated dUTP Nick-End Labeling (TUNEL) method were employed to test the pathological information and expression of the Sertoli cell surface marker in the testicular tissues of mice in each group. Results The sperm vitality, progressive sperm motility, and sperm morphology of patients who received SJGBW treatment were all improved (P<0.05). In the AZS group, the average sperm count, sperm vitality, pregnancy rate, and female mouse litters were all lower relative to mice in the control group. Following SJGBW treatment, the average sperm count, sperm vitality, pregnancy rate, and female mouse litters of mice in the AZS group were all significantly improved. The cytobiological experimental results showed that compared with the serum of normal male mice in the control group, the drug serum containing SJGBW could improve the cell vitality and proliferative ability of seminiferous epithelium Sertoli cells in AZS mice. Furthermore, the TUNEL results showed that the seminiferous tubule Sertoli cells and mesenchymal cells of the AZS mice exhibited the most significant apoptosis, which was alleviated following SJGBW treatment. Moreover, the levels of Sertoli cell marker, SOX9, and anti-apoptosis protein, Bcl2, in SJGBW-treated mice were both higher than that in AZS mice. Conclusions SJGBW can promote the development and maturation of germ cells by facilitating the proliferation of Sertoli cells in AZS patients, thereby improving the fertility of these patients.
Collapse
Affiliation(s)
- Handu Liu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Jianguo Xue
- Department of Andrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lipeng Li
- Department of Reproductive Medicine, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hui Mo
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| |
Collapse
|
131
|
Wu X, Zhou L, Shi J, Cheng CY, Sun F. Multiomics analysis of male infertility. Biol Reprod 2022; 107:118-134. [PMID: 35639635 DOI: 10.1093/biolre/ioac109] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/12/2022] [Accepted: 05/17/2022] [Indexed: 11/14/2022] Open
Abstract
Infertility affects 8-12% of couples globally, and the male factor is a primary cause in approximately 50% of couples. Male infertility is a multifactorial reproductive disorder, which can be caused by paracrine and autocrine factors, hormones, genes, and epigenetic changes. Recent studies in rodents and most notably in humans using multiomics approach have yielded important insights into understanding the biology of spermatogenesis. Nonetheless, the etiology and pathogenesis of male infertility are still largely unknown. In this review, we summarized and critically evaluated findings based on the use of advanced technologies to compare normal and obstructive azoospermia (OA) versus non-obstructive azoospermia (NOA) men, including whole-genome bisulfite sequencing (WGBS), single cell RNA-seq (scRNA-seq), whole exome sequencing (WES), and ATAC-seq. It is obvious that the multiomics approach is the method of choice for basic research and clinical studies including clinical diagnosis of male infertility.
Collapse
Affiliation(s)
- Xiaolong Wu
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China.,Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Liwei Zhou
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Jie Shi
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - C Yan Cheng
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China.,Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Fei Sun
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China.,Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| |
Collapse
|
132
|
Tang XJ, Xiao QH, Wang XL, He Y, Tian YN, Xia BT, Guo Y, Huang JL, Duan P, Tan Y. Single-Cell Transcriptomics-Based Study of Transcriptional Regulatory Features in the Non-Obstructive Azoospermia Testis. Front Genet 2022; 13:875762. [PMID: 35669193 PMCID: PMC9163961 DOI: 10.3389/fgene.2022.875762] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/04/2022] [Indexed: 11/18/2022] Open
Abstract
Non-obstructive azoospermia (NOA) is one of the most important causes of male infertility. Although many congenital factors have been identified, the aetiology in the majority of idiopathic NOA (iNOA) cases remains unknown. Herein, using single-cell RNA-Seq data sets (GSE149512) from the Gene Expression Omnibus (GEO) database, we constructed transcriptional regulatory networks (TRNs) to explain the mutual regulatory relationship and the causal relationship between transcription factors (TFs). We defined 10 testicular cell types by their marker genes and found that the proportion of Leydig cells (LCs) and macrophages (tMΦ) was significantly increased in iNOA testis. We identified specific TFs including LHX9, KLF8, KLF4, ARID5B and RXRG in iNOA LCs. In addition, we found specific TFs in iNOA tMΦ such as POU2F2, SPIB IRF5, CEBPA, ELK4 and KLF6. All these identified TFs are strongly engaged in cellular fate, function and homeostasis of the microenvironment. Changes in the activity of the above-mentioned TFs might affect the function of LCs and tMΦ and ultimately cause spermatogenesis failure. This study illustrate that these TFs play important regulatory roles in the occurrence and development of NOA.
Collapse
Affiliation(s)
- Xiao-juan Tang
- Department of Andrology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Qiao-hong Xiao
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Xue-lin Wang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Yan He
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
- Postgraduate Training Basement of Jinzhou Medicical University, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Ya-nan Tian
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
- Postgraduate Training Basement of Jinzhou Medicical University, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Bin-tong Xia
- Department of Urology Surgery, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Yang Guo
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Jiao-long Huang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Peng Duan
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Yan Tan
- Department of Andrology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
- Biomedical Engineering College, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
133
|
Marečková M, Massalha H, Lorenzi V, Vento-Tormo R. Mapping Human Reproduction with Single-Cell Genomics. Annu Rev Genomics Hum Genet 2022; 23:523-547. [PMID: 35567278 DOI: 10.1146/annurev-genom-120121-114415] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The trillions of cells in the human body develop as a result of the fusion of two extremely specialized cells: an oocyte and a sperm. This process is essential for the continuation of our species, as it ensures that parental genetic information is mixed and passed on from generation to generation. In addition to producing oocytes, the female reproductive system must provide the environment for the appropriate development of the fetus until birth. New genomic and computational tools offer unique opportunities to study the tight spatiotemporal regulatory mechanisms that are required for the cycle of human reproduction. This review explores how single-cell technologies have been used to build cellular atlases of the human reproductive system across the life span and how these maps have proven useful to better understand reproductive pathologies and dissect the heterogeneity of in vitro model systems. Expected final online publication date for the Annual Review of Genomics and Human Genetics, Volume 23 is October 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Magda Marečková
- Wellcome Sanger Institute, Cambridge, United Kingdom; .,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom;
| | - Hassan Massalha
- Wellcome Sanger Institute, Cambridge, United Kingdom; .,Theory of Condensed Matter Group, Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | | | | |
Collapse
|
134
|
Tang D, Li K, Lv M, Xu C, Geng H, Wang C, Cheng H, He X, Zhang Y, Cao Y. Altered mRNAs Profiles in the Testis of Patients With "Secondary Idiopathic Non-Obstructive Azoospermia". Front Cell Dev Biol 2022; 10:824596. [PMID: 35646930 PMCID: PMC9133692 DOI: 10.3389/fcell.2022.824596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Non-obstructive azoospermia (NOA) is the most severe form of male infertility. Currently, known causative factors, including congenital and several acquired causes only account for approximately 30% of NOA cases. The causes for NOA remain unclear for most patients, which is known as idiopathic (iNOA). However, whether iNOA is due to congenital defects or acquired abnormalities is a confusing problem due to the delayed diagnosis of this frustrating condition until the childbearing age. Therefore, we collected several cases with "secondary idiopathic NOA" and detected the altered mRNAs profiles in the testicular tissues to explore the possible molecular basis. Materials and Methods: In this study, several patients with a previous history of natural pregnancy with their partners before, who were diagnosed as iNOA based on the outcomes of routine semen analysis and multiple testis biopsies now, were enrolled. Some known risk factors and genetic factors were excluded. Therefore, we defined this phenotype as "secondary idiopathic NOA." To explore the possible molecular basis of this disease, we performed mRNA expression analysis through next-generation sequencing on three cases and other three patients with obstructive azoospermia as controls. Bioinformatics analyses were conducted to assess differentially expressed genes and possible biological mechanisms involved in the disease. Quantitative real-time reverse transcription polymerase chain reaction assays were applied to confirm the results in several selected mRNAs involved in stages and metabolism of Sertoli cells. Results: A series of mRNAs were found to be altered in testicular tissues between patients with "secondary idiopathic NOA" and controls, including 6,028 downregulated and 3,402 upregulated mRNAs. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genome (KEGG) analyses revealed a range of GO and KEGG terms, such as cellular process involved in reproduction, protein degradation, and absorption. Conclusion: The present study introduces a novel classification called "secondary idiopathic NOA." We provide a global view of the altered mRNAs involved in spermatogenetic failure in these cases. Regarding the limited samples, further studies should be taken to understand this new classification.
Collapse
Affiliation(s)
- Dongdong Tang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Kuokuo Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Mingrong Lv
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Chuan Xu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hao Geng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chao Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huiru Cheng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Xiaojin He
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Yan Zhang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, WuHan, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| |
Collapse
|
135
|
Nie X, Munyoki SK, Sukhwani M, Schmid N, Missel A, Emery BR, DonorConnect, Stukenborg JB, Mayerhofer A, Orwig KE, Aston KI, Hotaling JM, Cairns BR, Guo J. Single-cell analysis of human testis aging and correlation with elevated body mass index. Dev Cell 2022; 57:1160-1176.e5. [PMID: 35504286 PMCID: PMC9090997 DOI: 10.1016/j.devcel.2022.04.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/12/2022] [Accepted: 04/04/2022] [Indexed: 01/15/2023]
Abstract
Aging men display reduced reproductive health; however, testis aging is poorly understood at the molecular and genomic levels. Here, we utilized single-cell RNA-seq to profile over 44,000 cells from both young and older men and examined age-related changes in germline development and in the testicular somatic cells. Age-related changes in spermatogonial stem cells appeared modest, whereas age-related dysregulation of spermatogenesis and somatic cells ranged from moderate to severe. Altered pathways included signaling and inflammation in multiple cell types, metabolic signaling in Sertoli cells, hedgehog signaling and testosterone production in Leydig cells, cell death and growth in testicular peritubular cells, and possible developmental regression in both Leydig and peritubular cells. Remarkably, the extent of dysregulation correlated with body mass index in older but not in younger men. Collectively, we reveal candidate molecular mechanisms underlying the complex testicular changes conferred by aging and their possible exacerbation by concurrent chronic conditions such as obesity.
Collapse
Affiliation(s)
- Xichen Nie
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Sarah K Munyoki
- Department of Obstetrics, Gynecology and Reproductive Sciences and Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Meena Sukhwani
- Department of Obstetrics, Gynecology and Reproductive Sciences and Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Nina Schmid
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilians-University (LMU), Grosshaderner Strasse 9, Planegg, Munich, 82152, Germany
| | - Annika Missel
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilians-University (LMU), Grosshaderner Strasse 9, Planegg, Munich, 82152, Germany
| | - Benjamin R Emery
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | | | - Jan-Bernd Stukenborg
- NORDFERTIL Research Laboratory Stockholm, Childhood Cancer Research Unit, Bioclinicum J9:30, Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Solna 17164, Sweden
| | - Artur Mayerhofer
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilians-University (LMU), Grosshaderner Strasse 9, Planegg, Munich, 82152, Germany
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences and Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Kenneth I Aston
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - James M Hotaling
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Bradley R Cairns
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| | - Jingtao Guo
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
136
|
Yang CX, Yang YW, Mou Q, Chen L, Wang C, Du ZQ. Proteomic changes induced by ascorbic acid treatment on porcine immature Sertoli cells. Theriogenology 2022; 188:13-21. [DOI: 10.1016/j.theriogenology.2022.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/24/2022] [Accepted: 05/13/2022] [Indexed: 01/08/2023]
|
137
|
Peng T, Liao C, Ye X, Chen Z, Lan Y, Fu X, An G. Gonadotropins treatment prior to microdissection testicular sperm extraction in non-obstructive azoospermia: a single-center cohort study. Reprod Biol Endocrinol 2022; 20:61. [PMID: 35365173 PMCID: PMC8973804 DOI: 10.1186/s12958-022-00934-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Microdissection testicular sperm extraction (micro-TESE) in combination with ICSI can make paternity possible for non-obstructive azoospermia (NOA) patients. Testicular sperm can be successfully retrieved in nearly half of NOA patients. Nevertheless, not many convincing protocols are established to improve sperm retrieval rate (SRR). The goal of this study was to evaluate whether gonadotropins therapy before micro-TESE could improve sperm retrieval rate and affect the ICSI outcomes in non-obstructive azoospermia patients with hypergonadotropic hypogonadism. METHODS This retrospective cohort study included a total of 569 non-obstructive azoospermia men who underwent micro-TESE with or without 3-month of preoperative hCG / hCG plus highly purified urinary FSH (uFSH) between January 2016 and December 2019. The primary outcome was the sperm retrieval rate of micro-TESE. RESULTS Sperm was found in 27 patients among 395 NOA men who accepted preoperative gonadotropins treatment (6.8%, 27/395) in post-treatment semen analysis for ICSI. One hundred forty nine out of 542 patients could successfully obtain enough sperm for ICSI through the micro-TESE (overall SRR = 27.5%). There was a statistically significant difference in the SRR between the preoperative gonadotropins treatment and non-gonadotropins treatment groups (31.2%, 115/368 vs. 19.5%, 34/174, P = 0.006). In the multivariable analysis with IPTW according to the propensity score, there was a significant association between preoperative gonadotropins treatment and the SRR (OR, 1.59; 95% CI: 1.02-2.52; P = 0.042). No differences in the clinical pregnancy rate, live birth delivery rate, or miscarriage rate were observed between the two groups. CONCLUSION Preoperative gonadotropins therapy seems to have a role in improving SRR in NOA patients with hypergonadotropic hypogonadism. We found that gonadotropins therapy had no effect on ICSI clinical outcomes and live birth.
Collapse
Affiliation(s)
- Tianwen Peng
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Chen Liao
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Xin Ye
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Zhicong Chen
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Yu Lan
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Xin Fu
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Geng An
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China.
| |
Collapse
|
138
|
Wang S, Qian Z, Ge X, Li C, Xue M, Liang K, Ma R, Ouyang L, Zheng L, Jing J, Cao S, Zhang Y, Yang Y, Chen Y, Ma J, Yao B. LncRNA Tug1 maintains blood-testis barrier integrity by modulating Ccl2 expression in high-fat diet mice. Cell Mol Life Sci 2022; 79:114. [PMID: 35103851 PMCID: PMC11073184 DOI: 10.1007/s00018-022-04142-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 01/02/2023]
Abstract
Sertoli cells are essential for spermatogenesis in the testicular seminiferous tubules by forming blood-testis barrier (BTB) and creating a unique microenvironment for spermatogenesis. Many lncRNAs have been reported to participate in spermatogenesis. However, the role of long noncoding RNAs (lncRNAs) in Sertoli cells has rarely been examined. Herein, we found that a high-fat diet (HFD) decreased sperm quality, impaired BTB integrity and resulted in accumulation of saturated fatty acids (SFAs), especially palmitic acid (PA), in mouse testes. PA decreased the expression of tight junction (TJ)-related proteins, increased permeability and decreased transepithelial electrical resistance (TER) in primary Sertoli cells and TM4 cells. Moreover, lncRNA Tug1 was found to be involved in PA-induced BTB disruption by RNA-seq. Tug1 depletion distinctly impaired the TJs of Sertoli cells and overexpression of Tug1 alleviated the disruption of BTB integrity induced by PA. Moreover, Ccl2 was found to be a downstream target of Tug1, and decreased TJ-related protein levels and TER and increased FITC-dextran permeability in vitro. Furthermore, the addition of Ccl2 damaged BTB integrity after overexpression of Tug1 in the presence of PA. Mechanistically, we found that Tug1 could directly bind to EZH2 and regulate H3K27me3 occupancy in the Ccl2 promoter region by RNA immunoprecipitation and chromatin immunoprecipitation assays. Our study revealed an important role of Tug1 in the BTB integrity of Sertoli cells and provided a new view of the role of lncRNAs in male infertility.
Collapse
Affiliation(s)
- Shuxian Wang
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Zhang Qian
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Xie Ge
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Chuwei Li
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Mengqi Xue
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Kuan Liang
- Center of Reproductive Medicine, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, Jiangsu, China
| | - Rujun Ma
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Lei Ouyang
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Lu Zheng
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Jun Jing
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Siyuan Cao
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Yu Zhang
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Yang Yang
- Basic Medical Laboratory, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Yabing Chen
- Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, China
| | - Jinzhao Ma
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China.
| | - Bing Yao
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, 210002, Jiangsu, China.
- Center of Reproductive Medicine, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, Jiangsu, China.
| |
Collapse
|
139
|
Chen W, Li Z, Zhu ZJ, Wang YZ, Wang XB, Yao CC, Zhao LY, Zhang ZB, Wu Y. Novel mutation in ODF2 causes multiple morphological abnormalities of the sperm flagella in an infertile male. Asian J Androl 2022; 24:463-472. [PMID: 35102900 PMCID: PMC9491046 DOI: 10.4103/aja202183] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Numerous genes have been associated with multiple morphological abnormalities of the sperm flagella (MMAF), which cause severe asthenozoospermia and lead to male infertility, while the causes of approximately 50% of MMAF cases remain unclear. To reveal the genetic causes of MMAF in an infertile patient, whole-exome sequencing was performed to screen for pathogenic genes, and electron microscope was used to reveal the sperm flagellar ultrastructure. A novel heterozygous missense mutation in the outer dense fiber protein 2 (ODF2) gene was detected, which was inherited from the patient's mother and predicted to be potentially damaging. Transmission electron microscopy revealed that the outer dense fibers were defective in the patient's sperm tail, which was similar to that of the reported heterozygous Odf2 mutation mouse. Immunostaining of ODF2 showed severe ODF2 expression defects in the patient's sperm. Therefore, it was concluded that the heterozygous mutation in ODF2 caused MMAF in this case. To evaluate the possibility of assisted reproductive technology (ART) treatment for this patient, intracytoplasmic sperm injection (ICSI) was performed, with the help of a hypo-osmotic swelling test and laser-assisted immotile sperm selection (LAISS) for available sperm screening, and artificial oocyte activation with ionomycin was applied to improve the fertilization rate. Four ICSI cycles were performed, and live birth was achieved in the LAISS-applied cycle, suggesting that LAISS would be valuable in ART treatment for MMAF.
Collapse
|
140
|
Wright WW. The Regulation of Spermatogonial Stem Cells in an Adult Testis by Glial Cell Line-Derived Neurotrophic Factor. Front Endocrinol (Lausanne) 2022; 13:896390. [PMID: 35721702 PMCID: PMC9203831 DOI: 10.3389/fendo.2022.896390] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/11/2022] [Indexed: 12/05/2022] Open
Abstract
This review focuses on the in vivo regulation of spermatogonial stem cells (SSCs) in adult testes by glial cell line-derived neurotrophic factor (GDNF). To study adult mouse testes, we reversibly inhibited GDNF stimulation of SSCs via a chemical-genetic approach. This inhibition diminishes replication and increases differentiation of SSCs, and inhibition for 9 days reduces transplantable SSC numbers by 90%. With more sustained inhibition, all SSCs are lost, and testes eventually resemble human testes with Sertoli cell-only (SCO) syndrome. This resemblance prompted us to ask if GDNF expression is abnormally low in these infertile human testes. It is. Expression of FGF2 and FGF8 is also reduced, but some SCO testes contain SSCs. To evaluate the possible rebuilding of an SSC pool depleted due to inadequate GDNF signaling, we inhibited and then restored signaling to mouse SSCs. Partial rebuilding occurred, suggesting GDNF as therapy for men with SCO syndrome.
Collapse
|
141
|
Hofmann MC, McBeath E. Sertoli Cell-Germ Cell Interactions Within the Niche: Paracrine and Juxtacrine Molecular Communications. Front Endocrinol (Lausanne) 2022; 13:897062. [PMID: 35757413 PMCID: PMC9226676 DOI: 10.3389/fendo.2022.897062] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/25/2022] [Indexed: 12/22/2022] Open
Abstract
Male germ cell development depends on multiple biological events that combine epigenetic reprogramming, cell cycle regulation, and cell migration in a spatio-temporal manner. Sertoli cells are a crucial component of the spermatogonial stem cell niche and provide essential growth factors and chemokines to developing germ cells. This review focuses mainly on the activation of master regulators of the niche in Sertoli cells and their targets, as well as on novel molecular mechanisms underlying the regulation of growth and differentiation factors such as GDNF and retinoic acid by NOTCH signaling and other pathways.
Collapse
|
142
|
Liu H, Deng M, Zhu Y, Wu D, Tong X, Li L, Wang L, Xu F, Wang T. Establishment of an oligoasthenospermia mouse model based on TAp73 gene suppression. Animal Model Exp Med 2021; 4:351-358. [PMID: 34977486 PMCID: PMC8690982 DOI: 10.1002/ame2.12186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/20/2021] [Accepted: 10/20/2021] [Indexed: 11/11/2022] Open
Abstract
Background Oligoasthenospermia is one of the main causes of male infertility. Researchers usually use chemical drugs to directly damage germ cells to prepare oligoasthenospermia models, which disregards the adhesion and migration between spermatogenic cells and Sertoli cells. TAp73 is a critical regulator of the adhesin of germ cell; thus, we sought to explore a novel oligoasthenospermia model based on TAp73 gene suppression. Methods Mice in the Pifithrin-α group were injected intraperitoneally with 2.5 mg/kg Pifithrin-α (TAp73 inhibitor) daily for 30 consecutive days. Reproductive hormone levels and epididymal sperm quality, as well as the network morphology of Sertoli cells were tested. Results Sperm density, motility, and the relative protein and mRNA expression of TAp73 and Nectin 2 were obviously decreased in the Pifithrin-α group compared with the normal control group. No significant distinction was observed in the relative mRNA and protein expression of ZO-1. Furthermore, the tight junctions (TJs) and apical ectoplasmic specialization (ES) were destroyed in the Pifithrin-α group. Conclusion The above results indicate that we successfully established a new oligoasthenospermia mouse model. This study provides a foundation for further exploration of the roles of TAp73 genes during spermatogenesis and provides new research objects for further oligospermia research and future drug discovery.
Collapse
Affiliation(s)
- Hong‐Juan Liu
- School of Integrated Traditional Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Meng‐Yun Deng
- School of Integrated Traditional Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Yan‐Yan Zhu
- School of Integrated Traditional Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - De‐Ling Wu
- Anhui Province Key Laboratory of Chinese Medical FormulaSchool of PharmacyAnhui University of Chinese MedicineHefeiChina
| | - Xiao‐Hui Tong
- School of Integrated Traditional Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Li Li
- School of Integrated Traditional Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Lei Wang
- School of Integrated Traditional Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Fei Xu
- School of Integrated Traditional Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Tong‐Sheng Wang
- School of Integrated Traditional Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
- Anhui Province Key Laboratory of Chinese Medical FormulaSchool of PharmacyAnhui University of Chinese MedicineHefeiChina
| |
Collapse
|
143
|
Hu J, Othmane B, Yu A, Li H, Cai Z, Chen X, Ren W, Chen J, Zu X. 5mC regulator-mediated molecular subtypes depict the hallmarks of the tumor microenvironment and guide precision medicine in bladder cancer. BMC Med 2021; 19:289. [PMID: 34836536 PMCID: PMC8627095 DOI: 10.1186/s12916-021-02163-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/18/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Depicting the heterogeneity and functional characteristics of the tumor microenvironment (TME) is necessary to achieve precision medicine for bladder cancer (BLCA). Although classical molecular subtypes effectively reflect TME heterogeneity and characteristics, their clinical application is limited by several issues. METHODS In this study, we integrated the Xiangya cohort and multiple external BLCA cohorts to develop a novel 5-methylcytosine (5mC) regulator-mediated molecular subtype system and a corresponding quantitative indicator, the 5mC score. Unsupervised clustering was performed to identify novel 5mC regulator-mediated molecular subtypes. The principal component analysis was applied to calculate the 5mC score. Then, we correlated the 5mC clusters (5mC score) with classical molecular subtypes, immunophenotypes, clinical outcomes, and therapeutic opportunities in BLCA. Finally, we performed pancancer analyses on the 5mC score. RESULTS Two 5mC clusters, including 5mC cluster 1 and cluster 2, were identified. These novel 5mC clusters (5mC score) could accurately predict classical molecular subtypes, immunophenotypes, prognosis, and therapeutic opportunities of BLCA. 5mC cluster 1 (high 5mC score) indicated a luminal subtype and noninflamed phenotype, characterized by lower anticancer immunity but better prognosis. Moreover, 5mC cluster 1 (high 5mC score) predicted low sensitivity to cancer immunotherapy, neoadjuvant chemotherapy, and radiotherapy, but high sensitivity to antiangiogenic therapy and targeted therapies, such as blocking the β-catenin, FGFR3, and PPAR-γ pathways. CONCLUSIONS The novel 5mC regulator-based subtype system reflects many aspects of BLCA biology and provides new insights into precision medicine in BLCA. Furthermore, the 5mC score may be a generalizable predictor of immunotherapy response and prognosis in pancancers.
Collapse
Affiliation(s)
- Jiao Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Belaydi Othmane
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Anze Yu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Immunobiology & Transplant Science Center, Houston Methodist Research Institute, Texas Medical Center, Houston, TX, 77030, USA
| | - Huihuang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhiyong Cai
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xu Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute for Infection Prevention and Hospital Epidemiology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Wenbiao Ren
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Jinbo Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
144
|
Zhou R, Lv X, Chen T, Chen Q, Tian H, Yang C, Guo W, Liu C. Construction and external validation of a 5-gene random forest model to diagnose non-obstructive azoospermia based on the single-cell RNA sequencing of testicular tissue. Aging (Albany NY) 2021; 13:24219-24235. [PMID: 34738918 PMCID: PMC8610122 DOI: 10.18632/aging.203675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/28/2021] [Indexed: 01/02/2023]
Abstract
Non-obstructive azoospermia (NOA) is among the most severe factors for male infertility, but our understandings of the latent biological mechanisms remain insufficient. The single-cell RNA sequencing (scRNA-seq) data of 432 testicular cells isolated from the patient with NOA was analyzed, and the cell samples were grouped into 5 cell clusters. A sum of 455 cell markers was identified and then included in the protein-protein interaction network. The Top 5 most critical genes in the network, including CCT8, CDC6, PSMD1, RPS4X, RPL36A, were selected for the diagnosis model construction through the random forest (RF). The RF model was a strong classifier for NOA and obstructive azoospermia (OA), which was validated in the training cohort (n = 58, AUC = 1) and external validation cohort (n = 20, AUC = 0.9). We collected the seminal plasma samples and testicular biopsy samples from 20 OA and 20 NOA cases from the local hospital, and the gene expression was detected via Real-Time quantitative Polymerase Chain Reaction (RT-qPCR) and Immunohistochemistry. The RF model also exhibited high accuracy (AUC = 0.725) in the local cohort. In summary, a novel gene signature was developed and externally validated based on scRNA-seq analysis, providing some new biomarkers to uncover the underlying mechanisms and a promising clinical tool for diagnosis in NOA.
Collapse
Affiliation(s)
- Ranran Zhou
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xianyuan Lv
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Tianle Chen
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Qi Chen
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hu Tian
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Cheng Yang
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wenbin Guo
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Cundong Liu
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
145
|
Mahyari E, Guo J, Lima AC, Lewinsohn DP, Stendahl AM, Vigh-Conrad KA, Nie X, Nagirnaja L, Rockweiler NB, Carrell DT, Hotaling JM, Aston KI, Conrad DF. Comparative single-cell analysis of biopsies clarifies pathogenic mechanisms in Klinefelter syndrome. Am J Hum Genet 2021; 108:1924-1945. [PMID: 34626582 DOI: 10.1016/j.ajhg.2021.09.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/31/2021] [Indexed: 12/25/2022] Open
Abstract
Klinefelter syndrome (KS), also known as 47, XXY, is characterized by a distinct set of physiological abnormalities, commonly including infertility. The molecular basis for Klinefelter-related infertility is still unclear, largely because of the cellular complexity of the testis and the intricate endocrine and paracrine signaling that regulates spermatogenesis. Here, we demonstrate an analysis framework for dissecting human testis pathology that uses comparative analysis of single-cell RNA-sequencing data from the biopsies of 12 human donors. By comparing donors from a range of ages and forms of infertility, we generate gene expression signatures that characterize normal testicular function and distinguish clinically distinct forms of male infertility. Unexpectedly, we identified a subpopulation of Sertoli cells within multiple individuals with KS that lack transcription from the XIST locus, and the consequence of this is increased X-linked gene expression compared to all other KS cell populations. By systematic assessment of known cell signaling pathways, we identify 72 pathways potentially active in testis, dozens of which appear upregulated in KS. Altogether our data support a model of pathogenic changes in interstitial cells cascading from loss of X inactivation in pubertal Sertoli cells and nominate dosage-sensitive factors secreted by Sertoli cells that may contribute to the process. Our findings demonstrate the value of comparative patient analysis in mapping genetic mechanisms of disease and identify an epigenetic phenomenon in KS Sertoli cells that may prove important for understanding causes of infertility and sex chromosome evolution.
Collapse
|
146
|
Integration and gene co-expression network analysis of scRNA-seq transcriptomes reveal heterogeneity and key functional genes in human spermatogenesis. Sci Rep 2021; 11:19089. [PMID: 34580317 PMCID: PMC8476490 DOI: 10.1038/s41598-021-98267-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023] Open
Abstract
Spermatogenesis is a complex process of cellular division and differentiation that begins with spermatogonia stem cells and leads to functional spermatozoa production. However, many of the molecular mechanisms underlying this process remain unclear. Single-cell RNA sequencing (scRNA-seq) is used to sequence the entire transcriptome at the single-cell level to assess cell-to-cell variability. In this study, more than 33,000 testicular cells from different scRNA-seq datasets with normal spermatogenesis were integrated to identify single-cell heterogeneity on a more comprehensive scale. Clustering, cell type assignments, differential expressed genes and pseudotime analysis characterized 5 spermatogonia, 4 spermatocyte, and 4 spermatid cell types during the spermatogenesis process. The UTF1 and ID4 genes were introduced as the most specific markers that can differentiate two undifferentiated spermatogonia stem cell sub-cellules. The C7orf61 and TNP can differentiate two round spermatid sub-cellules. The topological analysis of the weighted gene co-expression network along with the integrated scRNA-seq data revealed some bridge genes between spermatogenesis's main stages such as DNAJC5B, C1orf194, HSP90AB1, BST2, EEF1A1, CRISP2, PTMS, NFKBIA, CDKN3, and HLA-DRA. The importance of these key genes is confirmed by their role in male infertility in previous studies. It can be stated that, this integrated scRNA-seq of spermatogenic cells offers novel insights into cell-to-cell heterogeneity and suggests a list of key players with a pivotal role in male infertility from the fertile spermatogenesis datasets. These key functional genes can be introduced as candidates for filtering and prioritizing genotype-to-phenotype association in male infertility.
Collapse
|
147
|
Vitrification with microinjection of single seminiferous tubules: an efficient cryopreservation approach for limited testicular tissue. Reprod Biomed Online 2021; 43:687-699. [PMID: 34556414 DOI: 10.1016/j.rbmo.2021.06.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/08/2021] [Accepted: 06/28/2021] [Indexed: 11/23/2022]
Abstract
RESEARCH QUESTION Is vitrification with microinjection of single seminiferous tubules an efficient cryopreservation approach for limited testicular tissue? DESIGN Testicular tissue from 10 patients with normal spermatogenesis were assigned to a fresh control group or one of the following cryopreservation procedures: uncontrolled slow freezing (USF) using either 1.5 or 2.1 M DMSO combined with sucrose and vitrification with or without single seminiferous tubules microinjection. RESULTS Single seminiferous tubules microinjected with cryoprotective agents (CPA) enhanced the penetration of CPA compared with CPA-treated testicular tissue fragments. Microinjection of seminiferous tubules (VLP) maintained tubule structural integrity and germ cell numbers, and reduced spermatogonial apoptosis after cryopreservation compared with vitrification without microinjection (apoptosis rate: VLP versus vitrification without microinjection, P = 0.047; VLP versus USF, P= 0.049). Freezing of single seminiferous tubules using 0.25-ml straws and traditional sperm freezing methods protected sperm retrieval and recovery rates, and the progressive motility index. CONCLUSIONS Vitrification of single seminiferous tubule with microinjection of low CPA concentration is an effective approach to testicular cryopreservation.
Collapse
|
148
|
Ji C, Wang Y, Wei X, Zhang X, Cong R, Yao L, Qin C, Song N. Potential of testis-derived circular RNAs in seminal plasma to predict the outcome of microdissection testicular sperm extraction in patients with idiopathic non-obstructive azoospermia. Hum Reprod 2021; 36:2649-2660. [PMID: 34477868 DOI: 10.1093/humrep/deab196] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION Do testis-derived circular RNAs (circRNAs) in seminal plasma have potential as biomarkers to predict the outcome of microdissection testicular sperm extraction (micro-TESE) in patients with idiopathic non-obstructive azoospermia (NOA)? SUMMARY ANSWER Testis-derived circRNAs in the seminal plasma can indeed be used for predicting the outcome of micro-TESE in patients with idiopathic NOA. WHAT IS KNOWN ALREADY Micro-TESE is an effective method to obtain sperm samples from patients with idiopathic NOA. However, its success rate is only 40-50% in such patients. STUDY DESIGN, SIZE, DURATION Six idiopathic NOA patients with different micro-TESE results were included as the discovery cohort. Their testicular tissues were used for extracting and sequencing circRNAs. Five circRNAs with the most significantly different expression levels were selected for further verification. PARTICIPANTS/MATERIALS, SETTING, METHODS Fifty-two patients with idiopathic NOA were included as the validation cohort. Preoperative seminal plasma samples of 52 patients with idiopathic NOA and 25 intraoperative testicular tissues were collected and divided into 'success' and 'failure' groups according to the results of micro-TESE. Quantitative real-time polymerase chain reaction was performed to verify differences in the expression levels of the selected circRNAs between the two groups in the testicular tissues and seminal plasma. MAIN RESULTS AND THE ROLE OF CHANCE Whether at the seminal plasma or testicular tissue level, the differences in the expression levels of the three circRNAs (hsa_circ_0000277, hsa_circ_0060394 and hsa_circ_0007773) between the success and failure groups were consistent with the sequencing results. A diagnostic receiver operating curve (ROC) analysis of the AUC indicated excellent diagnostic performance of these circRNAs in seminal plasma in predicting the outcome of micro-TESE (AUC values: 0.920, 0.928 and 0.891, respectively). On the basis of least absolute shrinkage and selection operator (LASSO) logistic regression, the three circRNAs were combined to construct a new prediction model. The diagnostic ROC curve analysis of the model showed an AUC value of 0.958. The expression levels of these circRNAs in seminal plasma using three normospermic volunteer samples remained stable after 48 h at room temperature. LARGE SCALE DATA NA. LIMITATIONS, REASONS FOR CAUTION This was a single-center retrospective study with relatively few cases. The functions of these circRNAs, as well as their relationship with spermatogenesis, have not yet been established. WIDER IMPLICATIONS OF THE FINDINGS Testis-derived circRNAs in seminal plasma can reflect the microenvironment of the testis and can be used as reliable biomarkers to screen patients with idiopathic NOA who might be suitable for micro-TESE. STUDY FUNDING/COMPETING INTEREST(S) This article was funded by the National Natural Science Foundation of China (Grant no. 81871151). There were no competing interests.
Collapse
Affiliation(s)
- Chengjian Ji
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yichun Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiyi Wei
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xingyu Zhang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Rong Cong
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Liangyu Yao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chao Qin
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ninghong Song
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Department of Urology, The Affiliated Kezhou People's Hospital of Nanjing Medical University, Xinjiang, China
| |
Collapse
|
149
|
Zhang Y, Liu T, Hu X, Wang M, Wang J, Zou B, Tan P, Cui T, Dou Y, Ning L, huang Y, Rao S, Wang D, Zhao X. CellCall: integrating paired ligand-receptor and transcription factor activities for cell-cell communication. Nucleic Acids Res 2021; 49:8520-8534. [PMID: 34331449 PMCID: PMC8421219 DOI: 10.1093/nar/gkab638] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/24/2021] [Accepted: 07/16/2021] [Indexed: 12/14/2022] Open
Abstract
With the dramatic development of single-cell RNA sequencing (scRNA-seq) technologies, the systematic decoding of cell-cell communication has received great research interest. To date, several in-silico methods have been developed, but most of them lack the ability to predict the communication pathways connecting the insides and outsides of cells. Here, we developed CellCall, a toolkit to infer inter- and intracellular communication pathways by integrating paired ligand-receptor and transcription factor (TF) activity. Moreover, CellCall uses an embedded pathway activity analysis method to identify the significantly activated pathways involved in intercellular crosstalk between certain cell types. Additionally, CellCall offers a rich suite of visualization options (Circos plot, Sankey plot, bubble plot, ridge plot, etc.) to present the analysis results. Case studies on scRNA-seq datasets of human testicular cells and the tumor immune microenvironment demonstrated the reliable and unique functionality of CellCall in intercellular communication analysis and internal TF activity exploration, which were further validated experimentally. Comparative analysis of CellCall and other tools indicated that CellCall was more accurate and offered more functions. In summary, CellCall provides a sophisticated and practical tool allowing researchers to decipher intercellular communication and related internal regulatory signals based on scRNA-seq data. CellCall is freely available at https://github.com/ShellyCoder/cellcall.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tianyuan Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xuesong Hu
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Mei Wang
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jing Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Bohao Zou
- Department of Statistics, University of California Davis, Davis, CA, USA
| | - Puwen Tan
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tianyu Cui
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yiying Dou
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lin Ning
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan huang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shuan Rao
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Dong Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyang Zhao
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, China
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
150
|
Aging, inflammation and DNA damage in the somatic testicular niche with idiopathic germ cell aplasia. Nat Commun 2021; 12:5205. [PMID: 34471128 PMCID: PMC8410861 DOI: 10.1038/s41467-021-25544-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 08/18/2021] [Indexed: 12/18/2022] Open
Abstract
Molecular mechanisms associated with human germ cell aplasia in infertile men remain undefined. Here we perform single-cell transcriptome profiling to highlight differentially expressed genes and pathways in each somatic cell type in testes of men with idiopathic germ cell aplasia. We identify immaturity of Leydig cells, chronic tissue inflammation, fibrosis, and senescence phenotype of the somatic cells, as well markers of chronic inflammation in the blood. We find that deregulated expression of parentally imprinted genes in myoid and immature Leydig cells, with relevant changes in the ratio of Lamin A/C transcripts and an active DNA damage response in Leydig and peritubular myoid cells are also indicative of senescence of the testicular niche. This study offers molecular insights into the pathogenesis of idiopathic germ cell aplasia. Molecular mechanisms associated with human germ cell aplasia in infertile men remain undefined. Here the authors perform single-cell transcriptome profiling to highlight differentially expressed genes and pathways in each somatic cell type in testes of men with idiopathic germ cell aplasia.
Collapse
|