1
|
Gong J, Lv Y, Meng Y, Zhang W, Jiang X, Xiao M. Effects of prenatal stress on reproductive function of male offspring through the KISS1 system. Endocr Connect 2024; 13:e240027. [PMID: 39140811 PMCID: PMC11466243 DOI: 10.1530/ec-24-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 08/14/2024] [Indexed: 08/15/2024]
Abstract
Prenatal stress can lead to the programming of the neuroendocrine system in male offspring, disrupting the hypothalamic testicular axis and adversely affecting the reproductive health of male offspring. This study aimed to determine the long-term effects of prenatal stress on the KISS1 system in male offspring and the effects on reproductive function in male offspring. Sixteen pregnant females were divided into a prenatal control group (PC, n = 8) and a prenatal stress group (PS, n = 8). The PS group was modeled with chronic unpredictable mild stress (CUMS) from day 1 of gestation to full-term delivery. Differences between the two groups in various maternal parameters, including glucocorticoid secretion, litter size, and the effects of male offspring birth weight, the KISS1 system, and reproductive function, were determined. Male offspring of PS dams had lower birth weights compared to prenatal controls.KISS1 gene expression is reduced at birth and in adult PS offspring, and its receptor KISS1-R protein is similarly reduced in PS offspring at birth and adulthood. In adulthood, PS male offspring show significantly reduced sex hormone production, altered testicular morphology, reduced maturation of their supporting cells, and decreased expression of connexin 43 (CX43), leading to an altered sperm microenvironment and reduced sperm quality. In conclusion, prenatal stress leads to adverse changes in the KISS1 system in male offspring and decreased reproductive function.
Collapse
Affiliation(s)
- Jian Gong
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yinjuan Lv
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yuhao Meng
- Hubei University of Chinese Medicine, Wuhan, China
| | | | | | - Min Xiao
- Laboratory Animal Center, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
2
|
Xiao H, Ding YL, Yang P, Chen Q, Huang HL, Chen X, Zhou HL, Tang SX. Association between anti-Müllerian hormone concentrations and sperm retrieval outcomes in patients with idiopathic nonobstructive azoospermia: a systematic review and meta-analysis. Asian J Androl 2024; 26:522-527. [PMID: 38748861 PMCID: PMC11449412 DOI: 10.4103/aja202419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/26/2024] [Indexed: 09/03/2024] Open
Abstract
ABSTRACT Microdissection testicular sperm extraction (mTESE) is commonly performed to retrieve sperm in the testes for assisted reproductive techniques in patients with idiopathic nonobstructive azoospermia (iNOA). However, the success rate of sperm retrieval varies among individuals. We aim to investigate the association between clinical parameters and sperm retrieval outcomes in patients with iNOA. We searched PubMed, EMBASE, and Web of Science from database inception to August 2, 2023. The main measure was whether sperm retrieval was successful in patients with iNOA who underwent mTESE. Pooled estimates of the sperm retrieval rate and weighted mean differences were calculated using random-effects models. The overall sperm retrieval rate was 36.8% (95% confidence interval [CI]: 27.5%-46.0%, I2 = 95.0%) in nine studies comprising 1892 patients with iNOA. No significant differences were found in age, testicular volume, serum total testosterone concentrations, or inhibin B concentrations between positive and negative sperm retrieval outcomes. Lower anti-Müllerian hormone concentrations in patients with iNOA were associated with a positive outcome of mTESE (weighted mean differences: -2.70; 95% CI: -3.94--1.46, I2 = 79.0%). In conclusion, this study shows a significant relationship between anti-Müllerian hormone and sperm retrieval outcomes in patients with iNOA, while age, testicular volume, total testosterone, and inhibin B show no significant association. These findings have important implications for assessing the potential success of sperm retrieval and selecting appropriate treatment strategies in patients with iNOA.
Collapse
Affiliation(s)
- Hong Xiao
- Department of Andrology and Sexual Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Holt R, Yahyavi SK, Wall-Gremstrup G, Jorsal MJ, Toft FB, Jørgensen N, Juul A, Blomberg Jensen M. Low-serum antimüllerian hormone is linked with poor semen quality in infertile men screened for participation in a randomized controlled trial. Fertil Steril 2024; 122:278-287. [PMID: 38522503 DOI: 10.1016/j.fertnstert.2024.03.018] [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: 11/29/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
OBJECTIVE To investigate possible associations between serum antimüllerian hormone (AMH) concentration and semen quality in infertile men. Studies investigating the associations between serum AMH concentration and semen quality in infertile men have shown conflicting results. DESIGN Infertile men were included during screening for participation in the First in Treating Male Infertility Study, a double-blinded, placebo-controlled, 1:1, single-center randomized controlled trial. SETTING Not applicable. PATIENTS At the screening visit, 400 participants produced a semen sample and had their serum analyzed for AMH concentration. INTERVENTION Not applicable. MAIN OUTCOME MEASURES Serum AMH concentration and semen quality. RESULTS All men were stratified according to serum AMH concentrations in quartiles (Q1-Q4). Men in the lowest quartile had a lower sperm concentration (1 × 106/mL) (Q1: 8.0 vs. Q2: 10.4 vs. Q3: 11.0 vs. Q4: 13.0), total sperm count (1 × 106) (Q1: 29.1 vs. Q2: 38.2 vs. Q3: 44.4 vs. Q4: 55.7), sperm motility (%) (Q1: 41 vs. Q2: 57 vs. Q3: 50 vs. Q4: 53), and progressive sperm motility (%) (Q1: 31 vs. Q2: 44 vs. Q3: 35 vs. Q4: 40) compared with the other quartiles. Moreover, men with a sperm concentration <2 million/mL had a lower serum AMH concentration compared with men having 2-16 × 106 /mL and >16 × 106/mL (31 pmol/L vs. 38 pmol/L vs. 43 pmol/L, respectively). In accordance, men with sperm motility <20% had a lower serum AMH concentration compared with men with sperm motility 20%-42%, and >42% (31 pmol/L vs. 43 pmol/L. vs. 39 pmol/L, respectively). CONCLUSION This study shows that low serum AMH concentration is associated with poor semen quality in infertile men, which implies that serum AMH concentration may have clinical value during the evaluation of male infertility. CLINICAL TRIAL REGISTRATION NUMBER NCT05212337.
Collapse
Affiliation(s)
- Rune Holt
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark; Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Sam Kafai Yahyavi
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark; Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Gustav Wall-Gremstrup
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Mads Joon Jorsal
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Frederikke Bay Toft
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark; Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Niels Jørgensen
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Martin Blomberg Jensen
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Giordano A, Pignolet B, Mascia E, Clarelli F, Sorosina M, Misra K, Bucciarelli F, Ferrè L, Moiola L, Liblau R, Filippi M, Esposito F. DNA Methylation in the Anti-Mullerian Hormone Gene and the Risk of Disease Activity in Multiple Sclerosis. Ann Neurol 2024; 96:289-301. [PMID: 38747444 DOI: 10.1002/ana.26959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 07/11/2024]
Abstract
OBJECTIVE Multiple sclerosis (MS) has a complex pathobiology, with genetic and environmental factors being crucial players. Understanding the mechanisms underlying heterogeneity in disease activity is crucial for tailored treatment. We explored the impact of DNA methylation, a key mechanism in the genetics-environment interplay, on disease activity in MS. METHODS Peripheral immune methylome profiling using Illumina Infinium MethylationEPIC BeadChips was conducted on 249 untreated relapsing-remitting MS patients, sampled at the start of disease-modifying treatment (DMT). A differential methylation analysis compared patients with evidence of disease activity (EDA) to those with no evidence of disease activity (NEDA) over 2 years from DMT start. Utilizing causal inference testing (CIT) and Mendelian randomization (MR), we sought to elucidate the relationships between DNA methylation, gene expression, genetic variation, and disease activity. RESULTS Four differentially methylated regions (DMRs) were identified between EDA and NEDA. Examining the influence of single nucleotide polymorphisms (SNPs), 923 variants were found to account for the observed differences in the 4 DMRs. Importantly, 3 out of the 923 SNPs, affecting DNA methylation in a DMR linked to the anti-Mullerian hormone (AMH) gene, were associated with disease activity risk in an independent cohort of 1,408 MS patients. CIT and MR demonstrated that DNA methylation in AMH acts as a mediator for the genetic risk of disease activity. INTERPRETATION This study uncovered a novel molecular pathway implicating the interaction between DNA methylation and genetic variation in the risk of disease activity in MS, emphasizing the role of sex hormones, particularly the AMH, in MS pathobiology. ANN NEUROL 2024;96:289-301.
Collapse
Affiliation(s)
- Antonino Giordano
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Neurology and MS Center, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Béatrice Pignolet
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, Toulouse, France
- Neurosciences Department, Toulouse University Hospital, Toulouse, France
| | - Elisabetta Mascia
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ferdinando Clarelli
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Melissa Sorosina
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Kaalindi Misra
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Florence Bucciarelli
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, Toulouse, France
| | - Laura Ferrè
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Neurology and MS Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Lucia Moiola
- Department of Neurology and MS Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Roland Liblau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, Toulouse, France
- Department of Immunology, Toulouse University Hospitals, Toulouse, France
| | - Massimo Filippi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Neurology and MS Center, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Federica Esposito
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Neurology and MS Center, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
5
|
Xiao H, Tang SX, Yao RJ, Ding YL, Yang P, Chen Q, Huang HL, Chen X, Zhou HL. Testicular volume is a noninvasive predictor of sperm retrieval failure in idiopathic nonobstructive azoospermia. Asian J Androl 2024; 26:421-425. [PMID: 38353461 PMCID: PMC11280197 DOI: 10.4103/aja202379] [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: 10/04/2023] [Accepted: 12/12/2023] [Indexed: 07/02/2024] Open
Abstract
We investigated the prognostic importance of noninvasive factors in predicting sperm retrieval failure in idiopathic nonobstructive azoospermia (iNOA). We studied 193 patients with nonobstructive azoospermia who underwent microsurgical testicular sperm extraction. The Chi-square test and Mann-Whitney U tests for clinical parameters and seminiferous tubule distribution were used for between-group comparisons. A logistic regression analysis was conducted to identify predictors of retrieval failure. Area under the receiver operating characteristic curve for each variable was evaluated, and the net clinical benefit was calculated using a clinical decision curve. Patients with iNOA had a lower sperm retrieval rate than those with known causes. Moreover, testicular volume was an independent factor affecting sperm extraction outcomes (odds ratio = 0.79, P < 0.05). The testicular volume cut-off value was 6.5 ml (area under the curve: 0.694). The patients with iNOA were categorized into two groups on the basis of the distribution of seminiferous tubules observed. The sperm retrieval rate and testicular volume were significantly different between the groups with a uniform or heterogeneous tubule distribution. There was also a significant association between a uniform tubule distribution and testicular volume. In conclusion, a testicular volume of more than 6.5 ml effectively predicts microsurgical testicular sperm extraction failure due to a uniform tubule distribution in patients with iNOA.
Collapse
Affiliation(s)
- Hong Xiao
- Department of Andrology and Sexual Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Song-Xi Tang
- Department of Andrology and Sexual Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Rui-Jie Yao
- Department of Andrology and Sexual Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Yi-Lang Ding
- Department of Andrology and Sexual Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Peng Yang
- Department of Andrology and Sexual Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Qiang Chen
- Department of Andrology and Sexual Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Hai-Lin Huang
- Department of Andrology and Sexual Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Xi Chen
- Department of Andrology and Sexual Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Hui-Liang Zhou
- Department of Andrology and Sexual Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
6
|
Li L, Lin W, Wang Z, Huang R, Xia H, Li Z, Deng J, Ye T, Huang Y, Yang Y. Hormone Regulation in Testicular Development and Function. Int J Mol Sci 2024; 25:5805. [PMID: 38891991 PMCID: PMC11172568 DOI: 10.3390/ijms25115805] [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: 04/07/2024] [Revised: 05/01/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
The testes serve as the primary source of androgens and the site of spermatogenesis, with their development and function governed by hormonal actions via endocrine and paracrine pathways. Male fertility hinges on the availability of testosterone, a cornerstone of spermatogenesis, while follicle-stimulating hormone (FSH) signaling is indispensable for the proliferation, differentiation, and proper functioning of Sertoli and germ cells. This review covers the research on how androgens, FSH, and other hormones support processes crucial for male fertility in the testis and reproductive tract. These hormones are regulated by the hypothalamic-pituitary-gonad (HPG) axis, which is either quiescent or activated at different stages of the life course, and the regulation of the axis is crucial for the development and normal function of the male reproductive system. Hormonal imbalances, whether due to genetic predispositions or environmental influences, leading to hypogonadism or hypergonadism, can precipitate reproductive disorders. Investigating the regulatory network and molecular mechanisms involved in testicular development and spermatogenesis is instrumental in developing new therapeutic methods, drugs, and male hormonal contraceptives.
Collapse
Affiliation(s)
- Lu Li
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Wanqing Lin
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Zhaoyang Wang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Rufei Huang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Huan Xia
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Ziyi Li
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Jingxian Deng
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Tao Ye
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Yadong Huang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China
| | - Yan Yang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China
| |
Collapse
|
7
|
Silveira JM, Cesar Dos Santos A, Calado de Brito DC, de Oliveira MF, Conley AJ, de Assis Neto AC. Morphohistometric and steroidogenic parameters during testicular and epididymal differentiation in cavy (Galea spixii) fetuses. Reprod Biol 2024; 24:100829. [PMID: 38039944 DOI: 10.1016/j.repbio.2023.100829] [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: 04/24/2023] [Revised: 10/30/2023] [Accepted: 11/17/2023] [Indexed: 12/03/2023]
Abstract
Sexual differentiation and steroidogenic mechanisms have an important impact on postnatal gonadal phenotypic development. Thus, establishing the activities that lead to male phenotypic development can provide a better understanding of this process. This study examined the prenatal development of cavies to establish morphological and histometric development patterns and protein and enzyme immunolocalization processes that are responsible for androgen synthesis in the testes and epididymis. Histological and histometric analyses of the diameter of the seminiferous cords and epididymal ducts of male fetuses on Days 25, 30, 40, and 50 were performed, as well as immunohistochemistry of the steroidogenic enzymes 5α-reductase and 17β-HSD, the androgen receptor, and the anti-Müllerian hormone (AMH). Our findings showed a cellular grouping of gonocytes from Day 30 onward that was characteristic of the seminiferous cord, which was not present in the lumen at any of the studied dates. From Day 50 onward, the differentiation of the three anatomical regions of the epididymis was evident, the head (caput), body (corpus), and tail (cauda), with tissue distinctions. Furthermore, the diameters of the seminiferous cords and epididymal ducts significantly increased with age. On Day 50, the tail showed the greatest diameter of the three regions. The Sertoli and Leydig cells exhibited AMH immunoreactivity at all dates. In addition, the Leydig cells and epididymal epithelial tissue were immunopositive for 5α-reductase, 17β-HSD, and the androgen receptor; therefore, these factors influenced the development and maintenance of the testis and epididymis during cavy prenatal development.
Collapse
Affiliation(s)
- Júlia Moreira Silveira
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Amilton Cesar Dos Santos
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | | | - Alan James Conley
- Department of Population Health & Reproduction, School of Veterinary Medicine, University of California, UC, Davis, CA, USA
| | - Antonio Chaves de Assis Neto
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
8
|
Dahiphale SM, Potdar J, Acharya N, Jyotsna G, Saloni, Desale R. Congenital Anomalies of the Female Genital Tract: A Comprehensive Review. Cureus 2024; 16:e56753. [PMID: 38654788 PMCID: PMC11037924 DOI: 10.7759/cureus.56753] [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: 12/21/2023] [Accepted: 03/23/2024] [Indexed: 04/26/2024] Open
Abstract
This comprehensive review provides an in-depth examination of congenital anomalies of the female genital tract, explicitly focusing on the American Society for Reproductive Medicine (ASRM) Müllerian Anomalies Classification. The classification system is crucial for standardizing communication and guiding accurate diagnoses in clinical practice. The review explores the diverse clinical presentations, etiological factors, and diagnostic modalities associated with these anomalies. Management strategies, ranging from conservative approaches to advanced reproductive technologies, are discussed in the context of individualized treatment plans based on the ASRM classification. The psychosocial impact of female genital tract anomalies is thoroughly examined, emphasizing the importance of holistic care and patient-centered approaches. Looking toward the future, the review outlines emerging research areas, including advances in diagnosis techniques, innovative treatment modalities, and genetic studies. It ultimately underscores the need for a comprehensive understanding of physical and psychosocial dimensions, offering insights for healthcare professionals to navigate this complex landscape and improve the lives of affected individuals.
Collapse
Affiliation(s)
- Swati M Dahiphale
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Jyotsana Potdar
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Neema Acharya
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Garapati Jyotsna
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Saloni
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Rahul Desale
- Radiodiagnosis, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| |
Collapse
|
9
|
Toraman E, Budak B, Bayram C, Sezen S, Mokhtare B, Hacımüftüoğlu A. Role of parthenolide in paclitaxel-induced oxidative stress injury and impaired reproductive function in rat testicular tissue. Chem Biol Interact 2024; 387:110793. [PMID: 37949423 DOI: 10.1016/j.cbi.2023.110793] [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: 08/23/2023] [Revised: 10/09/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
The chemotherapeutic agent paclitaxel (PTX) causes testicular toxicity due to oxidative stress. Parthenolide (PTL), the active ingredient of the Tanacetum parthenium plant, is used to treat inflammation, dizziness, and spasms. In the present study, we evaluated the therapeutic effect of PTL on PTX-induced testicular toxicity in rats and its role in reproductive function. To this end, 6 groups were formed: control, PTX, sham, T1, T2, and T3. After testicular toxicity was induced in rats with 8 mg/kg PTX, the rats were treated with 1 mg/kg, 2 mg/kg, and 4 mg/kg PTL for 14 days. GSH and MDA levels were measured in rat testicular tissue after the last dose of PTL was administered. To determine the damage caused by PTX to testicular tissue by detecting 8-OHdG and iNOS, sections were prepared and examined histopathologically and immunohistochemically. Furthermore, the gene expressions and enzymatic activities of SOD, CAT, GPx, GST, and GR were investigated in all groups. After PTL treatment, MDA, 8-OHdG, and iNOS levels decreased while GSH levels increased in testicular tissue. Increased levels of antioxidant genes and enzymes also reduced oxidative stress. Additionally, the expression levels of the Dazl, Ddx4, and Amh genes, which are involved in gametogenesis and sperm production, decreased in case of toxicity and increased with PTL treatment. The data from this study show that PTL may have a therapeutic effect in the treatment of testicular damage by eliminating the oxidative stress-induced damage caused by PTX in testicular tissue, providing an effective approach to alleviating testicular toxicity, and playing an important role in reproduction/sperm production, especially at a dose of 4 mg/kg.
Collapse
Affiliation(s)
- Emine Toraman
- Atatürk University, Science Faculty, Department of Molecular Biology and Genetics, Erzurum, Turkey.
| | - Büşra Budak
- Atatürk University, Faculty of Medicine, Department of Obstetrics and Gynaecology, Erzurum, Turkey
| | - Cemil Bayram
- Atatürk University, Faculty of Medicine, Department of Medical Pharmacology, Erzurum, Turkey
| | - Selma Sezen
- Ağrı İbrahim Çeçen University, Faculty of Medicine, Department of Medical Pharmacology, Ağrı, Turkey
| | - Behzad Mokhtare
- Atatürk University, Faculty of Veterinary Medicine, Department of Veterinary Pathology, Erzurum, Turkey
| | - Ahmet Hacımüftüoğlu
- Atatürk University, Faculty of Medicine, Department of Medical Pharmacology, Erzurum, Turkey
| |
Collapse
|
10
|
Liu Y, Liu Y, Chen S, Kong Z, Guo Y, Wang H. Prenatal exposure to acetaminophen at different doses, courses and time causes testicular dysplasia in offspring mice and its mechanism. CHEMOSPHERE 2023; 345:140496. [PMID: 37865203 DOI: 10.1016/j.chemosphere.2023.140496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 10/08/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
Epidemiological investigation suggested that the use of acetaminophen during pregnancy may cause offspring testicular dysplasia, but no systematic study has been conducted. In this study, Kunming mice were given acetaminophen at different doses (100/200/400 mg/kg.d), courses (single/multiple), time (second/third trimester) during pregnancy. Fetal blood and testes were collected on gestaional day 18 for detection. The results indicated abnormal testicular development in the PAcE (prenatal acetaminophen exposure) groups. The maximum diameter/cross-sectional area decreased, the interstitial space widened, and decreased proliferation/increased apoptosis were observed, especially in the high-dose, multi-course and second-trimester groups. Meanwhile, the serum testosterone level decreased in PAcE groups, and the steroid synthesis function in Leydig cells, Sertoli and spermatogenic cell function were inhibited, it was more significant in high-dose, multi-course and second-trimester groups. Furthermore, Wnt signal pathway was activated but Notch signal pathway was inhibited in the PAcE groups. Finally, in vitro experiment, acetaminophen could inhibit spermatogonial cell proliferation, enhance apoptosis, and change Wnt/Notch signal pathway. In conclusion, this study confirmed that PAcE can change fetal testicular development in a dose, course and time-dependent manner, and found that multicellular function impaired. This study provides theoretical and experimental basis for systematically elucidating the developmental toxicity of acetaminophen in testis.
Collapse
Affiliation(s)
- Yi Liu
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Yi Liu
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China; Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Sijia Chen
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Ziyu Kong
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Yu Guo
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Hui Wang
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
11
|
Ferré-Dolcet L, Bordogna M, Contiero B, Fontaine C, Bedin S, Romagnoli S. Anti-Müllerian Hormone Concentrations for Determining Resumption of Sertoli Cell Function following Removal of a 4.7 mg Deslorelin Implant in Tomcats. Animals (Basel) 2023; 13:2552. [PMID: 37627341 PMCID: PMC10451382 DOI: 10.3390/ani13162552] [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: 06/27/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Background: Deslorelin implant use in cats is a medical alternative to surgical sterilization, and due to its prolonged efficacy, its use has shown growing interest in the veterinary community. In the case of breeding facilities, its removal is often requested for the early restoration of testicular function. As anti-Müllerian hormones (AMH) in males is dependent of testosterone secretion, its assay may determine the restoration of testicular steroid secretion. An average of 3 weeks has been already described for tomcats' testicular function resumption after implant removal, but information about AMH concentrations in deslorelin-treated tomcats is lacking. Methods: Fourteen tomcats were treated for temporary suppression of fertility with a 4.7 mg deslorelin implant, which was surgically removed after 3, 6 or 9 months (n = 6, 4 and 4 tomcats, respectively). A general clinical and reproductive check with a gonadorelin stimulation test for testosterone determination was performed before deslorelin implant administration. After implant removal, tomcats' testicles were ultrasonographically checked for volume determination every 1-2 weeks with observation of the glans penis (presence or absence of spikes) and blood collection to assay both testosterone and AMH concentrations. Results: AMH concentrations increased significantly during the deslorelin treatment from 20.95 ± 4.97 ng/mL to 82.41 ± 14.59 ng/mL (p < 0.05). Following implant removal, AMH concentrations progressively decreased to pre-treatment levels, with a value of 28.42 ± 7.98 ng/mL on the third week post-removal where testosterone secretion was again detected. Conclusions: Even if a big variability of AMH concentrations exists between male individuals, resumption of tomcats' testicular function following a deslorelin treatment can be determined by AMH assay.
Collapse
Affiliation(s)
- Lluis Ferré-Dolcet
- Department of Animal Medicine, Production and Health, University of Padova, 35122 Padova, Italy (S.B.)
| | | | - Barbara Contiero
- Department of Animal Medicine, Production and Health, University of Padova, 35122 Padova, Italy (S.B.)
| | | | - Silvia Bedin
- Department of Animal Medicine, Production and Health, University of Padova, 35122 Padova, Italy (S.B.)
| | - Stefano Romagnoli
- Department of Animal Medicine, Production and Health, University of Padova, 35122 Padova, Italy (S.B.)
| |
Collapse
|
12
|
Pozzi E, Raffo M, Negri F, Boeri L, Saccà A, Belladelli F, Cilio S, Ventimiglia E, d’Arma A, Pagliardini L, Viganò P, Pontillo M, Lucianò R, Colecchia M, Montorsi F, Alfano M, Salonia A. Anti-Müllerian hormone predicts positive sperm retrieval in men with idiopathic non-obstructive azoospermia-findings from a multi-centric cross-sectional study. Hum Reprod 2023; 38:1464-1472. [PMID: 37322566 PMCID: PMC10482483 DOI: 10.1093/humrep/dead125] [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: 01/12/2023] [Revised: 05/24/2023] [Indexed: 06/17/2023] Open
Abstract
STUDY QUESTION Is it possible to identify a reliable marker of successful sperm retrieval (+SR) in men with idiopathic non-obstructive azoospermia (iNOA) undergoing microdissection testicular sperm extraction (mTESE)? SUMMARY ANSWER A higher likelihood of +SR during mTESE is observed in men with iNOA and lower preoperative serum anti-Müllerian hormone (AMH) levels, with good predictive accuracy achieved using an AMH threshold of <4 ng/ml. WHAT IS KNOWN ALREADY AMH has been previously linked to +SR in men with iNOA undergoing mTESE prior to ART. STUDY DESIGN, SIZE, DURATION A multi-centre cross-sectional study was carried out with a cohort of 117 men with iNOA undergoing mTESE at three tertiary-referral centres. PARTICIPANTS/MATERIALS, SETTING, METHODS Data from 117 consecutive white-European men with iNOA presenting for primary couple's infertility associated with a pure male factor at three centres were analysed. Descriptive statistics was applied to compare patients with negative (-SR) versus +SR at mTESE. Multivariate logistic regression models were fitted to predict +SR at mTESE, after adjusting for possible confounders. Diagnostic accuracy of the factors associated with +SR was assessed. Decision curve analyses were used to display the clinical benefit. MAIN RESULTS AND THE ROLE OF CHANCE Overall, 60 (51.3%) men had an -SR and 57 (48.7%) had a +SR at mTESE. Patients with +SR had lower levels of baseline AMH (P = 0.005) and higher levels of estradiol (E2) (P = 0.01). At multivariate logistic regression analysis, lower levels of AMH (odds ratio: 0.79; 95% CI: 0.64-0.93, P = 0.03) were associated with +SR at mTESE, after adjusting for possible confounders (e.g. age, mean testicular volume, FSH, and E2). A threshold of AMH <4 ng/ml achieved the highest accuracy for +SR at mTESE, with an AUC of 70.3% (95% CI: 59.8-80.7). Decision curve analysis displayed the net clinical benefit of using an AMH <4 ng/ml threshold. LIMITATIONS, REASONS FOR CAUTION There is a need for external validation in even larger cohorts, across different centres and ethnicities. Systematic reviews and meta-analysis to provide high level of evidence are lacking in the context of AMH and SR rates in men with iNOA. WIDER IMPLICATIONS OF THE FINDINGS Current findings suggest that slightly more than one in two men with iNOA had -SR at mTESE. Overall, men with iNOA with lower levels of AMH had a significantly higher percentage of successful SR at surgery. A threshold of <4 ng/ml for circulating AMH ensured satisfactory sensitivity, specificity, and positive predictive values in the context of +SR at mTESE. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by voluntary donations from the Urological Research Institute (URI). All authors declare no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Edoardo Pozzi
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Massimiliano Raffo
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- Urology Unit, ASST Spedali Civili di Brescia, Department of Medical and Surgical Specialties, Radiological Science and Public Health, University of Brescia, Brescia, Italy
| | - Fausto Negri
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- Urology Unit, ASST Spedali Civili di Brescia, Department of Medical and Surgical Specialties, Radiological Science and Public Health, University of Brescia, Brescia, Italy
| | - Luca Boeri
- Department of Urology, Foundation IRCCS Ca’ Granda -Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Antonino Saccà
- Department of Urology, AO Papa Giovanni XXIII, Bergamo, Italy
| | - Federico Belladelli
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Simone Cilio
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- Urology Unit, Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples “Federico II”, Naples, Italy
| | - Eugenio Ventimiglia
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessia d’Arma
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Luca Pagliardini
- Reproductive Sciences Laboratory, Obstetrics and Gynaecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Viganò
- Infertility Unit, Foundation IRCCS Ca’ Granda -Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Marina Pontillo
- Laboratory Medicine Service, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Roberta Lucianò
- Unit of Pathology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Maurizio Colecchia
- Laboratory Medicine Service, IRCCS Ospedale San Raffaele, Milan, Italy
- Unit of Pathology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Montorsi
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Salonia
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
13
|
Pereira SA, Oliveira FCB, Naulé L, Royer C, Neves FAR, Abreu AP, Carroll RS, Kaiser UB, Coelho MS, Lofrano-Porto A. Mouse Testicular Mkrn3 Expression Is Primarily Interstitial, Increases Peripubertally, and Is Responsive to LH/hCG. Endocrinology 2023; 164:bqad123. [PMID: 37585624 PMCID: PMC10449413 DOI: 10.1210/endocr/bqad123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
Studies in humans and mice support a role for Makorin RING finger protein 3 (MKRN3) as an inhibitor of gonadotropin-releasing hormone (GnRH) secretion prepubertally, and its loss of function is the most common genetic cause of central precocious puberty in humans. Studies have shown that the gonads can synthesize neuropeptides and express MKRN3/Mkrn3 mRNA. Therefore, we aimed to investigate the spatiotemporal expression pattern of Mkrn3 in gonads during sexual development, and its potential regulation in the functional testicular compartments by gonadotropins. Mkrn3 mRNA was detected in testes and ovaries of wild-type mice at all ages evaluated, with a sexually dimorphic expression pattern between male and female gonads. Mkrn3 expression was highest peripubertally in the testes, whereas it was lower peripubertally than prepubertally in the ovaries. Mkrn3 is expressed primarily in the interstitial compartment of the testes but was also detected at low levels in the seminiferous tubules. In vitro studies demonstrated that Mkrn3 mRNA levels increased in human chorionic gonadotropin (hCG)-treated Leydig cell primary cultures. Acute administration of a GnRH agonist in adult mice increased Mkrn3 expression in testes, whereas inhibition of the hypothalamic-pituitary-gonadal axis by chronic administration of GnRH agonist had the opposite effect. Finally, we found that hCG increased Mkrn3 mRNA levels in a dose-dependent manner. Taken together, our developmental expression analyses, in vitro and in vivo studies show that Mkrn3 is expressed in the testes, predominantly in the interstitial compartment, and that Mkrn3 expression increases after puberty and is responsive to luteinizing hormone/hCG stimulation.
Collapse
Affiliation(s)
- Sidney A Pereira
- Molecular Pharmacology Laboratory, School of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Fernanda C B Oliveira
- Molecular Pharmacology Laboratory, School of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
| | - Lydie Naulé
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Carine Royer
- Molecular Pharmacology Laboratory, School of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
| | - Francisco A R Neves
- Molecular Pharmacology Laboratory, School of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
| | - Ana Paula Abreu
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rona S Carroll
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michella S Coelho
- Molecular Pharmacology Laboratory, School of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
| | - Adriana Lofrano-Porto
- Molecular Pharmacology Laboratory, School of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
14
|
Lærkeholm Müller M, Busch AS, Ljubicic ML, Upners EN, Fischer MB, Hagen CP, Albrethsen J, Frederiksen H, Juul A, Andersson AM. Urinary concentration of phthalates and bisphenol A during minipuberty is associated with reproductive hormone concentrations in infant boys. Int J Hyg Environ Health 2023; 250:114166. [PMID: 37058994 DOI: 10.1016/j.ijheh.2023.114166] [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: 11/16/2022] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 04/16/2023]
Abstract
BACKGROUND The transient postnatal activation of the hypothalamic-pituitary-gonadal hormone axis is termed minipuberty and considered an important developmental period, which is highly sensitive to endocrine disruption. Here, we explore exposure-outcome associations during minipuberty between concentrations of potentially endocrine disrupting chemicals (EDCs) in urine of infant boys and their serum reproductive hormone concentrations. METHODS In total, 36 boys participating in the COPENHAGEN Minipuberty Study had data available for both urine biomarkers of target endocrine disrupting chemicals and reproductive hormones in serum from samples collected on the same day. Serum concentrations of reproductive hormones were measured by immunoassays or by LC-MS/MS. Urinary concentrations of metabolites of 39 non-persisting chemicals, including phthalates and phenolic compounds, were measured by LC-MS/MS. Nineteen chemicals had concentrations above the limit of detection in ≥50% of children and were included in data analysis. Associations of urinary phthalate metabolite and phenol concentrations (in tertiles) with hormone outcomes (age- and sex-specific SD-scores) were analysed by linear regression. Primarily, we focused on the EU regulated phthalates; butylbenzyl phthalate (BBzP), di-iso-butyl phthalate (DiBP), di-n-butyl phthalate (DnBP), and di-(2-ethylhexyl) phthalate (DEHP) as well as bisphenol A (BPA). Urinary metabolites of DiBP, DnBP and DEHP were summed and expressed as ∑DiBPm, ∑DnBPm and ∑DEHPm. RESULTS Compared to boys in the lowest ∑DnBPm tertile, urinary concentration of ∑DnBPm was associated with concurrent higher luteinizing hormone (LH) and anti-Müllerian hormone (AMH) SD-scores as well as lower testosterone/LH ratio in boys in the middle ∑DnBPm tertile (estimates (CI 95%) 0.79 (0.04; 1.54), 0.91 (0.13; 1.68), and -0.88 (-1.58;-0.19), respectively). Further, higher insulin-like peptide 3 (INSL3) SD-scores and lower DHEAS SD-score in boys in the highest ∑DnBPm tertile (0.91 (0.12; 1.70) and -0.85 (-1.51;-0.18), respectively) were observed. In addition, boys in the middle and highest ∑DEHPm tertile had higher LH (1.07 (0.35; 1.79) and 0.71 (-0.01; 1.43), respectively) and in the highest ∑DEHPm tertile also higher AMH (0.85 (0.10; 1.61)) concentration SD-scores, respectively. Boys in the highest BPA tertile had significantly higher AMH and lower DHEAS concentration compared to boys in the lowest BPA tertile (1.28 (0.54; 2.02) and -0.73 (-1.45; -0.01)), respectively. DISCUSSION Our findings indicate that exposure to chemicals with known or suspected endocrine disrupting potential, especially the EU-regulated DnBP, DEHP and BPA, may modify male reproductive hormone concentrations in infant boys suggesting that minipuberty is a critical window sensitive to endocrine disruption.
Collapse
Affiliation(s)
- Matilde Lærkeholm Müller
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Alexander Siegfried Busch
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; University of Münster, Department of General Pediatrics, Münster, Germany
| | - Marie Lindhardt Ljubicic
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Emmie N Upners
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Margit B Fischer
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Casper P Hagen
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Jakob Albrethsen
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Hanne Frederiksen
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anna-Maria Andersson
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
15
|
An K, Yao B, Tan Y, Kang Y, Su J. Potential Role of Anti-Müllerian Hormone in Regulating Seasonal Reproduction in Animals: The Example of Males. Int J Mol Sci 2023; 24:5874. [PMID: 36982948 PMCID: PMC10054328 DOI: 10.3390/ijms24065874] [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: 02/10/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Seasonal reproduction is a survival strategy by which animals adapt to environmental changes to improve their fitness. Males are often characterized by a significantly reduced testicular volume, indicating that they are in an immature state. Although many hormones, including gonadotropins, have played a role in testicular development and spermatogenesis, research on other hormones is insufficient. The anti-Müllerian hormone (AMH), which is a hormone responsible for inducing the regression of Müllerian ducts involved in male sex differentiation, was discovered in 1953. Disorders in AMH secretion are the main biomarkers of gonadal dysplasia, indicating that it may play a crucial role in reproduction regulation. A recent study has found that the AMH protein is expressed at a high level during the non-breeding period of seasonal reproduction in animals, implying that it may play a role in restricting breeding activities. In this review, we summarize the research progress on the AMH gene expression, regulatory factors of the gene's expression, and its role in reproductive regulation. Using males as an example, we combined testicular regression and the regulatory pathway of seasonal reproduction and attempted to identify the potential relationship between AMH and seasonal reproduction, to broaden the physiological function of AMH in reproductive suppression, and to provide new ideas for understanding the regulatory pathway of seasonal reproduction.
Collapse
Affiliation(s)
- Kang An
- Key Laboratory of Grassland Ecosystem, Ministry of Education, College of Grassland Science, Gansu Agricultural University, Lanzhou 730070, China
- Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou 730070, China
| | - Baohui Yao
- Key Laboratory of Grassland Ecosystem, Ministry of Education, College of Grassland Science, Gansu Agricultural University, Lanzhou 730070, China
- Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou 730070, China
| | - Yuchen Tan
- Key Laboratory of Grassland Ecosystem, Ministry of Education, College of Grassland Science, Gansu Agricultural University, Lanzhou 730070, China
- Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou 730070, China
| | - Yukun Kang
- Key Laboratory of Grassland Ecosystem, Ministry of Education, College of Grassland Science, Gansu Agricultural University, Lanzhou 730070, China
- Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou 730070, China
| | - Junhu Su
- Key Laboratory of Grassland Ecosystem, Ministry of Education, College of Grassland Science, Gansu Agricultural University, Lanzhou 730070, China
- Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
16
|
Reny SE, Mukherjee A, Mol PM. The curious case of testicular descent: factors controlling testicular descent with a note on cryptorchidism. AFRICAN JOURNAL OF UROLOGY 2023. [DOI: 10.1186/s12301-023-00342-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
Abstract
Background
The testicular descent is a uniquely complex process depending upon multiple factors like growth and reorganisation of the specific gonadal ligaments, hormones, etc., which interplay with each other. Though an unambiguous event, it is still laced with incredulity since the data interpretation were intermingled between different species creating more ambiguity in certain aspects of this process. In order to understand the aetiopathology of cryptorchidism the extensive study of the factors controlling the descent is necessitous.
Main body
Though testes originate in the abdomen, they migrate to an extra abdominal site the scrotum, which makes it vulnerable to pathological conditions associated with the descent. The hormones that play vital role in the first phase of descent are insulin-like hormone 3 (INSL3), Anti-müllerian hormone as well as testosterone, whereas androgens, genitofemoral nerve and its neurotransmitter calcitonin gene-related peptide (CGRP) influence the second phase. Despite the vast research regarding the complex nexus of events involving the descent there are disparities among the cross species studies. However all these discrepancies make testicular descent yet again fascinating and perplexing. Our aim is to provide a comprehensive review including recent advances which provides thorough coverage of anatomical and hormonal factors in the descent as well as cryptorchidism.
Conclusion
Though our understanding on testicular descent has evolved over the decades there still has obscurity surrounding it and the studies on the factors responsible for descent are becoming more intense with the time. Our knowledge on many factors such as INSL3 and CGRP is more established now; however, on the other hand the role of androgens still remains speculative. As the knowledge and understanding of the biological process of testicular descent increases it will pave ways to new treatment plans to treat cryptorchidism more effectively.
Collapse
|
17
|
Bahrami Z, Daeifarshbaf N, Amjadi F, Aflatoonian R. The effects of hormonal changes on sperm DNA integrity in oligoasthenoteratospermia individuals: A case-control study. Int J Reprod Biomed 2023; 20:999-1006. [PMID: 36819202 PMCID: PMC9928973 DOI: 10.18502/ijrm.v20i12.12560] [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: 02/28/2021] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 01/11/2023] Open
Abstract
Background Hormonal imbalance is one of the important etiological factors for Oligoasthenoteratospermias (OAT). Objective This study aimed to evaluate the effects of hormonal changes including prolactin, TSH, testosterone, luteinizing hormone, follicle-stimulating hormone, and anti-Mullerian hormone on sperm DNA fragmentation in normal men compared with OAT to design a clinical algorithm for the comprehensive study of male factor infertilities. Materials and Methods We consecutively selected 60 candidates referred to the infertility clinic to collect the semen and blood samples. Then, a terminal deoxynucleotidyl transferase dUTP nick end labeling test was performed to evaluate the sperm DNA fragmentation index (DFI). After semen analysis and DFI checking, they were classified into 4 groups consisting of normospermia and OAT men each with or without increased DFI. Hormone parameters were analyzed using enzyme-linked immunoassay. Results Follicle-stimulating hormone and luteinizing hormone levels showed positive correlations with DFI in a significant way (p ≤ 0.01), while testosterone and thyroid-stimulating hormone were associated with sperm concentration. Prolactin and anti-Mullerian hormone levels significantly correlated (p ≤ 0.01) with sperm concentration and DFI value simultaneously. Conclusion Decreased and increased levels of serum hormones could adversely affect semen profile and sperm DNA integrity which lead to severe male infertility. Although we investigated the effects of the main hormones related to male infertility on DNA damage, the role of these hormones on the fertilization rate and embryo quality needs to be evaluated in further studies.
Collapse
Affiliation(s)
- Zeinab Bahrami
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.,Laleh IVF Clinic, Laleh Hospital, Tehran, Iran
| | - Neda Daeifarshbaf
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Infertility Center, Imam Khomeini Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemehsadat Amjadi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Aflatoonian
- Laleh IVF Clinic, Laleh Hospital, Tehran, Iran.,Department of Endocrinology and Female Infertility at Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
18
|
Arato I, Giovagnoli S, Di Michele A, Bellucci C, Lilli C, Aglietti MC, Bartolini D, Gambelunghe A, Muzi G, Calvitti M, Eugeni E, Gaggia F, Baroni T, Mancuso F, Luca G. Nickel oxide nanoparticles exposure as a risk factor for male infertility: " In vitro" effects on porcine pre-pubertal Sertoli cells. Front Endocrinol (Lausanne) 2023; 14:1063916. [PMID: 37065743 PMCID: PMC10098343 DOI: 10.3389/fendo.2023.1063916] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/17/2023] [Indexed: 04/18/2023] Open
Abstract
Lately, nickel oxide nanoparticles (NiO NPs) have been employed in different industrial and biomedical fields. Several studies have reported that NiO NPs may affect the development of reproductive organs inducing oxidative stress and, resulting in male infertility. We investigated the in vitro effects of NiO NPs on porcine pre-pubertal Sertoli cells (SCs) which undergone acute (24 h) and chronic (from 1 up to 3 weeks) exposure at two subtoxic doses of NiO NPs of 1 μg/ml and 5 μg/ml. After NiO NPs exposure we performed the following analysis: (a) SCs morphological analysis (Light Microscopy); (b) ROS production and oxidative DNA damage, gene expression of antioxidant enzymes (c) SCs functionality (AMH, inhibin B Real-time PCR analysis and ELISA test); (d) apoptosis (WB analysis); (e) pro-inflammatory cytokines (Real-time PCR analysis), and (f) MAPK kinase signaling pathway (WB analysis). We found that the SCs exposed to both subtoxic doses of NiO NPs didn't sustain substantial morphological changes. NiO NPs exposure, at each concentration, reported a marked increase of intracellular ROS at the third week of treatment and DNA damage at all exposure times. We demonstrated, un up-regulation of SOD and HO-1 gene expression, at both concentrations tested. The both subtoxic doses of NiO NPs detected a down-regulation of AMH and inhibin B gene expression and secreted proteins. Only the 5 μg/ml dose induced the activation of caspase-3 at the third week. At the two subtoxic doses of NiO NPs a clear pro-inflammatory response was resulted in an up-regulation of TNF-α and IL-6 in terms of mRNA. Finally, an increased phosphorylation ratio of p-ERK1/2, p-38 and p-AKT was observed up to the third week, at both concentrations. Our results show the negative impact of subtoxic doses NiO NPs chronic exposure on porcine SCs functionality and viability.
Collapse
Affiliation(s)
- Iva Arato
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- *Correspondence: Iva Arato,
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | | | - Catia Bellucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Lilli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Desirée Bartolini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Angela Gambelunghe
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giacomo Muzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Mario Calvitti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Eugeni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesco Gaggia
- Internal Medicine Endocrine and Metabolic Sciences Unit, Santa Maria della Misericordia Hospital of Perugia, Perugia, Italy
| | - Tiziano Baroni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giovanni Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- International Biotechnological Center for Endocrine, Metabolic and Embryo-Reproductive Translational Research (CIRTEMER), Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Division of Medical Andrology and Endocrinology of Reproduction, Saint Mary Hospital, Terni, Italy
| |
Collapse
|
19
|
Germain N, Genteuil CD, Belleton G, Da Silva TL, Exbrayat C, Degas F, Hammour A, Gay A, Ravey B, Massoubre C, Galusca B. Continuous glucose monitoring assessment in patients suffering from anorexia nervosa reveals chronic prolonged mild hypoglycemia all over the nycthemeron. EUROPEAN EATING DISORDERS REVIEW 2022; 31:402-412. [PMID: 36541517 DOI: 10.1002/erv.2963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/04/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Anorexia nervosa (AN) is an eating disorder characterised by voluntary dietary restriction leading to severe undernutrition. Hypoglycaemia is mostly described through severe case reports and is always evaluated by fasting or post-meal blood glucose, showing nothing about hypoglycaemia's length or duration. The interest of continuous interstitial glucose monitoring (CGM), largely used in diabetes mellitus, has never been evaluated in AN patients. METHOD Glycaemia cycles in AN patients were assessed using CGM over 5 days and then analysed according to food intake. RESULTS Mean glycaemia was within normal range. 91% of the patients presented with at least one episode with glycaemia under 70 mg/dl. Within the 24 h, the percentage of time spent with a glycaemia under 70 mg/dl was of 20.82 ± 3.90% with a maximum of 52%. We found 2.52 ± 0.33 hypoglycaemia events per 24 h, including 21.11 ± 3.76% at night. CGM parameters correlated with cortisol and IGF1 plasma levels. Comparison with estimated carbohydrate intakes discriminated concordant and non-concordant estimations depending on patient. CONCLUSIONS AN patients display chronic prolonged mild hypoglycaemia all over the nycthemeron despite normal fasting glycaemia. Associated adaptive increased counter-regulatory hormones might protect AN patients from deeper hypoglycaemia. CGM allowed testing food intake self-estimation reliability of AN patients and could be a very useful biofeedback tool.
Collapse
Affiliation(s)
- Natacha Germain
- Department of Endocrinology Diabetes, Metabolism and Eating Disorders University Hospital of Saint‐Etienne Saint‐Etienne France
- TAPE Research Group Jean Monnet University of Saint‐Etienne Saint‐Etienne France
- Eating Disorder Reference Center University Hospital of Saint‐Etienne Saint‐Etienne France
| | - Clara Devin Genteuil
- Department of Endocrinology Diabetes, Metabolism and Eating Disorders University Hospital of Saint‐Etienne Saint‐Etienne France
- TAPE Research Group Jean Monnet University of Saint‐Etienne Saint‐Etienne France
- Eating Disorder Reference Center University Hospital of Saint‐Etienne Saint‐Etienne France
| | - Gwenaëlle Belleton
- Department of Endocrinology Diabetes, Metabolism and Eating Disorders University Hospital of Saint‐Etienne Saint‐Etienne France
- TAPE Research Group Jean Monnet University of Saint‐Etienne Saint‐Etienne France
- Eating Disorder Reference Center University Hospital of Saint‐Etienne Saint‐Etienne France
| | - Trecy Lopes Da Silva
- Department of Endocrinology Diabetes, Metabolism and Eating Disorders University Hospital of Saint‐Etienne Saint‐Etienne France
- TAPE Research Group Jean Monnet University of Saint‐Etienne Saint‐Etienne France
- Eating Disorder Reference Center University Hospital of Saint‐Etienne Saint‐Etienne France
| | - Chloé Exbrayat
- TAPE Research Group Jean Monnet University of Saint‐Etienne Saint‐Etienne France
| | - Fabien Degas
- Department of Endocrinology Diabetes, Metabolism and Eating Disorders University Hospital of Saint‐Etienne Saint‐Etienne France
- TAPE Research Group Jean Monnet University of Saint‐Etienne Saint‐Etienne France
- Eating Disorder Reference Center University Hospital of Saint‐Etienne Saint‐Etienne France
| | - Amira Hammour
- Department of Endocrinology Diabetes, Metabolism and Eating Disorders University Hospital of Saint‐Etienne Saint‐Etienne France
- TAPE Research Group Jean Monnet University of Saint‐Etienne Saint‐Etienne France
- Eating Disorder Reference Center University Hospital of Saint‐Etienne Saint‐Etienne France
| | - Aurélia Gay
- TAPE Research Group Jean Monnet University of Saint‐Etienne Saint‐Etienne France
- Eating Disorder Reference Center University Hospital of Saint‐Etienne Saint‐Etienne France
- Department of Psychiatry University Hospital of Saint‐Etienne Saint‐Etienne France
| | - Baptiste Ravey
- TAPE Research Group Jean Monnet University of Saint‐Etienne Saint‐Etienne France
- Eating Disorder Reference Center University Hospital of Saint‐Etienne Saint‐Etienne France
- Department of Psychiatry University Hospital of Saint‐Etienne Saint‐Etienne France
| | - Catherine Massoubre
- TAPE Research Group Jean Monnet University of Saint‐Etienne Saint‐Etienne France
- Eating Disorder Reference Center University Hospital of Saint‐Etienne Saint‐Etienne France
- Department of Psychiatry University Hospital of Saint‐Etienne Saint‐Etienne France
| | - Bogdan Galusca
- Department of Endocrinology Diabetes, Metabolism and Eating Disorders University Hospital of Saint‐Etienne Saint‐Etienne France
- TAPE Research Group Jean Monnet University of Saint‐Etienne Saint‐Etienne France
- Eating Disorder Reference Center University Hospital of Saint‐Etienne Saint‐Etienne France
| |
Collapse
|
20
|
Pryzhkova MV, Boers R, Jordan PW. Modeling Human Gonad Development in Organoids. Tissue Eng Regen Med 2022; 19:1185-1206. [PMID: 36350469 PMCID: PMC9679106 DOI: 10.1007/s13770-022-00492-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/08/2022] [Accepted: 09/17/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Our learning about human reproductive development is greatly hampered due to the absence of an adequate model. Animal studies cannot truthfully recapitulate human developmental processes, and studies of human fetal tissues are limited by their availability and ethical restrictions. Innovative three-dimensional (3D) organoid technology utilizing human pluripotent stem cells (hPSCs) offered a new approach to study tissue and organ development in vitro. However, a system for modeling human gonad development has not been established, thus, limiting our ability to study causes of infertility. METHODS In our study we utilized the 3D hPSC organoid culture in mini-spin bioreactors. Relying on intrinsic self-organizing and differentiation capabilities of stem cells, we explored whether organoids could mimic the development of human embryonic and fetal gonad. RESULTS We have developed a simple, bioreactor-based organoid system for modeling early human gonad development. Male hPSC-derived organoids follow the embryonic gonad developmental trajectory and differentiate into multipotent progenitors, which further specialize into testicular supporting and interstitial cells. We demonstrated functional activity of the generated cell types by analyzing the expression of cell type-specific markers. Furthermore, the specification of gonadal progenitors in organoid culture was accompanied by the characteristic architectural tissue organization. CONCLUSION This organoid system opens the opportunity for detailed studies of human gonad and germ cell development that can advance our understanding of sex development disorders. Implementation of human gonad organoid technology could be extended to modeling causes of infertility and regenerative medicine applications.
Collapse
Affiliation(s)
- Marina V Pryzhkova
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD, 21205, USA.
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Romina Boers
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD, 21205, USA
- Department of Molecular Cell Biology and Immunology, Amsterdam Universitair Medische Centra, 1117 HV, Amsterdam, The Netherlands
| | - Philip W Jordan
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD, 21205, USA.
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| |
Collapse
|
21
|
Hadlow NC, Brown SJ, Lim EM, Prentice D, Pettigrew S, Cronin SL, Prescott SL, Silva D, Yeap BB. Anti-Müllerian hormone concentration is associated with central adiposity and reproductive hormones in expectant fathers. Clin Endocrinol (Oxf) 2022; 97:634-642. [PMID: 35319116 PMCID: PMC9790283 DOI: 10.1111/cen.14725] [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: 01/10/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The role of the anti-Müllerian hormone (AMH) as an indicator of physical and reproductive health in men is unclear. We assessed the relationships between AMH and follicle-stimulating hormone (FSH), luteinizing hormone (LH), testosterone, and metabolic parameters, in a cohort of expectant fathers. DESIGN ORIGINS Project prospective cohort study. SETTING Community-dwelling men. PARTICIPANTS Partners of pregnant women attending antenatal appointments. MAIN OUTCOME MEASURES Serum AMH, FSH, LH, testosterone, and metabolic parameters. RESULTS In 485 expectant fathers, median age 33 years, median AMH was 40 pmol/L (quartiles 29, 56). AMH was inversely correlated with FSH, age, and body mass index (BMI) (correlation coefficients: -.32, -.24, and -.17 respectively). The age association was nonlinear, with peak AMH between 20 and 30 years, a decline thereafter, and somewhat steady levels after 45 years. The inverse association of AMH with FSH was log-linear and independent of age and BMI (β: -.07, SE: 0.01, p < .001). AMH was inversely correlated with waist circumference and directly associated with sex hormone-binding globulin. Testosterone was moderately correlated with AMH (correlation coefficient: .09, β: .011, SE: 0.004, p = .014): this association was mediated by an inverse relationship with BMI (mediated proportion 0.49, p < .001). CONCLUSIONS In reproductively active men, lower AMH is a biomarker for advancing age, and for poorer metabolic and reproductive health. The inverse association between AMH and FSH is independent of age and BMI, whereas the association of AMH and testosterone is mediated via BMI. The utility of AMH to predict reproductive and cardiometabolic outcomes in men warrants further investigation.
Collapse
Affiliation(s)
- Narelle C. Hadlow
- Medical SchoolUniversity of Western AustraliaPerthWestern AustraliaAustralia
- Biochemistry Department, Sonic HealthcareClinipath PathologyPerthWestern AustraliaAustralia
| | - Suzanne J. Brown
- Department of Endocrinology and DiabetesSir Charles Gairdner HospitalPerthWestern AustraliaAustralia
| | - Ee Mun Lim
- Medical SchoolUniversity of Western AustraliaPerthWestern AustraliaAustralia
- Department of Endocrinology and DiabetesSir Charles Gairdner HospitalPerthWestern AustraliaAustralia
- Biochemistry Department, PathWest Laboratory MedicineSir Charles Gairdner HospitalPerthWestern AustraliaAustralia
| | - David Prentice
- Perron Institute for Neurological and Translational SciencePerthWestern AustraliaAustralia
| | - Simone Pettigrew
- George Institute for Global HealthUniversity of New South WalesSydneyNew South WalesAustralia
| | - Sophie L. Cronin
- Medical SchoolUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Susan L. Prescott
- Medical SchoolUniversity of Western AustraliaPerthWestern AustraliaAustralia
- The ORIGINS ProjectTelethon Kids InstitutePerthWestern AustraliaAustralia
- Department of ImmunologyPerth Children's HospitalPerthWestern AustraliaAustralia
| | - Desiree Silva
- Medical SchoolUniversity of Western AustraliaPerthWestern AustraliaAustralia
- The ORIGINS ProjectTelethon Kids InstitutePerthWestern AustraliaAustralia
- Department of PaediatricsJoondalup Health CampusPerthWestern AustraliaAustralia
- School of Medical and Health SciencesEdith Cowan UniversityPerthWestern AustraliaAustralia
| | - Bu B. Yeap
- Medical SchoolUniversity of Western AustraliaPerthWestern AustraliaAustralia
- Department of Endocrinology and DiabetesFiona Stanley HospitalPerthWestern AustraliaAustralia
| |
Collapse
|
22
|
Quiarato Lozano AF, Marques Tavares B, Villela E Silva P, Franco de Barros JW, Kempinas WDG. Reproductive development of male rats exposed in utero to stress and/or sertraline. Toxicol Sci 2022; 190:189-203. [PMID: 36161332 DOI: 10.1093/toxsci/kfac100] [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: 11/14/2022] Open
Abstract
Despite increased prescription of sertraline during pregnancy, little is known about its action on reproductive development. Therefore, the present study aimed to investigate the impact that stress, associated or not with sertraline, causes on the reproductive development of male rats. Pregnant Wistar rats were divided into 4 groups (n = 16/group): CO - received filtered water; SE - received 20 mg/kg sertraline; ST - submitted to restraint stress and received filtered water; SS - submitted to restraint stress and received sertraline. The treatment was carried out from gestational days (GD) 13 to 20. The animals were euthanized on GD 20 (n = 8/group), postnatal day (PND) 45 (n = 8/group) and PND 110 (n = 8/group). The testes and epididymis were analyzed histologically, and immunohistochemistry was performed on the testes by proliferating cell nuclear antigen (PCNA) and the Wilms tumor protein (Wt1). Sperm quality was also analyzed on PND 110. The evolution of body weight, anogenital distance (AGD), and puberty installation day were also verified. Statistical analysis: Two-way ANOVA or Kruskal-Wallis test (p ≤ 0.05). Fetal testes presented a large number of acidophilic cells in the sertraline-exposed groups. The SS group also showed a decrease in the nuclear volume of Leydig cells. This same group showed low expression of PCNA and Wt1, decreased weight of the testes and epididymis, lower AGD, and delayed puberty installation. The adulthood groups exposed to sertraline presented alterations in sperm morphology and motility. The results demonstrated that prenatal exposure to sertraline compromises the development of the rat reproductive system.
Collapse
Affiliation(s)
- Ana Flávia Quiarato Lozano
- Laboratory of Reproductive and Developmental Biology and Toxicology, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Bruna Marques Tavares
- Laboratory of Reproductive and Developmental Biology and Toxicology, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Patrícia Villela E Silva
- Laboratory of Reproductive and Developmental Biology and Toxicology, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Jorge Willian Franco de Barros
- Laboratory of Reproductive and Developmental Biology and Toxicology, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Wilma De Grava Kempinas
- Laboratory of Reproductive and Developmental Biology and Toxicology, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, SP, Brazil
| |
Collapse
|
23
|
Petrusová J, Manning J, Kubovčiak J, Kolář M, Filipp D. Two complementary approaches for efficient isolation of Sertoli cells for transcriptomic analysis. Front Cell Dev Biol 2022; 10:972017. [PMID: 36158203 PMCID: PMC9495933 DOI: 10.3389/fcell.2022.972017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022] Open
Abstract
Sertoli cells (SCs) are the only somatic cells that reside in seminiferous tubules of testis. They directly interact with and support the development of germ cells, thus have an indispensable role in the process of spermatogenesis. SCs first appear in a proliferative state and then, with the initiation of the first wave of spermatogenesis, progress to a mature “nurturing” state which supports lifelong continuous sperm production. During this development, the SC transcriptome must adapt rapidly as obstacles in SC maturation often result in deficiencies in male fertility. Due to its importance in spermatogenesis, a reliable, rapid, and precise method for the isolation of high purity, viable and unadulterated SC has been largely missing. We have developed an improved method for the preparation of a testicular single cell suspension comprised of two alternative protocols to separate SCs from the rest of the testicular cells by FACS. The first sorting scheme is based on their co-expression of surface specific markers, FSHr and Occludin-1, while the second focuses on the co-staining of SCs with FSHr-specific antibody and Hoechst 33342, which discriminates DNA content of testicular cells. The entire procedure can be completed in less than 3 h which permits the analysis of the development-related transcriptional profile of these cells. Notably, our comparative study showed that this method resulted in a SC transcriptome that is largely comparable to SCs which were briskly isolated due to their cell-specific expression of fluorescent protein. Interestingly, we also show that SCs sorted as FSHr+Occludin+ cells contained a tangible portion of transcripts from all types of testicular germ cells. Sorting of SCs according to their 2C DNA content significantly reduced the presence of these transcripts, thus seems to be the most suitable approach for accurate determination of the SC transcriptome. We believe that these novel approaches for the isolation of SCs will assist researchers in the elucidation of their function as well as their role in spermatogenesis and disorders related to male infertility.
Collapse
Affiliation(s)
- Jana Petrusová
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jasper Manning
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jan Kubovčiak
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Michal Kolář
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Dominik Filipp
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
- *Correspondence: Dominik Filipp,
| |
Collapse
|
24
|
Satarug S, Gobe GC, Vesey DA. Multiple Targets of Toxicity in Environmental Exposure to Low-Dose Cadmium. TOXICS 2022; 10:toxics10080472. [PMID: 36006151 PMCID: PMC9412446 DOI: 10.3390/toxics10080472] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/06/2022] [Accepted: 08/12/2022] [Indexed: 05/06/2023]
Abstract
Dietary assessment reports and population surveillance programs show that chronic exposure to low levels of environmental cadmium (Cd) is inevitable for most people, and adversely impacts the health of children and adults. Based on a risk assessment model that considers an increase in the excretion of β2-microglobulin (β2M) above 300 μg/g creatinine to be the "critical" toxicity endpoint, the tolerable intake level of Cd was set at 0.83 µg/kg body weight/day, and a urinary Cd excretion rate of 5.24 µg/g creatinine was considered to be the toxicity threshold level. The aim of this review is to draw attention to the many other toxicity endpoints that are both clinically relevant and more appropriate to derive Cd exposure limits than a β2M endpoint. In the present review, we focus on a reduction in the glomerular filtration rate and diminished fecundity because chronic exposure to low-dose Cd, reflected by its excretion levels as low as 0.5 µg/g creatinine, have been associated with dose-dependent increases in risk of these pathological symptoms. Some protective effects of the nutritionally essential elements selenium and zinc are highlighted. Cd-induced mitochondrial dysfunction is discussed as a potential mechanism underlying gonadal toxicities and infertility.
Collapse
Affiliation(s)
- Soisungwan Satarug
- Kidney Disease Research Collaborative, Translational Research Institute, Brisbane 4102, Australia
- Correspondence:
| | - Glenda C. Gobe
- Kidney Disease Research Collaborative, Translational Research Institute, Brisbane 4102, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia
- NHMRC Centre of Research Excellence for CKD QLD, UQ Health Sciences, Royal Brisbane and Women’s Hospital, Brisbane 4029, Australia
| | - David A. Vesey
- Kidney Disease Research Collaborative, Translational Research Institute, Brisbane 4102, Australia
- Department of Nephrology, Princess Alexandra Hospital, Brisbane 4075, Australia
| |
Collapse
|
25
|
Goktepe O, Balcioglu E, Baran M, Cengiz O, Ceyhan A, Suna PA, Bolat D, Yalcin B, Yay A. Protective effects of melatonin on female rat ovary treated with nonylphenol. Biotech Histochem 2022; 98:13-19. [PMID: 35611760 DOI: 10.1080/10520295.2022.2075566] [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: 11/02/2022] Open
Abstract
We investigated using histochemistry and immunohistochemistry ovarian damage caused by nonylphenol (NP) and the protective effect of melatonin treatment of NP induced ovarian damage. We used 21 female rats divided randomly into three groups: control, NP and melatonin + NP. Histopathological examination of the ovaries, and counting and classification of follicles were performed using Masson's trichrome staining. Expression of anti-Mullerian hormone (AMH), Bax, Bcl-2 and caspase-3 was detected in the ovaries using immunohistochemistry. Melatonin had an ameliorative effect on NP induced follicular atresia and absence of corpora lutea. More follicles were observed in the ovaries of animals treated with melatonin prior to treatment with NP. AMH immunoreactivity was significantly lower in the NP group than in the melatonin + NP group. NP increased immunostaining for Bax, Bcl-2 and caspase-3. Melatonin significantly reduced the increased expression of Bax, Bcl-2 and caspase-3 due to NP exposure. We found that pretreatment with melatonin is beneficial for protecting the ovaries from damage by NP.
Collapse
Affiliation(s)
- Ozge Goktepe
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Esra Balcioglu
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey.,Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| | - Munevver Baran
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Ozge Cengiz
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Ayse Ceyhan
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Pinar Alisan Suna
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Demet Bolat
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Betul Yalcin
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Arzu Yay
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey.,Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| |
Collapse
|
26
|
Gültiken N, Yarim M, Aslan S, Gürler H, Yarim GF, Tuncay M, İnal S, Schäfer-Somi S. Expression of Anti-Müllerian Hormone and Its Type 2 Receptor in the Ovary of Pregnant and Cyclic Domestic Cats. Animals (Basel) 2022; 12:877. [PMID: 35405866 PMCID: PMC8997022 DOI: 10.3390/ani12070877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 01/26/2023] Open
Abstract
To evaluate the expression of AMH and its receptor AMHRII, ovaries of 33 p cats were investigated by western blot and immunohistochemistry. After ovariohysterectomy, the cats were grouped according to pregnancy stages and ovarian/placental endocrine activity: group I (n = 3, 24−29 days), II (n = 8, 32−40 days), III (n = 4, 41−46 days), IV (n = 6, 53−61 days) and according to cycle stages: V (n = 6, interestrus) and VI (n = 6, estrus). Serum progesterone- and AMH-concentration was measured. Follicle numbers did not differ between groups. The number of corpora lutea was higher in pregnant cats than in the non-pregnant cats. Serum AMH concentration was at maximum between day 30 and 50 of gestation, and was higher than in non-pregnant cats, then decreased towards term (p < 0.05). In the ovaries, AMH immunopositivity was observed in granulosa cells of secondary and antral follicles, and in interstitial cells of corpora lutea; highest percentage of immunopositive areas was detected in group III (p < 0.05). A positive correlation between the number of corpora lutea and the positive AMH signals in ovarian tissue was determined (r2 = 0.832, p < 0.05); however, only during mid-gestation (group II). Expression of AMHRII was in close co-localization with AMH and strong in the interstitial cells surrounding follicles undergoing atresia. AMHRII expression did not differ between pregnant groups but was higher compared to estrus cats (p ˂ 0.05). We conclude that AMH and AMHRII expression in the feline ovary is comparable to other species. The high serum AMH concentration and ovarian AMHRII expression between day 30 and 50 of gestation are probably related to ovarian activity and follicular atresia.
Collapse
Affiliation(s)
- Nilgün Gültiken
- Department of Obstetrics and Gynecology, University of Ondokuz Mayis, Samsun 55200, Turkey; (N.G.); (H.G.); (M.T.)
| | - Murat Yarim
- Department of Pathology, University of Ondokuz Mayis, Samsun 55200, Turkey; (M.Y.); (S.İ.)
| | - Selim Aslan
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Near East University, Nicosia 99138, Cyprus;
| | - Hande Gürler
- Department of Obstetrics and Gynecology, University of Ondokuz Mayis, Samsun 55200, Turkey; (N.G.); (H.G.); (M.T.)
| | - Gul Fatma Yarim
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Ondokuz Mayis, Samsun 55200, Turkey;
| | - Müge Tuncay
- Department of Obstetrics and Gynecology, University of Ondokuz Mayis, Samsun 55200, Turkey; (N.G.); (H.G.); (M.T.)
| | - Sinem İnal
- Department of Pathology, University of Ondokuz Mayis, Samsun 55200, Turkey; (M.Y.); (S.İ.)
| | - Sabine Schäfer-Somi
- Platform for Artificial Insemination and Embryo Transfer, University of Veterinary Medicine, 1210 Vienna, Austria
| |
Collapse
|
27
|
Domain G, Buczkowska J, Kalak P, Wydooghe E, Banchi P, Pascottini OB, Niżański W, Van Soom A. Serum Anti-Müllerian Hormone: A Potential Semen Quality Biomarker in Stud Dogs? Animals (Basel) 2022; 12:ani12030323. [PMID: 35158647 PMCID: PMC8833318 DOI: 10.3390/ani12030323] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 01/03/2023] Open
Abstract
Anti-Müllerian hormone (AMH) has been suggested to be involved in spermatogenesis. The aim of this study was to investigate the relationship between blood serum AMH concentration and semen quality in dogs. Moreover, this study sought to find the optimal cut-off point value of serum AMH with the greatest sensitivity and specificity to predict semen quality. Forty-five clinically healthy dogs were included in the study and their age as well as the following semen parameters were determined and correlated to serum AMH concentration: total sperm output, normal morphology, plasma membrane integrity, total motility, progressive motility, and velocity parameters. Statistical analysis for correlations were performed using Spearman’s correlation coefficients. Moderate negative associations were found between serum AMH and semen total motility (r = −0.38, p = 0.01), progressive motility (r = −0.36, p = 0.01), and normal morphology (r = −0.36, p= 0.02). Based on these associations, an AMH concentration of 5.54 µg/L was found to be the optimal cut-off point value to obtain the greatest summation of sensitivity (86%) and specificity (63%) to predict semen quality. The serum AMH assay may therefore be a potential hormonal marker to predict which dogs would require further semen analysis. Future research is however needed to confirm these preliminary results.
Collapse
Affiliation(s)
- Guillaume Domain
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (E.W.); (P.B.); (O.B.P.); (A.V.S.)
- Correspondence:
| | - Justyna Buczkowska
- Department of Reproduction and Clinic of Farm Animals, University of Environmental Science, Grundwaldzki Square 49, 50-357 Wroclaw, Poland; (J.B.); (P.K.); (W.N.)
| | - Patrycja Kalak
- Department of Reproduction and Clinic of Farm Animals, University of Environmental Science, Grundwaldzki Square 49, 50-357 Wroclaw, Poland; (J.B.); (P.K.); (W.N.)
| | - Eline Wydooghe
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (E.W.); (P.B.); (O.B.P.); (A.V.S.)
| | - Penelope Banchi
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (E.W.); (P.B.); (O.B.P.); (A.V.S.)
| | - Osvaldo Bogado Pascottini
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (E.W.); (P.B.); (O.B.P.); (A.V.S.)
- Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Wojciech Niżański
- Department of Reproduction and Clinic of Farm Animals, University of Environmental Science, Grundwaldzki Square 49, 50-357 Wroclaw, Poland; (J.B.); (P.K.); (W.N.)
| | - Ann Van Soom
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (E.W.); (P.B.); (O.B.P.); (A.V.S.)
| |
Collapse
|
28
|
Cannarella R, Mancuso F, Arato I, Lilli C, Bellucci C, Gargaro M, Curto R, Aglietti MC, La Vignera S, Condorelli RA, Luca G, Calogero AE. Sperm-carried IGF2 downregulated the expression of mitogens produced by Sertoli cells: A paracrine mechanism for regulating spermatogenesis? Front Endocrinol (Lausanne) 2022; 13:1010796. [PMID: 36523595 PMCID: PMC9744929 DOI: 10.3389/fendo.2022.1010796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/07/2022] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Insulin-like growth factor 2 (IGF2) mRNA has been found in human and mouse spermatozoa. It is currently unknown whether the IGF2 protein is expressed in human spermatozoa and, if so, its possible role in the cross-talk between germ and Sertoli cells (SCs) during spermatogenesis. METHODS To accomplish this, we analyzed sperm samples from four consecutive Caucasian men. Furthermore, to understand its role during the spermatogenetic process, porcine SCs were incubated with increasing concentrations (0.33, 3.33, and 10 ng/mL) of recombinant human IGF2 (rhIGF2) for 48 hours. Subsequently, the experiments were repeated by pre-incubating SCs with the non-competitive insulin-like growth factor 1 receptor (IGF1R) inhibitor NVP-AEW541. The following outcomes were evaluated: 1) Gene expression of the glial cell-line derived neurotrophic factor (GDNF), fibroblast growth factor 2 (FGF2), and stem cell factor (SCF) mitogens; 2) gene and protein expression of follicle-stimulating hormone receptor (FSHR), anti-Müllerian hormone (AMH), and inhibin B; 3) SC proliferation. RESULTS We found that the IGF2 protein was present in each of the sperm samples. IGF2 appeared as a cytoplasmic protein localized in the equatorial and post-acrosomal segment and with a varying degree of expression in each cell. In SCs, IGF2 significantly downregulated GDNF gene expression in a concentration-dependent manner. FGF2 and SCF were downregulated only by the highest concentration of IGF2. Similarly, IGF2 downregulated the FSHR gene and FSHR, AMH, and inhibin B protein expression. Finally, IGF2 significantly suppressed the SC proliferation rate. All these findings were reversed by pre-incubation with NVP-AEW541, suggesting an effect mediated by the interaction of IGF2 with the IGFR. CONCLUSION In conclusion, sperm IGF2 seems to downregulate the expression of mitogens, which are known to be physiologically released by the SCs to promote gonocyte proliferation and spermatogonial fate adoption. These findings suggest the presence of paracrine regulatory mechanisms acting on the seminiferous epithelium during spermatogenesis, by which germ cells can influence the amount of mitogens released by the SCs, their sensitivity to FSH, and their rate of proliferation.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
- *Correspondence: Rossella Cannarella,
| | - Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Iva Arato
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Lilli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Catia Bellucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marco Gargaro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Roberto Curto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Maria C. Aglietti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A. Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giovani Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Aldo E. Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
29
|
Bertho S, Neyroud AS, Brun T, Jaillard S, Bonnet F, Ravel C. Anti-Müllerian hormone: A function beyond the Müllerian structures. Morphologie 2021; 106:252-259. [PMID: 34924282 DOI: 10.1016/j.morpho.2021.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 10/19/2022]
Abstract
The anti-Müllerian hormone (AMH) is a heterodimeric glycoprotein belonging to the TGFb superfamily implicated in human embryonic development. This hormone was first described as allowing regression of the epithelial embryonic Müllerian structures in males, which would otherwise differentiate into the uterus and fallopian tubes. It activates a signaling pathway mediated by two transmembrane receptors. Binding of AMH to its receptor induces morphological changes leading to the degeneration of Müllerian ducts. Recently, new data has shown the role played by this hormone on structures other than the genital tract. If testicular AMH expression decreases in humans over the course of a lifetime, synthesis may persist in other tissues in adulthood. The mechanisms underlying its production have been unveiled. The aim of this review is to describe the different pathways in which AMH has been identified and plays a pivotal role.
Collapse
Affiliation(s)
- S Bertho
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France.
| | - A S Neyroud
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France; Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000 Rennes, France
| | - T Brun
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France
| | - S Jaillard
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France; Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000 Rennes, France
| | - F Bonnet
- CHU Rennes, Service d'Endocrinologie, 35000 Rennes, France
| | - C Ravel
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France; Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000 Rennes, France
| |
Collapse
|
30
|
Ho EV, Shi C, Cassin J, He MY, Nguyen RD, Ryan GE, Tonsfeldt KJ, Mellon PL. Reproductive Deficits Induced by Prenatal Antimüllerian Hormone Exposure Require Androgen Receptor in Kisspeptin Cells. Endocrinology 2021; 162:6371276. [PMID: 34529765 PMCID: PMC8507963 DOI: 10.1210/endocr/bqab197] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Indexed: 11/19/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common reproductive disorder characterized by elevated androgens and antimüllerian hormone (AMH). These hormones remain elevated throughout pregnancy, and potential effects of hormone exposure on offspring from women with PCOS remain largely unexplored. Expanding on recent reports of prenatal AMH exposure in mice, we have fully characterized the reproductive consequences of prenatal AMH (pAMH) exposure throughout the lifespan of first- and second-generation offspring of both sexes. We also sought to elucidate mechanisms underlying pAMH-induced reproductive effects. There is a known reciprocal relationship between AMH and androgens, and in PCOS and PCOS-like animal models, androgen feedback is dysregulated at the level of the hypothalamus. Kisspeptin neurons express androgen receptors and play a critical role in sexual development and function. We therefore hypothesized that pAMH-induced reproductive phenotypes would be mediated by androgen signaling at the level of kisspeptin cells. We tested the pAMH model in kisspeptin-specific androgen receptor knockout (KARKO) mice and found that virtually all pAMH-induced phenotypes assayed are eliminated in KARKO offspring compared to littermate controls. By demonstrating the necessity of androgen receptor in kisspeptin cells to induce pAMH phenotypes, we have advanced understanding of the interactions between AMH and androgens in the context of prenatal exposure, which could have significant implications for children of women with PCOS.
Collapse
Affiliation(s)
- Emily V Ho
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Chengxian Shi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Jessica Cassin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Michelle Y He
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Ryan D Nguyen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Genevieve E Ryan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Karen J Tonsfeldt
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
- Correspondence: Pamela L. Mellon, PhD, Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093-0674, USA.
| |
Collapse
|
31
|
Dard R, Moreau M, Parizot E, Ghieh F, Brehier L, Kassis N, Serazin V, Lamaziere A, Racine C, di Clemente N, Vialard F, Janel N. DYRK1A Overexpression in Mice Downregulates the Gonadotropic Axis and Disturbs Early Stages of Spermatogenesis. Genes (Basel) 2021; 12:1800. [PMID: 34828406 PMCID: PMC8621272 DOI: 10.3390/genes12111800] [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] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022] Open
Abstract
Down syndrome (DS) is the most common chromosomal disorder. It is responsible for intellectual disability (ID) and several medical conditions. Although men with DS are thought to be infertile, some spontaneous paternities have been reported. The few studies of the mechanism of infertility in men with DS are now dated. Recent research in zebrafish has indicated that overexpression of DYRK1A (the protein primarily responsible for ID in DS) impairs gonadogenesis at the embryonic stage. To better ascertain DYRK1A's role in infertility in DS, we investigated the effect of DYRK1A overexpression in a transgenic mouse model. We found that overexpression of DYRK1A impairs fertility in transgenic male mice. Interestingly, the mechanism in mice differs slightly from that observed in zebrafish but, with disruption of the early stages of spermatogenesis, is similar to that seen in humans. Unexpectedly, we observed hypogonadotropic hypogonadism in the transgenic mice.
Collapse
Affiliation(s)
- Rodolphe Dard
- Laboratoire Processus Dégénératifs, Stress et Vieillissement, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Université de Paris, 75205 Paris, France; (M.M.); (E.P.); (N.K.); (N.J.)
- Université Paris-Saclay, UVSQ, INRAE, ENVA, BREED, 78350 Jouy-en-Josas, France; (F.G.); (L.B.); (F.V.)
- Département de Génétique, CHI de Poissy St Germain en Laye, 78300 Poissy, France;
| | - Manon Moreau
- Laboratoire Processus Dégénératifs, Stress et Vieillissement, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Université de Paris, 75205 Paris, France; (M.M.); (E.P.); (N.K.); (N.J.)
| | - Estelle Parizot
- Laboratoire Processus Dégénératifs, Stress et Vieillissement, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Université de Paris, 75205 Paris, France; (M.M.); (E.P.); (N.K.); (N.J.)
- Université Paris-Saclay, UVSQ, INRAE, ENVA, BREED, 78350 Jouy-en-Josas, France; (F.G.); (L.B.); (F.V.)
| | - Farah Ghieh
- Université Paris-Saclay, UVSQ, INRAE, ENVA, BREED, 78350 Jouy-en-Josas, France; (F.G.); (L.B.); (F.V.)
| | - Leslie Brehier
- Université Paris-Saclay, UVSQ, INRAE, ENVA, BREED, 78350 Jouy-en-Josas, France; (F.G.); (L.B.); (F.V.)
| | - Nadim Kassis
- Laboratoire Processus Dégénératifs, Stress et Vieillissement, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Université de Paris, 75205 Paris, France; (M.M.); (E.P.); (N.K.); (N.J.)
| | - Valérie Serazin
- Département de Génétique, CHI de Poissy St Germain en Laye, 78300 Poissy, France;
| | - Antonin Lamaziere
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université-INSERM, 75012 Paris, France; (A.L.); (C.R.); (N.d.C.)
| | - Chrystèle Racine
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université-INSERM, 75012 Paris, France; (A.L.); (C.R.); (N.d.C.)
| | - Nathalie di Clemente
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université-INSERM, 75012 Paris, France; (A.L.); (C.R.); (N.d.C.)
| | - François Vialard
- Université Paris-Saclay, UVSQ, INRAE, ENVA, BREED, 78350 Jouy-en-Josas, France; (F.G.); (L.B.); (F.V.)
- Département de Génétique, CHI de Poissy St Germain en Laye, 78300 Poissy, France;
| | - Nathalie Janel
- Laboratoire Processus Dégénératifs, Stress et Vieillissement, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Université de Paris, 75205 Paris, France; (M.M.); (E.P.); (N.K.); (N.J.)
| |
Collapse
|
32
|
Anti-Müllerian hormone, testosterone, and insulin-like peptide 3 as biomarkers of Sertoli and Leydig cell function during deslorelin-induced testicular downregulation in the dog. Theriogenology 2021; 175:100-110. [PMID: 34534687 DOI: 10.1016/j.theriogenology.2021.08.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 07/10/2021] [Accepted: 08/13/2021] [Indexed: 12/17/2022]
Abstract
The role of anti-Müllerian hormone (AMH) and insulin-like peptide 3 (INSL3) in male infertility is not fully understood. We used the downregulated testis as a model of gonadotropin-dependent infertility. Serum testosterone and AMH concentrations were studied in five adult male Beagles implanted (day 0) with 4.7 mg deslorelin (Suprelorin®, Virbac) (DES group). Testicular expression of LH receptor (LHR) and androgen receptor (AR), AMH, type 2 AMH receptor (AMHR2), INSL3 and its receptor (RXFP2) was evaluated 112 days (16 weeks) after deslorelin treatment by qPCR and immunohistochemistry, and compared to untreated adult (CON, n = 6) and prepubertal (PRE, n = 8) dogs. Serum testosterone concentration decreased significantly by the onset of aspermia on study day 14 (four dogs) or day 21 (one dog), and was baseline on day 105 (week 15). In contrast, serum AMH started to increase only after the onset of aspermia and reached the maximum detectable concentration of the assay by day 49-105 in individual dogs. Testicular LHR gene expression in DES was lower than in CON and PRE (P < 0.0001), while AR gene expression in DES was similar to CON and significantly higher than PRE (P < 0.0001). Testicular AMH expression in DES was intermediate compared to the lowest mRNA levels found in CON and the highest in PRE (P ≤ 0.006). AMHR2 gene expression was similar between groups. AMH protein was detected in Sertoli cells only, while AMHR2 immunoreactivity was principally detected in Leydig cells which appeared to be increased in DES. INSL3 and RXFP2 gene expression was significantly downregulated in the DES testis along with noticeably weak Leydig cell immunosignals compared to CON. In conclusion, deslorelin treatment caused testicular LH insensitivity without affecting androgen sensitivity, and de-differentiation of Sertoli and Leydig cells. In DES, upregulation of the AMH-AMHR2 feed-back loop and downregulation of the INSL3-RXFP2 feed-forward loop are paracrine-autocrine mechanisms that may additionally regulate testosterone production independent of gonadotropins. Our results support AMH and INSL3 as unique biomarkers and paracrine-autocrine regulators of testis function involved in the intimate interplay between Sertoli and Leydig cells.
Collapse
|
33
|
Pan Q, Kay T, Depincé A, Adolfi M, Schartl M, Guiguen Y, Herpin A. Evolution of master sex determiners: TGF-β signalling pathways at regulatory crossroads. Philos Trans R Soc Lond B Biol Sci 2021; 376:20200091. [PMID: 34247498 DOI: 10.1098/rstb.2020.0091] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
To date, more than 20 different vertebrate master sex-determining genes have been identified on different sex chromosomes of mammals, birds, frogs and fish. Interestingly, six of these genes are transcription factors (Dmrt1- or Sox3- related) and 13 others belong to the TGF-β signalling pathway (Amh, Amhr2, Bmpr1b, Gsdf and Gdf6). This pattern suggests that only a limited group of factors/signalling pathways are prone to become top regulators again and again. Although being clearly a subordinate member of the sex-regulatory network in mammals, the TGF-β signalling pathway made it to the top recurrently and independently. Facing this rolling wave of TGF-β signalling pathways, this review will decipher how the TGF-β signalling pathways cope with the canonical sex gene regulatory network and challenge the current evolutionary concepts accounting for the diversity of sex-determining mechanisms. This article is part of the theme issue 'Challenging the paradigm in sex chromosome evolution: empirical and theoretical insights with a focus on vertebrates (Part I)'.
Collapse
Affiliation(s)
- Qiaowei Pan
- Department of Ecology and Evolution, University of Lausanne, 1015 Lausanne, Switzerland
| | - Tomas Kay
- Department of Ecology and Evolution, University of Lausanne, 1015 Lausanne, Switzerland
| | | | - Mateus Adolfi
- University of Würzburg, Developmental Biochemistry, Biocenter, 97074 Würzburg, Germany
| | - Manfred Schartl
- University of Würzburg, Developmental Biochemistry, Biocenter, 97074 Würzburg, Germany.,Xiphophorus Genetic Stock Center, Texas State University, San Marcos, TX 78666, USA
| | - Yann Guiguen
- INRAE, UR 1037 Fish Physiology and Genomics, 35000 Rennes, France
| | - Amaury Herpin
- INRAE, UR 1037 Fish Physiology and Genomics, 35000 Rennes, France.,State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, People's Republic of China
| |
Collapse
|
34
|
Xu H, Zhang M, Zhang H, Alpadi K, Wang L, Li R, Qiao J. Clinical Applications of Serum Anti-Müllerian Hormone Measurements in Both Males and Females: An Update. Innovation (N Y) 2021; 2:100091. [PMID: 34557745 PMCID: PMC8454570 DOI: 10.1016/j.xinn.2021.100091] [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: 11/04/2020] [Accepted: 01/03/2021] [Indexed: 12/17/2022] Open
Abstract
Infertility is one of the most common non-communicable diseases, affecting both men and women equally. Ovarian reserve, the number of primordial follicles in the ovaries is believed to be the most important determinants for female fertility. Anti-Müllerian hormone (AMH) secreted from granulosa cells of growing follicles is recognized as the most important biomarker for ovarian reserve. Ovarian reserve models have been developed using AMH and other hormonal indicators, thus childbearing plans and reproductive choices could be arranged by women. In assisted reproductive technology cycles, measurement of AMH helps to predict ovarian response and guide recombinant follicle-stimulating hormone dosing in women. Serum AMH level is increasingly being recognized as a potential surrogate marker for polycystic ovarian morphology, one of the criteria for diagnosis of polycystic ovarian syndrome. AMH is also secreted by Sertoli cells of testes in men, and AMH measurements in the prediction of surgical sperm recovery rate in men have also been investigated. AMH levels are significantly higher in boys than in girls before puberty. Therefore, serum levels of AMH in combination with testosterone is used for the differential diagnosis of disorders of sex development, anorchia, non-obstructive azoospermia, and persistent Müllerian duct syndrome. Recently, serum AMH measurements have also been used in fertility preservation programs in oncofertility, screening for granulosa cell tumors, and prediction of menopause applications. In this review, we will focus on clinical application of AMH in fertility assessments for healthy men and women, as well as for cancer patients. Anti-Müllerian hormone (AMH) plays a key role in models assessing ovarian reserve AMH is used for the differential diagnosis of disorders of sex development AMH provides a molecular marker for related fertility and infertility disorders An international standard will aid in the development of various AMH assays
Collapse
Affiliation(s)
- Huiyu Xu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, P.R. China.,National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, P.R. China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, P.R. China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, P.R. China
| | - Mengqian Zhang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, P.R. China.,National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, P.R. China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, P.R. China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, P.R. China
| | - Hongxian Zhang
- Department of Urology, Peking University Third Hospital, Beijing 100191, P.R. China
| | | | - Lina Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, P.R. China.,National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, P.R. China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, P.R. China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, P.R. China
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, P.R. China.,National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, P.R. China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, P.R. China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, P.R. China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, P.R. China.,National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, P.R. China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, P.R. China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, P.R. China
| |
Collapse
|
35
|
Sharma A, Minhas S, Dhillo WS, Jayasena CN. Male infertility due to testicular disorders. J Clin Endocrinol Metab 2021; 106:e442-e459. [PMID: 33295608 PMCID: PMC7823320 DOI: 10.1210/clinem/dgaa781] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Indexed: 12/13/2022]
Abstract
CONTEXT Male infertility is defined as the inability to conceive following 1 year of regular unprotected intercourse. It is the causative factor in 50% of couples and a leading indication for assisted reproductive techniques (ART). Testicular failure is the most common cause of male infertility, yet the least studied to date. EVIDENCE ACQUISITION The review is an evidence-based summary of male infertility due to testicular failure with a focus on etiology, clinical assessment, and current management approaches. PubMed-searched articles and relevant clinical guidelines were reviewed in detail. EVIDENCE SYNTHESIS/RESULTS Spermatogenesis is under multiple levels of regulation and novel molecular diagnostic tests of sperm function (reactive oxidative species and DNA fragmentation) have since been developed, and albeit currently remain as research tools. Several genetic, environmental, and lifestyle factors provoking testicular failure have been elucidated during the last decade; nevertheless, 40% of cases are idiopathic, with novel monogenic genes linked in the etiopathogenesis. Microsurgical testicular sperm extraction (micro-TESE) and hormonal stimulation with gonadotropins, selective estrogen receptor modulators, and aromatase inhibitors are recently developed therapeutic approaches for men with the most severe form of testicular failure, nonobstructive azoospermia. However, high-quality clinical trials data is currently lacking. CONCLUSIONS Male infertility due to testicular failure has traditionally been viewed as unmodifiable. In the absence of effective pharmacological therapies, delivery of lifestyle advice is a potentially important treatment option. Future research efforts are needed to determine unidentified factors causative in "idiopathic" male infertility and long-term follow-up studies of babies conceived through ART.
Collapse
Affiliation(s)
- Aditi Sharma
- Section of Endocrinology and Investigative Medicine, Imperial College London, UK
| | - Suks Minhas
- Department of Urology, Charing Cross Hospital, London, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, UK
| | - Channa N Jayasena
- Section of Endocrinology and Investigative Medicine, Imperial College London, UK
| |
Collapse
|
36
|
Qin W, Wang B, Yang L, Yuan Y, Xiong X, Li J, Yin S. Molecular cloning, characterization, and function analysis of the AMH gene in Yak (Bos grunniens) Sertoli cells. Theriogenology 2021; 163:1-9. [PMID: 33476894 DOI: 10.1016/j.theriogenology.2021.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 10/22/2022]
Abstract
Sertoli cells (SCs) are important testicular somatic cells that carry out various functions in spermatogenesis. Understanding the biological mechanisms underlying SC development may facilitate the understanding of animal reproduction. Anti-Mullerian hormone (AMH) is a dimeric glycoprotein produced by SCs and plays essential roles in spermatogenesis. In this study, we cloned the coding sequence of the yak AMH, predicated the structure of AMH protein, analyzed AMH expression in the testis at different stages, and studied the functions of AMH in yak SCs. The open reading frame (ORF) of the yak AMH contained 1728 bp and encoded 575 amino acids. Structural analysis revealed that the yak AMH protein had a highly conserved transforming growth factor-β (TGF-β) domain. The mRNA expression level for the AMH gene in yak testis increased significantly from the fetal stage to calf stage, then decreased with the increase of age. The highest expression was found in calf stage. Cell proliferation was depressed in AMH-deficient SCs. Expression of several genes involved in SC proliferation and development, including PCNA, BCL-2, BAX, CASP3, AR and AMHR2 were altered after knockdown of AMH. Also, three SC-secreted factors essential for spermatogenesis, SCF, GDNF and ABP, were repressed at the transcription level after AMH knockdown in yak SCs. Moreover, supplementation with exogenous AMH protein partially rescued SC proliferation, and the expression of PCNA, BCL-2, AR and AMHR2 after AMH gene interference. This research provided theoretical basis for understanding the mechanism by which AMH regulates yak spermatogenesis and might give new insights in improving yak reproductive performance in the future.
Collapse
Affiliation(s)
- Wenchang Qin
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu, Sichuan, 610041, China; College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, 610041, China
| | - Bin Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu, Sichuan, 610041, China; College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, 610041, China
| | - Liuqing Yang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu, Sichuan, 610041, China; College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, 610041, China
| | - YuJie Yuan
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu, Sichuan, 610041, China; College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, 610041, China
| | - Xianrong Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu, Sichuan, 610041, China; College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, 610041, China
| | - Jian Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu, Sichuan, 610041, China; College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, 610041, China
| | - Shi Yin
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu, Sichuan, 610041, China; College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, 610041, China; Key Laboratory of Modem Technology (Southwest Minzu University), State Ethnic Affairs Commission, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
37
|
Molecular insights into hormone regulation via signaling pathways in Sertoli cells: With discussion on infertility and testicular tumor. Gene 2020; 753:144812. [DOI: 10.1016/j.gene.2020.144812] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/17/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023]
|
38
|
da Rosa LA, Escott GM, Simonetti RB, da Silva JCD, Werlang ICR, Goldani MZ, de Fraga LS, Loss EDS. Role of non-classical effects of testosterone and epitestosterone on AMH balance and testicular development parameters. Mol Cell Endocrinol 2020; 511:110850. [PMID: 32387527 DOI: 10.1016/j.mce.2020.110850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 04/02/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022]
Abstract
Testosterone (T) and its 17-α epimer, epitestosterone (EpiT), are described as having non-classical effects in addition to their classical androgen actions via the intracellular androgen receptor (iAR). The actions of these androgens play an essential role in triggering factors that shift Sertoli cells from the proliferation phase to the maturation phase. This process is essential for successful spermatogenesis and normal fertility. The aim of this work was to investigate the difference between T and EpiT effects in normal and in chemically castrated Wistar rats. We also tested the effects of these hormones when the iAR-dependent pathways were inhibited by the antiandrogen flutamide. Rats were chemically castrated on postnatal day (pnd) 5 using EDS, a cytotoxic agent that promotes apoptosis of Leydig cells, reducing androgen levels. Then, animals received replacement with T or EpiT and were treated or not with flutamide from pnd 6 to pnd 13 or 20 and were euthanized on pnd 14 and 21. Animals treated with EpiT and flutamide had lower body weight overall. Epididymis weight was also reduced in animals treated with EpiT and flutamide. Flutamide per se reduced epididymis weight at both ages (pnd 14 and 21). Testicular weight and the testicular/body weight ratio were reduced in EDS animals, and flutamide further reduced this weight in animals which received T replacement. EDS administration reduced mRNA levels of both AMH (anti-Müllerian hormone) and its receptor, AMHR2, at pnd 14. In the testes of flutamide-treated animals, EpiT reduced AMH, and both T and EpiT replacement diminished AMHR2 mRNA expression also on pnd 14. EDS decreased iAR expression, and androgen replacement did not change this effect on pnd 21. In rats receiving flutamide, only those also receiving T and EpiT replacement exhibited decreased iAR expression. An increase in connexin 43 expression was observed in animals treated with EpiT without flutamide, whereas in rats treated with flutamide, both hormones were ineffective to increase connexin 43 expression reduced by EDS. Our results suggest that EpiT has an antiandrogen effect on androgen-sensitive tissues such as the epididymis. Nonetheless, the effects of T and EpiT on testicular development parameters are similar. Both hormones may act through their iAR-independent non-classical pathway, regulating AMH and AMHR2, as well as iAR expression.
Collapse
Affiliation(s)
- Luciana Abreu da Rosa
- Laboratório de Endocrinologia Experimental e Eletrofisiologia, Departamento de Fisiologia, Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Pediatria Translacional/Núcleo de Estudos em Saúde da Criança e do Adolescente (NESCA)/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Gustavo Monteiro Escott
- Laboratório de Endocrinologia Experimental e Eletrofisiologia, Departamento de Fisiologia, Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Médicas: Endocrinologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Rajla Bressan Simonetti
- Laboratório de Endocrinologia Experimental e Eletrofisiologia, Departamento de Fisiologia, Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Jessica Caroline Dias da Silva
- Laboratório de Endocrinologia Experimental e Eletrofisiologia, Departamento de Fisiologia, Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Isabel Cristina Ribas Werlang
- Laboratório de Pediatria Translacional/Núcleo de Estudos em Saúde da Criança e do Adolescente (NESCA)/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Marcelo Zubaran Goldani
- Laboratório de Pediatria Translacional/Núcleo de Estudos em Saúde da Criança e do Adolescente (NESCA)/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Luciano Stürmer de Fraga
- Laboratório de Endocrinologia Experimental e Eletrofisiologia, Departamento de Fisiologia, Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Eloísa da Silveira Loss
- Laboratório de Endocrinologia Experimental e Eletrofisiologia, Departamento de Fisiologia, Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
39
|
Rajeswari JJ, Hatef A, Unniappan S. Nesfatin-1-like peptide suppresses hypothalamo-pituitary-gonadal mRNAs, gonadal steroidogenesis, and oocyte maturation in fish†. Biol Reprod 2020; 103:802-816. [PMID: 32542346 DOI: 10.1093/biolre/ioaa106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/02/2020] [Accepted: 06/11/2020] [Indexed: 12/16/2022] Open
Abstract
Nucleobindin (Nucb)-1 and Nucb2 are DNA and Ca2+ binding proteins with multiple functions in vertebrates. Prohormone convertase-mediated processing of Nucb2 results in the production of biologically active nesfatin-1. Nesfatin-1 is involved in the regulation of reproduction in many vertebrates, including fish. Our lab originally reported a nesfatin-1-like peptide (Nlp) encoded in Nucb1 that exhibits nesfatin-1-like metabolic effects. We hypothesized that Nlp has a suppressive role in the reproductive physiology of fish. In this research, whether Nlp regulates reproductive hormones and oocyte maturation in fish were determined. Single intraperitoneal (IP) injection of goldfish Nlp (50 ng/g body weight) suppressed salmon and chicken gonadotropin-releasing hormone (sgnrh and cgnrh2), gonadotropin-inhibiting hormone (gnih) and its receptor (gnihr), and kisspeptin and brain aromatase mRNA expression in the hypothalamus of both male and female goldfish. In the pituitary, Nlp decreased mRNAs encoding lhb, fshb and kisspeptin and its receptor, while a significant increase in gnih and gnihr was observed. In the gonads, lh (only in male fish) and fsh receptor mRNAs were also significantly downregulated in Nlp-injected fish. Sex-specific modulation of gnih, gnihr, and kisspeptin system in the gonads was also observed. Nlp decreased sex steroidogenic enzyme encoding mRNAs and circulating levels of testosterone and estradiol. In addition, incubation of zebrafish ovarian follicles with Nlp resulted in a reduction in oocyte maturation. These results provide evidence for a robust role for Nlp in regulating reproductive hormones in goldfish and oocyte maturation in zebrafish, and these effects resemble that of nesfatin-1.
Collapse
Affiliation(s)
- Jithine Jayakumar Rajeswari
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Canada
| | - Azadeh Hatef
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Canada.,Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Canada
| |
Collapse
|
40
|
Seminal Plasma Anti-Müllerian Hormone: A Potential AI-Boar Fertility Biomarker? BIOLOGY 2020; 9:biology9040078. [PMID: 32290279 PMCID: PMC7236007 DOI: 10.3390/biology9040078] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 12/26/2022]
Abstract
The anti-Müllerian hormone (AMH), a Sertoli cell-secreted glycoprotein that is present in seminal plasma (SP), is considered as a marker of spermatogenesis in humans. This study aimed to evaluate the presence of this hormone in boar SP, together with its putative relationship with sperm quality, function, and in vivo fertility parameters in liquid-stored semen samples. The concentration of SP-AMH was assessed in 126 ejaculates from artificial insemination (AI)-boars (n = 92) while using a commercial Enzyme-Linked ImmunoSorbent Assay (ELISA) kit with monoclonal antibodies specific for Sus scrofa AMH (CEA228Po, Cloud-clone). Sperm quality (concentration, motility, viability, and acrosome damage) and functionality (membrane lipid disorder and intracellular H2O2 generation) were assessed in semen samples at 0 and 72 h of liquid-storage. In addition, fertility parameters from 3113 sows inseminated with the AI-boars were recorded in terms of farrowing rate, litter size, number of stillbirths per litter, and the duration of pregnancy over a 12-month period. The results revealed that the SP-AMH concentration varied widely among boar ejaculates, with no differences among breeds. Moreover, the SP-AMH concentration proved to be a good predictive biomarker for sperm concentration (p ˂ 0.05), but poor for other sperm quality, functionality, and in vivo fertility parameters of liquid-stored semen samples from AI-boars.
Collapse
|
41
|
Sansone A, Isidori AM, Kliesch S, Schlatt S. Immunohistochemical characterization of the anti-Müllerian hormone receptor type 2 (AMHR-2) in human testes. Endocrine 2020; 68:215-221. [PMID: 32026338 PMCID: PMC7160062 DOI: 10.1007/s12020-020-02210-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/17/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE In males, AMH is secreted by immature Sertoli cells; following exposure to endogenous androgens, Sertoli cells undergo a process of maturation which ultimately inhibits AMH expression to undetectable levels in the serum. However, expression of AMH receptor (AMHR-2) has never been studied in human testes, and high intratubular concentrations of AMH have been reported in recent literature. We therefore assessed expression of AMHR-2 in several testicular tissue samples by immunohistochemistry (IHC). METHODS The IHC method was first validated on tissue samples from healthy human testis (n = 2) and from marmoset ovary (n = 1). The same method was then used for assessment on testicular histopathology specimens from patients with mixed atrophy (MA, n = 2), spermatogenetic arrest (SA, n = 2), Sertoli cell-only syndrome (SCO, n = 1), Klinefelter syndrome (KS, n = 1), and nonseminomatous germ cell tumors (NSGCT, n = 1). Tissue samples from two subjects at different pubertal stages (AndroProtect (AP), aged 5 and 14 years) with hematological malignancies were also retrieved. RESULTS In adult men, AMHR-2 was expressed on peritubular mesenchymal cells, with patterns closely mirroring α-smooth muscle actin expression. Similar patterns were preserved in almost all conditions; however, in nonseminomatous germ cell tumors the tissue architecture was lost, including AMHR-2 expression. More positive and diffuse staining was observed in tissue samples from prepubertal testes. CONCLUSIONS In specimens from both healthy and affected testes, AMHR-2 expression appears weaker in adult than in prepubertal tissue sections. The persistence of AMHR-2 expression seemingly hints at a possible effect of intratesticular AMH on the tubular walls.
Collapse
Affiliation(s)
- A Sansone
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149, Münster, Germany
- Department of Experimental Medicine, Food Science and Endocrinology, Sapienza University of Rome, 00161, Rome, Italy
| | - A M Isidori
- Department of Experimental Medicine, Food Science and Endocrinology, Sapienza University of Rome, 00161, Rome, Italy
| | - S Kliesch
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149, Münster, Germany
| | - S Schlatt
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149, Münster, Germany.
| |
Collapse
|
42
|
Xiong L, Yang M, Zheng K, Wang Z, Gu S, Tong J, Liu J, Shah NA, Nie L. Comparison of Adult Testis and Ovary MicroRNA Expression Profiles in Reeves' Pond Turtles ( Mauremys reevesii) With Temperature-Dependent Sex Determination. Front Genet 2020; 11:133. [PMID: 32194623 PMCID: PMC7061903 DOI: 10.3389/fgene.2020.00133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Some differentially expressed genes (DEGs) that encode key enzymes involved in steroidogenic biosynthesis (CYP19A1) and key molecules related to gonadal functions (DMRT1, SOX9, AMH, FOXL2, WNT4, RSPO2, and GDF9) have been identified in adult gonadal RNA-seq studies of Reeves' pond turtle (Mauremys reevesii) with temperature-dependent sex determination (TSD). Gonadal functional maintenance and gametogenesis comprises a highly regulated and coordinated biological process, and increasing evidence indicates that microRNAs (miRNAs) may be involved in this dynamic program. However, it is not clear how the regulatory network comprising miRNAs changes the expression levels of these genes. In this study, miRNA sequencing of adult testis and ovary tissues from M. reevesii detected 25 known and 379 novel miRNAs, where 60 miRNAs were differentially expressed in the testis and ovary. A total of 1,477 target genes based on the differentially expressed miRNAs were predicted, where 221 target genes also exhibited differential expression. To verify the accuracy of the sequencing data, 10 differentially expressed miRNAs were validated by quantitative reverse transcription real-time PCR, and were found to be consistent with the transcriptome sequencing results. Moreover, several miRNA/target gene pairs, i.e., mre-let-7a-5p/mre-let-7e-5p and CYP19A1, mre-miR-200a-3p and DMRT1, mre-miR-101-3p and SOX9, and mre-miR-138-5p and AMH were identified. To explore the regulatory role of miRNAs, we conducted target gene enrichment analysis of the miRNAs and 221 target genes in the regulatory network. The signaling pathways related to gonadal functional maintenance and gametogenesis based on the DEGs and target genes were then compared. Our findings provide crucial information to facilitate further research into the regulatory mechanisms involving miRNAs in turtle species with TSD.
Collapse
Affiliation(s)
- Lei Xiong
- Life Science College, Provincial Key Lab of the Conservation and Exploitation Research of Biological Resources in Anhui, Anhui Normal University, Wuhu, China.,Biochemistry Department, Wannan Medical College, Wuhu, China
| | - Mengli Yang
- Life Science College, Provincial Key Lab of the Conservation and Exploitation Research of Biological Resources in Anhui, Anhui Normal University, Wuhu, China
| | - Kai Zheng
- Life Science College, Provincial Key Lab of the Conservation and Exploitation Research of Biological Resources in Anhui, Anhui Normal University, Wuhu, China
| | - Ziming Wang
- Life Science College, Provincial Key Lab of the Conservation and Exploitation Research of Biological Resources in Anhui, Anhui Normal University, Wuhu, China
| | - Shengli Gu
- Life Science College, Provincial Key Lab of the Conservation and Exploitation Research of Biological Resources in Anhui, Anhui Normal University, Wuhu, China.,Biochemistry Department, Wannan Medical College, Wuhu, China
| | - Jiucui Tong
- Life Science College, Provincial Key Lab of the Conservation and Exploitation Research of Biological Resources in Anhui, Anhui Normal University, Wuhu, China.,Biochemistry Department, Wannan Medical College, Wuhu, China
| | - Jianjun Liu
- Life Science College, Provincial Key Lab of the Conservation and Exploitation Research of Biological Resources in Anhui, Anhui Normal University, Wuhu, China
| | - Nadar Ali Shah
- Life Science College, Provincial Key Lab of the Conservation and Exploitation Research of Biological Resources in Anhui, Anhui Normal University, Wuhu, China
| | - Liuwang Nie
- Life Science College, Provincial Key Lab of the Conservation and Exploitation Research of Biological Resources in Anhui, Anhui Normal University, Wuhu, China
| |
Collapse
|
43
|
Zhao X, Wang Y, Ma M, Zhang W, Si J, Wang W, Lin Z, Han G, Chen CD, Tong M, Kuang Y, Lyu Q. A new panel containing specific spermatogenesis markers to identify spermatogenic cells in nonobstructive azoospermia patients. Acta Biochim Biophys Sin (Shanghai) 2019; 51:656-659. [PMID: 31132083 DOI: 10.1093/abbs/gmz056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Indexed: 01/27/2023] Open
Affiliation(s)
- Xinxi Zhao
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yiqin Wang
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Meng Ma
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Zhang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiqiang Si
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Wang
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhen Lin
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Gang Han
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Charlie Degui Chen
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Minghan Tong
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qifeng Lyu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
Rodprasert W, Virtanen HE, Mäkelä JA, Toppari J. Hypogonadism and Cryptorchidism. Front Endocrinol (Lausanne) 2019; 10:906. [PMID: 32010061 PMCID: PMC6974459 DOI: 10.3389/fendo.2019.00906] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 12/11/2019] [Indexed: 01/24/2023] Open
Abstract
Congenital cryptorchidism (undescended testis) is one of the most common congenital urogenital malformations in boys. Prevalence of cryptorchidism at birth among boys born with normal birth weight ranges from 1.8 to 8.4%. Cryptorchidism is associated with a risk of low semen quality and an increased risk of testicular germ cell tumors. Testicular hormones, androgens and insulin-like peptide 3 (INSL3), have an essential role in the process of testicular descent from intra-abdominal position into the scrotum in fetal life. This explains the increased prevalence of cryptorchidism among boys with diseases or syndromes associated with congenitally decreased secretion or action of androgens, such as patients with congenital hypogonadism and partial androgen insensitivity syndrome. There is evidence to support that cryptorchidism is associated with decreased testicular hormone production later in life. It has been shown that cryptorchidism impairs long-term Sertoli cell function, but may also affect Leydig cells. Germ cell loss taking place in the cryptorchid testis is proportional to the duration of the condition, and therefore early orchiopexy to bring the testis into the scrotum is the standard treatment. However, the evidence for benefits of early orchiopexy for testicular endocrine function is controversial. The hormonal treatments using human chorionic gonadotropin (hCG) or gonadotropin-releasing hormone (GnRH) to induce testicular descent have low success rates, and therefore they are not recommended by the current guidelines for management of cryptorchidism. However, more research is needed to assess the effects of hormonal treatments during infancy on future male reproductive health.
Collapse
Affiliation(s)
- Wiwat Rodprasert
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- The Population Research Centre, University of Turku, Turku, Finland
- *Correspondence: Wiwat Rodprasert
| | - Helena E. Virtanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- The Population Research Centre, University of Turku, Turku, Finland
| | - Juho-Antti Mäkelä
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- The Population Research Centre, University of Turku, Turku, Finland
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- The Population Research Centre, University of Turku, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| |
Collapse
|