1
|
Wang K, Suo Y, Shen D, Shi Y, Jin X, Li Y, Li C. Improvement in Heat Stress-Induced Damage to Sperm Quality Following Fecal Microbiota Transplantation from L-Arginine-Treated Mice. Animals (Basel) 2025; 15:796. [PMID: 40150325 PMCID: PMC11939313 DOI: 10.3390/ani15060796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/02/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Heat stress has become a significant concern in animal husbandry, as it adversely affects reproductive performance, particularly sperm quality, through mechanisms that are not fully understood. This study aimed to investigate the protective effects of L-arginine against heat stress-induced sperm damage and explore its potential mechanisms through the modulation of the intestinal microbiota. This study consisted of two experiments. First, in a heat-stressed mouse model, L-arginine was administered to evaluate its effects on the reproductive health of heat-stressed mice. In the second experiment, by transplanting L-arginine-induced changes in the gut microbiota into heat-stressed mice, the protective effects of the microbiota on the sperm of heat-stressed mice were assessed. The findings revealed a significant amelioration of decreased sperm quality and testicular injury induced by heat stress. Post heat stress, mice supplemented with L-arginine presented an increase in seminal vesicle gland weight and index, partial alleviation of testicular tissue morphology, and a substantial increase in testosterone concentration (p < 0.05). Additionally, L-arginine upregulated the expression of testosterone synthesis genes and the mRNA levels of sperm generation-related genes, including 3β-HSD, Stra8, WT1, and Gdnf (p < 0.05). Concurrently, L-arginine-induced microbial communities mitigated heat stress-induced decreases in sperm quality and testicular injury, coupled with increases in the mRNA expression levels of Cyp17a1, 17β-HSD, Plzf, and Gdnf (p < 0.05). Furthermore, there was a reduction in the expression of proinflammatory factors, namely, NFκB, MyD88, TNF-α, and TGF-β3 (p < 0.05). In conclusion, L-arginine may influence the ratio of beneficial bacteria to harmful bacteria in the intestinal microbiota, thereby reducing inflammation caused by heat stress, maintaining intestinal health, and influencing the microenvironment for spermatogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Chunmei Li
- Research Centre for Livestock Environmental Control and Smart Production, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (K.W.); (Y.S.); (D.S.); (Y.S.); (X.J.); (Y.L.)
| |
Collapse
|
2
|
Delgouffe E, Silva SM, Chalmel F, Cools W, Raets C, Tilleman K, T'Sjoen G, Baert Y, Goossens E. Partial rejuvenation of the spermatogonial stem cell niche after gender-affirming hormone therapy in trans women. eLife 2025; 13:RP94825. [PMID: 39773877 PMCID: PMC11706602 DOI: 10.7554/elife.94825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Although the impact of gender-affirming hormone therapy (GAHT) on spermatogenesis in trans women has already been studied, data on its precise effects on the testicular environment is poor. Therefore, this study aimed to characterize, through histological and transcriptomic analysis, the spermatogonial stem cell niche of 106 trans women who underwent standardized GAHT, comprising estrogens and cyproterone acetate. A partial dedifferentiation of Sertoli cells was observed, marked by the co-expression of androgen receptor and anti-Müllerian hormone which mirrors the situation in peripubertal boys. The Leydig cells also exhibited a distribution analogous to peripubertal tissue, accompanied by a reduced insulin-like factor 3 expression. Although most peritubular myoid cells expressed alpha-smooth muscle actin 2, the expression pattern was disturbed. Besides this, fibrosis was particularly evident in the tubular wall and the lumen was collapsing in most participants. A spermatogenic arrest was also observed in all participants. The transcriptomic profile of transgender tissue confirmed a loss of mature characteristics - a partial rejuvenation - of the spermatogonial stem cell niche and, in addition, detected inflammation processes occurring in the samples. The present study shows that GAHT changes the spermatogonial stem cell niche by partially rejuvenating the somatic cells and inducing fibrotic processes. These findings are important to further understand how estrogens and testosterone suppression affect the testis environment, and in the case of orchidectomized testes as medical waste material, their potential use in research.
Collapse
Affiliation(s)
- Emily Delgouffe
- Biology of the Testis (BITE) Laboratory, Genetics, Reproduction and Development (GRAD) Research Group, Vrije Universiteit BrusselBrusselsBelgium
| | - Samuel Madureira Silva
- Biology of the Testis (BITE) Laboratory, Genetics, Reproduction and Development (GRAD) Research Group, Vrije Universiteit BrusselBrusselsBelgium
| | - Frédéric Chalmel
- Inserm, EHESP, Institut de Recherche en Santé, Environnement et Travail (IRSET), Université de RennesRennesFrance
| | - Wilfried Cools
- Core facility, Support for Quantitative and Qualitative Research (SQUARE), Vrije Universiteit BrusselBrusselsBelgium
| | - Camille Raets
- Core facility, Support for Quantitative and Qualitative Research (SQUARE), Vrije Universiteit BrusselBrusselsBelgium
| | - Kelly Tilleman
- Department for Reproductive Medicine, Ghent University HospitalGhentBelgium
| | - Guy T'Sjoen
- Department of Endocrinology and Center for Sexology and Gender, Ghent University HospitalGhentBelgium
| | - Yoni Baert
- Biology of the Testis (BITE) Laboratory, Genetics, Reproduction and Development (GRAD) Research Group, Vrije Universiteit BrusselBrusselsBelgium
- In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit BrusselBrusselsBelgium
| | - Ellen Goossens
- Biology of the Testis (BITE) Laboratory, Genetics, Reproduction and Development (GRAD) Research Group, Vrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
3
|
Murugesh V, Ritting M, Salem S, Aalam SMM, Garcia J, Chattha AJ, Zhao Y, Knapp DJHF, Kalthur G, Granberg CF, Kannan N. Puberty Blocker and Aging Impact on Testicular Cell States and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.23.586441. [PMID: 38585884 PMCID: PMC10996503 DOI: 10.1101/2024.03.23.586441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Spermatogonial stem cell (SSC) acquisition of meiotogenetic state during puberty to produce genetically diverse gametes is blocked by drugs collectively referred as 'puberty blocker' (PB). Investigating the impact of PB on juvenile SSC state and function is challenging due to limited tissue access and clinical data. Herein, we report largest clinically annotated juvenile testicular biorepository with all children with gender dysphoria on chronic PB treatment highlighting shift in pediatric patient demography in US. At the tissue level, we report mild-to-severe sex gland atrophy in PB treated children. We developed most extensive integrated single-cell RNA dataset to date (>100K single cells; 25 patients), merging both public and novel (52 month PB-treated) datasets, alongside innovative computational approach tailed for germ cells and evaluated the impact of PB and aging on SSC. We report novel constitutional ranges for each testicular cell type across the entire age spectrum, distinct effects of treatments on prepubertal vs adult SSC, presence of spermatogenic epithelial cells exhibiting post-meiotic-state, irrespective of age, puberty status, or PB treatment. Further, we defined distinct effects of PB and aging on testicular cell lineage composition, and SSC meiotogenetic state and function. Using single cell data from prepubertal and young adult, we were able to accurately predict sexual maturity based both on overall cell type proportions, as well as on gene expression patterns within each major cell type. Applying these models to a PB-treated patient that they appeared pre-pubertal across the entire tissue. This combined with the noted gland atrophy and abnormalities from the histology data raise a potential concern regarding the complete 'reversibility' and reproductive fitness of SSC. The biorepository, data, and research approach presented in this study provide unique opportunity to explore the impact of PB on testicular reproductive health.
Collapse
Affiliation(s)
- Varshini Murugesh
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Megan Ritting
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Salem Salem
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Joaquin Garcia
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Asma J Chattha
- Department of Pediatrics, Mayo Clinic, Rochester, MN, USA
| | - Yulian Zhao
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
| | - David JHF Knapp
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Institut de Recherche en Immunologie et Cancérologie, and Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montreal, QC, Canada
- Senior authors
| | - Guruprasad Kalthur
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
- Senior authors
| | | | - Nagarajan Kannan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Rochester, MN, USA
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, USA
- Senior authors
- Lead contact
| |
Collapse
|
4
|
Kvernebo Sunnergren K, Dahlgren J, Ankarberg-Lindgren C. Mini review shows that a testicular volume of 3 mL was the most reliable clinical sign of pubertal onset in males. Acta Paediatr 2023; 112:2300-2306. [PMID: 37410401 DOI: 10.1111/apa.16899] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 07/07/2023]
Abstract
AIM We aimed to evaluate aspects of pubertal development to identify the most reliable clinical sign of pubertal onset in males. METHODS We performed a mini review of the literature. RESULTS In 1951 Reynolds and Wines categorised pubic hair growth and genital development in five stages by visual inspection. Today the Tanner scale is used to assess the five stages of pubertal development, The second genital stage, characterised by enlargement of the scrotum defines pubertal onset in males. Testicular volume may be evaluated by using a calliper or by ultrasound scan. The Prader orchidometer, described in 1966, offers a method for evaluating testicular growth by palpation. Pubertal onset is commonly defined as testicular volume >3 or ≥4 mL. The development of sensitive laboratory methods has enabled studies analysing hormonal activity in the hypothalamus-pituitary-gonadal axis. We review the relationships between physical and hormonal signs of puberty. We also discuss the results of studies assessing different aspects of pubertal development with a focus on identifying the most reliable clinical sign of pubertal onset in males. CONCLUSION A substantial amount of evidence supports testicular volume of 3 mL as the most reliable clinical sign of male pubertal onset.
Collapse
Affiliation(s)
- Kjersti Kvernebo Sunnergren
- Department of Pediatrics, Göteborg Pediatric Growth Research Center (GP-GRC), Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Child and Adolescent Psychiatry, Ryhov County Hospital, Jönköping, Sweden
| | - Jovanna Dahlgren
- Department of Pediatrics, Göteborg Pediatric Growth Research Center (GP-GRC), Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Endocrinology, Region Västra Götaland, Sahlgrenska University Hospital, Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Carina Ankarberg-Lindgren
- Department of Pediatrics, Göteborg Pediatric Growth Research Center (GP-GRC), Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
5
|
Albrethsen J, Østergren PB, Norup PB, Sønksen J, Fode M, Kistorp C, Nordsborg NB, Solheim SA, Mørkeberg J, Main KM, Juul A. Serum Insulin-like Factor 3, Testosterone, and LH in Experimental and Therapeutic Testicular Suppression. J Clin Endocrinol Metab 2023; 108:2834-2839. [PMID: 37235781 DOI: 10.1210/clinem/dgad291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Abstract
BACKGROUND Serum insulin-like factor 3 (INSL3) is a Leydig cell biomarker, but little is known about the circulating concentration of INSL3 during hypothalamus-pituitary-testicular suppression. AIM To study the concomitant changes in serum concentrations of INSL3, testosterone, and LH during experimental and therapeutic testicular suppression. METHODS We included serum samples from 3 different cohorts comprising subjects before and after testicular suppression: (1) 6 healthy young men who were treated with androgens (Sustanon, Aspen Pharma, Dublin, Ireland); 2) 10 transgender girls (male sex assigned at birth) who were treated with 3-monthly GnRH agonist injections (Leuprorelinacetat, Abacus Medicine, Copenhagen, Denmark); and (3) 55 patients with prostate cancer who were randomized to surgical castration (bilateral subcapsular orchiectomy) or treatment with GnRH agonist (Triptorelin, Ipsen Pharma, Kista, Sweden). Serum INSL3 and testosterone concentrations were quantified in stored serum samples using validated liquid chromatography-tandem mass spectrometry methodologies, and LH was measured by an ultrasensitive immunoassay. RESULTS The circulating concentrations of INSL3, testosterone, and LH decreased during experimental testicular suppression in healthy young men by Sustanon injections and subsequently returned to baseline levels after release of suppression. All 3 hormones decreased during therapeutic hormonal hypothalamus-pituitary-testicular suppression in transgender girls and in patients with prostate cancer. CONCLUSION INSL3 resembles testosterone as a sensitive marker of testicular suppression and reflects Leydig cell function, also during exposure to exogenous testosterone. Serum INSL3 measurements may complement testosterone as a Leydig cell marker in male reproductive disorders, during therapeutic testicular suppression as well as in surveillance of illicit use of androgens.
Collapse
Affiliation(s)
- Jakob Albrethsen
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
- International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet and University of Copenhagen, 2100 Copenhagen, Denmark
| | - Peter Busch Østergren
- Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Endocrinology, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
| | - Pernille Badsberg Norup
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
- International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet and University of Copenhagen, 2100 Copenhagen, Denmark
| | - Jens Sønksen
- Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Endocrinology, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
| | - Mikkel Fode
- Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Endocrinology, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
| | - Caroline Kistorp
- Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Nutrition, Exercise and Sports, University of Copenhagen, 2100 Copenhagen, Denmark
| | | | - Sara Amalie Solheim
- Science and Research, Anti Doping Denmark, 2660 Brøndby, Denmark
- Department of Urology, Copenhagen University Hospital - 2730 Herlev and Gentofte, Copenhagen, Denmark
| | - Jakob Mørkeberg
- Department of Urology, Copenhagen University Hospital - 2730 Herlev and Gentofte, Copenhagen, Denmark
| | - Katharina M Main
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
- International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet and University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
- International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet and University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
6
|
The Klinefelter Syndrome and Testicular Sperm Retrieval Outcomes. Genes (Basel) 2023; 14:genes14030647. [PMID: 36980920 PMCID: PMC10048758 DOI: 10.3390/genes14030647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Klinefelter syndrome (KS), caused by the presence of an extra X chromosome, is the most prevalent chromosomal sexual anomaly, with an estimated incidence of 1:500/1000 per male live birth (karyotype 47,XXY). High stature, tiny testicles, small penis, gynecomastia, feminine body proportions and hair, visceral obesity, and testicular failure are all symptoms of KS. Endocrine (osteoporosis, obesity, diabetes), musculoskeletal, cardiovascular, autoimmune disorders, cancer, neurocognitive disabilities, and infertility are also outcomes of KS. Causal theories are discussed in addition to hormonal characteristics and testicular histology. The retrieval of spermatozoa from the testicles for subsequent use in assisted reproduction treatments is discussed in the final sections. Despite testicular atrophy, reproductive treatments allow excellent results, with rates of 40–60% of spermatozoa recovery, 60% of clinical pregnancy, and 50% of newborns. This is followed by a review on the predictive factors for successful sperm retrieval. The risks of passing on the genetic defect to children are also discussed. Although the risk is low (0.63%) when compared to the general population (0.5–1%), patients should be informed about embryo selection through pre-implantation genetic testing (avoids clinical termination of pregnancy). Finally, readers are directed to a number of reviews where they can enhance their understanding of comprehensive diagnosis, clinical care, and fertility preservation.
Collapse
|
7
|
Holt R, Yahyavi SK, Kooij I, Andreassen CH, Andersson AM, Juul A, Jørgensen N, Blomberg Jensen M. Low serum anti-Müllerian hormone is associated with semen quality in infertile men and not influenced by vitamin D supplementation. BMC Med 2023; 21:79. [PMID: 36855109 PMCID: PMC9976369 DOI: 10.1186/s12916-023-02782-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 02/13/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Anti-Müllerian hormone (AMH) is released by testicular Sertoli cells and of great importance during fetal male sexual development, but less is known about the role of circulating AMH during adulthood. In vitro studies have shown that vitamin D may induce AMH transcription, but a controlled trial investigating the possible effect of vitamin D on serum AMH has not been conducted in men. METHODS A single-center, double-blinded, randomized placebo-controlled clinical trial (NCT01304927) conducted in Copenhagen, Denmark. A total of 307 infertile men were included and randomly assigned (1:1) to a single dose of 300,000 IU cholecalciferol followed by 1400 IU cholecalciferol + 500 mg of calcium daily (n = 151) or placebo (n = 156) for 150 days. Difference in serum AMH was a predefined secondary endpoint. Explorative outcomes were associations between serum AMH and gonadal function in infertile men. The primary endpoint was difference in semen quality and has previously been published. RESULTS Infertile men in the lowest AMH tertile had significantly lower sperm concentration (∆T3-1 16 mill/mL (228%); P < 0.001), sperm count (∆T3-1 55 million (262%); P < 0.001), motile sperm count (∆T3-1 28 million (255%); P < 0.001), progressive motile sperm count (∆T3-1 18 million (300%); P < 0.001), testis size (∆T3-1 2.7 mL (16%); P < 0.001), serum inhibin B (∆T3-1 72 pg/mL (59%); P < 0.001), inhibin B/FSH ratio (∆T3-1 48 (145%); P < 0.001), and higher FSH (∆T3-1 2.6 (38%); P < 0.001) than the tertile of infertile men with highest serum AMH. Vitamin D supplementation had no effect on serum AMH compared with placebo treatment. CONCLUSIONS In infertile men, low serum AMH is associated with severely impaired gonadal function illustrated by poor semen quality and lower testosterone/LH ratio. Serum AMH in infertile men was not influenced by vitamin D supplementation.
Collapse
Affiliation(s)
- Rune Holt
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Sam Kafai Yahyavi
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Ireen Kooij
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Christine Hjorth Andreassen
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Anna-Maria Andersson
- 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
| | - 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
| | - Martin Blomberg Jensen
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark. .,Division of Bone and Mineral Research, HSDM/HMS, Harvard University, Boston, USA.
| |
Collapse
|
8
|
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: 4.7] [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
|
9
|
Understanding the Underlying Molecular Mechanisms of Meiotic Arrest during In Vitro Spermatogenesis in Rat Prepubertal Testicular Tissue. Int J Mol Sci 2022; 23:ijms23115893. [PMID: 35682573 PMCID: PMC9180380 DOI: 10.3390/ijms23115893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 12/10/2022] Open
Abstract
In vitro spermatogenesis appears to be a promising approach to restore the fertility of childhood cancer survivors. The rat model has proven to be challenging, since germ cell maturation is arrested in organotypic cultures. Here, we report that, despite a meiotic entry, abnormal synaptonemal complexes were found in spermatocytes, and in vitro matured rat prepubertal testicular tissues displayed an immature phenotype. RNA-sequencing analyses highlighted up to 600 differentially expressed genes between in vitro and in vivo conditions, including genes involved in blood-testis barrier (BTB) formation and steroidogenesis. BTB integrity, the expression of two steroidogenic enzymes, and androgen receptors were indeed altered in vitro. Moreover, most of the top 10 predicted upstream regulators of deregulated genes were involved in inflammatory processes or immune cell recruitment. However, none of the three anti-inflammatory molecules tested in this study promoted meiotic progression. By analysing for the first time in vitro matured rat prepubertal testicular tissues at the molecular level, we uncovered the deregulation of several genes and revealed that defective BTB function, altered steroidogenic pathway, and probably inflammation, could be at the origin of meiotic arrest.
Collapse
|
10
|
Adamczewska D, Słowikowska-Hilczer J, Walczak-Jędrzejowska R. The Fate of Leydig Cells in Men with Spermatogenic Failure. Life (Basel) 2022; 12:570. [PMID: 35455061 PMCID: PMC9028943 DOI: 10.3390/life12040570] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/25/2022] [Accepted: 04/08/2022] [Indexed: 11/18/2022] Open
Abstract
The steroidogenic cells in the testicle, Leydig cells, located in the interstitial compartment, play a vital role in male reproductive tract development, maintenance of proper spermatogenesis, and overall male reproductive function. Therefore, their dysfunction can lead to all sorts of testicular pathologies. Spermatogenesis failure, manifested as azoospermia, is often associated with defective Leydig cell activity. Spermatogenic failure is the most severe form of male infertility, caused by disorders of the testicular parenchyma or testicular hormone imbalance. This review covers current progress in knowledge on Leydig cells origin, structure, and function, and focuses on recent advances in understanding how Leydig cells contribute to the impairment of spermatogenesis.
Collapse
Affiliation(s)
| | | | - Renata Walczak-Jędrzejowska
- Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, 92-213 Lodz, Poland; (D.A.); (J.S.-H.)
| |
Collapse
|
11
|
Inhibin-B and FSH Are Good Indicators of Spermatogenesis but Not the Best Indicators of Fertility. Life (Basel) 2022; 12:life12040511. [PMID: 35455002 PMCID: PMC9030543 DOI: 10.3390/life12040511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 01/19/2023] Open
Abstract
Biochemical markers of spermatogenesis and fertility assessment are important in the practical management of infertile males and the determination of an individual’s prognosis. We performed an analysis on 100 males with a male infertility factor. The following study inclusion parameters were analyzed: seminogram, FSH, LH, testosterone, estradiol, prolactin, TSH, and inhibin B concentrations. The patients were subsequently treated by reproductive endocrinologists in accordance with AUA/ASRM and EAU guidelines. The reproductive status was evaluated over a period of 3 years. We found a strong correlation of sperm count with inhibin B (r = 0.74, p < 0.001) and FSH concentration levels (r = −0.46, p < 0.001). Among 95 patients at follow-up, pregnancies occurred for 59 of their partners (48 spontaneous, 5 after IVF−ET, and 6 after IUI). Thirty-six patients remained childless despite the therapy. Sperm count and inhibin B level were the best predictors of natural fertilization (ROC AUC: 0.86 and 0.84; cut-off: 2.7 mln/mL and 45 pg/mL). Although inhibin B and FSH can be used to evaluate spermatogenesis and fertility, the initial sperm concentration appeared to be the best predictor of success. Pregnancy was achieved in a surprisingly large proportion of patients with a very low concentration of inhibin B and a low initial sperm count. It is noteworthy that 81% of the pregnancies were achieved without medically assisted reproduction.
Collapse
|
12
|
Walczak-Jędrzejowska R, Forma E, Oszukowska E, Bryś M, Marchlewska K, Kula K, Słowikowska-Hilczer J. Expression of G-Protein-Coupled Estrogen Receptor ( GPER) in Whole Testicular Tissue and Laser-Capture Microdissected Testicular Compartments of Men with Normal and Aberrant Spermatogenesis. BIOLOGY 2022; 11:biology11030373. [PMID: 35336747 PMCID: PMC8945034 DOI: 10.3390/biology11030373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 01/23/2023]
Abstract
Simple Summary Nowadays, there is no doubt that estrogens play an important role in male reproduction, affecting testicular cell differentiation, proliferation, apoptosis and metabolism. It is also widely believed that intratesticular balance of androgens and estrogens is crucial for the testicular development and function and that the increased testicular estrogen production may be associated with spermatogenic failure. There is also growing epidemiological evidence that the exposure of men to endocrine disruptors demonstrating estrogenic activity (xenoestrogens) may lead to impairment of male fertility via interference with estrogen signaling pathways. Besides the two classical nuclear estrogen receptors, the membrane-bound G protein-coupled estrogen receptor (GPER) was described in human testicular tissue. However, there are little data on its expression in testes with disturbed spermatogenesis. In this study, we investigated the GPER expression pattern in biopsies of azoospermic men with complete and aberrant spermatogenesis. Our results showed an increased expression of the GPER in testes with impaired spermatogenesis. Moreover, they indicate a possible involvement of estrogen signaling through GPER in disturbed function of Sertoli cells—the cells that support spermatogenic process. Abstract In this study, we retrospectively investigated GPER expression in biopsies of azoospermic men with complete (obstructive azoospermia—OA) and aberrant spermatogenesis (nonobstructive azoospermia—NOA). Each biopsy was histologically evaluated with morphometry. The testicular GPER expression was analyzed by the immunohistochemistry and RT-PCR technique in the whole testicular tissue and in seminiferous tubules and Leydig cells after laser-capture microdissection. In laser-microdissected compartments, we also analyzed transcriptional expression of selected Leydig (CYP17A1, HSD17B3, StAR) and Sertoli cell (AMH, SCF, BMP4) function markers. Immunohistochemical staining revealed expression of GPER in the cytoplasm of Leydig and Sertoli cells. Its stronger intensity was observed in Sertoli cells of NOA biopsies. The RT-PCR analysis of the GPER mRNA level unequivocally showed its increased expression in seminiferous tubules (i.e., Sertoli cells), not Leydig cells in NOA biopsies. This increased expression correlated positively with the transcriptional level of AMH—a marker of Sertoli cell immaturity, as well as FSH serum level in NOA but not in the OA group. Our results clearly demonstrate altered GPER expression in testes with primary spermatogenic impairment that might be related to Sertoli cell maturity/function.
Collapse
Affiliation(s)
- Renata Walczak-Jędrzejowska
- Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, Pomorska Str. 251, 92-213 Lodz, Poland; (K.M.); (K.K.); (J.S.-H.)
- Correspondence: ; Tel.: +48-42-272-53-91
| | - Ewa Forma
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska Str. 141/143, 90-236 Lodz, Poland; (E.F.); (M.B.)
| | - Elżbieta Oszukowska
- II Clinic of Urology, Medical University of Lodz, Pabianicka Str. 62, 93-513 Lodz, Poland;
| | - Magdalena Bryś
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska Str. 141/143, 90-236 Lodz, Poland; (E.F.); (M.B.)
| | - Katarzyna Marchlewska
- Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, Pomorska Str. 251, 92-213 Lodz, Poland; (K.M.); (K.K.); (J.S.-H.)
| | - Krzysztof Kula
- Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, Pomorska Str. 251, 92-213 Lodz, Poland; (K.M.); (K.K.); (J.S.-H.)
| | - Jolanta Słowikowska-Hilczer
- Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, Pomorska Str. 251, 92-213 Lodz, Poland; (K.M.); (K.K.); (J.S.-H.)
| |
Collapse
|
13
|
Jensen CFS, Wang D, Mamsen LS, Giwercman A, Jørgensen N, Fode M, Ohl D, Dong L, Hildorf SE, Pors SE, Fedder J, Ntemou E, Andersen CY, Sønksen J. Sertoli and Germ Cells Within Atrophic Seminiferous Tubules of Men With Non-Obstructive Azoospermia. Front Endocrinol (Lausanne) 2022; 13:825904. [PMID: 35721721 PMCID: PMC9201000 DOI: 10.3389/fendo.2022.825904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Infertile men with non-obstructive azoospermia (NOA) have impaired spermatogenesis. Dilated and un-dilated atrophic seminiferous tubules are often present in the testes of these patients, with the highest likelihood of active spermatogenesis in the dilated tubules. Little is known about the un-dilated tubules, which in NOA patients constitute the majority. To advance therapeutic strategies for men with NOA who fail surgical sperm retrieval we aimed to characterize the spermatogonial stem cell microenvironment in atrophic un-dilated tubules. METHODS Testis biopsies approximately 3x3x3 mm3 were obtained from un-dilated areas from 34 patients. They were classified as hypospermatogenesis (HS) (n=5), maturation arrest (MA) (n=14), and Sertoli cell only (SCO) (n= 15). Testis samples from five fertile men were included as controls. Biopsies were used for histological analysis, RT-PCR analysis and immunofluorescence of germ and Sertoli cell markers. RESULTS Anti-Müllerian hormone mRNA and protein expression was increased in un-dilated tubules in all three NOA subtypes, compared to the control, showing an immature state of Sertoli cells (p<0.05). The GDNF mRNA expression was significantly increased in MA (P=0.0003). The BMP4 mRNA expression showed a significant increase in HS, MA, and SCO (P=0.02, P=0.0005, P=0.02, respectively). The thickness of the tubule wall was increased 2.2-fold in the SCO-NOA compared to the control (p<0.05). In germ cells, we found the DEAD-box helicase 4 (DDX4) and melanoma-associated antigen A4 (MAGE-A4) mRNA and protein expression reduced in NOA (MAGE-A: 46% decrease in HS, 53% decrease in MA, absent in SCO). In HS-NOA, the number of androgen receptor positive Sertoli cells was reduced 30% with a similar pattern in mRNA expression. The γH2AX expression was increased in SCO as compared to HS and MA. However, none of these differences reached statistical significance probably due to low number of samples. CONCLUSIONS Sertoli cells were shown to be immature in un-dilated tubules of three NOA subtypes. The increased DNA damage in Sertoli cells and thicker tubule wall in SCO suggested a different mechanism for the absence of spermatogenesis from SCO to HS and MA. These results expand insight into the differences in un-dilated tubules from the different types of NOA patients.
Collapse
Affiliation(s)
- Christian Fuglesang Skjødt Jensen
- Department of Urology, Herlev and Gentofte Hospital, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Danyang Wang
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Linn Salto Mamsen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Aleksander Giwercman
- Department of Translational Medicine and Reproductive Medicine Centre, Lunds University and Skane University Hospital, Malmö, Sweden
| | - Niels Jørgensen
- Department of Growth and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Mikkel Fode
- Department of Urology, Herlev and Gentofte Hospital, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Dana Ohl
- Department of Urology, University of Michigan, Ann Arbor, MI, United States
| | - Lihua Dong
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Simone Engmann Hildorf
- Centre of Andrology & Fertility Clinic, Department D, Odense University Hospital, Odense C, Denmark
| | - Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Jens Fedder
- Centre of Andrology & Fertility Clinic, Department D, Odense University Hospital, Odense C, Denmark
| | - Elissavet Ntemou
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
- *Correspondence: Elissavet Ntemou,
| | - Claus Yding Andersen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Jens Sønksen
- Department of Urology, Herlev and Gentofte Hospital, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Nguyen Hoai B, Hoang L, Tran D, Nguyen Cao T, Doan Tien L, Sansone A, Jannini EA. Ultrasonic testicular size of 24,440 adult Vietnamese men and the correlation with age and hormonal profiles. Andrologia 2021; 54:e14333. [PMID: 34961956 DOI: 10.1111/and.14333] [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: 10/26/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/28/2022] Open
Abstract
Testicular size is an important parameter to investigate male reproductive and sexual functions. However, diagnosis and treatments are hindered by the lack of country-specific standard values for testicular volume. We conducted the present retrospective chart review study on 24,440 men who sought consultation at Andrology and Sexual Medicine-Hanoi Medical University Hospital to provide a reference range of testicular volume and to determine the correlations between testicular volume, age and hormonal profiles. These men were classified into groups being healthy fathers, hypogonadal men, unexplained infertile men, men with unknown fertility, testicular pathologies and other andrological condition groups. Hypogonadal men and unexplained infertile men had significantly smaller testicular sizes compared with healthy fathers. The mean value of testicular volume of healthy subjects was 13.64 ± 3.44 ml (left testis: 13.94 ± 3.72 ml; right testis: 13.34 ± 3.61 ml; p < 0.001). Testicular size of Vietnamese men was negatively correlated with LH and FSH (Rho = -0.16 and -0.33, p < 0.001) and positively correlated with testosterone after adjusting for confounding factors. Testicular volume was independent of the subject's age and smoking habits.
Collapse
Affiliation(s)
- Bac Nguyen Hoai
- Department of Andrology and Sexual Medicine, Hanoi Medical University's Hospital, Hanoi, Vietnam
| | - Long Hoang
- Department of Urology, Hanoi Medical University's Hospital, Hanoi, Vietnam
| | - Duc Tran
- Department of Urology, 108 Military Central Hospital, Hanoi, Vietnam
| | - Thang Nguyen Cao
- Department of Andrology and Sexual Medicine, Hanoi Medical University's Hospital, Hanoi, Vietnam
| | - Luu Doan Tien
- Radiology Center, Hanoi Medical University's Hospital, Hanoi, Vietnam
| | - Andrea Sansone
- Chair of Endocrinology and Sexual Medicine (ENDOSEX), Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Emmanuele A Jannini
- Chair of Endocrinology and Sexual Medicine (ENDOSEX), Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Honorary Chair of Sexual Medicine, Hanoi Medical University, Hanoi, Vietnam
| |
Collapse
|
15
|
Sansone A, Kliesch S, Dugas M, Sandhowe-Klaverkamp R, Isidori AM, Schlatt S, Zitzmann M. Serum concentrations of dihydrotestosterone are associated with symptoms of hypogonadism in biochemically eugonadal men. J Endocrinol Invest 2021; 44:2465-2474. [PMID: 33811609 PMCID: PMC8502125 DOI: 10.1007/s40618-021-01561-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/23/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE Symptoms of hypogonadism are often reported by subjects with normal serum testosterone (T) levels. We aimed to assess the association between clinical symptoms in andrological outpatients and sex steroids levels. METHODS This is a retrospective cross-sectional cohort study in an Academic clinic and research unit. International Index of Erectile Function (IIEF, EF domain) and Aging Males Symptoms scale (AMS) questionnaires were completed by 635 and 574 men, respectively (mean age: 47.3 ± 13.9 and 47.4 ± 13.8 years, p = 0.829), free of interfering medications with complaints possibly related to hypogonadism. RESULTS Serum total/free T as well as dihydro-T (DHT) was associated with IIEF-EF and AMS scores in the overall population using univariate analyses. Multivariate approaches revealed DHT concentrations in subjects with normal T levels (n = 416, Total T > 12 nmol/L) to be significant predictors of AMS scores. A 0.1 nmol/l serum DHT increase within the eugonadal range was associated with a 4.67% decrease in odds of having worse symptoms (p = 0.011). In men with biochemical hypogonadism (Total T < 12 nmol/L), total and free T rather than DHT were associated with AMS results. This association was not found for IIEF-EF scores. Indirect effects of age and BMI were seen for relations with hormone concentrations but not questionnaire scores. CONCLUSION DHT can be associated with symptoms of hypogonadism in biochemically eugonadal men. Serum DHT measurement might be helpful once the diagnosis of hypogonadism has been ruled out but should not be routinely included in the primary diagnostic process.
Collapse
Affiliation(s)
- A Sansone
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Münster, Germany.
- Chair of Endocrinology and Medical Sexology (ENDOSEX), Department of Systems Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy.
| | - S Kliesch
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Münster, Germany
| | - M Dugas
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - R Sandhowe-Klaverkamp
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Münster, Germany
| | - A M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - S Schlatt
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Münster, Germany
| | - M Zitzmann
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Münster, Germany
| |
Collapse
|
16
|
Sinopidis X, Kostopoulou E, Rojas-Gil AP, Panagidis A, Kourea E, Skiadopoulos S, Georgiou G, Spiliotis BE. Association of antimullerian hormone with the size of the appendix testis, the androgen and estrogen receptors and their expression in the appendix testis, in congenital cryptorchidism. J Pediatr Endocrinol Metab 2021; 34:1247-1255. [PMID: 34265880 DOI: 10.1515/jpem-2021-0240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/20/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Antimullerian hormone (AMH) causes regression of the mullerian ducts in the male fetus. The appendix testis (AT) is a vestigial remnant of mullerian duct origin, containing both androgen (AR) and estrogen (ER) receptors. The role of both AMH and AT in testicular descent is yet to be studied. We investigated the possible association of AMH with AT size, the AR and ER, and their expression in the AT, in congenital cryptorchidism. METHODS A total of 26 patients with congenital unilateral cryptorchidism and 26 controls with orthotopic testes were investigated, and 21 ATs were identified in each group. AMH and insulin-like three hormone (INSL3) concentrations were measured with spectrophotometry. AR and ER receptor expression was assessed with immunohistochemistry using monoclonal antibodies R441 for AR and MAB463 for ER. For the estimation of receptor expression, the Allred Score method was used. RESULTS AMH concentrations did not present significant differences between patients with congenital cryptorchidism and the controls. Also, no correlation was found between AMH, INSL3, and AT length. Allred scores did not present significant differences. However, expression percentiles and intensity for both receptors presented significant differences. Three children with cryptorchidism and the highest AMH levels also had the highest estrogen receptor scores in the AT. CONCLUSIONS No association was found between AMH and the studied major parameters. However, higher AMH concentrations, in combination with higher estrogen receptor scores in the AT, may play a role in cryptorchidism in some children. Larger population samples are needed to verify this observation.
Collapse
Affiliation(s)
- Xenophon Sinopidis
- Department of Pediatric Surgery, University of Patras School of Medicine, Patras, Greece
| | - Eirini Kostopoulou
- Research Laboratory of the Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Patras School of Medicine, Patras, Greece
| | - Andrea Paola Rojas-Gil
- Faculty of Health Sciences, Department of Nursing, University of Peloponnese, Tripoli, Greece
| | | | - Eleni Kourea
- Department of Pathology, University of Patras School of Medicine, Patras, Greece
| | - Spyros Skiadopoulos
- Department of Medical Physics, University of Patras School of Medicine, Patras, Greece
| | - George Georgiou
- Department of Pediatric Surgery, Children's Hospital, Patras, Greece
| | - Bessie E Spiliotis
- Research Laboratory of the Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Patras School of Medicine, Patras, Greece
| |
Collapse
|
17
|
Krysiak R, Kowalcze K, Okopień B. Hypothalamic-pituitary-gonadal axis and sexual functioning in metformin-treated men after discontinuation of testosterone replacement therapy: A pilot study. J Clin Pharm Ther 2021; 46:1764-1775. [PMID: 34490645 DOI: 10.1111/jcpt.13528] [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: 05/29/2021] [Revised: 08/02/2021] [Accepted: 08/26/2021] [Indexed: 11/28/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Metformin was found to reduce elevated gonadotropin levels. The aim of the present study was to determine whether metformin modulates the impact of discontinuation of testosterone therapy on hypothalamic-pituitary-gonadal axis activity and sexual function in men with low testosterone levels. METHODS The study included 28 men with late-onset hypogonadism (defined according to the criteria of the European Male Aging Study group) receiving testosterone undecanoate (120 mg in three equal doses), 12 of whom had been treated with oral metformin (1.7-3 g daily). Both testosterone and metformin had been administered for at least six months before enrolment. In all patients, testosterone replacement required to be discontinued. The control group included 16 testosterone- and metformin-treated men with late-onset hypogonadism who during the entire study period continued their treatment. Glucose homeostasis markers, as well as plasma levels of insulin, gonadotropins, testosterone, calculated bioavailable testosterone, dehydroepiandrosterone-sulphate, oestradiol, thyrotropin, free thyroxine, prolactin, insulin-growth factor-1 and cortisol were measured at the beginning of the study and four months later. Moreover, at the beginning and the end of the study, all enrolled patients completed a questionnaire assessing their sexual functioning (IIEF-15). RESULTS AND DISCUSSION Discontinuation of testosterone therapy resulted in a decrease in total testosterone and bioavailable testosterone (by 42% and 45% in metformin-treated patients, and by 52% and 54% in metformin-naïve patients), as well as impaired all aspects of male sexual function. Changes in bioavailable testosterone, as well as in erectile function, orgasmic function and sexual desire were less pronounced if subjects received metformin. Only in metformin-naïve men, follow-up FSH and LH levels were higher than at baseline (by 75% and 62%). Moreover, discontinuation of testosterone therapy in metformin-naïve men increased glycated haemoglobin, as well as worsened insulin sensitivity. There were no differences between baseline and follow-up levels of the remaining hormones. In metformin-naïve subjects, the increase in gonadotropin levels correlated with the changes in testosterone levels and insulin sensitivity. No effect on glucose homeostasis markers, hormone levels and sexual functioning was observed in the control group. WHAT IS NEW AND CONCLUSION The obtained results suggest that metformin treatment mitigates the unfavourable effect of discontinuation of testosterone treatment on hypothalamic-pituitary-testicular axis activity and sexual function in men with late-onset hypogonadism.
Collapse
Affiliation(s)
- Robert Krysiak
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Katowice, Poland
| | - Karolina Kowalcze
- Department of Pediatrics in Bytom, School of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Bogusław Okopień
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
18
|
Aging, inflammation and DNA damage in the somatic testicular niche with idiopathic germ cell aplasia. Nat Commun 2021; 12:5205. [PMID: 34471128 PMCID: PMC8410861 DOI: 10.1038/s41467-021-25544-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 08/18/2021] [Indexed: 12/18/2022] Open
Abstract
Molecular mechanisms associated with human germ cell aplasia in infertile men remain undefined. Here we perform single-cell transcriptome profiling to highlight differentially expressed genes and pathways in each somatic cell type in testes of men with idiopathic germ cell aplasia. We identify immaturity of Leydig cells, chronic tissue inflammation, fibrosis, and senescence phenotype of the somatic cells, as well markers of chronic inflammation in the blood. We find that deregulated expression of parentally imprinted genes in myoid and immature Leydig cells, with relevant changes in the ratio of Lamin A/C transcripts and an active DNA damage response in Leydig and peritubular myoid cells are also indicative of senescence of the testicular niche. This study offers molecular insights into the pathogenesis of idiopathic germ cell aplasia. Molecular mechanisms associated with human germ cell aplasia in infertile men remain undefined. Here the authors perform single-cell transcriptome profiling to highlight differentially expressed genes and pathways in each somatic cell type in testes of men with idiopathic germ cell aplasia.
Collapse
|
19
|
Jeminiwa BO, Knight RC, Abbot KL, Pondugula SR, Akingbemi BT. Gonadal sex steroid hormone secretion after exposure of male rats to estrogenic chemicals and their combinations. Mol Cell Endocrinol 2021; 533:111332. [PMID: 34038751 PMCID: PMC9310441 DOI: 10.1016/j.mce.2021.111332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 11/21/2022]
Abstract
Environmental chemicals can interfere with the endocrine axis hence they are classified as endocrine disrupting chemicals (EDCs). Bisphenol S (BPS) is used in the manufacture of consumer products because of its superior thermal stability and is thought to be a safe replacement chemical for its analog bisphenol A (BPA). However, the safety profile of these compounds alone or in the presence of other EDCs is yet to be fully investigated. Also, the estrogenic chemical 17α-ethinyl estradiol (EE2) and a constituent of female oral contraceptives for women, is present in water supplies. To simulate concurrent exposure of the population to chemical mixtures, we investigated the effects of BPA, BPS, EE2, and their combinations on sex steroid secretion in the growing male rat gonad. Prepubertal and pubertal male rats at 21 and 35 days of age were provided test chemicals in drinking water (parts per billion) for 14 days. At termination of exposure, some individual chemical effects were modified by exposure to chemical combinations. Single chemical exposures markedly decreased androgen secretion but their combination (e.g., BPA + BPS + EE2) caused the opposite effect, i.e., increased Leydig cell T secretion. Also, the test chemicals acting alone or in combination increased testicular and Leydig cell 17β-estradiol (E2) secretion. Chemical-induced changes in T and E2 secretion were associated with altered testicular expression of the cholesterol side-chain cleavage (Cyp11a1) and 17β-hydroxysteroid dehydrogenase (Hsd17β) enzyme protein. Additional studies are warranted to understand the mechanisms by which single and chemical combinations impact function of testicular cells and disrupt their paracrine regulation.
Collapse
Affiliation(s)
- B O Jeminiwa
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - R C Knight
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - K L Abbot
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - S R Pondugula
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - B T Akingbemi
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
20
|
Schneider F, Dabel J, Sandhowe-Klaverkamp R, Neuhaus N, Schlatt S, Kliesch S, Wistuba J. Serum and intratesticular inhibin B, AMH, and spermatogonial numbers in trans women at gender-confirming surgery: An observational study. Andrology 2021; 9:1781-1789. [PMID: 34085780 DOI: 10.1111/andr.13059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/30/2021] [Accepted: 05/23/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Anti-Müllerian hormone and inhibin B are produced by Sertoli cells. Anti-Müllerian hormone secretion indicates an immature Sertoli cell state. Inhibin B serves as a marker of male fertility. Identification of markers reflecting the presence of germ cells is of particular relevance in trans persons undergoing gender-affirming hormone therapy in order to offer individualized fertility preservation methods. OBJECTIVES Serum and intratesticular inhibin B and anti-Müllerian hormone values were assessed and related to clinical features, laboratory values, and germ cell numbers. MATERIALS AND METHODS Twenty-two trans women from three clinics were included. As gender-affirming hormone therapy, 10-12.5 mg of cyproterone acetate plus estrogens were administered. Height, weight, age, medication, and treatment duration were inquired by questionnaires. Serum luteinizing hormone, follicle-stimulating hormone, testosterone, and estradiol were measured by immuno-assays. Serum and intratesticular inhibin B and anti-Müllerian hormone were measured by commercially available ELISAs. Spermatogonia were quantified as spermatogonia per cubic millimeter testicular tissue applying a morphometric analysis of two independent testicular cross-sections per individual after MAGEA4 immunostaining. RESULTS Patients with high inhibin B levels presented with a higher number of spermatogonia (*p < 0.05). Furthermore, mean serum inhibin B was associated with low age (*p < 0.05), low follicle-stimulating hormone (*p < 0.05), and low testosterone (*p < 0.05). Serum anti-Müllerian hormone, however, was not related to spermatogonial numbers. It correlated with high testosterone (*p < 0.05) and high follicle-stimulating hormone (*p < 0.05) only. High intratesticular inhibin B was accompanied by high luteinizing hormone (*p < 0.05), high follicle-stimulating hormone (**p < 0.01), and high testosterone levels (**p < 0.01). Higher the intratesticular anti-Müllerian hormone levels, the longer gender-affirming hormone therapy was administered (*p < 0.05). DISCUSSION AND CONCLUSION Serum inhibin B levels indicate the presence of spermatogonia, whereas anti-Müllerian hormone seems not to be a reliable marker concerning germ cell abundance.
Collapse
Affiliation(s)
- Florian Schneider
- Institute of Reproductive and Regenerative Medicine, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany.,Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Jennifer Dabel
- Institute of Reproductive and Regenerative Medicine, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Reinhild Sandhowe-Klaverkamp
- Institute of Reproductive and Regenerative Medicine, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Nina Neuhaus
- Institute of Reproductive and Regenerative Medicine, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Stefan Schlatt
- Institute of Reproductive and Regenerative Medicine, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Sabine Kliesch
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Joachim Wistuba
- Institute of Reproductive and Regenerative Medicine, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
21
|
Cargnelutti F, Di Nisio A, Pallotti F, Sabovic I, Spaziani M, Tarsitano MG, Paoli D, Foresta C. Effects of endocrine disruptors on fetal testis development, male puberty, and transition age. Endocrine 2021; 72:358-374. [PMID: 32757113 PMCID: PMC8128728 DOI: 10.1007/s12020-020-02436-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/23/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE Endocrine disruptors (EDs) are exogenous substances able to impair endocrine system; consequently, they may cause numerous adverse effects. Over the last years, particular focus has been given to their harmful effects on reproductive system, but very little is known, especially in males. The aim of this review is to discuss the detrimental effects of EDs exposure on fetal testis development, male puberty, and transition age. METHODS A search for the existing literature focusing on the impact of EDs on fetal testis development, male puberty, andrological parameters (anogenital distance, penile length, and testicular volume), and testicular cancer with particular regard to pubertal age provided the most current information available for this review. Human evidence-based reports were given priority over animal and in vitro experimental results. Given the paucity of available articles on this subject, all resources were given careful consideration. RESULTS Information about the consequences associated with EDs exposure in the current literature is limited and often conflicting, due to the scarcity of human studies and their heterogeneity. CONCLUSIONS We conclude that current evidence does not clarify the impact of EDs on human male reproductive health, although severe harmful effects had been reported in animals. Despite controversial results, overall conclusion points toward a positive association between exposure to EDs and reproductive system damage. Further long-term studies performed on wide number of subjects are necessary in order to identify damaging compounds and remove them from the environment.
Collapse
Affiliation(s)
- Francesco Cargnelutti
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Andrea Di Nisio
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy
| | - Francesco Pallotti
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Iva Sabovic
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy
| | - Matteo Spaziani
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Maria Grazia Tarsitano
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Donatella Paoli
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" University of Rome, Viale del Policlinico 155, 00161, Rome, Italy.
| | - Carlo Foresta
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy
| |
Collapse
|
22
|
Sansone A, Schubert M, Tüttelmann F, Krallmann C, Zitzmann M, Kliesch S, Gromoll J. Pituitary response to GnRH stimulation tests in different FSHB-211 G/T genotypes. Hum Reprod 2021; 36:1376-1382. [PMID: 33704441 DOI: 10.1093/humrep/deab033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 01/14/2021] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Does pituitary response to a GnRH stimulation test differ according to the different FSHB-211 G/T genotypes? SUMMARY ANSWER The promoter polymorphism FSHB-211 G > T affects the pituitary response to exogenous GnRH stimulation by reducing FSH and increasing LH outputs. WHAT IS KNOWN ALREADY The FSHB-211 G > T single nucleotide polymorphism (SNP) is known to affect pituitary FSH output by impairing the transcriptional activity of FSHB. STUDY DESIGN, SIZE, DURATION This was a cross-sectional, retrospective study on 67 male subjects (mean age: 24.6 ± 10.3 years) undergoing a GnRH stimulation test for diagnostic purposes in cases of secondary hypogonadism. PARTICIPANTS/MATERIALS, SETTING, METHODS A GnRH stimulation test was performed by administering an i.v. bolus of 100 µg of the GnRH-analogue gonadorelin acetate to all patients, with blood samples drawn from the cubital vein immediately prior to injection (T0) and 30 (T1) and 45 minutes (T2) after. Clinical and genetic data were retrieved from a computerized database. Linear longitudinal mixed-effect models were used to assess the effects of SNP genotype on FSH and LH levels over time via additive and recessive models. MAIN RESULTS AND THE ROLE OF CHANCE An overall marked increase in serum FSH and LH following administration i.v. of 100 µg of an LHRH-analogue was found (P < 0.0001 for linear trend, both models). Peak levels of LH were significantly higher in TT carriers than in GT and GG carriers (P = 0.012); no significant between-groups difference was found concerning stimulated FSH levels. In both the additive and recessive model, the main effect of T allele(s) did not reach statistical significance concerning FSH levels (P = 0.9502 and P = 0.8576, respectively), yet interaction effects over time demonstrated an attenuated response in T-allele carriers compared to the GG-allele carriers (P = 0.0219 and P = 0.0276). Main and interaction effects for LH were significant in both the additive (P = 0.0022 and P = 0.0013, respectively) and recessive model (P = 0.0025 and P = 0.0016, respectively). LIMITATIONS, REASONS FOR CAUTION Given the retrospective nature of the study and the small number of TT carriers, results should be interpreted with caution. WIDER IMPLICATIONS OF THE FINDINGS The FSHB c.-211G>T polymorphism might result in an impaired response to endogenous, as well as exogenous, GnRH stimulation. This finding might contribute to the clinical phenotype of reduced testicular volume and sperm count for patients carrying one or two T alleles. STUDY FUNDING/COMPETING INTEREST(S) Parts of the study were supported by the German Research Foundation (CRU326 Male Germ Cells). On behalf of all authors, the corresponding author states that there is no conflict of interest. TRIAL REGISTRATION NUMBER NA.
Collapse
Affiliation(s)
- Andrea Sansone
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany.,Chair in Endocrinology and Medical Sexology (ENDOSEX), Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Maria Schubert
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Claudia Krallmann
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Michael Zitzmann
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Jörg Gromoll
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| |
Collapse
|
23
|
Lei T, Blois SM, Freitag N, Bergmann M, Bhushan S, Wahle E, Huang ACC, Chen HL, Hartmann MF, Wudy SA, Liu FT, Meinhardt A, Fijak M. Targeted disruption of galectin 3 in mice delays the first wave of spermatogenesis and increases germ cell apoptosis. Cell Mol Life Sci 2021; 78:3621-3635. [PMID: 33507326 PMCID: PMC11072302 DOI: 10.1007/s00018-021-03757-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/22/2020] [Accepted: 01/06/2021] [Indexed: 12/22/2022]
Abstract
Galectin 3 is a multifunctional lectin implicated in cellular proliferation, differentiation, adhesion, and apoptosis. This lectin is broadly expressed in testicular somatic cells and germ cells, and is upregulated during testicular development. Since the role of galectin 3 in testicular function remains elusive, we aimed to characterize the role of galectin 3 in testicular physiology. We found that galectin 3 transgenic mice (Lgals3-/-) exhibited significantly decreased testicular weight in adulthood compared to controls. The transgenic mice also exhibited a delay to the first wave of spermatogenesis, a decrease in the number of germ cells at postnatal day 5 (P5) and P15, and defective Sertoli cell maturation. Mechanistically, we found that Insulin-like-3 (a Leydig cell marker) and enzymes involved in steroid biosynthesis were significantly upregulated in adult Lgals3-/- testes. These observations were accompanied by increased serum testosterone levels. To determine the underlying causes of the testicular atrophy, we monitored cellular apoptosis. Indeed, adult Lgals3-/- testicular cells exhibited an elevated apoptosis rate that is likely driven by downregulated Bcl-2 and upregulated Bax and Bak expression, molecules responsible for live/death cell balance. Moreover, the percentage of testicular macrophages within CD45+ cells was decreased in Lgals3-/- mice. These data suggest that galectin 3 regulates spermatogenesis initiation and Sertoli cell maturation in part, by preventing germ cells from undergoing apoptosis and regulating testosterone biosynthesis. Going forward, understanding the role of galectin 3 in testicular physiology will add important insights into the factors governing the development of germ cells and steroidogenesis and delineate novel biomarkers of testicular function.
Collapse
Affiliation(s)
- Tao Lei
- Department of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, Aulweg 123, 35385, Giessen, Germany
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sandra M Blois
- Department of Obstetrics and Fetal Medicine, AG Glycoimmunology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251, Hamburg, Germany
- Experimental and Clinical Research Center, A Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, The Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Nancy Freitag
- Department of Obstetrics and Fetal Medicine, AG Glycoimmunology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251, Hamburg, Germany
- Experimental and Clinical Research Center, A Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, The Charité Universitätsmedizin Berlin, Berlin, Germany
- Division of General Internal and Psychosomatic Medicine, Berlin Institute of Health, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin, Germany
| | - Martin Bergmann
- Institute of Veterinary Anatomy, Histology, and Embryology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Sudhanshu Bhushan
- Department of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, Aulweg 123, 35385, Giessen, Germany
| | - Eva Wahle
- Department of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, Aulweg 123, 35385, Giessen, Germany
| | | | - Hung-Lin Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Michaela F Hartmann
- Steroid Research and Mass Spectrometry Unit, Pediatric Endocrinology and Diabetology, Center of Child and Adolescent Medicine, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Stefan A Wudy
- Steroid Research and Mass Spectrometry Unit, Pediatric Endocrinology and Diabetology, Center of Child and Adolescent Medicine, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, Aulweg 123, 35385, Giessen, Germany
| | - Monika Fijak
- Department of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, Aulweg 123, 35385, Giessen, Germany.
| |
Collapse
|
24
|
Yang H, Wei H, Shen L, Kumar C S, Chen Q, Chen Y, Kumar SA. A novel stop-loss DAX1 variant affecting its protein-interaction with SF1 precedes the adrenal hypoplasia congenital with rare spontaneous precocious puberty and elevated hypothalamic-pituitary-gonadal/adrenal axis responses. Eur J Med Genet 2021; 64:104192. [PMID: 33766795 DOI: 10.1016/j.ejmg.2021.104192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/20/2021] [Accepted: 03/14/2021] [Indexed: 11/29/2022]
Abstract
The case study unveils the likely mechanism of a novel stop-loss DAX1 variant preceding the prolonged precocious puberty in the adrenal hypoplasia congenital (AHC) boy. A boy aged five years and nine months initially examined for the primary adrenal insufficiency symptoms. Next-generation sequencing confirmed the X-linked inheritance of a novel stop-loss DAX1 variant: c.1411T>C/p.Ter471Gln associated with AHC in the patient. The patient was subjected to a brief clinical follow-up from 11 to 15.1 years of age. The effect of the mutant-DAX1 variant (p.Ter471Gln) on DAX1-steroidogenic factor 1 (SF1) (protein-protein) interaction was studied by protein-protein docking using the ClusPro-online tool. At 5.9 yrs of age, the patient exhibited precocious puberty with the secondary sexual characteristics of Tanner 2 stage (of 9-14 yrs of age). The patient showed primary adrenal insufficiency with diminished cortisol concentrations at blood serum (25 ng/ml) and urine (3.55 μg/24 h) levels. Upon steroidal exposure, the patient showed normalized serum cortisol levels of 45-61 ng/ml. However, the precocious puberty got prolonged with the increased penis length of 8.5 cm and the bone age of 18 yrs old during the follow-up. The patient showed increased basal serum adrenocorticotropic hormone (110->2000 pg/ml) and follicle-stimulating hormone (18.4-22.3 mIU/ml) concentrations. Following an elevated hypothalamic-pituitary-gonadal axis activity witnessed upon gonarellin stimulation. Protein-protein docking confirmed a weaker interaction between the mutant-DAX1 (p.Ter471Gln) protein and the wild-SF1 protein. Overall, we hypothesize the weakened mutant-DAX1-SF1 (protein-protein) interaction could govern the prolonged precocious puberty augmented with the elevated hypothalamic-pituitary-gonadal/adrenal axis responses via SF1-induced neuronal nitric oxide synthetase activation in the patient.
Collapse
Affiliation(s)
- Haihua Yang
- Department of Endocrinology, Metabolism and Genetics, Henan Children's Hospital (aka. Children's Hospital Affiliated to Zhengzhou University), No-33, Longhu Waihuan East Road, Zhengzhou, 450018, China
| | - Haiyan Wei
- Department of Endocrinology, Metabolism and Genetics, Henan Children's Hospital (aka. Children's Hospital Affiliated to Zhengzhou University), No-33, Longhu Waihuan East Road, Zhengzhou, 450018, China.
| | - Linghua Shen
- Department of Endocrinology, Metabolism and Genetics, Henan Children's Hospital (aka. Children's Hospital Affiliated to Zhengzhou University), No-33, Longhu Waihuan East Road, Zhengzhou, 450018, China
| | - Selvaa Kumar C
- School of Biotechnology and Bioinformatics, D. Y. Patil Deemed to Be University, Sector-15, CBD Belapur. Navi Mumbai, 400614, India
| | - Qiong Chen
- Department of Endocrinology, Metabolism and Genetics, Henan Children's Hospital (aka. Children's Hospital Affiliated to Zhengzhou University), No-33, Longhu Waihuan East Road, Zhengzhou, 450018, China
| | - Yongxing Chen
- Department of Endocrinology, Metabolism and Genetics, Henan Children's Hospital (aka. Children's Hospital Affiliated to Zhengzhou University), No-33, Longhu Waihuan East Road, Zhengzhou, 450018, China
| | - Senthil Arun Kumar
- Department of Endocrinology, Metabolism and Genetics, Henan Children's Hospital (aka. Children's Hospital Affiliated to Zhengzhou University), No-33, Longhu Waihuan East Road, Zhengzhou, 450018, China.
| |
Collapse
|
25
|
Gualtieri P, Tarsitano MG, DE Santis GL, Romano L, Esposito E, DE Lorenzo A. Obesity in childhood: how to improve male adolescence incoming. Minerva Endocrinol (Torino) 2021; 47:358-370. [PMID: 33759441 DOI: 10.23736/s2724-6507.21.03224-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Earlier or delayed puberty can be determined by numerous causes, but the exact mechanisms are not fully known. Anyway, those may be independent from the hypothalamicpituitary-gonadal axis involvement. Certainly, obesity is one of the main factors. In fact, obesity and infertility are strongly linked. For this reason, we want to analyse the relationship between puberty and obesity and give an updated state-of-the-art starting by discussing a 14-year-old obese boy's clinical case. EVIDENCE ACQUISITION Literature data are conflicting. Main criticisms are related to study design and evaluation criteria. Indeed, examined populations are not homogeneous by age, Tanner stage and BMI classification. The obesity epidemic is involved in earlier puberty, and puberty seems to be anticipated in all BMI groups. Very few studies evaluate the level of adiposity in the diagnosis of obesity. However, the role of the adipose tissue is crucial for hormone synthesis and regulation. Therefore, fat mass age-related and not simply BMI has to be considered by clinicians for appropriate diagnosis. EVIDENCE SYNTHESIS Regarding the clinical case, in three months our patient recovered delayed pubertal development following an anti-inflammatory and antioxidant Mediterranean Diet. Loss of weight, as in decrease of fat mass but saving of lean mass, increased testicular volume and testosterone levels occurred. CONCLUSIONS Puberty depends on several factors, including obesity. Further studies are needed to evaluate age groups, Tanner stage, diet and lifestyle, ethnicity and above all the fat/lean mass ratio. Lack of adequate tools could hinder a clinician's ability to recognize when or if therapeutic intervention is needed.
Collapse
Affiliation(s)
- Paola Gualtieri
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy -
| | | | - Gemma L DE Santis
- School of Specialization in Food Science, University of Rome Tor Vergata, Rome, Italy
| | - Lorenzo Romano
- School of Specialization in Food Science, University of Rome Tor Vergata, Rome, Italy
| | - Ernesto Esposito
- Direzione Generale Dipartimento Politiche della Persona, Regione Basilicata, Potenza, Italy
| | - Antonino DE Lorenzo
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
26
|
Kohva E, Varimo T, Huopio H, Tenhola S, Voutilainen R, Toppari J, Miettinen PJ, Vaaralahti K, Viinamäki J, Backman JT, Hero M, Raivio T. Anti-Müllerian hormone and letrozole levels in boys with constitutional delay of growth and puberty treated with letrozole or testosterone. Hum Reprod 2021; 35:257-264. [PMID: 31958337 PMCID: PMC7048712 DOI: 10.1093/humrep/dez231] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/15/2019] [Indexed: 12/14/2022] Open
Abstract
STUDY QUESTION Does treatment of constitutional delay of growth and puberty (CDGP) in boys with aromatase inhibitor letrozole (Lz) or conventional low-dose testosterone (T) have differing effects on developing seminiferous epithelium? SUMMARY ANSWER Anti-Müllerian hormone (AMH) declined similarly in both treatment groups, and the two Sertoli cell-derived markers (AMH and inhibin B (iB)) exhibited differing responses to changes in gonadotrophin milieu. WHAT IS KNOWN ALREADY Boys with CDGP may benefit from puberty-inducing medication. Peroral Lz activates gonadotrophin secretion, whereas intramuscular low-dose T may transiently suppress gonadotrophins and iB. STUDY DESIGN, SIZE, DURATION Sera of 28 boys with CDGP who participated in a randomised, controlled, open-label trial at four paediatric centres in Finland between August 2013 and January 2017 were analysed. The patients were randomly assigned to receive either Lz (2.5 mg/day) (n = 15) or T (1 mg/kg/month) (n = 13) for 6 months. PARTICIPANTS/MATERIALS, SETTING, METHODS The 28 patients were at least 14 years of age, showed first signs of puberty, wanted medical attention for CDGP and were evaluated at 0, 3, 6 and 12 months of visits. AMH levels were measured with an electrochemiluminescence immunoassay and Lz levels with liquid chromatography coupled with tandem mass spectrometry. MAIN RESULTS AND THE ROLE OF CHANCE AMH levels decreased in both treatment groups during the 12-month follow-up (P < 0.0001). Between 0 and 3 months, the changes in gonadotrophin levels (increase in the Lz group, decrease in the T group) correlated strongly with the changes in levels of iB (FSH vs iB, r = 0.55, P = 0.002; LH vs iB, r = 0.72, P < 0.0001), but not with the changes in AMH (P = NS). At 12 months, AMH levels did not differ between the groups (P = NS). Serum Lz levels (range, 124-1262 nmol/L) were largely explained by the Lz dose per weight (at 3 months r = 0.62, P = 0.01; at 6 months r = 0.52, P = 0.05). Lz levels did not associate with changes in indices of hypothalamic-pituitary-gonadal axis activity or Sertoli cell markers (in all, P = NS). LIMITATIONS, REASONS FOR CAUTION The original trial was not blinded for practical reasons and included a limited number of participants. WIDER IMPLICATIONS OF THE FINDINGS In early puberty, treatment-induced gonadotrophin stimulus was unable to counteract the androgen-mediated decrease in AMH, while changes in iB levels were associated with changes in gonadotrophin levels. AMH decreased similarly in both groups during the treatment, reassuring safety of developing seminiferous epithelium in both treatment approaches. Since a fixed dose of Lz induced variable serum Lz levels with a desired puberty-promoting effect in all boys, more research is needed to aim at a minimal efficient dose per weight. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the Academy of Finland, the Foundation for Pediatric Research, the Emil Aaltonen Foundation, Sigrid Juselius Foundation and Helsinki University Hospital Research Funds. The authors have nothing to disclose. TRIAL REGISTRATION NUMBER NCT01797718.
Collapse
Affiliation(s)
- E Kohva
- Children's Hospital, Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland
| | - T Varimo
- Children's Hospital, Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland
| | - H Huopio
- Department of Pediatrics, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - S Tenhola
- Department of Pediatrics, Kymenlaakso Central Hospital, Kotka, Finland
| | - R Voutilainen
- Department of Pediatrics, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - J Toppari
- Department of Pediatrics, Turku University Hospital and Institute of Biomedicine, Research Centre for Integrated Physiology and Pharmacology, University of Turku, Turku, Finland
| | - P J Miettinen
- Children's Hospital, Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland
| | - K Vaaralahti
- Children's Hospital, Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland
| | - J Viinamäki
- Department of Clinical Pharmacology, and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J T Backman
- Department of Clinical Pharmacology, and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - M Hero
- Children's Hospital, Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland
| | - T Raivio
- Children's Hospital, Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland.,Translational Stem Cell Biology and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
27
|
Zhao JJ, Kang CM, Zhang P, Zheng L. Performance characteristics of the Mindray chemiluminescence anti-Müllerian hormone assay. J Clin Lab Anal 2021; 35:e23734. [PMID: 33660884 PMCID: PMC8059718 DOI: 10.1002/jcla.23734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aim of this study was to define the performance characteristics of the Mindray chemiluminescence assay for anti-Müllerian hormone (AMH) detection. DESIGNS AND METHODS Intra-assay and total imprecision, analytical sensitivity, linearity, and interference were compared between the Mindray and Roche assays using pools of human serum according to Clinical and Laboratory Standards Institute protocols. Additionally, male and female reference intervals were established using serum specimens collected from otherwise healthy groups and patients with polycystic ovary syndrome (PCOS). RESULTS The intra-assay and total imprecision percent coefficients of variation for low and high AMH serum levels were 2.74%/ 3.01% and 5.41%/5.35% respectively. The limits of blank, detection, and quantitation were 0.007, 0.01, and 0.03 ng/ml, respectively. The assay displayed good linearity over the range of 0.01-23 ng/ml. The coefficient of determination (R2 ) of the Mindray versus Roche assays was 0.9713 with 411 samples with AMH concentrations ranging from 0.014 to 22.1 ng/ml. The slope and intercept of the regression equation were 0.9687 and 0.3419, respectively. There was no significant interference from triglycerides (up to 3000 mg/dl), bilirubin (up to 50 mg/dl), hemoglobin (up to 500 mg/dl), or total protein (up to 10 g/dl). Reference intervals showed the expected decrease in serum AMH levels with age in healthy women and increased levels in women with PCOS. CONCLUSION The Mindray AMH assay demonstrated acceptable analytical performance under routine conditions and is suitable for determining AMH levels in serum samples.
Collapse
Affiliation(s)
- Jing-Jing Zhao
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chun-Min Kang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Zhang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Zhao X, Ji M, Wen X, Chen D, Huang F, Guan X, Tian J, Xie J, Shao J, Wang J, Huang L, Lin H, Ye L, Chen H. Effects of Midazolam on the Development of Adult Leydig Cells From Stem Cells In Vitro. Front Endocrinol (Lausanne) 2021; 12:765251. [PMID: 34867807 PMCID: PMC8632869 DOI: 10.3389/fendo.2021.765251] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/04/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Midazolam is a neurological drug with diverse functions, including sedation, hypnosis, decreased anxiety, anterograde amnesia, brain-mediated muscle relaxation, and anticonvulsant activity. Since it is frequently used in children and adolescents for extended periods of time, there is a risk that it may affect their pubertal development. Here, we report a potential effect of the drug on the development of Leydig cells (LCs), the testosterone (T)-producing cells in the testis. METHODS Stem LCs (SLCs), isolated from adult rat testes by a magnetic-activated cell sorting technique, were induced to differentiate into LCs in vitro for 3 weeks. Midazolam (0.1-30 μM) was added to the culture medium, and the effects on LC development were assayed. RESULTS Midazolam has dose-dependent effects on SLC differentiation. At low concentrations (0.1-5 μM), the drug can mildly increase SLC differentiation (increased T production), while at higher concentrations (15-30 μM), it inhibits LC development (decreased T production). T increases at lower levels may be due to upregulations of scavenger receptor class b Member 1 (SCARB1) and cytochrome P450 17A1 (CYP17A1), while T reductions at higher levels of midazolam could be due to changes in multiple steroidogenic proteins. The uneven changes in steroidogenic pathway proteins, especially reductions in CYP17A1 at high midazolam levels, also result in an accumulation of progesterone. In addition to changes in T, increases in progesterone could have additional impacts on male reproduction. The loss in steroidogenic proteins at high midazolam levels may be mediated in part by the inactivation of protein kinase B/cAMP response element-binding protein (AKT/CREB) signaling pathway. CONCLUSION Midazolam has the potential to affect adult Leydig cell (ALC) development at concentrations comparable with the blood serum levels in human patients. Further studies are needed to test the effects on human cells.
Collapse
Affiliation(s)
- Xingyi Zhao
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Minpeng Ji
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin Wen
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dan Chen
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fu Huang
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoju Guan
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Tian
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiajia Xie
- Department of Pharmacology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingjing Shao
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiexia Wang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Luoqi Huang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Han Lin
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Leping Ye
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- *Correspondence: Haolin Chen, ; Leping Ye,
| | - Haolin Chen
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Pharmacology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Haolin Chen, ; Leping Ye,
| |
Collapse
|
29
|
Abstract
Sarcopenia describes low muscle mass and strength associated with ageing, whilst reduced physical performance indicates the severity of the condition. It can happen independently of other medical conditions and can be a key feature of the frailty phenotype. Frailty is a syndrome of increased vulnerability to incomplete resolution of homeostasis, following a stressor event. Researchers have described the implications of hypothalamic pituitary dysregulation in the pathogenesis of both entities. This review summarizes the recent evidence in this area as well as other endocrine factors such as insulin resistance and vitamin D status and outlines current research priorities. We conducted searches to PubMed and Embase databases for articles, reviews and studies reporting new data on the interaction between hormones of the endocrine system and frailty and/ or sarcopenia in the last 5 years. Interventional studies, cohort studies, case-control studies and animal studies were included. Clinical trials register was also searched to identify ongoing relevant studies. Studies have given us insights into the complex relationships between factors such as anabolic hormones, glucocorticoids and vitamin D on muscle strength and performance and their involvement in ageing phenotypes. However, robust randomized controlled trials are needed to consolidate existing evidence in humans and inform clinical practice. Current evidence supports hormone replacement in patients with confirmed deficiencies, to optimize health and prevent complications. Hormone replacement has limited use for age-related conditions. Current interest is focused on muscle/bone/fat interactions and health outcomes in "sarcopenic obesity." A life-course approach to improving 'health-span' is advocated. Lifestyle factors such as nutrition and physical activity have important interactions with body composition, physical function and metabolic outcomes. Large-scale clinical trials will determine the efficacy and long-term safety of hormone supplementation in the management of sarcopenia and frailty.
Collapse
Affiliation(s)
- Vicky Kamwa
- Musculoskeletal Endocrinology Research Group, Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK - .,Academic Metabolic Bone Unit, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK - .,Institute of Inflammation and Ageing, The University of Birmingham, Birmingham, UK -
| | - Carly Welch
- Institute of Inflammation and Ageing, The University of Birmingham, Birmingham, UK
| | - Zaki K Hassan-Smith
- Musculoskeletal Endocrinology Research Group, Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK.,Academic Metabolic Bone Unit, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
30
|
Mustieles V, D'Cruz SC, Couderq S, Rodríguez-Carrillo A, Fini JB, Hofer T, Steffensen IL, Dirven H, Barouki R, Olea N, Fernández MF, David A. Bisphenol A and its analogues: A comprehensive review to identify and prioritize effect biomarkers for human biomonitoring. ENVIRONMENT INTERNATIONAL 2020; 144:105811. [PMID: 32866736 DOI: 10.1016/j.envint.2020.105811] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/24/2020] [Accepted: 05/07/2020] [Indexed: 05/21/2023]
Abstract
Human biomonitoring (HBM) studies have demonstrated widespread and daily exposure to bisphenol A (BPA). Moreover, BPA structural analogues (e.g. BPS, BPF, BPAF), used as BPA replacements, are being increasingly detected in human biological matrices. BPA and some of its analogues are classified as endocrine disruptors suspected of contributing to adverse health outcomes such as altered reproduction and neurodevelopment, obesity, and metabolic disorders among other developmental and chronic impairments. One of the aims of the H2020 European Human Biomonitoring Initiative (HBM4EU) is the implementation of effect biomarkers at large scales in future HBM studies in a systematic and standardized way, in order to complement exposure data with mechanistically-based biomarkers of early adverse effects. This review aimed to identify and prioritize existing biomarkers of effect for BPA, as well as to provide relevant mechanistic and adverse outcome pathway (AOP) information in order to cover knowledge gaps and better interpret effect biomarker data. A comprehensive literature search was performed in PubMed to identify all the epidemiologic studies published in the last 10 years addressing the potential relationship between bisphenols exposure and alterations in biological parameters. A total of 5716 references were screened, out of which, 119 full-text articles were analyzed and tabulated in detail. This work provides first an overview of all epigenetics, gene transcription, oxidative stress, reproductive, glucocorticoid and thyroid hormones, metabolic and allergy/immune biomarkers previously studied. Then, promising effect biomarkers related to altered neurodevelopmental and reproductive outcomes including brain-derived neurotrophic factor (BDNF), kisspeptin (KiSS), and gene expression of nuclear receptors are prioritized, providing mechanistic insights based on in vitro, animal studies and AOP information. Finally, the potential of omics technologies for biomarker discovery and its implications for risk assessment are discussed. To the best of our knowledge, this is the first effort to comprehensively identify bisphenol-related biomarkers of effect for HBM purposes.
Collapse
Affiliation(s)
- Vicente Mustieles
- University of Granada, Center for Biomedical Research (CIBM), Spain; Instituto de Investigación Biosanitaria (ibs. GRANADA), Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), Spain.
| | - Shereen Cynthia D'Cruz
- Univ Rennes, EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Stephan Couderq
- Evolution des Régulations Endocriniennes, Département "Adaptation du Vivant", UMR 7221 MNHN/CNRS, Sorbonne Université, Paris 75006, France
| | | | - Jean-Baptiste Fini
- Evolution des Régulations Endocriniennes, Département "Adaptation du Vivant", UMR 7221 MNHN/CNRS, Sorbonne Université, Paris 75006, France
| | - Tim Hofer
- Section of Toxicology and Risk Assessment, Norwegian Institute of Public Health, P.O. Box 222 Skøyen, NO-0213 Oslo, Norway
| | - Inger-Lise Steffensen
- Section of Toxicology and Risk Assessment, Norwegian Institute of Public Health, P.O. Box 222 Skøyen, NO-0213 Oslo, Norway
| | - Hubert Dirven
- Section of Toxicology and Risk Assessment, Norwegian Institute of Public Health, P.O. Box 222 Skøyen, NO-0213 Oslo, Norway
| | - Robert Barouki
- University Paris Descartes, ComUE Sorbonne Paris Cité, Paris, France. Institut national de la santé et de la recherche médicale (INSERM, National Institute of Health & Medical Research) UMR S-1124, Paris, France
| | - Nicolás Olea
- University of Granada, Center for Biomedical Research (CIBM), Spain; Instituto de Investigación Biosanitaria (ibs. GRANADA), Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), Spain
| | - Mariana F Fernández
- University of Granada, Center for Biomedical Research (CIBM), Spain; Instituto de Investigación Biosanitaria (ibs. GRANADA), Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), Spain.
| | - Arthur David
- Univ Rennes, EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France.
| |
Collapse
|
31
|
Ham J, You S, Lim W, Song G. Bifenthrin impairs the functions of Leydig and Sertoli cells in mice via mitochondrion-endoplasmic reticulum dysregulation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 266:115174. [PMID: 32683091 DOI: 10.1016/j.envpol.2020.115174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/17/2020] [Accepted: 07/02/2020] [Indexed: 06/11/2023]
Abstract
Bifenthrin (BF) is a synthetic insecticide that is widely used in fields, resulting in an increase in its exposure to animals. However, reports on the toxic effects of BF on mammalian species and the underlying mechanism are still lacking. Here, we elucidated the mechanism underlying the toxic effects of BF on mouse reproduction using cell lines of immature mouse Leydig (TM3) and Sertoli (TM4) cells, which are constituent cells of testes. Our results show that BF suppressed the proliferation and viability of TM3 and TM4 cells. Additionally, treatment with BF induced cell cycle arrest, apoptotic cell death, and DNA fragmentation. Mitochondrial dysfunction and disruption of calcium homeostasis were observed in BF-treated TM3 and TM4 cells. Further, bifenthrin modulated unfolded protein response and mitochondrion-associated membrane and mitogen-activated protein kinase (MAPK)/phosphoinositide 3-kinase (PI3K) signaling pathways. The expression of the mRNAs related to cell cycle progression, steroidogenesis, and spermatogenesis was downregulated by BF, suggestive of testicular toxicity. Taken together, these results demonstrate the intracellular mechanism of action of BF to involve antiproliferative and apoptotic effects and testicular dysfunction in mouse testis.
Collapse
Affiliation(s)
- Jiyeon Ham
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Seungkwon You
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| | - Whasun Lim
- Department of Food and Nutrition, Kookmin University, Seoul, 02707, Republic of Korea.
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
32
|
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: 2.4] [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]
|
33
|
New insights into anti-Müllerian hormone role in the hypothalamic-pituitary-gonadal axis and neuroendocrine development. Cell Mol Life Sci 2020; 78:1-16. [PMID: 32564094 PMCID: PMC7867527 DOI: 10.1007/s00018-020-03576-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/08/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022]
Abstract
Research into the physiological actions of anti-Müllerian hormone (AMH) has rapidly expanded from its classical role in male sexual differentiation to the regulation of ovarian function, routine clinical use in reproductive health and potential use as a biomarker in the diagnosis of polycystic ovary syndrome (PCOS). During the past 10 years, the notion that AMH could act exclusively at gonadal levels has undergone another paradigm shift as several exciting studies reported unforeseen AMH actions throughout the Hypothalamic–Pituitary–Gonadal (HPG) axis. In this review, we will focus on these findings reporting novel AMH actions across the HPG axis and we will discuss their potential impact and significance to better understand human reproductive disorders characterized by either developmental alterations of neuroendocrine circuits regulating fertility and/or alterations of their function in adult life. Finally, we will summarize recent preclinical studies suggesting that elevated levels of AMH may potentially be a contributing factor to the central pathophysiology of PCOS and other reproductive diseases.
Collapse
|
34
|
Albrethsen J, Johannsen TH, Jørgensen N, Frederiksen H, Sennels HP, Jørgensen HL, Fahrenkrug J, Petersen JH, Linneberg A, Nordkap L, Bang AK, Andersson AM, Juul A. Evaluation of Serum Insulin-like Factor 3 Quantification by LC-MS/MS as a Biomarker of Leydig Cell Function. J Clin Endocrinol Metab 2020; 105:5811414. [PMID: 32211773 DOI: 10.1210/clinem/dgaa145] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/23/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND The peptide hormone insulin-like factor 3 (INSL3) is a marker for Leydig cell function and the clinical use of serum INSL3 measurements has been suggested by several groups. AIM (1) To establish a reference range for liquid chromatography-tandem mass spectrometry (LC-MS/MS) of serum INSL3 in healthy boys and men; and (2) to compare the associations of serum INSL3 and testosterone (T) to pubertal stage, lifestyle factors, diurnal variation, body composition, and human chorionic gonadotropin (hCG) stimulation. RESULTS In a reference range based on LC-MS/MS analysis of serum from 1073 boys and men, INSL3 increased from levels close to the detection limit (0.03 µg/L) in prepubertal boys to a maximum mean level of 1.3 µg/L (95% CI, 0.9-2.7) in young men (19-40 years of age) and decreased slightly in older men (0.1 µg/L per decade). Serum T, but not INSL3, was associated with body mass index or body fat percentage and with alcohol consumption. Smoking was positively associated with serum T, but negatively associated with INSL3. There were significant diurnal variations in both INSL3 and T in men (P < 0.001), but serum INSL3 varied substantially less, compared with serum T (± 11% vs ± 26%). Mean serum INSL3 increased after hCG stimulation, but less than T (+ 17% vs + 53%). In both healthy men and in patients suspected of testicular failure, baseline serum INSL3 was more closely associated to the hCG-induced increase in serum T than baseline T itself. CONCLUSION Measurement of serum INSL3 by LC-MS/MS has promise as a marker of testicular disorders.
Collapse
Affiliation(s)
- Jakob Albrethsen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Centre for Research and Research Training in Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Trine Holm Johannsen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Centre for Research and Research Training in Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Niels Jørgensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Centre for Research and Research Training in Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Hanne Frederiksen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Centre for Research and Research Training in Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Henriette P Sennels
- Department of Clinical Biochemistry, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Loevendahl Jørgensen
- Department of Clinical Biochemistry, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jan Fahrenkrug
- Department of Clinical Biochemistry, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen Holm Petersen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Biostatistics, University of Copenhagen, Denmark
| | - Allan Linneberg
- Center for Clinical Research and Disease Prevention, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Loa Nordkap
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Centre for Research and Research Training in Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anne Kirstine Bang
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Centre for Research and Research Training in Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anna-Maria Andersson
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Centre for Research and Research Training in Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Centre for Research and Research Training in Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
35
|
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: 0.6] [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
|