1
|
Luther PM, Spillers NJ, Talbot NC, Sinnathamby ES, Ellison D, Kelkar RA, Ahmadzadeh S, Shekoohi S, Kaye AD. Testosterone replacement therapy: clinical considerations. Expert Opin Pharmacother 2024; 25:25-35. [PMID: 38229462 DOI: 10.1080/14656566.2024.2306832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/15/2024] [Indexed: 01/18/2024]
Abstract
INTRODUCTION As an increasingly popular therapeutic option, testosterone replacement therapy (TRT) has gained significant notoriety for its health benefits in indicated populations, such as those suffering from hypogonadism. AREAS COVERED Benefits such as improved libido, muscle mass, cognition, and quality of life have led to widened public interest in testosterone as a health supplement. No therapy exists without side effects; testosterone replacement therapy has been associated with side effects such as an increased risk of polycythemia, benign prostate hypertrophy (BPH), prostate cancer, gynecomastia, testicular atrophy, and infertility. Testosterone replacement therapy is often accompanied by several prophylactic co-therapies aimed at reducing the prevalence of these side effects. Literature searches for sections on the clinical benefits and risks associated with TRT were performed to include clinical trials, meta-analyses, and systematic reviews from the last 10 years. EXPERT OPINION Data from clinical studies over the last decade suggest that the benefits of this therapy outweigh the risks and result in overall increased quality of life and remission of symptoms related to hypogonadism. With this in mind, the authors of this review suggest that carefully designed clinical trials are warranted for the investigation of TRT in symptomatic age-related hypogonadism.
Collapse
Affiliation(s)
- Patrick M Luther
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | - Noah J Spillers
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | - Norris C Talbot
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | - Evan S Sinnathamby
- School of Medicine, LSU Health Sciences Center New Orleans, New Orleans, LA, USA
| | - Dakota Ellison
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | - Rucha A Kelkar
- School of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Shahab Ahmadzadeh
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | - Alan D Kaye
- Departments of Anesthesiology and Pharmacology, Toxicology, and Neurosciences, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| |
Collapse
|
2
|
Burns K, Phillips C, Brannigan R, Franklin A, Howell J, Schmidt D, Sopfe J, Appiah LC, Anazodo A. Male pediatric, adolescent, and young adult reproductive survivorship. Pediatr Blood Cancer 2023; 70 Suppl 5:e28823. [PMID: 37381156 DOI: 10.1002/pbc.28823] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 06/30/2023]
Abstract
As pediatric, adolescent, and young adult cancer survival rates increase, emphasis is placed on reducing late effects, including reproductive complications and potential impact to fertility. Male survivors are at risk of abnormalities in sperm, hormone deficiencies, and sexual dysfunction. This can impact one's progression into puberty and ability to have a biological child and impacts quality of life following treatment. Access to reproductive care is important and requires patient assessment and appropriate referral to reproductive specialists. This review addresses reproductive complications associated with therapy, standard-of-care testing, and therapeutic interventions. The psychologic impact on psychosexual functioning is also addressed.
Collapse
Affiliation(s)
- Karen Burns
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Christine Phillips
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Robert Brannigan
- Department of Urology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Anna Franklin
- Center for Cancer and Blood Disorders, Children's Hospital of Colorado, Aurora, Colorado
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Jonathan Howell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Deb Schmidt
- Division of Oncology, Children's Hospital of Wisconsin, Milwaukee, Wisconsin
| | - Jenna Sopfe
- Center for Cancer and Blood Disorders, Children's Hospital of Colorado, Aurora, Colorado
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Leslie Coker Appiah
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Denver, Colorado
- Department of Surgery, Pediatric and Adolescent Gynecology, Children's Hospital Colorado, Denver, Colorado
| | - Antoinette Anazodo
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, New South Wales, Australia
- Nelune Comprehensive Cancer Centre, Randwick, New South Wales, Australia
- School of Women's and Children's, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
Esposito M, Salerno M, Calvano G, Agliozzo R, Ficarra V, Sessa F, Favilla V, Cimino S, Pomara C. Impact of anabolic androgenic steroids on male sexual and reproductive function: a systematic review. Panminerva Med 2023; 65:43-50. [PMID: 35146992 DOI: 10.23736/s0031-0808.22.04677-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Anabolic-androgenic steroids (AASs) are a complex cluster of synthetic derivatives of testosterone. AAS abuse is considered a major public health issue since it has increased among young/adolescent males. The use of steroids has a prevalence rate of 14% in young athletes and 30-75% in professional athletes or bodybuilders. AASs simulate the testosterone mechanism, binding the intracellular androgen receptor, and dysregulating the normal hypothalamic-pituitary-gonadal axis in the same way as exogenous testosterone. Abuse can produce several side effects on organs, such as the genital system. The physio-pathological mechanisms that cause AAS abuse-related, genital system disorders in humans are still not completely known. EVIDENCE ACQUISITION This study focuses on the effect of AASs on the male reproductive organs in humans and animals. EVIDENCE SYNTHESIS A systematic review was performed using SCOPUS, PubMed, Google Scholar, and Web of Sciences database up to 31 December 2021 using the keywords: "anabolic-androgenic steroids," "erectile dysfunction," "spermatogenesis" and "infertility;" (anabolic agents) "erectile dysfunction," "spermatogenesis" and "infertility." The review of the literature identified 66 articles published until 2021. Sixty-two articles were included. The use of AASs induces testicular atrophy and azoospermia known as "anabolic steroid-induced hypogonadism." Anabolic steroid induced infertility is characterized by oligo or azoospermia and abnormalities in sperm motility and morphology. Although sperm quality recovers in most cases within 4 months of stopping anabolic steroid abuse, the negative consequences on spermatogenesis can take up to 3 years to disappear. Human studies reported a positive correlation between AAS abuse in athletes and an increase in morphologically abnormal spermatozoa. Animal studies showed the destruction of Leydig cells and testicular atrophy in animals treated with cycles of AASs. CONCLUSIONS The present review of the literature highlights how little is known about the action of AASs on the male genital system. However, although their use is prohibited in many countries, the black market for these substances is still very frequent. The scientific landscape still has a lot to invest in the research of AAS on the male genital system to make young people even more aware of the negative aspects of these substances, contributing to the reduction of these products in an inappropriate way.
Collapse
Affiliation(s)
- Massimiliano Esposito
- Unit of Legal Medicine, Department of Medical, Surgical and Advanced Technologies, "G.F. Ingrassia, " University of Catania, Catania, Italy
| | - Monica Salerno
- Unit of Legal Medicine, Department of Medical, Surgical and Advanced Technologies, "G.F. Ingrassia, " University of Catania, Catania, Italy
| | - Gianluca Calvano
- Section of Urology, Department of Surgery, University of Catania, Catania, Italy
| | - Roberta Agliozzo
- Section of Urology, Department of Surgery, University of Catania, Catania, Italy
| | - Vincenzo Ficarra
- Section of Urology, Department of Human and Pediatric Pathology Gaetano Barresi, University of Messina, Messina, Italy
| | - Francesco Sessa
- Unit of Legal Medicine, Department of Medical, Surgical and Advanced Technologies, "G.F. Ingrassia, " University of Catania, Catania, Italy
| | - Vincenzo Favilla
- Section of Urology, Department of Human and Pediatric Pathology Gaetano Barresi, University of Messina, Messina, Italy
| | - Sebastiano Cimino
- Section of Urology, Department of Surgery, University of Catania, Catania, Italy
| | - Cristoforo Pomara
- Unit of Legal Medicine, Department of Medical, Surgical and Advanced Technologies, "G.F. Ingrassia, " University of Catania, Catania, Italy -
| |
Collapse
|
4
|
Chao HH, Zhang Y, Dong PY, Gurunathan S, Zhang XF. Comprehensive review on the positive and negative effects of various important regulators on male spermatogenesis and fertility. Front Nutr 2023; 9:1063510. [PMID: 36726821 PMCID: PMC9884832 DOI: 10.3389/fnut.2022.1063510] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/23/2022] [Indexed: 01/17/2023] Open
Abstract
With the increasing global incidence of infertility, the influence of environmental factors, lifestyle habits, and nutrients on reproductive health has gradually attracted the attention of researchers. The quantity and quality of sperm play vital roles in male fertility, and both characteristics can be affected by external and internal factors. In this review, the potential role of genetic, environmental, and endocrine factors; nutrients and trace elements in male reproductive health, spermatozoa function, and fertility potency and the underlying mechanisms are considered to provide a theoretical basis for clinical treatment of infertility.
Collapse
Affiliation(s)
- Hu-He Chao
- Development Center for Medical Science and Technology, National Health Commission of the People's Republic of China, Beijing, China
| | - Ye Zhang
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China
| | - Pei-Yu Dong
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | | | - Xi-Feng Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China,*Correspondence: Xi-Feng Zhang ✉ ; ✉
| |
Collapse
|
5
|
The Mechanism and Experimental Validation of Forsythoside A in the Treatment of Male Infertility Were Analyzed Based on Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7723358. [PMID: 36248414 PMCID: PMC9560825 DOI: 10.1155/2022/7723358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/15/2022]
Abstract
Chinese medicine extracts are currently the hotspot of new drug research and development. Herein, we report the mechanism of action of the traditional Chinese medicine extract Forsythiaside A in the treatment of male infertility and experimental verification. We first obtained 95 intersection genes between the target protein of Forsythiaside A and the target genes of male infertility and screened 13 key genes. In molecular docking, Forsythiaside A can each have a higher total docking score with 12 key genes and have a better combination. These 95 intersection genes are mainly related to biological processes such as response to peptide hormone, response to oxidative stress, and participation in the oxidative stress of the forkhead box O (FoxO) signaling pathway. Therefore, we use ornidazole to induce an experimental model of oligoasthenospermia in rats and use different concentrations of Forsythiaside A to intervene. We proved that the semen quality and superoxide dismutase (SOD) activities of model group rats were significantly lower than those of the blank group, and semen quality and SOD activities of the low-dose group and high-dose group were significantly higher than those of the model group. The malondialdehyde (MDA) level of model group rats was significantly higher than that of blank group, while the MDA levels of the low-dose group and high-dose group were significantly lower than that of the model group. Forsythoside A is a potential drug substance for male infertility and improves the semen quality, MDA levels, and SOD activities of rats with oligoasthenospermia.
Collapse
|
6
|
Nazarian A, Azarbayjani MA, Atashak S, Peeri M. Effects of resistance training, palm pollen grain extracts, and testosterone injection on luteinizing hormone receptors, claudin-1, cingulin, and zonula occludens in the prostate tissues of adult male rats. Andrologia 2022; 54:e14394. [PMID: 35226967 DOI: 10.1111/and.14394] [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: 08/03/2021] [Revised: 01/17/2022] [Accepted: 02/01/2022] [Indexed: 11/27/2022] Open
Abstract
The present study aimed to investigate the effects of resistance training, Phoenix dactylifera extract, and testosterone enanthate injection on luteinizing hormone receptor, claudin-1, cingulin, and zonula occludens in the prostate tissues of adult rats. 30 male rats were divided into six groups: (1) control, (2) resistance training, (3) Phoenix dactylifera extract, (4) testosterone enanthate, (5) resistance training+Phoenix dactylifera extract, and (6) resistance training + testosterone enanthate. After completing the treatments and resistance training, all rats were sacrificed via anaesthesia. The results showed that resistance training, Phoenix dactylifera, and testosterone enanthate significantly increased the luteinizing hormone receptor, claudin-1, cingulin, and zonula occludens gene expression levels in the prostate. The resistance training treatment, along with Phoenix dactylifera + testosterone enanthate, exerted synergic effects on the prostate luteinizing hormone receptor levels and claudin-1 gene expression. In conclusion, Phoenix dactylifera, as a natural compound with fewer side effects than testosterone injection, can be used to enhance athletic performance. Besides, considering the potential benefits of Phoenix dactylifera, it can be considered in the treatment of testosterone deficiency; however, further research is needed.
Collapse
Affiliation(s)
- Aram Nazarian
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | | | - Sirvan Atashak
- Department of Exercise Physiology, Mahabad Branch, Islamic Azad University, Mahabad, Iran
| | - Maghsoud Peeri
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
7
|
Whitaker DL, Geyer-Kim G, Kim ED. Anabolic steroid misuse and male infertility: management and strategies to improve patient awareness. Expert Rev Endocrinol Metab 2021; 16:109-122. [PMID: 33973822 DOI: 10.1080/17446651.2021.1921574] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022]
Abstract
Introduction: Anabolic androgenic steroid use is an uncommon but important cause of male infertility. As paternal age and anabolic steroid use increases, providers are more likely than ever to encounter men with infertility and prior or concurrent anabolic steroid use. In this review, we outline the background, epidemiology and pathophysiology of anabolic steroid induced male infertility and provide recommendations regarding the diagnosis, management, and future prevention of this condition.Areas covered: Male reproductive physiology is a tightly regulated process that can be influenced by exogenous sources such as anabolic steroids and selective androgen receptor modulators (SARMs). Data suggest that a combination of selective estrogen receptor modulators (SERMs), human chorionic gonadotropin (hCG), aromatase inhibitors (AIs), and recombinant follicle-stimulating hormone (rFSH) may lead to spermatogenesis recovery.Expert opinion: Anabolic steroid and SARM users continue to exhibit lack of understanding regarding the potential side effects of their use on male fertility. Current literature suggests that spermatogenesis can be safely recovered using a combination of SERMs, hCG, AIs and rFSH although additional studies are necessary. While anabolic steroid prevention strategies have largely been focused on the individual level, further investigation is necessary and should be approached in a socioecological manner.
Collapse
Affiliation(s)
- Dustin L Whitaker
- Department of Urology, University of Tennessee Graduate School of Medicine, Knoxville, Tennessee, United States
| | - Gabriella Geyer-Kim
- Department of Urology, University of Tennessee Graduate School of Medicine, Knoxville, Tennessee, United States
| | - Edward D Kim
- Department of Urology, University of Tennessee Graduate School of Medicine, Knoxville, Tennessee, United States
| |
Collapse
|
8
|
Adeldust H, Farzinpour A, Farshad A, Rostamzadeh J, López Béjar M. Effect of orally administrated letrozole on reproduction performance and gene expression of FOXJ1, LPR2 and PVRL3 in reproductive tract in aged roosters. Theriogenology 2020; 161:131-139. [PMID: 33310231 DOI: 10.1016/j.theriogenology.2020.11.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 12/28/2022]
Abstract
letrozole is an aromatase inhibitor that stops the production of estrogen through interrupting the entrance of hormone androgen into a small amount of estrogen. Therefore, the current study was developed to estimate orally administrated Letrozole on the reproductive performance and relative abundance of Foxj1, PVRL3, and LPR2 mRNA in aged roosters. Fifty-five-week old ROSS 308 breeder roosters (n = 18) were orally treated using letrozole. Primarily, the body weight of the animals was recorded, and they were randomly classified into three groups (n = 6 birds/group) receiving different doses of Letrozole, including 0, 0.015, and 0.03 mg/kg body weight/day for three weeks. At the end of the trial, seminal traits, plasma, testicular hormone levels (testosterone, estradiol, and FSH), histopathological studies, in vitro fertility, and relative abundance of testis PVRL3, epidydimal Foxj1, and LPR2 mRNA were evaluated. Based on the results, the sperm quality variables were statistically higher in the 0.03 group compared to the controls. Greater histologic parameters, such as diameter of seminiferous tubules, thickness of seminiferous epithelium, categorized epididymal region, and in vitro fertility rates were estimated for the treated groups(p < 0.001). Plasma and testicular testosterone, estradiol concentrations, and plasma FSH levels were significantly influenced by letrozll treatment (p < 0.001). Relative mRNA transcript abundance increased for PVRL3 and decreased for Foxj1 and LPR2 in treated groups. Overall, aromatase inhibitors can enhance the reproductive performance of aged commercial broiler breeder roosters. However, it can impact endocytosis and ciliogenesis events via reducing estradiol.
Collapse
Affiliation(s)
- Hamideh Adeldust
- Department of Animal Science, Faculty of Agriculture, Pasdaran St, Sanandaj, Kurdistan, 66177-15175, Iran
| | - Amjad Farzinpour
- Department of Animal Science, Faculty of Agriculture, Pasdaran St, Sanandaj, Kurdistan, 66177-15175, Iran.
| | - Abbas Farshad
- Department of Animal Science, Faculty of Agriculture, Pasdaran St, Sanandaj, Kurdistan, 66177-15175, Iran
| | - Jalal Rostamzadeh
- Department of Animal Science, Faculty of Agriculture, Pasdaran St, Sanandaj, Kurdistan, 66177-15175, Iran
| | - Manel López Béjar
- Department of Health and Animal Anatomy, Universitat Autònoma de Barcelona, Plaça Cívica, 08193, Bellaterra, Barcelona, Spain
| |
Collapse
|
9
|
Comparative Study of the Steroidogenic Effects of Human Chorionic Gonadotropin and Thieno[2,3-D]pyrimidine-Based Allosteric Agonist of Luteinizing Hormone Receptor in Young Adult, Aging and Diabetic Male Rats. Int J Mol Sci 2020; 21:ijms21207493. [PMID: 33050653 PMCID: PMC7590010 DOI: 10.3390/ijms21207493] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022] Open
Abstract
Low-molecular-weight agonists of luteinizing hormone (LH)/human chorionic gonadotropin (hCG) receptor (LHCGR), which interact with LHCGR transmembrane allosteric site and, in comparison with gonadotropins, more selectively activate intracellular effectors, are currently being developed. Meanwhile, their effects on testicular steroidogenesis have not been studied. The purpose of this work is to perform a comparative study of the effects of 5-amino-N-tert-butyl-4-(3-(1-methylpyrazole-4-carboxamido)phenyl)-2-(methylthio)thieno[2,3-d] pyrimidine-6-carboxamide (TP4/2), a LHCGR allosteric agonist developed by us, and hCG on adenylyl cyclase activity in rat testicular membranes, testosterone levels, testicular steroidogenesis and spermatogenesis in young (four-month-old), aging (18-month-old) and diabetic male Wistar rats. Type 1 diabetes was caused by a single streptozotocin (50 mg/kg) injection. TP4/2 (20 mg/kg/day) and hCG (20 IU/rat/day) were administered for 5 days. TP4/2 was less effective in adenylyl cyclase stimulation and ability to activate steroidogenesis when administered once into rats. On the 3rd–5th day, TP4/2 and hCG steroidogenic effects in young adult, aging and diabetic rats were comparable. Unlike hCG, TP4/2 did not inhibit LHCGR gene expression and did not hyperstimulate the testicular steroidogenesis system, moderately increasing steroidogenic proteins gene expression and testosterone production. In aging and diabetic testes, TP4/2 improved spermatogenesis. Thus, during five-day administration, TP4/2 steadily stimulates testicular steroidogenesis, and can be used to prevent androgen deficiency in aging and diabetes.
Collapse
|
10
|
Xia K, Chen H, Wang J, Feng X, Gao Y, Wang Y, Deng R, Wu C, Luo P, Zhang M, Wang C, Zhang Y, Zhang Y, Liu G, Tu X, Sun X, Li W, Ke Q, Deng C, Xiang AP. Restorative functions of Autologous Stem Leydig Cell transplantation in a Testosterone-deficient non-human primate model. Theranostics 2020; 10:8705-8720. [PMID: 32754273 PMCID: PMC7392013 DOI: 10.7150/thno.46854] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
Rationale: Stem Leydig cells (SLCs) transplantation can restore testosterone production in rodent models and is thus a potential solution for treating testosterone deficiency (TD). However, it remains unknown whether these favorable effects will be reproduced in more clinically relevant large-animal models. Therefore, we assessed the feasibility, safety and efficacy of autologous SLCs transplantation in a testosterone-deficient non-human primate (NHP) model. Methods: Cynomolgus monkey SLCs (CM-SLCs) were isolated from testis biopsies of elderly (> 19 years) cynomolgus monkeys by flow cytometry. Autologous CM-SLCs were injected into the testicular interstitium of 7 monkeys. Another 4 monkeys were injected the same way with cynomolgus monkey dermal fibroblasts (CM-DFs) as controls. The animals were then examined for sex hormones, semen, body composition, grip strength, and exercise activity. Results: We first isolated CD271+ CM-SLCs which were confirmed to expand continuously and show potential to differentiate into testosterone-producing Leydig cells (LCs) in vitro. Compared with CM-DFs transplantation, engraftment of autologous CM-SLCs into elderly monkeys could significantly increase the serum testosterone level in a physiological pattern for 8 weeks, without any need for immunosuppression. Importantly, CM-SLCs transplantation recovered spermatogenesis and ameliorated TD-related symptoms, such as those related to body fat mass, lean mass, bone mineral density, strength and exercise capacity. Conclusion: For the first time, our short-term observations demonstrated that autologous SLCs can increase testosterone levels and ameliorate relevant TD symptoms in primate models. A larger cohort with long-term follow-up will be required to assess the translational potential of autologous SLCs for TD therapy.
Collapse
|
11
|
Zhang C, Xie Y, Chen H, Lv L, Yao J, Zhang M, Xia K, Feng X, Li Y, Liang X, Sun X, Deng C, Liu G. FOXO4-DRI alleviates age-related testosterone secretion insufficiency by targeting senescent Leydig cells in aged mice. Aging (Albany NY) 2020; 12:1272-1284. [PMID: 31959736 PMCID: PMC7053614 DOI: 10.18632/aging.102682] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/25/2019] [Indexed: 12/22/2022]
Abstract
Male late-onset hypogonadism is an age-related disease, the core mechanism of which is dysfunction of senescent Leydig cells. Recent studies have shown that elimination of senescent cells can restore proper homeostasis to aging tissue. In the present study, we found that the fork head box O (FOXO) transcription factor FOXO4 was specially expressed in human Leydig cells and that its translocation to the nucleus in the elderly was related to decreased testosterone synthesis. Using hydrogen peroxide-induced senescent TM3 Leydig cells as an in vitro model, we observed that FOXO4 maintains the viability of senescent Leydig cells and suppresses their apoptosis. By disrupting the FOXO4-p53 interaction, FOXO4-DRI, a specific FOXO4 blocker, selectively induced p53 nuclear exclusion and apoptosis in senescent Leydig cells. In naturally aged mice, FOXO4-DRI improved the testicular microenvironment and alleviated age-related testosterone secretion insufficiency. These findings reveal the therapeutic potential of FOXO4-DRI for the treatment of male late-onset hypogonadism.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China.,Reproductive Medicine Research Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Yun Xie
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510000, China
| | - Haicheng Chen
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Linyan Lv
- Reproductive Medicine Research Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou 510000, China
| | - Jiahui Yao
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Min Zhang
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Kai Xia
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Xin Feng
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Yanqing Li
- Reproductive Medicine Research Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Xiaoyan Liang
- Reproductive Medicine Research Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Xiangzhou Sun
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Chunhua Deng
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Guihua Liu
- Reproductive Medicine Research Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| |
Collapse
|
12
|
Ni W, Liu K, Hou G, Pan C, Wu S, Zheng J, Cao J, Chen Q, Huang X. Diurnal variation in sperm DNA fragmentation: analysis of 11,382 semen samples from two populations and in vivo animal experiments. Chronobiol Int 2019; 36:1455-1463. [PMID: 31418296 DOI: 10.1080/07420528.2019.1649275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Circadian rhythms have been found in some reproductive functions phenotypes but remain unclear for sperm DNA fragmentation index (DFI). The present study aims to investigate the diurnal variation of DFI in mice model and men sperm. Adult male mice were sacrificed for sperm DFI with Sperm Chromatin Structure Assay (SCSA) in 24 hours at 6 evenly distributed time points. A cosinor pattern of DFI was observed with a nadir at zeitgeber time 10 AM. In a community population with 630 semen samples collected between 8 AM and 20 PM, the temporal variation of DFI also fit a cosinor pattern with a - 343° acrophase and a nadir at 11 AM (P = .031). In a reproductive-medical-center dataset of 10752 semen samples collected between 7 AM and 11 AM, the decreasing trend of DFI was also confirmed. For the males with multiple samples, intra-individual comparison between different timepoints was performed, and each consecutive hour after 7 AM was also associated with 2.5 (95% CI: -1.0, 5.9)% lower DFI by SCSA or 4.9 (1.9, 7.8)% lower DFI by SCD. Our study reveals a daily diurnal variance in sperm DFI which may suggest a practical approach to get more qualified sperms for natural or assisted reproduction. Abbreviations: BMI, Body mass index; DFI, DNA fragmentation index; MARHCS, Male Reproductive Health in the Chongqing College Students; RMC, Reproductive Medical Center; SCD, Sperm Chromatin Dispersion; SCSA, Sperm Chromatin Structure Assay.
Collapse
Affiliation(s)
- Wuhua Ni
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
| | - Kun Liu
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Guizhong Hou
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Chengshuang Pan
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
| | - Suichun Wu
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
| | - Jiujia Zheng
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
| | - Jia Cao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Qing Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Xuefeng Huang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
| |
Collapse
|
13
|
Guo H, Du X, Zhang Y, Wu J, Wang C, Li M, Hua X, Zhang XA, Yan J. Specific miRNA-G Protein-Coupled Receptor Networks Regulate Sox9a/Sox9b Activities to Promote Gonadal Rejuvenation in Zebrafish. Stem Cells 2019; 37:1189-1199. [DOI: 10.1002/stem.3040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/17/2019] [Accepted: 05/04/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Huiping Guo
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
| | - Xinlu Du
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
| | - Ying Zhang
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
| | - Jiacheng Wu
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
| | - Chenghui Wang
- Department of Aquaculture; Shanghai Ocean University; Lingang New City, Shanghai People's Republic of China
| | - Mingyou Li
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources; Ministry of Education; Shanghai People's Republic of China
| | - Xianxin Hua
- Department of Cancer Biology; University of Pennsylvania Perelman School of Medicine; Philadelphia, Pennsylvania USA
| | - Xin A. Zhang
- Stephenson Cancer Center and Department of Physiology; The University of Oklahoma Health Sciences Center; Oklahoma City Oklahoma USA
| | - Jizhou Yan
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources; Ministry of Education; Shanghai People's Republic of China
| |
Collapse
|
14
|
|
15
|
Zhao N, Lu XL, Li JT, Zhang JM. Treatment of idiopathic oligozoospermia with combined human chorionic gonadotropin/human menopausal gonadotrophin: A randomised, double-blinded, placebo-controlled clinical study. Andrologia 2019; 51:e13271. [PMID: 30891813 DOI: 10.1111/and.13271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/13/2019] [Accepted: 02/26/2019] [Indexed: 01/26/2023] Open
Abstract
To evaluate whether hCG/hMG therapy has beneficial effects on idiopathic oligozoospermia in Chinese infertility population. The patients were randomly divided into the treatment group receiving hCG/hMG for 3 months and the placebo group receiving placebo for 3 months. Semen and biochemical analysis was performed, and DNA fragmentation as well as spermatid concentration was evaluated. Administration of hCG/hMG for 3 months could significantly improve sperm concentration, rate of forward motile spermatozoa, total motile sperm count, the percentage of sperm with normal morphology and the rate of spontaneous pregnancy in medium- and higher-level inhibin B group respectively. Moreover, in medium- and higher-level inhibin B group, sperm DNA fragmentation index and spermatid concentration were significantly declined respectively at the end of treatment. However, there were no significant differences in lower-level inhibin B group before and after treatment in term of seminal parameters, DNA fragmentation and spermatid concentration. HCG/hMG therapy for 3 months has a beneficial effect on a part of male with idiopathic oligozoospermia, and the efficacy of hCG/hMG therapy is associated with the inhibin B level.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Andrology, People's Hospital of Liaocheng City, Liaocheng City, China
| | - Xi-Lan Lu
- Center for Reproductive Medicine, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Jun-Tao Li
- Center for Reproductive Medicine, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | | |
Collapse
|
16
|
Grubić Kezele T. Cryptozoospermia after treatment with clomiphene citrate following long-term use of intramuscular testosterone undecanoate depot injection (Nebido®). Horm Mol Biol Clin Investig 2019; 39:/j/hmbci.ahead-of-print/hmbci-2018-0078/hmbci-2018-0078.xml. [PMID: 30870141 DOI: 10.1515/hmbci-2018-0078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/10/2019] [Indexed: 11/15/2022]
Abstract
Objective To illustrate the importance of treatment duration with intramuscular testosterone undecanoate (Nebido®) for the final spermatogenesis recovery after treatment cessation. Also, to show a subsequent poor efficacy of the selective estrogen receptor modulator (SERM) clomiphene citrate (CC) in treating steroid-induced azoospermia following Nebido® cessation and describe that initial oligozoospermia, existing before starting Nebido®, largely contributes to that treatment outcome. Methodology Setting: Department of Human Reproduction and Department of Endocrinology, Clinical Hospital Center Rijeka, Rijeka, and Department of Endocrinology, Clinical Hospital Center Sestre milosrdnice, Zagreb, Croatia. PATIENT A male patient having been diagnosed with primary hypogonadotropic hypogonadism, oligozoospermia and low testosterone (T) level, was treated with intramuscular testosterone undecanoate (TU) depot 1 g (Nebido®) to prevent further progression of testosterone deficiency symptoms (low mood, energy and concentration, fatigue, muscle weakness). INTERVENTIONS Stopping Nebido® and treatment with CC 50 mg per day 5 days per week for 3-6 month to recover spermatogenesis. MAIN OUTCOME MEASURES T levels and semen analyses. Results Semen analyses did not return to values before taking Nebido® 1 year after cessation nor after 3 months of treatment with CC. Values of T, follicle stimulating hormone (FSH) and luteinizing hormone (LH) dropped even more than before starting Nebido®, after 1 year of cessation. Conclusions Here we describe a case of initially idiopathic gonadal failure with subsequent secondary gonadal failure and infertility resulting from testosterone replacement therapy (TRT) treatment, and poor spermatogenesis recovery outcome of CC used post Nebido® cessation.
Collapse
Affiliation(s)
- Tanja Grubić Kezele
- Department of Physiology, Immunology and Pathophysiology, University of Rijeka Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia, Phone +385-51-651 238, Mobile Phone: +385-91-755-06-47, Fax +385-51-675 699
| |
Collapse
|
17
|
Mora Rodríguez JA, Porchia LM, Camargo F, López-Bayghen E. The use of insulin-like growth factor 1 improved the parameters of the seminogram in a patient with severe oligoasthenoteratozoospermia. SAGE Open Med Case Rep 2019; 7:2050313X19834154. [PMID: 30858973 PMCID: PMC6404043 DOI: 10.1177/2050313x19834154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 02/05/2019] [Indexed: 11/26/2022] Open
Abstract
Male patients suffering from oligoasthenoteratozoospermia typically failed to
achieve pregnancy, even with assisted reproductive technologies. Growth hormone
and insulin-like growth factor 1 have been shown to regulate sperm quality
parameters; therefore, the insulin-like growth factor 1 supplement could improve
sperm parameters. Here, we determine the effect insulin-like growth factor 1 has
on sperm parameters in a patient suffering from oligoasthenoteratozoospermia. A
47-year-old male was administered once a day 1.5 IU of insulin-like growth
factor 1 by intradermal injection for 2 months. Seminogram analysis was
performed before and after. Treatment with insulin-like growth factor 1 resulted
in a 15.5-fold improvement in sperm concentration (1.1 × 106 vs 18.3
× 106 per mL), 71.4% change in volume (0.7 vs 1.2 mL), increased
progressive motility (2% vs 43%), and the total volume of sperm with progressive
motility (0% vs 23.6%). Here, we show that administering a daily dose of
insulin-like growth factor 1 can improve sperm quality parameters.
Collapse
Affiliation(s)
- José Arturo Mora Rodríguez
- Laboratorio de Fertilización In Vitro and Laboratorio de Investigación y Diagnóstico Molecular, Instituto de Infertilidad y Genética México SC, Ingenes, México City, México
| | - Leonardo M Porchia
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México City, México
| | - Felipe Camargo
- Laboratorio de Fertilización In Vitro and Laboratorio de Investigación y Diagnóstico Molecular, Instituto de Infertilidad y Genética México SC, Ingenes, México City, México
| | - Esther López-Bayghen
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México City, México
| |
Collapse
|
18
|
|
19
|
Pan MM, Hockenberry MS, Kirby EW, Lipshultz LI. Male Infertility Diagnosis and Treatment in the Era of In Vitro Fertilization and Intracytoplasmic Sperm Injection. Med Clin North Am 2018; 102:337-347. [PMID: 29406062 DOI: 10.1016/j.mcna.2017.10.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
As assisted reproductive technologies use increases, the evaluation of male factor infertility has often become overlooked. However, male evaluation remains critically important, with benefits seen in overall health, as well as in natural and assisted pregnancy and birth rates. A comprehensive assessment of the male partner should be offered to all couples seeking infertility care.
Collapse
Affiliation(s)
- Michael M Pan
- Scott Department of Urology, Baylor College of Medicine, 6624 Fannin Street #1700, Houston, TX 77030, USA.
| | - Mark S Hockenberry
- Scott Department of Urology, Center for Reproductive Medicine and Surgery, Baylor College of Medicine, 6624 Fannin Street #1700, Houston, TX 77030, USA
| | - Edgar W Kirby
- Scott Department of Urology, Center for Reproductive Medicine and Surgery, Baylor College of Medicine, 6624 Fannin Street #1700, Houston, TX 77030, USA
| | - Larry I Lipshultz
- Scott Department of Urology, Center for Reproductive Medicine and Surgery, Baylor College of Medicine, 6624 Fannin Street #1700, Houston, TX 77030, USA
| |
Collapse
|
20
|
Xu W, Zhu Q, Zhang B, Liu S, Dai X, Gao C, Gao L, Cui Y. Protective effect of calretinin on testicular Leydig cells via the inhibition of apoptosis. Aging (Albany NY) 2018; 9:1269-1279. [PMID: 28437248 PMCID: PMC5425126 DOI: 10.18632/aging.101226] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/17/2017] [Indexed: 12/11/2022]
Abstract
The core mechanism of Late-onset hypogonadism (LOH) is the deficiency of androgen due to the functional and quantitative decline of testicular Leydig cells. Here we explored the protective effect of calretinin, a Ca2+-binding protein, on Leydig cells. We found in MLTC-1 cells transfected with LV-calb2, the cell viability and optical density (OD) were higher (p<0.05), cells in the S phase of the cell cycle were increased (p<0.01) and p-ERK1/2 and p-AKT levels were significantly higher (p<0.01 and p<0.05), while in R2C cells transfected with LV-siRNA-calb2, all of the results mentioned above were adverse (p<0.05). The cell apoptotic index after calretinin over-expressed was significantly lower (p<0.001), while the expression levels of mitochondria-related apoptotic factors such as cleaved caspase-9 and cytochrome C (cyto C) were lower and ratio of Bcl2/Bax was higher (p<0.05). After calretinin down-regulated, the apoptotic index was higher (p<0.05), while the expression levels of mitochondria-related apoptotic factors were higher and the ratio of Bcl2/Bax was lower (p<0.05). Therefore, calretinin increases Leydig cell viability and proliferation, possibly via ERK1/2 and AKT pathways, and suppresses apoptosis possibly via the mitochondria-related apoptotic pathway, which could be beneficial in understanding the pathophysiology of LOH and could lead to the study of new treatments.
Collapse
Affiliation(s)
- Wendan Xu
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Qian Zhu
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Bei Zhang
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Shan Liu
- Center of Reproductive Medicine, Bethune International Peace Hospital, Hebei Shijiazhuang, China
| | - Xiaonan Dai
- Nanjing Maternal and Child Care Service Center, Nanjing Medical University, Nanjing, 210005, China
| | - Chao Gao
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Li Gao
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Yugui Cui
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
21
|
Maresch CC, Stute DC, Alves MG, Oliveira PF, de Kretser DM, Linn T. Diabetes-induced hyperglycemia impairs male reproductive function: a systematic review. Hum Reprod Update 2017; 24:86-105. [DOI: 10.1093/humupd/dmx033] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 10/16/2017] [Indexed: 12/30/2022] Open
Affiliation(s)
- Constanze C Maresch
- Clinical Research Unit, Centre of Internal Medicine, Justus-Liebig-University, Giessen, Germany
- Hudson Institute of Medical Research and Department of Anatomy & Developmental Biology, Monash University, Melbourne, Australia
| | - Dina C Stute
- Clinical Research Unit, Centre of Internal Medicine, Justus-Liebig-University, Giessen, Germany
| | - Marco G Alves
- Department of Microscopy, Laboratory of Cell Biology, and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Pedro F Oliveira
- Department of Microscopy, Laboratory of Cell Biology, and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- Department of Genetics, Faculty of Medicine, University of Porto, Porto, Portugal
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| | - David M de Kretser
- Hudson Institute of Medical Research and Department of Anatomy & Developmental Biology, Monash University, Melbourne, Australia
| | - Thomas Linn
- Clinical Research Unit, Centre of Internal Medicine, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
22
|
Al-Alami ZM, Shraideh ZA, Taha MO. Rosmarinic acid reverses the effects of metronidazole-induced infertility in male albino rats. Reprod Fertil Dev 2017; 29:1910-1920. [PMID: 27876113 DOI: 10.1071/rd16174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 10/22/2016] [Indexed: 12/16/2023] Open
Abstract
Rosmarinic acid (RA) is a natural antioxidant that has many biological activities. In the present study we investigated the potential of RA to reverse the negative effects of the widely used antibiotic and antiprotozoal agent metronidazole (MTZ), which is known to induce reversible male infertility. Two doses of RA (5 and 15mg kg-1) were studied in sexually mature rats with and without MTZ-induced infertility. Rats were intraperitoneally injected with 5mg kg-1 RA or 15mg kg-1 RA (in distilled water) and, 45min later, they were intraperitoneally injected with 40mg kg-1 MTZ (in distilled water). Cauda epididymidal sperm suspensions were used to assess sperm count, motility and morphology. Histological and ultrastructural studies were performed on the testes and cauda epididymidis. In rats in which infertility was not induced, neither dose of RA affected the parameters assessed. However, in sexually mature rats in which infertility was induced by 40mg kg-1 MTZ, RA at both 5 and 15mg kg-1 ameliorated the damaging effects of MTZ on final bodyweight (30 days later), sperm motility and morphology. Only 5mg kg-1 RA, and not 15mg kg-1 RA, improved the harmful effects of MTZ on the sperm count and testis ultrastructure. The findings of the present study have considerable clinical implications and suggest a possible use for RA to reverse the negative effects of MTZ on male fertility, the male reproductive system and spermatogenesis.
Collapse
Affiliation(s)
- Zina M Al-Alami
- Faculty of Pharmacy, Isra University, PO Box 22, Amman 11622, Jordan
| | - Ziad A Shraideh
- Department of Biological Sciences, Faculty of Science, The University of Jordan, Amman 11942, Jordan
| | - Mutasem O Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
23
|
Use of Exogenous Testosterone for the Treatment of Male Factor Infertility: A Survey of Nigerian Doctors. Int J Reprod Med 2017; 2017:4607623. [PMID: 28951884 PMCID: PMC5603118 DOI: 10.1155/2017/4607623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/20/2017] [Indexed: 11/22/2022] Open
Abstract
Background Though exogenous testosterone is known for its contraceptive effects in men, it is sometimes prescribed by medical practitioners for the treatment of male factor infertility in the mistaken belief that exogenous testosterone improves sperm count. The aim of this study was to evaluate the scope of testosterone use in the treatment of male factor infertility by medical practitioners in Lagos, Nigeria. Methods A survey using a structured questionnaire was carried out amongst doctors attending a regular Continuing Medical Education (CME) programme in Lagos, Nigeria. Results There were 225 respondents. Most of the respondents (69.8%, n = 157) indicated that exogenous testosterone increases sperm count. Only 22 respondents (9.8%) indicated (correctly) that exogenous testosterone decreases sperm count. Seventy-seven respondents (34.2%) had prescribed some form of exogenous testosterone in the treatment of male factor infertility. The vast majority of respondents who had prescribed testosterone (81.8%, n = 63) thought exogenous testosterone increases sperm count. There was no statistically significant difference in the pattern of prescription across the respondents' specialty (p = 0.859) or practice type (p = 0.747). Conclusion The misuse of exogenous testosterone for the treatment of male infertility was common amongst the respondents, with most of them wrongly believing that exogenous testosterone increases sperm count.
Collapse
|
24
|
Rezaie Agdam H, Razi M, Amniattalab A, Malekinejad H, Molavi M. Co-Administration of Vitamin E and Testosterone Attenuates The Atrazine-Induced Toxic Effects on Sperm Quality and Testes in Rats. CELL JOURNAL 2017; 19:292-305. [PMID: 28670522 PMCID: PMC5412788 DOI: 10.22074/cellj.2016.490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 07/27/2016] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Atrazine (ATZ) as a widely used herbicide is considered as a potent endocrine disrupter which adversely affects reproductive systems in both genders. This study aimed to assess the effects of testosterone (T)- and vitamin E (VitE)- alone and their coadministration on testicular function and sperm parameters after exposure to ATZ in rats. MATERIALS AND METHODS In this experimental study, the rats (n=30) are assigned into the following 5 groups: control-sham group (n=6) receiving corn oil, ATZ group (n=6) receiving 200 mg/kg ATZ alone, ATZ+VitE group (n=6) receiving 150 mg/kg ATZ+VitE, ATZ+T group (n=6) receiving 400 µg/kg ATZ+T, and ATZ+VitE+T group (n=6) receiving ATZ+VitE+T for 48 consecutive days. Total antioxidant capacity (TAC), total thiol molecules (TTM), and malondialdehyde (MDA) were analyzed. Serum levels of T, luteinizing hormone (LH), and inhibin-B (IN-B) were also determined. Histological examination and sperm analysis were performed. The data were analyzed using Graph-Pad Prism software version 2.01. RESULTS Co-administration of VitE and T significantly (P<0.05) increased ATZ-decreased TAC and TTM levels and reduced ATZ-increased MDA content. T and VitE significantly (P<0.05) increased serum levels of ATZ-reduced T (1.94 ± 0.96), IN-B (122.10 ± 24.33) and LH (0.40 ± 0.10). The T+VitE animals showed a reduction in apoptotic cells and an increase in Leydig cells steroidogenesis. Co-administration of T and VitE significantly (P<0.05) reduced the ATZ-induced DNA disintegrity and chromatin de-condensation. VitE and T protected germinal cells RNA and protein contents against ATZ-induced damages. CONCLUSION T and VitE in simultaneous form of administration were able to normalize the ATZ-induced derangements through promoting antioxidant capacity and endocrine function.
Collapse
Affiliation(s)
| | - Mazdak Razi
- Department of Comparative Histology, Faculty of Veterinary Medicine, Urmia, Iran
| | - Amir Amniattalab
- Department of Pathology, Faculty of Veterinary Medicine, Islamic Azad University, Urmia Branch, Urmia, Iran
| | - Hassan Malekinejad
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Urmia, Iran
| | | |
Collapse
|
25
|
Letrozole, an aromatase inhibitor, reduces post-peak age-related regression of rooster reproductive performance. Anim Reprod Sci 2017; 183:110-117. [PMID: 28578791 DOI: 10.1016/j.anireprosci.2017.05.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/05/2017] [Accepted: 05/19/2017] [Indexed: 11/21/2022]
Abstract
This study was designed to evaluate orally administrated Letrozole (Lz) on reproductive performance, plasma testosterone and estradiol concentrations and relative abundance of mRNA of GnRH, FSH and LH in roosters. Ross 308 roosters (n=32) that were 40-weeks of age were individually housed and received a basal standard diet supplemented different amounts of capsulated Lz [0 (Lz-0), 0.5 (Lz-0.5), 1 (Lz-1) or 1.5 (Lz-1.5), mg Lz/bird/day] for 12 weeks. Sperm quality variables and plasma testosterone and estradiol concentrations were assessed from the first to the tenth week of the treatment period. Semen samples from the 11th to 12th week were used for artificial insemination and eggs were collected and allotted to assess fertility and hatchability rates. Relative abundance of hypothalamic and pituitary GnRH, LH and FSH mRNA was evaluated at the end of 12th week. The results indicated that total and forward sperm motility as well as egg hatchability rate were greater in the Lz-0.5 group. Greater sperm concentrations, ejaculate volume, sperm plasma membrane integrity, testis index and fertility rates were recorded for both Lz-0.5 and Lz-1 groups compared with the Lz-0 group (P<0.05). Body weight, percentage of sperm abnormalities, and sperm plasma membrane functionality were not affected by treatment. Testosterone and estradiol concentrations were negatively related with greater testosterone concentrations in the Lz-1.5 group which had lesser estradiol concentrations. Relative mRNA transcript abundance for GnRH, LH and FSH was Lz dose responsive being greater in the treated groups; however, this trend plateaued for GnRH and for the relative abundance of both LH and FSH mRNA was less in the Lz-1.5 group than the other treatment groups. It is concluded that Lz may be an effective treatment to improve age related post-peak reproductive performance of roosters.
Collapse
|
26
|
Zang ZJ, Tang HF, Tuo Y, Xing WJ, Ji SY, Gao Y, Deng CH. Effects of velvet antler polypeptide on sexual behavior and testosterone synthesis in aging male mice. Asian J Androl 2017; 18:613-9. [PMID: 26608944 PMCID: PMC4955189 DOI: 10.4103/1008-682x.166435] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Twenty-four-month-old male C57BL/6 mice with low serum testosterone levels were used as a late-onset hypogonadism (LOH) animal model for examining the effects of velvet antler polypeptide (VAP) on sexual function and testosterone synthesis. These mice received VAP for 5 consecutive weeks by daily gavage at doses of 100, 200, or 300 mg kg−1 body weight per day (n = 10 mice per dose). Control animals (n = 10) received the same weight-based volume of vehicle. Sexual behavior and testosterone levels in serum and interstitial tissue of testis were measured after the last administration of VAP. Furthermore, to investigate the mechanisms of how VAP affects sexual behavior and testosterone synthesis in vivo, the expression of steroidogenic acute regulatory protein (StAR), cytochrome P450 cholesterol side-chain cleavage enzyme (P450scc), and 3β-hydroxysteroid dehydrogenase (3β-HSD) in Leydig cells was also measured by immunofluorescence staining and quantitative real-time PCR. As a result, VAP produced a significant improvement in the sexual function of these aging male mice. Serum testosterone level and intratesticular testosterone (ITT) concentration also increased in the VAP-treated groups. The expression of StAR, P450scc, and 3β-HSD was also found to be enhanced in the VAP-treated groups compared with the control group. Our results suggested that VAP was effective in improving sexual function in aging male mice. The effect of velvet antler on sexual function was due to the increased expression of several rate-limiting enzymes of testosterone synthesis (StAR, P450scc, and 3β-HSD) and the following promotion of testosterone synthesis in vivo.
Collapse
Affiliation(s)
- Zhi-Jun Zang
- Department of Infertility and Sexual Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong-Feng Tang
- Department of Dermatology, Shunde First People's Hospital Affiliated to Southern Medical University, Foshan, China
| | - Ying Tuo
- Department of Histopathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei-Jie Xing
- Center for Reproductive Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Su-Yun Ji
- Department of Dermatology, Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Yong Gao
- Reproductive Medicine Centre and Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chun-Hua Deng
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
27
|
Zang ZJ, Ji SY, Zhang YN, Gao Y, Zhang B. Effects of Saikokaryukotsuboreito on Spermatogenesis and Fertility in Aging Male Mice. Chin Med J (Engl) 2017; 129:846-53. [PMID: 26996482 PMCID: PMC4819307 DOI: 10.4103/0366-6999.178972] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background: Aspermia caused by exogenous testosterone limit its usage in late-onset hypogonadism (LOH) patients desiring fertility. Saikokaryukotsuboreito (SKRBT) is reported to improve serum testosterone and relieve LOH-related symptoms. However, it is unclear whether SKRBT affects fertility. We aimed to examine the effects of SKRBT on spermatogenesis and fertility in aging male mice. Methods: Thirty aging male mice were randomly assigned to three groups. Mice were orally administered with phosphate-buffer solution or SKRBT (300 mg/kg, daily) or received testosterone by subcutaneous injections (10 mg/kg, every 3 days). Thirty days later, each male mouse was mated with two female mice. All animals were sacrificed at the end of 90 days. Intratesticular testosterone (ITT) levels, quality of sperm, expression of synaptonemal complex protein 3 (SYCP3), and fertility were assayed. Results: In the SKRBT-treated group, ITT, quality of sperm, and expression of SYCP3 were all improved compared with the control group (ITT: 85.50 ± 12.31 ng/g vs. 74.10 ± 11.45 ng/g, P = 0.027; sperm number: [14.94 ± 4.63] × 106 cells/ml vs. [8.79 ± 4.38] × 106 cells/ml, P = 0.002; sperm motility: 43.16 ± 9.93% vs. 33.51 ± 6.98%, P = 0.015; the number of SYCP3-positive cells/tubule: 77.50 ± 11.01 ng/ml vs. 49.30 ± 8.73 ng/ml, P < 0.001; the expression of SYCP3 protein: 1.23 ± 0.09 vs. 0.84 ± 0.10, P < 0.001), but fertility was not significantly changed (P > 0.05, respectively). In the testosterone-treated group, ITT, quality of sperm, and expression of SYCP3 were markedly lower than the control group (ITT: 59.00 ± 8.67, P = 0.005; sperm number: [4.34 ± 2.45] × 106 cells/ml, P = 0.018; sperm motility: 19.53 ± 7.69%, P = 0.001; the number of SYCP3-positive cells/tubule: 30.00 ± 11.28, P < 0.001; the percentage of SYCP3-positive tubules/section 71.98 ± 8.88%, P = 0.001; the expression of SYCP3 protein: 0.71 ± 0.09, P < 0.001), and fertility was also suppressed (P < 0.05, respectively). Conclusion: SKRBT had no adverse effect on fertility potential in aging male mice.
Collapse
Affiliation(s)
- Zhi-Jun Zang
- Department of Infertility and Sexual Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | | | | | | | | |
Collapse
|
28
|
McBride JA, Coward RM. Recovery of spermatogenesis following testosterone replacement therapy or anabolic-androgenic steroid use. Asian J Androl 2017; 18:373-80. [PMID: 26908067 PMCID: PMC4854084 DOI: 10.4103/1008-682x.173938] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The use of testosterone replacement therapy (TRT) for hypogonadism continues to rise, particularly in younger men who may wish to remain fertile. Concurrently, awareness of a more pervasive use of anabolic-androgenic steroids (AAS) within the general population has been appreciated. Both TRT and AAS can suppress the hypothalamic-pituitary-gonadal (HPG) axis resulting in diminution of spermatogenesis. Therefore, it is important that clinicians recognize previous TRT or AAS use in patients presenting for infertility treatment. Cessation of TRT or AAS use may result in spontaneous recovery of normal spermatogenesis in a reasonable number of patients if allowed sufficient time for recovery. However, some patients may not recover normal spermatogenesis or tolerate waiting for spontaneous recovery. In such cases, clinicians must be aware of the pathophysiologic derangements of the HPG axis related to TRT or AAS use and the pharmacologic agents available to reverse them. The available agents include injectable gonadotropins, selective estrogen receptor modulators, and aromatase inhibitors, but their off-label use is poorly described in the literature, potentially creating a knowledge gap for the clinician. Reviewing their use clinically for the treatment of hypogonadotropic hypogonadism and other HPG axis abnormalities can familiarize the clinician with the manner in which they can be used to recover spermatogenesis after TRT or AAS use.
Collapse
Affiliation(s)
| | - Robert M Coward
- Department of Urology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7235, USA
| |
Collapse
|
29
|
Abstract
Vasectomy is a safe and effective method of contraception used by 42–60 million men worldwide. Approximately 3%–6% of men opt for a vasectomy reversal due to the death of a child or divorce and remarriage, change in financial situation, desire for more children within the same marriage, or to alleviate the dreaded postvasectomy pain syndrome. Unlike vasectomy, vasectomy reversal is a much more technically challenging procedure that is performed only by a minority of urologists and places a larger financial strain on the patient since it is usually not covered by insurance. Interest in this procedure has increased since the operating microscope became available in the 1970s, which consequently led to improved patency and pregnancy rates following the procedure. In this clinical update, we discuss patient evaluation, variables that may influence reversal success rates, factors to consider in choosing to perform vasovasostomy versus vasoepididymostomy, and the usefulness of vasectomy reversal to alleviate postvasectomy pain syndrome. We also review the use of robotics for vasectomy reversal and other novel techniques and instrumentation that have emerged in recent years to aid in the success of this surgery.
Collapse
Affiliation(s)
| | - Ryan P Smith
- Department of Urology, University of Virginia, Charlottesville, VA 22908-0422, USA
| |
Collapse
|
30
|
|
31
|
Zang Z, Gao Y, Ji S, Jiang M. Effects of Guhanyangshengjing Tablet on Testosterone Synthesis and Expression of SYCP3 in the Testis of Aging Male Rats. Chin Med 2016. [DOI: 10.4236/cm.2016.72006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
32
|
He J, Cui J, Wang R, Gao L, Gao X, Yang L, Zhang Q, Cao J, Yu W. Exposure to Hypoxia at High Altitude (5380 m) for 1 Year Induces Reversible Effects on Semen Quality and Serum Reproductive Hormone Levels in Young Male Adults. High Alt Med Biol 2015; 16:216-22. [PMID: 26288097 DOI: 10.1089/ham.2014.1046] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jiang He
- Institute of Clinical Medicine, Urumqi General Hospital of Lanzhou Command, PLA, Urumqi, Xinjiang, China
| | - Jianhua Cui
- Institute of Mountain Sickness, The 18th Hospital of PLA, Yecheng, Xinjiang, China
| | - Rui Wang
- Institute of Clinical Medicine, Urumqi General Hospital of Lanzhou Command, PLA, Urumqi, Xinjiang, China
| | - Liang Gao
- Institute of Mountain Sickness, The 18th Hospital of PLA, Yecheng, Xinjiang, China
| | - Xiaokang Gao
- Institute of Clinical Medicine, Urumqi General Hospital of Lanzhou Command, PLA, Urumqi, Xinjiang, China
| | - Liu Yang
- Institute of Clinical Medicine, Urumqi General Hospital of Lanzhou Command, PLA, Urumqi, Xinjiang, China
| | - Qiong Zhang
- Institute of Clinical Medicine, Urumqi General Hospital of Lanzhou Command, PLA, Urumqi, Xinjiang, China
| | - Jinjun Cao
- Institute of Clinical Medicine, Urumqi General Hospital of Lanzhou Command, PLA, Urumqi, Xinjiang, China
| | - Wuzhong Yu
- Institute of Clinical Medicine, Urumqi General Hospital of Lanzhou Command, PLA, Urumqi, Xinjiang, China
| |
Collapse
|
33
|
|
34
|
Hsiao CH, Ji ATQ, Chang CC, Cheng CJ, Lee LM, Ho JHC. Local injection of mesenchymal stem cells protects testicular torsion-induced germ cell injury. Stem Cell Res Ther 2015; 6:113. [PMID: 26025454 PMCID: PMC4449584 DOI: 10.1186/s13287-015-0079-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 12/22/2014] [Accepted: 04/10/2015] [Indexed: 11/30/2022] Open
Abstract
Introduction Testicular torsion is a urological emergency and infertility is a common complication due to ischemic injury. Surgical reduction and orchiopexy is indicated, but to date there is no effective method for restoration of spermatogenesis. The effects of mesenchymal stem cells (MSCs) on acute tissue injury have been demonstrated, and the abilities of paracrine support, differentiation and immune-modulation may benefit to testicular torsion-induced infertility. We investigate the therapeutic efficacy and the mechanisms of MSCs in testicular torsion-induced germ cell injury when injected locally. Methods Six to eight-week-old Sprague–Dawley rats received surgical 720 degree torsion for 3 hours, followed by detorsion on the left testis. 20 μl of phosphate-buffered saline (PBS) without or with 3 x 104 MSCs from human orbital fat tissues (OFSCs) were given for 10 rats, respectively, via local injection into the left testis 30 minutes before detorsion. 20 μl of PBS injection for 6 rats with surgical exposure without torsion served as sham control. Histopathology with Johnsen’s score analysis, Western blot analysis for superoxide dismutase 2, Bax, Caspase-3, human insulin growth factor-1 and human stem cell factor, malondialdehyde (MDA) assay in testis and plasma, hormones level including testosterone, follicle-stimulating hormone (FSH) and luteinizing hormone (LH) by ELISA Kits, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and fluorescence staining for P450, Sox-9 and VASA were performed. Results Animals were sacrificed and bilateral orchiectomy was performed 7 days after torsion-detorsion. Local injections of OFSCs prevented torsion-induced infertility judging from Johnsen's score. TUNEL assay and Western blot analysis on caspase 3 and Bax demonstrated that OFSCs prevented ischemic/reperfusion induced intrinsic apoptosis. MDA assay revealed that OFSCs significantly reduced the oxidative stress in the damaged testicular tissues. After the OFSC injection, serum testosterone secretion was increased, while the elevation of FSH triggered by testicular injury was balanced. OFSCs also produced stem cell factor in the damaged testis. Immunofluorescence staining revealed that most transplanted cells surrounded the Leydig cells. Some of transplanted cells differentiated into p450 expressing cells within 7 days. Conclusions Local injection of allogenic MSCs before surgical detorsion is a simple, clinical friendly procedure to rescue torsion-induced infertility.
Collapse
Affiliation(s)
- Chi-Hao Hsiao
- Department of Urology, Wan Fang Hospital, Taipei Medical University, #111, Section 3, Hsing-Long Road, Taipei, 116, Taiwan. .,Graduate Institute of Clinical Medicine, Taipei Medical University, #250 Wu-Hsing Street, Taipei, 110, Taiwan.
| | - Andrea Tung-Qian Ji
- Center for Stem Cell Research, Wan Fang Hospital, Taipei Medical University, #111, Section 3, Hsing-Long Road, Taipei, 116, Taiwan.
| | - Chih-Cheng Chang
- Graduate Institute of Clinical Medicine, Taipei Medical University, #250 Wu-Hsing Street, Taipei, 110, Taiwan. .,Department of Pulmonary Medicine, Shuang Ho Hospital, Taipei Medical University, #291, Zhongzheng Road, Zhonghe District, New Taipei, 235, Taiwan.
| | - Chien-Jui Cheng
- Department of Pathology, College of Medicine, Taipei Medical University, #250, Wu-Hsing Street, Taipei, Taiwan. .,Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, #250, Wu-Hsing Street, Taipei, 110, Taiwan.
| | - Liang-Ming Lee
- Department of Urology, Wan Fang Hospital, Taipei Medical University, #111, Section 3, Hsing-Long Road, Taipei, 116, Taiwan.
| | - Jennifer Hui-Chun Ho
- Graduate Institute of Clinical Medicine, Taipei Medical University, #250 Wu-Hsing Street, Taipei, 110, Taiwan. .,Center for Stem Cell Research, Wan Fang Hospital, Taipei Medical University, #111, Section 3, Hsing-Long Road, Taipei, 116, Taiwan.
| |
Collapse
|
35
|
Sato N, Hasegawa T, Hasegawa Y, Arisaka O, Ozono K, Amemiya S, Kikuchi T, Tanaka H, Harada S, Miyata I, Tanaka T. Treatment situation of male hypogonadotropic hypogonadism in pediatrics and proposal of testosterone and gonadotropins replacement therapy protocols. Clin Pediatr Endocrinol 2015; 24:37-49. [PMID: 26019400 PMCID: PMC4436555 DOI: 10.1297/cpe.24.37] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 10/27/2014] [Indexed: 11/15/2022] Open
Abstract
Male hypogonadotropic hypogonadism (MHH), a disorder associated with infertility, is
treated with testosterone replacement therapy (TRT) and/or gonadotropins replacement
therapy (GRT) (TRT and GRT, together with HRT hormone replacement therapy). In Japan,
guidelines have been set for treatment during adolescence. Due to the risk of rapid
maturation of bone age, low doses of testosterone or gonadotropins have been used.
However, the optimal timing and methods of therapeutic intervention have not yet been
established. The objective of this study was to investigate the current situation of
treatment for children with MHH in Japan and to review a primary survey involving
councilors of the Japanese Society for Pediatric Endocrinology and a secondary survey
obtained from 26 facilities conducting HRT. The subjects were 55 patients with MHH who
reached their adult height after HRT. The breakdown of the patients is as follows: 7
patients with Kallmann syndrome, 6 patients with isolated gonadotropin deficiency, 18
patients with acquired hypopituitarism due to intracranial and pituitary tumor, 22
patients with classical idiopathic hypopituitarism due to breech delivery, and 2 patients
with CHARGE syndrome. The mean age at the start of HRT was 15.7 yrs and mean height was
157.2 cm. The mean age at reaching adult height was 19.4 yrs, and the mean adult height
was 171.0 cm. The starting age of HRT was later than the normal pubertal age and showed a
significant negative correlation with pubertal height gain, but it showed no correlation
with adult height. As for spermatogenesis, 76% of the above patients treated with hCG-rFSH
combined therapy showed positive results, though ranging in levels; impaired
spermatogenesis was observed in some with congenital MHH, and favorable spermatogenesis
was observed in all with acquired MHH. From the above, we propose the establishment of a
treatment protocol for the start low-dose testosterone or low-dose gonadotropins by
dividing subjects into two groups to determine different treatment protocols, acquired and
congenital MHH, and to conduct them at a timing closer to the onset of puberty, namely, at
a timing near entrance to junior high school. We also propose a new HRT protocol using
preemptive FSH therapy prior to GRT aimed at achieving future fertility in patients with
congenital MHH.
Collapse
Affiliation(s)
- Naoko Sato
- Study Group of Treatment for MHH ; Tanaka Growth Clinic, Tokyo, Japan
| | - Tomonobu Hasegawa
- Study Group of Treatment for MHH ; Pharmaceutical Affairs Committee, the Japanese Society for Pediatric Endocrinology ; Department of Pediatrics, Keio University Hospital, Tokyo, Japan
| | - Yukihiro Hasegawa
- Study Group of Treatment for MHH ; Division of Endocrinology and Metabolism, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Osamu Arisaka
- Pharmaceutical Affairs Committee, the Japanese Society for Pediatric Endocrinology ; Department of Pediatrics, Dokkyo Medical University, Tochigi, Japan
| | - Keiichi Ozono
- Pharmaceutical Affairs Committee, the Japanese Society for Pediatric Endocrinology ; Department of Pediatrics, Osaka University, Osaka, Japan
| | - Shin Amemiya
- Pharmaceutical Affairs Committee, the Japanese Society for Pediatric Endocrinology ; Department of Pediatrics, Saitama Medical University, Saitama, Japan
| | - Toru Kikuchi
- Pharmaceutical Affairs Committee, the Japanese Society for Pediatric Endocrinology ; Department of Pediatrics, Niigata University, Niigata, Japan ; Present: Department of Pediatrics, Saitama Medical University, Saitama, Japan
| | - Hiroyuki Tanaka
- Pharmaceutical Affairs Committee, the Japanese Society for Pediatric Endocrinology ; Study Group of Treatment for MHH
| | - Shohei Harada
- Pharmaceutical Affairs Committee, the Japanese Society for Pediatric Endocrinology ; Division of Clinical Practice Policy, National Institute for Child Health and Development, Tokyo, Japan
| | - Ichiro Miyata
- Pharmaceutical Affairs Committee, the Japanese Society for Pediatric Endocrinology ; Department of Pediatrics, Jikei University School of Medicine, Tokyo, Japan
| | - Toshiaki Tanaka
- Study Group of Treatment for MHH ; Pharmaceutical Affairs Committee, the Japanese Society for Pediatric Endocrinology ; Tanaka Growth Clinic, Tokyo, Japan
| |
Collapse
|
36
|
Karavolos S, Reynolds M, Panagiotopoulou N, McEleny K, Scally M, Quinton R. Male central hypogonadism secondary to exogenous androgens: a review of the drugs and protocols highlighted by the online community of users for prevention and/or mitigation of adverse effects. Clin Endocrinol (Oxf) 2015; 82:624-32. [PMID: 25333666 DOI: 10.1111/cen.12641] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 09/23/2014] [Accepted: 10/17/2014] [Indexed: 11/28/2022]
Abstract
Androgen- or anabolic steroid-induced hypogonadism (ASIH) is no longer confined to professional athletes; its prevalence amongst young men and teenagers using androgens and/or anabolic steroids (AASs) is rising fast, and those affected can experience significant symptoms. Clinicians are increasingly encountering demanding, well-informed men affected by ASIH, yet lacking authoritative information on the subject may struggle to project a credible message. In this article, we overview the methods and drugs that men use in an attempt to counteract ASIH (with a view to either preventing its onset, or reversing it once it has developed) and summarize the scientific evidence underpinning these. The main channel for obtaining these drugs is the Internet, where they can be readily sourced without a valid prescription. An Internet search using relevant terms revealed a huge number of websites providing advice on how to buy and use products to counteract ASIH. Drugs arising repeatedly in our search included human chorionic gonadotrophin (hCG), selective oestrogen receptor modulators (SERMs) and aromatase inhibitors (AIs). The quality and accuracy of the online information was variable, but review of medical literature also highlighted a lack of scientific data to guide clinical practice. It is important for clinicians to be aware of the AAS user's self-treatment strategies with regard to ASIH side-effect mitigation. By ensuring that they are well-informed, clinicians are more likely to retain the credibility and trust of AAS users, who will in turn likely be more open to engage with appropriate management.
Collapse
Affiliation(s)
- Stamatios Karavolos
- Newcastle Fertility Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | | | | | | | | |
Collapse
|
37
|
Pan M, Ramasamy R, Kovac JR. Is semen analysis necessary prior to the commencement of testosterone supplementation therapy in men of reproductive age? Can Urol Assoc J 2015; 8:446-7. [PMID: 25553161 DOI: 10.5489/cuaj.2259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A semen analysis (SA) should be performed on all men of reproductive age prior to the commencement of testosterone supplementation therapy (TST). A baseline SA has numerous benefits including the ability to unmask occult azoospermia, act as a baseline measure of reproductive function, and provide a recovery target for management of TST-induced testicular dysfunction. Physicians treating men of reproductive age with TST should incorporate SA as part of their initial treatment protocol.
Collapse
Affiliation(s)
- Michael Pan
- Scott Department of Urology, Baylor College of Medicine, Houston, TX
| | - Ranjith Ramasamy
- Scott Department of Urology, Baylor College of Medicine, Houston, TX
| | | |
Collapse
|
38
|
Anogenital distance as a measure of human male fertility. J Assist Reprod Genet 2014; 32:479-84. [PMID: 25533333 DOI: 10.1007/s10815-014-0410-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/11/2014] [Indexed: 10/24/2022] Open
Abstract
PURPOSE In humans, recent studies have correlated anogenital distance (AGD) in adult men to testicular function. While studies of a group of men suggest an association, the utility of AGD in an infertility evaluation remains uncertain. We sought to determine the utility of AGD to predict male fertility. METHODS Between 2010 and 2011, men were recruited at a urology clinic to participate. AGD was measured using digital calipers in men being evaluated at a urology clinic. ANOVA and ROC analyses were used to determine correlations between AGD, fatherhood status, and semen parameters. RESULTS In all, 473 men were included in the analysis with a mean age of 43 ± 13 years. Anogenital distance was significantly longer in men with higher sperm concentration, total sperm count, and total motile sperm count. In order to evaluate the discriminating ability of AGD, ROC curves were created comparing AGD and total testis volume. The area under the curve (AUC) was significantly larger for total testis volume compared to AGD when evaluating fertility (0.71 vs 0.63, p = 0.02). Similarly, there was a trend towards a higher AUC for testis volume compared to AGD for sperm concentration and total sperm count. Stratification of men with long/short AGD and large/small testes also did not improve the predictive value of AGD. CONCLUSIONS While AGD is associated with sperm production on a population level, at the individual level the distinction based AGD alone cannot accurately estimate the efficiency of spermatogenesis.
Collapse
|
39
|
Coward RM, Mata DA, Smith RP, Kovac JR, Lipshultz LI. Vasectomy Reversal Outcomes in Men Previously on Testosterone Supplementation Therapy. Urology 2014; 84:1335-40. [DOI: 10.1016/j.urology.2014.06.081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/31/2014] [Accepted: 06/16/2014] [Indexed: 12/19/2022]
|
40
|
Huang R, Zhu WJ, Li J, Gu YQ. The changes of stage distribution of seminiferous epithelium cycle and its correlations with Leydig cell stereological parameters in aging men. Pathol Res Pract 2014; 210:991-6. [PMID: 24951242 DOI: 10.1016/j.prp.2014.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/22/2014] [Accepted: 05/15/2014] [Indexed: 11/15/2022]
Abstract
PURPOSE To evaluate the changes of stage distribution of seminiferous epithelium cycle and its correlations with Leydig cell stereological parameters in aging men. METHODS Point counting method was used to analyze the stereological parameters of Leydig cells. The stage number of seminiferous epithelium cycle was calculated in the same testicular tissue samples which were used for Leydig cell stereological analysis. RESULTS The aging group had shown more severe pathological changes as well as higher pathologic scores than the young group. Compared with the control group, the volume density (VV) and surface density (NA) of Leydig cells in the aging group were increased significantly. The stage number of seminiferous epithelium cycle in the aging group was decreased coincidently compared to the young group. Leydig cell Vv in the young group has a positive relationship with stages I, II, III, V and VI of seminiferous epithelium cycle, and Leydig cell NA and numerical density (NV) were positively related to stage IV. However, only the correlation between NV and stage II was found in the aging group. CONCLUSIONS The stage number of seminiferous epithelium cycle was decreased in aging testes. Changes in the stage distribution in aging testes were related to the Leydig cell stereological parameters which presented as a sign of morphological changes.
Collapse
Affiliation(s)
- Rui Huang
- Department of Developmental and Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, People's Republic of China
| | - Wei-Jie Zhu
- Department of Developmental and Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, People's Republic of China.
| | - Jing Li
- Department of Pathophysiology, Medical College, Jinan University, Guangzhou, People's Republic of China
| | - Yi-Qun Gu
- National Research Institute for Family Planning, Beijing, People's Republic of China.
| |
Collapse
|
41
|
Rahnema CD, Lipshultz LI, Crosnoe LE, Kovac JR, Kim ED. Anabolic steroid-induced hypogonadism: diagnosis and treatment. Fertil Steril 2014; 101:1271-9. [PMID: 24636400 DOI: 10.1016/j.fertnstert.2014.02.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/26/2014] [Accepted: 02/04/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To develop an understanding of hypogonadal men with a history of anabolic-androgenic steroid (AAS) use and to outline recommendations for management. DESIGN Review of published literature and expert opinions. Intended as a meta-analysis, but no quality studies met the inclusion criteria. SETTING Not applicable. PATIENT(S) Men seeking treatment for symptomatic hypogonadism who have used nonprescribed AAS. INTERVENTION(S) History and physical examination followed by medical intervention if necessary. MAIN OUTCOME MEASURES(S) Serum testosterone and gonadotropin levels, symptoms, and fertility restoration. RESULT(S) Symptomatic hypogonadism is a potential consequence of AAS use and may depend on dose, duration, and type of AAS used. Complete endocrine and metabolic assessment should be conducted. Management strategies for anabolic steroid-associated hypogonadism (ASIH) include judicious use of testosterone replacement therapy, hCG, and selective estrogen receptor modulators. CONCLUSION(S) Although complications of AAS use are variable and patient specific, they can be successfully managed. Treatment of ASIH depends on the type and duration of AAS use. Specific details regarding a patient's AAS cycle are important in medical management.
Collapse
Affiliation(s)
- Cyrus D Rahnema
- University of Tennessee Graduate School of Medicine, Knoxville, Tennessee
| | - Larry I Lipshultz
- Scott Department of Urology, Baylor College of Medicine, Houston, Texas
| | - Lindsey E Crosnoe
- University of Tennessee Graduate School of Medicine, Knoxville, Tennessee
| | - Jason R Kovac
- Scott Department of Urology, Baylor College of Medicine, Houston, Texas
| | - Edward D Kim
- University of Tennessee Graduate School of Medicine, Knoxville, Tennessee.
| |
Collapse
|