1
|
Chang X, Li D, Guo Y, Sheng X, Wang X, Xing K, Xiao L, Lv X, Long C, Qi X. α-Linolenic acid promotes testosterone synthesis by improving mitochondrial function in primary rooster Leydig cells. Theriogenology 2025; 232:9-19. [PMID: 39504870 DOI: 10.1016/j.theriogenology.2024.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024]
Abstract
The present study aimed to investigate the direct effects of α-Linolenic acid (ALA) on the in vitro production of testosterone and the expression of key enzymes and proteins related to steroidogenesis in Leydig cells of roosters. METHODS Purified primary Leydig cells isolated from 65-week-old roosters were purified and treated with different concentrations of ALA treatments: (0 μm/L [control], solvent control group (DMSO), 20 μM/L, 40 μM/L, and 80 μM/L) and cell counting-8 (CCK-8) for cell viability assay, Enzyme-linked immunosorbent assay (ELISA) kit for the determination of testosterone in cell supernatants, quantitative (real-time) PCR, and analysis of activities of antioxidants catalase (CAT), superoxide dismutase (SOD) and malondialdehyde (MDA), evaluation of mitochondrial membrane potential, pro- and anti-apoptotic proteins/genes Bcl-2, Bcl-2-associated X protein (Bax), apoptosis-inducing factor (AIF) were done respectively. RESULTS Our results showed that ALA significantly increased testosterone secretion in primary rooster Leydig cells (P < 0.05), and 40 μM/L is the optimal dose. Leydig cells supplemented with ALA (20, 40, 80 μM) increased the expression of key enzymes and proteins 3β-hydroxysteroid dehydrogenase (3β-HSD), steroidogenic acute regulatory protein (StAR), cholesterol side-chain cleavage enzyme (P450scc) concerning steroidogenesis, enhanced antioxidant capability, improved mitochondrial biogenesis, and markedly improved the mitochondrial membrane potential (P < 0.05). Furthermore, the expression of the apoptosis-suppressive gene Bcl-2 was significantly increased, but Bax and AIF expression was decreased in the ALA group compared to that in the control group (P < 0.05). CONCLUSION ALA promoted testosterone production, enhanced steroidogenic enzyme expression, improved mitochondrial function, and antioxidant capacity, and reduced apoptosis in primary rooster Leydig cells, with 40 μM/L identified as the optimal concentration.
Collapse
Affiliation(s)
- Xuerui Chang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Danyang Li
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Yong Guo
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Xihui Sheng
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Xiangguo Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Kai Xing
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Longfei Xiao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Xueze Lv
- Department of Livestock and Poultry Products Testing, Beijing General Station of Animal Husbandry, Beijing, 100107, China
| | - Cheng Long
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China.
| | - Xiaolong Qi
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China.
| |
Collapse
|
2
|
Mahlouji M, Alavi SMH, Ghasemi J, Jalili AH, Mozanzadeh MT, Zhang S, Shazada NE, Butts IAE, Hoseinifar SH, Linhart O. Crude Oil-Induced Reproductive Disorders in Male Goldfish: Testicular Histopathology, Sex Steroid Hormones, and Sperm Swimming Kinematics. J Appl Toxicol 2024. [PMID: 39721597 DOI: 10.1002/jat.4745] [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: 10/24/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
Crude oil contamination has been shown to impair reproduction in aquatic animals through carcinogenic and genotoxic properties. Here, we assessed the endocrine-disrupting function of crude oil on male reproductive system based on testicular histology, sex steroid hormones, and fertility endpoints in adult male goldfish (Carassius auratus), which were exposed to 0.02- to 2-mg/L crude oil for 21 days (Experiment #1) or to 5- to 250-mg/L crude oil for 9 days (Experiment #2). The crude oil contained 0.22-mg/L nickel (Ni), 1.10-mg/L vanadium (V), and 12.87-mg/L polycyclic aromatic hydrocarbons (PAHs). Twenty-four hours after adding crude oil, the sum of PAHs ranged from 0.30 to 2.28 μg/L in the aquaria containing 0.02- and 250-mg/L crude oil, respectively. Water analyses for heavy metals in Experiment #2 showed high concentrations (mg/L) of Ni (0.07-0-09) and V (0.10-0.21). For both experiments, exposure to crude oil did not impact gonadosomatic index; however, testes showed histopathological defects including hyperplasia or hypertrophy of Sertoli cells, depletion of the Leydig cells, necrosis of germ cells, and fibrosis of lobular wall. In Experiment #1, sperm production and motility, testosterone (T), and 17β-estradiol (E2) were not significantly different among treatments. In Experiment #2, the number of spermiating males decreased by ~50% following exposure to 250-mg/L crude oil. Sperm production, motility kinematics, T, and the T/E2 ratio significantly decreased in males exposed to ≥ 50-mg/L crude oil; however, E2 remained unchanged. Results show crude oil-induced imbalance of sex steroid hormones disrupts spermatogenesis resulting in diminished sperm production and motility.
Collapse
Affiliation(s)
- Mahboubeh Mahlouji
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | | | | | - Amir Hossein Jalili
- Gas Research Division, Research Institute of Petroleum Industry, Tehran, Iran
| | - Mansour Torfi Mozanzadeh
- South of Iran Aquaculture Research Centre, Iranian Fisheries Science Research Institute, Agricultural Research, Education and Extension Organization, Ahwaz, Iran
| | - Songpei Zhang
- South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| | - Nururshopa Eskander Shazada
- South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| | - Ian A E Butts
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, Alabama, USA
| | - Seyed Hossein Hoseinifar
- Department of Fisheries, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Otomar Linhart
- South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| |
Collapse
|
3
|
Vasetska A, Packeiser EM, Körber H, Aslan S, Ay S, Findik M, Binli F, Selçuk M, Speiser-Fontaine C, Goericke-Pesch S. Molecular response of canine testis to GnRH agonist: Insights into AR, HIF-1α, and HSPs expression during arrest and recovery of spermatogenesis. Cell Stress Chaperones 2024; 30:9-21. [PMID: 39631561 PMCID: PMC11719361 DOI: 10.1016/j.cstres.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024] Open
Abstract
Slow-release gonadotropin-releasing hormone (GnRH) agonist implants are frequently used for contraception in male dogs. Although the effects are fully reversible, there is still concern about the safety of the implant's mode of action. Addressing this, we investigated cellular stress and androgen receptor (AR) signaling during downregulation and recovery. Testicular tissues were sampled from dogs castrated at different time points after GnRH implant removal and compared with untreated controls. AR, hypoxia-inducible factor 1 (HIF1A), heat shock proteins heat shock protein 72 (HSP72), heat shock protein 73 (heat shock cognate, HSPA8) (HSP73), heat shock protein A2 (HSPA2), heat shock protein 90 alpha (inducible isoform) (HSP90AA1), and heat shock protein 90 beta (constitutive isoform) (HSP90AB1) were investigated by quantitative real-time polymerase chain reaction and AR, HSP72, HSP73, and HSP90 immunohistochemically. While AR, HIF1A, and HSP70 were upregulated at gene expression level, HSPA8, HSPA2, and HSP90AA1 expression were downregulated during spermatogenic arrest; HSP90AB1 expression did not change. Immunohistochemistry verified AR-expression in Sertoli, peritubular, and Leydig cells, occasionally also in spermatogonia. Stress-inducible HSP72 was occasionally detected, while constitutive HSP73 and HSP90 were abundantly expressed by germ cells. Our results were similar to studies on seasonal breeders such as pine voles, geese, fish, and soft-shelled turtles. Accordingly, GnRH implants did not impose additional cellular stress on testicular cells when compared with natural recrudescence. Since comparative data on HIF1α are scarce, we cannot draw conclusions about hypoxic conditions.
Collapse
Affiliation(s)
- Anastasiia Vasetska
- Unit for Reproductive Medicine - Clinic for Small Animals, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Eva-Maria Packeiser
- Unit for Reproductive Medicine - Clinic for Small Animals, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Hanna Körber
- Unit for Reproductive Medicine - Clinic for Small Animals, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Selim Aslan
- Department of Obstetrics and Gynaecology - Faculty of Veterinary Medicine, Near East University, Nicosia, Cyprus
| | - Serhan Ay
- Department of Obstetrics and Gynaecology - Faculty of Veterinary Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Murat Findik
- Department of Obstetrics and Gynaecology - Faculty of Veterinary Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Firdevs Binli
- Department of Obstetrics and Gynaecology - Faculty of Veterinary Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Murat Selçuk
- Department of Reproduction and Artificial Insemination - Faculty of Veterinary Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | | | - Sandra Goericke-Pesch
- Unit for Reproductive Medicine - Clinic for Small Animals, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany.
| |
Collapse
|
4
|
Negri F, Pozzi E, Corsini C, Raffo M, Belladelli F, Bertini A, Cattafi F, Ventimiglia E, Matloob R, Saccà A, Boeri L, d'Arma A, Montorsi F, Salonia A. Short-Term Hormonal Changes Following Microdissection Testicular Sperm Extraction among Men with Non-Obstructive Azoospermia: Findings from a Large Longitudinal Prospective Multicentric Study. World J Mens Health 2024; 42:42.e102. [PMID: 39743218 DOI: 10.5534/wjmh.240184] [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: 07/21/2024] [Revised: 09/21/2024] [Accepted: 10/09/2024] [Indexed: 01/04/2025] Open
Abstract
PURPOSE We aimed to investigate possible hormonal changes following microdissection testicular sperm extraction (mTESE) in men with non-obstructive azoospermia (NOA) across three referral centers. MATERIALS AND METHODS We prospectively analyzed data from 102 consecutive NOA men. Patients with prior hormonal therapies were excluded. Preoperative serum hormone levels (total testosterone [tT], luteinizing hormone [LH], follicle-stimulating hormone [FSH], and 17β-estradiol) were collected, with repeat measurements at 3-month post-surgery. We divided the cohort into two groups using a tT cut-off value of 3 ng/mL: 1) men who kept eugonadal status; and, 2) men who were initially eugonadal but became testosterone deficient (TD) after surgery. RESULTS Overall, median (interquartile range [IQR]) age was 37 years (32-40 years). Positive sperm retrieval during mTESE was observed in 34 (33.3%) patients, and 48 (47.1%) underwent bilateral mTESE. Compared to baseline, 3-month postoperative median (IQR) hormonal levels were as follows: tT: 3.71 ng/mL (2.76-5.24 ng/mL) vs. 4.27 ng/mL (3.25-6.07 ng/mL), p=0.32; FSH: 22.0 mIU/mL (12.65-31.47 mIU/mL) vs. 19.5 mIU/mL (11.63-25.8 mIU/mL), p=0.25; LH: 9.0 mIU/mL (5.11-12.4 mIU/mL) vs. 7.6 mIU/mL (5.04-13.4 mIU/mL), p=0.73, respectively. Twelve (13.5%) eugonadal men at baseline showed TD after mTESE. Median (IQR) tT values at baseline and 3-month follow-up were compared between those who preserved eugonadal status after surgery and those who became TD after surgery: baseline levels were 4.46 ng/mL (4.1-6.27 ng/mL) vs. 4.14 ng/mL (3.24-4.98 ng/mL), p=0.09; and, 3-month follow-up levels were 4.58 ng/mL (3.58-5.56 ng/mL) vs. 2.51 ng/mL (2.31-2.76 ng/mL), p<0.001, respectively. Men who developed TD had lower testicular volume (TV) (6 [4-10] vs. 10 [8-12.25] Prader, p=0.001) and karyotype abnormalities (4 [33.3] vs. 1 [1.3], p=0.006). CONCLUSIONS This multicentric study shows that mTESE in men with NOA does not significantly impact short-time postoperative follow-up tT, LH, and FSH levels. A substantial proportion of men who were initially eugonadal demonstrated tT suggestive for TD at 3-month follow-up. These men had lower TV at baseline and abnormal karyotype.
Collapse
Affiliation(s)
- Fausto Negri
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Urology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Edoardo Pozzi
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Urology, IRCCS Ospedale San Raffaele, Milan, Italy
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Christian Corsini
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Urology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimiliano Raffo
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Urology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Federico Belladelli
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Urology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessandro Bertini
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Urology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Cattafi
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Urology, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Rayan Matloob
- Department of Urology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Antonino Saccà
- Department of Urology, AO Papa Giovanni XXIII, Bergamo, Italy
| | - Luca Boeri
- Department of Urology, Fondazione IRCCS Ca' Granda Ospedale Maggiore, Milan, Italy
| | - Alessia d'Arma
- Department of Urology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Montorsi
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Urology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Salonia
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Urology, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
5
|
Kakati R, Adhikari K, Saikia Q, Hazarika A. Assessment of reproductive, genotoxic, and cytotoxic effects of leachate-contaminated water in male mice. Heliyon 2024; 10:e40126. [PMID: 39583846 PMCID: PMC11582426 DOI: 10.1016/j.heliyon.2024.e40126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/26/2024] Open
Abstract
Leachate-contaminated water (LCS) poses significant health risks due to its heavy metal content and altered physicochemical properties. This study examined the physicochemical parameters and heavy metal levels in LCS and assessed its reproductive toxicity, genotoxicity, and cytotoxic effects in exposed mice. Groups of mice (n = 5) were orally administered 100 μL of 30 % and 70 % LCS (v/v) twice daily for 35 days. Drinking water served as a negative control, and cyclophosphamide (Cyp) (20 mg/kg bw) as a positive control. On day 36, the mice were weighed, sacrificed, and their testicular weight, sperm count, sperm morphology, viability, acrosome integrity, and serum testosterone were examined. Oxidative stress in the testes, histopathological changes, and serum markers for liver and kidney function (SGOT, SGPT, and creatinine) were also assessed. Genotoxic effects were evaluated using a micronuclei (MN) assay. Analysis of the leachate showed altered physicochemical parameters and elevated heavy metal levels. Exposure to LCS led to a significant decrease in relative testis weight, sperm count, normal sperm morphology, viability, acrosome integrity, and serum testosterone levels. It also caused a notable increase in MDA levels and a decrease in catalase (CAT), superoxide dismutase (SOD), and glutathione (GSH) levels, along with histological changes in the testes of LCS-treated mice compared to controls. Additionally, there was a significant rise in MN formation in RBCs and elevated levels of liver enzymes and creatinine, indicating liver and renal toxicity. Histological alterations in the liver and kidneys were also observed in LCS-exposed mice. These findings suggest that LCS induces reproductive toxicity, genotoxicity, and cytotoxicity in male subjects.
Collapse
Affiliation(s)
- Ranjit Kakati
- Department of Zoology, Gauhati University, Guwahati, India
| | - Kamal Adhikari
- Department of Zoology, Tihu College, Tihu, Nalbari, Assam, India
| | - Queen Saikia
- Department of Zoology, Mangaldai College, Mangaldai, Darrang, Assam, India
| | - Ajit Hazarika
- Tyagbir Hem Baruah College, Jamugurihat, Sonitpur, Assam, India
| |
Collapse
|
6
|
Chernykh V, Solovova O, Sorokina T, Shtaut M, Sedova A, Bliznetz E, Ismagilova O, Beskorovainaya T, Shchagina O, Polyakov A. CAG n Polymorphic Locus of Androgen Receptor ( AR) Gene in Russian Infertile and Fertile Men. Int J Mol Sci 2024; 25:12183. [PMID: 39596257 PMCID: PMC11594939 DOI: 10.3390/ijms252212183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
The androgen receptor (AR) is critical for mediating the effects of androgens. The polymorphic CAGn locus in exon 1 of the AR gene is associated with several diseases, including spinal and bulbar muscular atrophy (SBMA), prostate cancer, and male infertility. This study evaluated the CAGn locus in 9000 infertile Russian men and 286 fertile men (control group). The CAGn locus was analyzed using the amplified fragment length polymorphism method. In the infertile cohort, the number of CAG repeats ranged from 6 to 46, with a unimodal distribution. The number of CAG repeats in infertile and fertile men was 22.15 ± 0.93 and 22.02 ± 1.36, respectively. In infertile men, variants with 16 to 29 repeats were present in 97% of the alleles. A complete mutation (≥42 CAG repeats) was found in three patients, while three others had 39-41 repeats. The incidence of SBMA was 1:3000 infertile men. Significant differences (p < 0.05) were observed between infertile and fertile men in alleles with 21, 24 and 25 repeats. This study revealed certain differences in the CAGn polymorphic locus of the AR gene in Russian infertile and fertile men and determined the frequency of SBMA in infertile patients.
Collapse
Affiliation(s)
- Vyacheslav Chernykh
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.S.); (T.S.); (M.S.); (A.S.); (E.B.); (O.I.); (T.B.); (O.S.); (A.P.)
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of the Russian Federation, 117997 Moscow, Russia
| | - Olga Solovova
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.S.); (T.S.); (M.S.); (A.S.); (E.B.); (O.I.); (T.B.); (O.S.); (A.P.)
| | - Tatyana Sorokina
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.S.); (T.S.); (M.S.); (A.S.); (E.B.); (O.I.); (T.B.); (O.S.); (A.P.)
| | - Maria Shtaut
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.S.); (T.S.); (M.S.); (A.S.); (E.B.); (O.I.); (T.B.); (O.S.); (A.P.)
| | - Anna Sedova
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.S.); (T.S.); (M.S.); (A.S.); (E.B.); (O.I.); (T.B.); (O.S.); (A.P.)
| | - Elena Bliznetz
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.S.); (T.S.); (M.S.); (A.S.); (E.B.); (O.I.); (T.B.); (O.S.); (A.P.)
| | - Olga Ismagilova
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.S.); (T.S.); (M.S.); (A.S.); (E.B.); (O.I.); (T.B.); (O.S.); (A.P.)
| | - Tatiana Beskorovainaya
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.S.); (T.S.); (M.S.); (A.S.); (E.B.); (O.I.); (T.B.); (O.S.); (A.P.)
| | - Olga Shchagina
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.S.); (T.S.); (M.S.); (A.S.); (E.B.); (O.I.); (T.B.); (O.S.); (A.P.)
| | - Aleksandr Polyakov
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.S.); (T.S.); (M.S.); (A.S.); (E.B.); (O.I.); (T.B.); (O.S.); (A.P.)
| |
Collapse
|
7
|
Saez Lancellotti TE, Avena MV, Funes AK, Bernal-López MR, Gómez-Huelgas R, Fornes MW. Exploring the impact of lipid stress on sperm cytoskeleton: insights and prospects. Nat Rev Urol 2024:10.1038/s41585-024-00952-1. [PMID: 39528754 DOI: 10.1038/s41585-024-00952-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
The decline in male fertility correlates with the global rise in obesity and dyslipidaemia, representing significant public health challenges. High-fat diets induce metabolic alterations, including hypercholesterolaemia, hepatic steatosis and atherosclerosis, with detrimental effects on testicular function. Testicular tissue, critically dependent on lipids for steroidogenesis, is particularly vulnerable to these metabolic disruptions. Excessive lipid accumulation within the testes, including cholesterol, triglycerides and specific fatty acids, disrupts essential sperm production processes such as membrane formation, maturation, energy metabolism and cell signalling. This leads to apoptosis, impaired spermatogenesis, and abnormal sperm morphology and function, ultimately compromising male fertility. During spermiogenesis, round spermatids undergo extensive reorganization, including the formation of the acrosome, manchette and specialized filamentous structures, which are essential for defining the final sperm cell shape. In this Perspective, we examine the impact of high-fat diets on the cytoskeleton of spermatogenic cells and its consequences to identify the mechanisms underlying male infertility associated with dyslipidaemia. Understanding these processes may facilitate the development of therapeutic strategies, such as dietary interventions or natural product supplementation, that aim to address infertility in men with obesity and hypercholesterolaemia. The investigation of cytoskeleton response to lipid stress extends beyond male reproduction, offering insights with broader implications.
Collapse
Affiliation(s)
- Tania E Saez Lancellotti
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina.
- Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina.
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain.
| | - María V Avena
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Abi K Funes
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María-Rosa Bernal-López
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Ricardo Gómez-Huelgas
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel W Fornes
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
8
|
Asghar MA, Tang S, Wan B, Han H, Wong LP, Zhang X, Zhao Q. Understanding the impact of valproate on male fertility: insights from preclinical and clinical meta-analysis. BMC Pharmacol Toxicol 2024; 25:69. [PMID: 39334455 PMCID: PMC11438246 DOI: 10.1186/s40360-024-00791-1] [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: 05/06/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Valproic acid (VPA) is a widely used antiepileptic drug (AED) often prescribed as a first-line treatment for many idiopathic and symptomatic generalized epilepsies. Several studies have highlighted the side effects of VPA on male fertility and reproductive factors in males, although the specific underlying etiology of these abnormalities is not clear. The present systematic review and meta-analysis aimed to assess the preclinical and clinical evidence concerning the impact of VPA on male fertility and reproductive factors. METHODS The scientific literature was reviewed for eligibility using PubMed, Web of Science, and PsycINFO, encompassing preclinical and clinical studies. Factors related to male fertility and reproduction, such as differences in sperm count, sperm motility, and the percentage of abnormal sperm, were compared between the experimental groups treated with VPA (in both preclinical and clinical) and the control groups using the Standardized Mean Difference (SMD) with 95% confidence intervals (CIs). Additionally, differences in follicle-stimulating hormone (FSH) and luteinizing hormone (LH) were explicitly assessed in clinical studies. RESULTS Male fertility data were extracted from 7 preclinical studies (112 animals) and 5 clinical studies (274 male individuals). The results of animal studies found that the sperm count (SMD = -2.28, 95% CI: -3.39 to -1.18, P = 0.335) and sperm motility (SMD = -2.32, 95% CI: -3.34 to -1.30, P = 0.368) were decreased in the treated groups compared to the control groups. The percentage of abnormal sperm (SMD = 3.27, 95% CI: 1.98 to 4.56, P = 0.019) was significantly increased, while a non-significant reduction was revealed in the weight of the testis (SMD = -2.73, 95% CI: -4.23 to -1.23, P = 0.673) in treated groups. The outcomes of clinical studies indicated a non-significant decrease in sperm count (SMD = -0.78, 95% CI: -1.58 to 0.03, P = 0.286) among patients with epilepsy treated with VPA compared to control subjects. However, a significant reduction in sperm motility (SMD = -1.62, 95% CI: -2.81 to -0.43, P = 0.033 was observed. The percentage of abnormal sperm showed a non-significant increase (SMD = 0.93, 95% CI: -0.97 to 2.84, P = 0.616) after being treated with VPA. Furthermore, there was a non-significant reduction in the levels of FSH (SMD = -1.32, 95% CI: -2.93 to 0.29, P = 0.198) and LH (SMD = -0.96, 95% CI: -1.95 to 0.04, P = 0.211) observed in clinical participants. CONCLUSION This meta-analysis of both preclinical and clinical studies revealed that VPA causes a significant reduction in male fertility and reproductive factors among male patients with epilepsy. Clinical neurologists should be more cautious when prescribing VPA, especially to young male adult patients with epilepsy.
Collapse
Affiliation(s)
| | - Shixin Tang
- College of Public Health, Chongqing Medical University, Chongqing, PR China
| | - Bing Wan
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China
| | - Hang Han
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China
| | - Li Ping Wong
- Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Xiao Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China
| | - Qinjian Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
9
|
Abarikwu SO, Coimbra JLP, Campolina-Silva G, Rocha ST, Costa VV, Lacerda SMSN, Costa GMJ. Acute effects of atrazine on the immunoexpressions of sertoli and germ cells molecular markers, cytokines, chemokines, and sex hormones levels in mice testes and epididymides. CHEMOSPHERE 2024; 363:142852. [PMID: 39019188 DOI: 10.1016/j.chemosphere.2024.142852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 07/02/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024]
Abstract
Atrazine is currently one of the most commonly used agrochemicals in the United States and elsewhere. Here, we studied the immunoexpression of molecular markers of mammalian testicular functions: androgen receptor (AR), promyelocytic leukemia zinc finger (PLZF), GDNF family receptor alpha-1 (GFRA1), VASA/DDX4 (DEAD-Box Helicase 4) as well as the levels of intratesticular and intra-epididymal estradiol (E2) and dihydrotestosterone (DHT), tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), interleukins (IL-1β and IL-6, IL-10) and testicular chemokines (CXCL-1, CCL-2 and CCL3) in BalB/c mice after a sub-acute gavage treatment with a gonado-toxin, atrazine (50 mg/kg body wt.) for three days. We found high numbers of AR immunopositive Sertoli cells and low numbers of GFRA1, PLZF and VASA/DDX4-positive germ cells in the seminiferous tubule regions of the testes. While TNF-α level in the testes fell and remained unchanged in the epididymides, IFN-γ levels in the testes remained constant but increased in the epididymides. E2 and DHT concentrations remained unaltered in the testes but were changed in the epididymides. There were no significant changes in the levels of interleukins in the testis and epididymis. Intratesticular chemokines were also not significantly altered, except for CCL-4, which was increased in the testis. Light microscopy of the epididymis showed detached epithelium and some detached cells in the lumen. It is concluded that atrazine changed the inflammatory status of the gonads and highlighted Sertoli and undifferentiated spermatogonia as important targets for atrazine's toxic effects in the testis of mice. Concerning the epididymis, atrazine altered the epididymal hormonal concentrations and promoted histopathological modifications in its parenchyma.
Collapse
Affiliation(s)
- Sunny O Abarikwu
- Reproductive Biology and Molecular Toxicology Research Group, Department of Biochemistry, University of Port Harcourt, Choba, Nigeria.
| | - John L P Coimbra
- Laboratório de Biologia Celular, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Minas Gerais, Brazil
| | | | - Samuel Tadeu Rocha
- Laboratório de Biologia Celular, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Minas Gerais, Brazil
| | - Vivian Vasconcelos Costa
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Minas Gerais, Brazil
| | - Samyra M S N Lacerda
- Laboratório de Biologia Celular, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Minas Gerais, Brazil
| | - Guilherme M J Costa
- Laboratório de Biologia Celular, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Minas Gerais, Brazil
| |
Collapse
|
10
|
Boukari O, Ghoghbane S, Khemissi W, Lassili T, Tebourbi O, Rhouma KB, Sakly M, Hallegue D. Phycocyanin alleviates alcohol-induced testicular injury in male Wistar rats. Clin Exp Reprod Med 2024; 51:102-111. [PMID: 38229438 PMCID: PMC11140253 DOI: 10.5653/cerm.2023.06422] [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: 08/05/2023] [Revised: 09/25/2023] [Accepted: 10/10/2023] [Indexed: 01/18/2024] Open
Abstract
OBJECTIVE Given the noteworthy implications of alcohol consumption and its association with male infertility, there has been a notable focus on investigating natural alternatives to mitigate its adverse effects. Thus, this study was conducted to assess the potential protective effect of phycocyanin extract derived from the blue algae Arthrospira (Spirulina) platensis against ethanol-induced oxidative stress, disturbances in testicular morphology, and alterations in sperm production. METHODS Male rats were divided into four groups (five rats each): the control group received a saline solution, the ethanol exposed group (EtOH) was subjected to intraperitoneal injections of 10 mL/kg of ethanol solution at a concentration of 38% (v/v), the phycocyanin alone treated group (P) received oral administration of phycocyanin at a dosage of 50 mg/kg, and the phycocyanin-cotreated group (PE) was given oral phycocyanin followed by ethanol injections. All treatments were administered over a period of 14 days. RESULTS Our findings demonstrated that ethanol exposure induced reproductive toxicity, characterized by reduced sperm production and viability, alterations in testicular weight and morphology, increased lipid peroxidation levels, and elevated oxidative enzyme activity. In addition, the ethanol-intoxicated group showed perturbations in serum biochemical parameters. However, the simultaneous exposure to ethanol and phycocyanin exhibited a counteractive effect against ethanol toxicity. CONCLUSION The results showed that supplementation of phycocyanin prevented oxidative and testicular morphological damage-induced by ethanol and maintained normal sperm production, and viability.
Collapse
Affiliation(s)
- Oumayma Boukari
- Laboratory of Integrated Physiology, Faculty of Sciences of Bizerte, University of Carthage, Bizerte, Tunisia
| | - Soumaya Ghoghbane
- Laboratory of Integrated Physiology, Faculty of Sciences of Bizerte, University of Carthage, Bizerte, Tunisia
| | - Wahid Khemissi
- Laboratory of Integrated Physiology, Faculty of Sciences of Bizerte, University of Carthage, Bizerte, Tunisia
| | - Thalja Lassili
- Laboratory of Human and Experimental Pathological Anatomy, Pasteur Institute, Tunis, Tunisia
| | - Olfa Tebourbi
- Laboratory of Integrated Physiology, Faculty of Sciences of Bizerte, University of Carthage, Bizerte, Tunisia
| | - Khemais Ben Rhouma
- Laboratory of Integrated Physiology, Faculty of Sciences of Bizerte, University of Carthage, Bizerte, Tunisia
| | - Mohsen Sakly
- Laboratory of Integrated Physiology, Faculty of Sciences of Bizerte, University of Carthage, Bizerte, Tunisia
| | - Dorsaf Hallegue
- Laboratory of Integrated Physiology, Faculty of Sciences of Bizerte, University of Carthage, Bizerte, Tunisia
| |
Collapse
|
11
|
Li L, Lin W, Wang Z, Huang R, Xia H, Li Z, Deng J, Ye T, Huang Y, Yang Y. Hormone Regulation in Testicular Development and Function. Int J Mol Sci 2024; 25:5805. [PMID: 38891991 PMCID: PMC11172568 DOI: 10.3390/ijms25115805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/01/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
The testes serve as the primary source of androgens and the site of spermatogenesis, with their development and function governed by hormonal actions via endocrine and paracrine pathways. Male fertility hinges on the availability of testosterone, a cornerstone of spermatogenesis, while follicle-stimulating hormone (FSH) signaling is indispensable for the proliferation, differentiation, and proper functioning of Sertoli and germ cells. This review covers the research on how androgens, FSH, and other hormones support processes crucial for male fertility in the testis and reproductive tract. These hormones are regulated by the hypothalamic-pituitary-gonad (HPG) axis, which is either quiescent or activated at different stages of the life course, and the regulation of the axis is crucial for the development and normal function of the male reproductive system. Hormonal imbalances, whether due to genetic predispositions or environmental influences, leading to hypogonadism or hypergonadism, can precipitate reproductive disorders. Investigating the regulatory network and molecular mechanisms involved in testicular development and spermatogenesis is instrumental in developing new therapeutic methods, drugs, and male hormonal contraceptives.
Collapse
Affiliation(s)
- Lu Li
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Wanqing Lin
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Zhaoyang Wang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Rufei Huang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Huan Xia
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Ziyi Li
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Jingxian Deng
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Tao Ye
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Yadong Huang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China
| | - Yan Yang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China
| |
Collapse
|
12
|
Kamińska A, Lustofin S, Brzoskwinia M, Duliban M, Cyran-Gryboś J, Bilińska B, Hejmej A. Androgens and Notch signaling cooperate in seminiferous epithelium to regulate genes related to germ cell development and apoptosis. Reprod Biol 2024; 24:100878. [PMID: 38490111 DOI: 10.1016/j.repbio.2024.100878] [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: 10/04/2023] [Revised: 03/01/2024] [Accepted: 03/02/2024] [Indexed: 03/17/2024]
Abstract
It was reported previously that in adult males disruption of both androgen and Notch signaling impairs spermatid development and germ cell survival in rodent seminiferous epithelium. To explain the molecular mechanisms of these effects, we focused on the interaction between Notch signaling and androgen receptor (AR) in Sertoli cells and investigate its role in the control of proteins involved in apical ectoplasmic specializations, actin remodeling during spermiogenesis, and induction of germ cell apoptosis. First, it was revealed that in rat testicular explants ex vivo both testosterone and Notch signaling modulate AR expression and cooperate in the regulation of spermiogenesis-related genes (Nectin2, Afdn, Arp2, Eps8) and apoptosis-related genes (Fasl, Fas, Bax, Bcl2). Further, altered expression of these genes was found following exposure of Sertoli cells (TM4 cell line) and germ cells (GC-2 cell line) to ligands for Notch receptors (Delta-like1, Delta-like4, and Jagged1) and/or Notch pathway inhibition. Finally, direct interactions of Notch effector, Hairy/enhancer-of-split related with YRPW motif protein 1, and the promoter of Ar gene or AR protein were revealed in TM4 Sertoli cells. In conclusion, Notch pathway activity in Sertoli and germ cells regulates genes related to germ cell development and apoptosis acting both directly and indirectly by influencing androgen signaling in Sertoli cells.
Collapse
Affiliation(s)
- Alicja Kamińska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
| | - Sylwia Lustofin
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
| | - Małgorzata Brzoskwinia
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
| | - Michał Duliban
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
| | - Joanna Cyran-Gryboś
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Barbara Bilińska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
| | - Anna Hejmej
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
13
|
Zhou Y, Wei Z, Tan J, Sun H, Jiang H, Gao Y, Zhang H, Schroyen M. Alginate oligosaccharide extends the service lifespan by improving the sperm metabolome and gut microbiota in an aging Duroc boars model. Front Cell Infect Microbiol 2023; 13:1308484. [PMID: 38116132 PMCID: PMC10728478 DOI: 10.3389/fcimb.2023.1308484] [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] [Received: 10/06/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Introduction Alginate oligosaccharide (AOS), as a natural non-toxic plant extract, has been paid more attention in recent years due to its strong antioxidant, anti-inflammatory, and even anti-cancer properties. However, the mechanism by which AOS affects animal reproductive performance is still unclear. Methods The purpose of this study is to use multi-omics technology to analyze the effects of AOS in extending the service lifespan of aging boars. Results The results showed that AOS can significantly improve the sperm motility (p < 0.05) and sperm validity rate (p < 0.001) of aging boars and significantly reduce the abnormal sperm rate (p < 0.01) by increasing the protein levels such as CatSper 8 and protein kinase A (PKA) for semen quality. At the same time, AOS significantly improved the testosterone content in the blood of boars (p < 0.01). AOS significantly improved fatty acids such as adrenic acid (p < 0.05) and antioxidants such as succinic acid (p < 0.05) in sperm metabolites, significantly reducing harmful substances such as dibutyl phthalate (p < 0.05), which has a negative effect on spermatogenesis. AOS can improve the composition of intestinal microbes, mainly increasing beneficial bacteria Enterobacter (p = 0.1262) and reducing harmful bacteria such as Streptococcus (p < 0.05), Prevotellaceae_UCG-001 (p < 0.05), and Prevotellaceae_NK3B31_group (p < 0.05). Meanwhile, short-chain fatty acids in feces such as acetic acid (p < 0.05) and butyric acid (p < 0.05) were significantly increased. Spearman correlation analysis showed that there was a close correlation among microorganisms, sperm metabolites, and sperm parameters. Discussion Therefore, the data indicated that AOS improved the semen quality of older boars by improving the intestinal microbiota and sperm metabolome. AOS can be used as a feed additive to solve the problem of high elimination rate in large-scale boar studs.
Collapse
Affiliation(s)
- Yexun Zhou
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Zeou Wei
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jiajian Tan
- YangXiang Joint Stock Company, Animal Nutrition Institute, Guigang, China
| | - Haiqing Sun
- YangXiang Joint Stock Company, Animal Nutrition Institute, Guigang, China
| | - Haidi Jiang
- YangXiang Joint Stock Company, Animal Nutrition Institute, Guigang, China
| | - Yang Gao
- College of Life Science, Baicheng Normal University, Baicheng, Jilin, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Martine Schroyen
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| |
Collapse
|
14
|
Fan Q, He R, Li Y, Gao P, Huang R, Li R, Zhang J, Li H, Liang X. Studying the effect of hyperoside on recovery from cyclophosphamide induced oligoasthenozoospermia. Syst Biol Reprod Med 2023; 69:333-346. [PMID: 37578152 DOI: 10.1080/19396368.2023.2241600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023]
Abstract
Oligoasthenozoospermia is becoming a serious problem, but effective prevention or treatment is lacking. Hyperoside, one of the main active ingredients in traditional Chinese medicine, may be effective in the treatment of oligoasthenozoospermia. In this study, we used cyclophosphamide (CTX: 50 mg/kg) to establish a mouse model of Oligoasthenozoospermia to investigate the therapeutic effect of hyperoside (30 mg/kg) on CTX-induced oligoasthenozoospermia. All mice were divided into four groups: blank control group (Control), treatment control group (Hyp), disease group (CTX) and treatment group (CTX + H). Mice body weight, testicular weight, sperm parameters and testicular histology were used to assess the reproductive capacity of mice and to explore the underlying mechanism of hyperoside in the treatment of oligoasthenozoospermia by assessing hormone levels, protein levels of molecules related to hormone synthesis and transcript levels of important genes related to spermatogenesis. Treatment with hyperoside significantly improved sperm density, sperm viability and testicular function compared to untreated oligoasthenozoospermia mice. In mechanism, treatment with hyperoside resulted in significant improvement in pathological changes in spermatogenic tubules, with an increase in testosterone production, and upregulations of Protein Kinase CAMP-Activated Catalytic Subunit Beta (PRKACB), Steroidogenic Acute Regulatory Protein (STAR), and Cytochrome P450 Family 17 Subfamily A Member 1 (CYP17A1) for testosterone production. Hyperoside also promoted the cell cycle of germ cells and up-regulated meiosis and spermatogenesis-related genes, including DNA Meiotic Recombinase 1 (Dmc1), Ataxia telangiectasia mutated (Atm) and RAD21 Cohesin Complex Component (Rad21). In conclusion, hyperoside exerted protective effects on oligoasthenozoospermia mice by regulating testosterone production, meiosis and sperm maturation of germ cells.
Collapse
Affiliation(s)
- Qigang Fan
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ruifen He
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yi Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Pu Gao
- Second Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Runchun Huang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Rong Li
- Second Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Jiayu Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Hongli Li
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Key Laboratory for Gynecologic Oncology Gansu Province, Lanzhou, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Key Laboratory for Gynecologic Oncology Gansu Province, Lanzhou, China
| |
Collapse
|
15
|
Bonetto V, Magnelli V, Sabbatini M, Caprì F, van Loon JJWA, Tavella S, Masini MA. The importance of gravity vector on adult mammalian organisms: Effects of hypergravity on mouse testis. PLoS One 2023; 18:e0282625. [PMID: 37773950 PMCID: PMC10540970 DOI: 10.1371/journal.pone.0282625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/17/2023] [Indexed: 10/01/2023] Open
Abstract
In the age of space exploration, the effect of hypergravity on human physiology is a relatively neglected topic. However, astronauts have several experiences of hypergravity during their missions. The main disturbance of altered gravity can be imputed to cell cytoskeleton alteration and physiologic homeostasis of the body. Testis has proved to be a particularly sensible organ, subject to environmental alteration and physiological disturbance. This makes testis an organ eligible for investigating the alteration following exposure to altered gravity. In our study, mice were exposed to hypergravity (3g for 14 days) in the Large Diameter Centrifuge machine (ESA, Netherland). We have observed a morphological alteration of the regular architecture of the seminiferous tubules of testis as well as an altered expression of factors involved in the junctional complexes of Sertoli cells, responsible for ensuring the morpho-functional integrity of the organ. The expression of key receptors in physiological performance, such as Androgen Receptors and Interstitial Cells Stimulating Hormone receptors, was found lower expressed. All these findings indicate the occurrence of altered physiological organ performance such as the reduction of the spermatozoa number and altered endocrine parameters following hypergravity exposure.
Collapse
Affiliation(s)
- Valentina Bonetto
- Department of Science and Technology Innovation, University of Eastern Piedmont (UPO), Alessandria (AL), Italy
| | - Valeria Magnelli
- Department of Science and Technology Innovation, University of Eastern Piedmont (UPO), Alessandria (AL), Italy
| | - Maurizio Sabbatini
- Department of Science and Technology Innovation, University of Eastern Piedmont (UPO), Alessandria (AL), Italy
| | - Flavia Caprì
- Department of Science and Technology Innovation, University of Eastern Piedmont (UPO), Alessandria (AL), Italy
| | - Jack J. W. A. van Loon
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam Movement Sciences & Amsterdam Bone Center (ABC), Amsterdam UMC Location Vrije Universiteit Amsterdam & Academic Center for Dentistry Amsterdam (ACTA), Amsterdam, The Netherlands
- Life Support and Physical Sciences Section (TEC-MMG), European Space Agency (ESA), European Space Research and Technology Centre (ESTEC), Noordwijk, The Netherland
| | - Sara Tavella
- Department of Experimental Medicine, University of Genoa, Genoa (GE), Italy
| | - Maria Angela Masini
- Department of Science and Technology Innovation, University of Eastern Piedmont (UPO), Alessandria (AL), Italy
| |
Collapse
|
16
|
Hong Y, Zhou X, Li Q, Chen J, Wei Y, Wang S, Zheng X, Zhao J, Yu C, Pei J, Zhang J, Long C, Shen L, Wu S, Wei G. Wnt10a downregulation contributes to MEHP-induced disruption of self-renewal and differentiation balance and proliferation inhibition in GC-1 cells: Insights from multiple transcriptomic profiling. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 333:122091. [PMID: 37364752 DOI: 10.1016/j.envpol.2023.122091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/06/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023]
Abstract
Di (2-ethylhexyl) phthalate (DEHP), one of phthalic acid esters, has been widely used in daily products. Its main metabolite, mono (2-ethylhexyl) phthalate (MEHP) was reported to possess higher testicular toxicity than DEHP. To explore the precise mechanism in MEHP-induced testis damage, multiple transcriptomic sequencing was employed in spermatogonia cell line GC-1 cells treated with MEHP (0, 100, and 200 μM) for 24 h. Integrative omics analysis and empirical validation revealed that Wnt signaling pathway was downregulated and wnt10a, one of hub genes, may be the key player in this process. Similar results were observed in DEHP-exposed rats. MEHP-induced disturbance of self-renewal and differentiation was dose-dependent. Moreover, self-renewal proteins were downregulated; the differentiation level was stimulated. Meanwhile, GC-1 proliferation was decreased. Stable transformation strain of wnt10a overexpression GC-1 cell line constructed from lentivirus was utilized in this study. The upregulation of Wnt10a significantly reversed the dysfunction of self-renewal and differentiation and promoted the cell proliferation. Finally, retinol, predicted to be useful in CONNECTIVITY MAP (cMAP), failed to rescue the damage caused by MEHP. Cumulatively, our findings revealed that the downregulation of Wnt10a induced the imbalance of self-renew and differentiation, and inhibition of cell proliferation in GC-1 cells after MEHP exposure.
Collapse
Affiliation(s)
- Yifan Hong
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Xiazhu Zhou
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Qi Li
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Jing Chen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Yuexin Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Siyuan Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Xiangqin Zheng
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Jie Zhao
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Chengjun Yu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Jun Pei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Jie Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Chunlan Long
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Lianju Shen
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Shengde Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China.
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| |
Collapse
|
17
|
Huang X, Gao Y, Zhang Y, Wang J, Zheng N. Strontium Chloride Improves Reproductive Function and Alters Gut Microbiota in Male Rats. Int J Mol Sci 2023; 24:13922. [PMID: 37762223 PMCID: PMC10531462 DOI: 10.3390/ijms241813922] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Strontium (Sr) is an essential trace element in the human body and plays an important role in regulating male reproductive health. Recent studies have shown that gut flora plays a key role in maintaining spermatogenesis, as well as testicular health, through the gut-testis axis. At present, it is unclear whether gut microbiota can mediate the effects of Sr on sperm quality, and what the underlying mechanisms may be. We investigated the effects of different concentrations of strontium chloride (SrCl2) solutions (0, 50, 100, and 200 mg/kg BW) on reproductive function and gut microbiota in male Wistar rats (6-8 weeks, 250 ± 20 g). All the animals were euthanized after 37 days of treatment. The Sr-50 group significantly increased sperm concentration, sperm motility, and sperm viability in rats. After Sr treatment, serum and testicular testosterone (T) and Sr levels increased in a dose-dependent manner with increasing Sr concentration. At the same time, we also found that testicular marker enzymes (ACP, LDH) and testosterone marker genes (StAR, 3β-HSD, and Cyp11a1) increased significantly in varying degrees after Sr treatment, while serum NO levels decreased significantly in a dose-dependent manner. Further investigation of intestinal flora showed that SrCl2 affected the composition of gut microbiome, but did not affect the richness and diversity of gut microbiota. Sr treatment reduced the number of bacteria with negative effects on reproductive health, such as Bacteroidetes, Tenericutes, Romboutsia, Ruminococcaceae_UCG_014, Weissella, and Eubacterium_coprostanoligenes_group, and added bacteria with negative effects on reproductive health, such as Jeotgalicoccus. To further explore the Sr and the relationship between the gut microbiota, we conducted a Spearman correlation analysis, and the results showed that the gut microbiota was closely correlated with Sr content in serum and testicular tissue, sex hormone levels, and testicular marker enzymes. Additionally, gut microbiota can also regulate each other and jointly maintain the homeostasis of the body's internal environment. However, we found no significant correlation between intestinal flora and sperm quality in this study, which may be related to the small sample size of our 16S rDNA sequencing. In conclusion, the Sr-50 group significantly increased T levels and sperm quality, and improved the levels of testicular marker enzymes and testosterone marker genes in the rats. Sr treatment altered the gut flora of the rats. However, further analysis of the effects of gut microbiota in mediating the effects of SrCl2 on male reproductive function is needed. This study may improve the current understanding of the interaction between Sr, reproductive health, and gut microbiota, providing evidence for the development of Sr-rich foods and the prevention of male fertility decline.
Collapse
Affiliation(s)
- Xulai Huang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Laboratory of Quality and Safety Risk Assessment for Dairy Products, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yanan Gao
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Laboratory of Quality and Safety Risk Assessment for Dairy Products, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yangdong Zhang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Laboratory of Quality and Safety Risk Assessment for Dairy Products, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jiaqi Wang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Laboratory of Quality and Safety Risk Assessment for Dairy Products, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Laboratory of Quality and Safety Risk Assessment for Dairy Products, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
18
|
Zhang Y, Liu Y, Teng Z, Wang Z, Zhu P, Wang Z, Liu F, Liu X. Human umbilical cord mesenchymal stem cells (hUC-MSCs) alleviate paclitaxel-induced spermatogenesis defects and maintain male fertility. Biol Res 2023; 56:47. [PMID: 37574561 PMCID: PMC10424423 DOI: 10.1186/s40659-023-00459-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/28/2023] [Indexed: 08/15/2023] Open
Abstract
Chemotherapeutic drugs can cause reproductive damage by affecting sperm quality and other aspects of male fertility. Stem cells are thought to alleviate the damage caused by chemotherapy drugs and to play roles in reproductive protection and treatment. This study aimed to explore the effects of human umbilical cord mesenchymal stem cells (hUC-MSCs) on alleviating paclitaxel (PTX)-induced spermatogenesis and male fertility defects. An in vivo PTX-induced mice model was constructed to evaluate the reproductive toxicity and protective roles of hUC-MSCs in male fertility improvement. A 14 day PTX treatment regimen significantly attenuated mice spermatogenesis and sperm quality, including affecting spermatogenesis, reducing sperm counts, and decreasing sperm motility. hUC-MSCs treatment could significantly improve sperm functional indicators. Mating experiments with normal female mice and examination of embryo development at 7.5 days post-coitum (dpc) showed that hUC-MSCs restored male mouse fertility that was reduced by PTX. In IVF experiments, PTX impaired sperm fertility and blastocyst development, but hUC-MSCs treatment rescued these indicators. hUC-MSCs' protective role was also displayed through the increased expression of the fertility-related proteins HSPA2 and HSPA4L in testes with decreased expression in the PTX-treated group. These changes might be related to the PTX-induced decreases in expression of the germ cell proliferation protein PCNA and the meiosis proteins SYCP3, MLH1, and STRA8, which were restored after hUC-MSCs treatment. In the PTX-treated group, the expression of testicular antioxidant proteins SIRT1, NRF2, CAT, SOD1, and PRDX6 was significantly decreased, but hUC-MSCs could maintain these expressions and reverse PTX-related increases in BAX/BCL2 ratios. hUC-MSCs may be a promising agent with antioxidant and anti-apoptosis characteristics that can maintain sperm quality following chemotherapy treatment.
Collapse
Affiliation(s)
- YuSheng Zhang
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - YaNan Liu
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Zi Teng
- Shandong Stem Cell Engineering Technology Research Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - ZeLin Wang
- Shandong Stem Cell Engineering Technology Research Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Peng Zhu
- Shandong Stem Cell Engineering Technology Research Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - ZhiXin Wang
- Shandong Stem Cell Engineering Technology Research Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - FuJun Liu
- School of Bioscience and Technology, Weifang Medical University, Weifang, China.
- Shandong Stem Cell Engineering Technology Research Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China.
| | - XueXia Liu
- Shandong Stem Cell Engineering Technology Research Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China.
| |
Collapse
|
19
|
Zhang G, Devo P, O'Leary VB, Ovsepian SV. Ageing perspective on cognitive outcomes from reproductive hormone adjustments. Heliyon 2023; 9:e19050. [PMID: 37664737 PMCID: PMC10470197 DOI: 10.1016/j.heliyon.2023.e19050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/20/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023] Open
Abstract
In addition to primary reproductive functions, gonadal hormones play an important role in an array of neural mechanisms across the human lifespan. The ageing-related decline in their activity has been linked to the deterioration of cognitive functions in otherwise healthy women, associated with menopause transition, contributing to higher incidents of post-menopause dementia. Given the growing utility of gonadal steroids for birth control, as well as for compensatory treatment of menopause and oophorectomy symptoms, and adjuvant transgender therapy, their long-term effects on neural mechanisms warrant comprehensive assessment. In this article, we present an ageing perspective on the cognitive outcomes from contraceptive and replacement therapeutic use of gonadal hormones and discuss their effects on the risk of developing Alzheimer's and Parkinson's dementia. Despite rising data supporting the ameliorative effects of reproductive hormones on cognitive facilities, their impact varies depending on study design and type of intervention, thus, implying dynamic neuro-endocrine interactions with complex compensatory mechanisms. Elucidating differential effects of reproductive hormone adjustments on cognition with underlying mechanisms is expected not only to shed light on important aspects of brain ageing and dementia but to facilitate their use in personalized medicine with improved safety margins and therapeutic outcomes.
Collapse
Affiliation(s)
- Grace Zhang
- Faculty of Health Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
- Bader College, Herstmonceux Castle, Hailsham, BN27 1RN, United Kingdom
| | - Perry Devo
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, ME4 4TB, United Kingdom
| | - Valerie B. O'Leary
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruská 87, 10000 Prague, Czech Republic
| | - Saak V. Ovsepian
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, ME4 4TB, United Kingdom
| |
Collapse
|
20
|
Akhigbe RE, Afolabi OA, Ajayi AF. L-Arginine abrogates maternal and pre-pubertal codeine exposure-induced impaired spermatogenesis and sperm quality by modulating the levels of mRNA encoding spermatogenic genes. Front Endocrinol (Lausanne) 2023; 14:1180085. [PMID: 37529606 PMCID: PMC10390314 DOI: 10.3389/fendo.2023.1180085] [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: 04/20/2023] [Accepted: 06/13/2023] [Indexed: 08/03/2023] Open
Abstract
Introduction Although, codeine has been demonstrated to lower sperm quality; the effects of maternal and prepubertal codeine exposure on male offspring is yet to be reported. In addition, the effect of arginine on codeine-induced decline in sperm quality has not been explored. This study investigated the impact of maternal and prepubertal codeine exposure on spermatogenesis and sperm quality in F1 male Wistar rats to study the effect that codeine may have during recreational use in humans. Also, the effect of arginine supplementation on codeine-induced alteration in spermatogenesis and sperm quality was evaluated. Methods Female rats were treated with either 0.5 ml distilled water or codeine orally for eight weeks, and then mated with male rats (female:male, 2:1). The F1 male offsprings of both cohorts were weaned at 3 weeks old and administered distilled water, codeine, arginine, or codeine with arginine orally for eight weeks. Results Prepubertal codeine exposure in rats whose dams (female parents) were exposed to codeine delayed puberty and reduced the weight at puberty. Prepubertal codeine exposure exacerbated maternal codeine exposure-induced reduced total and daily spermatid production, sperm count, sperm motility, and normal sperm form, as well as impaired sperm plasma membrane integrity and increased not intact acrosome and damaged sperm DNA integrity. These perturbations were accompanied by a decrease in mRNA levels encoding spermatogenic genes, testicular testosterone and androgen receptor (AR) concentrations, and upregulation of sperm 8-hydroxydeoxyguanosine (8OHdG). Prepubertal arginine supplementation mitigated codeine-induced alterations. Discussion This study provides novel experimental evidence that maternal and prepubertal codeine exposure reprogramed spermatogenesis and sperm quality of male FI generation by decreasing mRNA levels encoding spermatogenic genes and AR via oxidative stress-mediated signaling, which was abrogated by prepubertal arginine supplementation.
Collapse
Affiliation(s)
- Roland Eghoghosoa Akhigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | - Oladele A. Afolabi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Ayodeji Folorusho Ajayi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
21
|
Yan Y, Tian J, Wang Y, Li Y, Zhang C, Zhang S, Lin P, Peng R, Zhao C, Zhuang L, Lai B, Zhou L, Zhang G, Li H. Transcriptomic Heterogeneity of Skin Across Different Anatomic Sites. J Invest Dermatol 2023; 143:398-407.e5. [PMID: 36122800 DOI: 10.1016/j.jid.2022.08.053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/05/2022] [Accepted: 08/26/2022] [Indexed: 02/05/2023]
Abstract
Multiomic studies, including RNA sequencing, single-cell RNA sequencing, and epigenomics, can provide insight into the connection between anatomically heterogeneous gene expression profile of the skin and dermatoses-predisposed sites, in which RNA sequencing is essential. Therefore, in this study, 159 skin samples collected mainly from discarded normal skin tissue during surgical treatment for benign skin tumors were used for RNA sequencing. On the basis of cluster analysis, the skin was divided into four regions, with each region showing specific physiological characteristics through differentially expressed gene analysis. The results showed that the head and neck region, perineum, and palmoplantar area were closely associated with lipid metabolism, hormone metabolism, blood circulation, and related neural regulation, respectively. Transcription factor enrichment indicated that different regions were associated with the development of adjacent tissues. Specifically, the head and neck region, trunk and extremities, perineum, and palmoplantar area were associated with the central nervous, axial, urogenital, and vascular systems, respectively. The results were imported into an open website (https://dermvis.github.io/) for retrieval. Our transcriptomic data elucidated that human skin exhibits transcriptomic heterogeneity reflecting physiological and developmental variation at different anatomic sites and provided guidance for further studies on skin development and dermatoses predisposed sites.
Collapse
Affiliation(s)
- Yicen Yan
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China
| | - Jie Tian
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Yang Wang
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China
| | - Yurong Li
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China
| | - Chong Zhang
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China
| | - Shenxi Zhang
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China
| | - Pingping Lin
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China
| | - Rui Peng
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China
| | - Chunxia Zhao
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China
| | - Le Zhuang
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
| | - Binbin Lai
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; Institute of Medical Technology, Peking University Health Science Center, Beijing, China; Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Liang Zhou
- National Institute of Health Data Science, Peking University, Beijing, China
| | - Guohong Zhang
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; Pathology Department, Shantou University Medical College, Guangdong, China
| | - Hang Li
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China.
| |
Collapse
|
22
|
Abdelfattah MG, Hussein MT, Ragab SMM, Khalil NSA, Attaai AH. The effects of Ginger (Zingiber officinale) roots on the reproductive aspects in male Japanese Quails (Coturnix coturnix japonica). BMC Vet Res 2023; 19:34. [PMID: 36737791 PMCID: PMC9896824 DOI: 10.1186/s12917-023-03576-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 01/13/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The Japanese quail is considered one of the most significant species in the poultry industry. However, the high male-to-female ratio results in the aggressive behavior of males. Dietary strategies that improve the properties of semen could reduce the number of males required to maintain optimal fertility and reduce aggressive behavior. Therefore, this study aims to provide insight into the possible improving efm fect of ginger roots on the reproductive aspects of Japanese male quails. RESULTS To achieve this objective, powder of Ginger roots was administrated to 2 groups of quails (10, and 15 g/Kg feed) from 7 days until 70 days of age. Some males were reared singly in cages (n = 40 for each group) to assess sperm quality and other males (n = 32 for each group) were raised with females to assess fertility and sperm-egg penetration. Additionally, biochemical tests and histological examination were also performed. When compared to the control group, dietary inclusion of Ginger at a dose of 15 g caused more improvement in ejaculate volume, sperm concentration, motility, viability and sperm-egg penetration. Whereas, the motility and fertility percentages of sperms were equipotent in both doses. Dose-dependent increases were found in the cloacal gland area and volume, as well as foam production and weight. Both doses resulted in a significant reduction in plasma total cholesterol along with an elevation cin plasma testosterone and lipid peroxides. The comparison between all groups concerning nitric oxide, catalase, superoxide dismutase, and total antioxidant capacity revealed the absence of significant difference. Morphologically, the diameter of the seminiferous tubules and the height of germinal epithelium significantly increased especially in the higher dose of Ginger. CONCLUSIONS Ginger roots especially at a dose of 15 gm/kg feed was effective in improving male reproductive performance. These findings are of utmost importance in encouraging the addition of Ginger roots in ration formulation in male quails.
Collapse
Affiliation(s)
| | - Manal T. Hussein
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Sohair M. M. Ragab
- Laboratory of Physiology, Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Nasser S. Abou Khalil
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Abdelraheim H. Attaai
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
- Department of Anatomy and Histology, School of Veterinary Medicine, Badr University, New Nasser City, West of Assiut, Assiut, Egypt
| |
Collapse
|
23
|
Saikia Q, Hazarika A, Kalita JC. Isoliquiritigenin ameliorates paroxetine-induced sexual dysfunction in male albino mice. Reprod Toxicol 2023; 117:108341. [PMID: 36740106 DOI: 10.1016/j.reprotox.2023.108341] [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: 09/26/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Paroxetine (PRX), a widely prescribed antidepressant, often leads to sexual dysfunction. The available management options such as sildenafil (SDF), are associated with side effects. The present study investigates the fertility-boosting properties of isoliquiritigenin (ISL) on PRX-induced sexual dysfunction in male mice. We allocated fertile mice into six different groups (n = 5): group I- DMSO; group II- PRX; group III- co-administered PRX and SDF; group IV- ISL alone; group V- co-administered PRX and ISL (low dose); and, group VI- co-administered PRX and ISL (high dose). 14 days post treatment, animals were sacrificed, and the weights of the testis and epididymis were evaluated. Furthermore, sperm parameters, testicular and epididymal antioxidant levels, serum testosterone and nitric oxide (NO) levels, histoarchitecture of testis and epididymis, and markers of cellular toxicity were assessed. Results revealed that the PRX administration reduced organ weights, sperm count, intact acrosome, catalase (CAT), superoxide dismutase (SOD), glutathione (GSH), serum testosterone, and NO levels, and increased sperm abnormalities and MDA levels (a biomarker for lipid peroxidation). Additionally, we observed damage in the testis and epididymis. The toxicity biomarker study revealed a higher concentration of SGOT, SGPT, and ALP enzymes in the PRX-treated group. However, the co-administration of PRX with ISL ameliorated the adverse effect of PRX on the parameters mentioned above. The PRX+ISL (high) results were almost at par with the PRX+SDF group. The group that received ISL alone showed overall improvements. In conclusion, our comprehensive panel of tests indicates that ISL could be helpful in managing sexual dysfunction.
Collapse
Affiliation(s)
- Queen Saikia
- Department of Zoology, Gauhati University, Guwahati, Assam, India.
| | - Ajit Hazarika
- Tyagbir Hem Baruah College, Jamugurihat, Sonitpur, Assam, India
| | | |
Collapse
|
24
|
Concessao PL, Bairy KL, Raghavendra AP. Ameliorating effect of Mucuna pruriens seed extract on sodium arsenite-induced testicular toxicity and hepato-renal histopathology in rats. Vet World 2023; 16:82-93. [PMID: 36855363 PMCID: PMC9967728 DOI: 10.14202/vetworld.2023.82-93] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 11/23/2022] [Indexed: 01/13/2023] Open
Abstract
Background and Aim A significant cause of arsenic poisoning is polluted groundwater. Arsenic poisoning results in the suppression of spermatogenesis and the liver and kidneys are vulnerable to the toxic effects as well. Mucuna pruriens has been identified to have fertility-enhancing and anti-lipid peroxidation properties. Based on these properties of M. pruriens, this study aimed to investigate the efficacy of M. pruriens seed extract in reducing sodium arsenite-induced testicular impairment and hepato-renal histopathology in rats. Materials and Methods The study was divided into two groups; short-term (45 days) and long-term (90 days) treatment groups and each group was divided into nine subgroups. Subgroups 1 and 2 served as normal and N-acetyl cysteine (NAC) controls, respectively. Subgroups 3-9 received sodium arsenite in the drinking water (50 mg/L). Subgroup-4 received NAC (210 mg/kg body weight [BW]) orally once daily. Subgroups 5-7 received aqueous seed extract of M. pruriens (350, 530, and 700 mg/kg BW, respectively) orally once daily. Subgroups 8 and 9 received a combination of NAC and aqueous seed extract (350 and 530 mg/kg BW, respectively) orally once daily. Following the treatment, animals were sacrificed and sperm parameters and DNA damage were evaluated. Testis, liver, and kidneys were analyzed for histopathology. Results Sodium arsenite-induced a significant reduction in sperm parameters and increase in the abnormal architecture of spermatozoa. Histology revealed tissue necrosis. The M. pruriens seed extract ameliorated the damaging effects of sodium arsenite with respect to tissue architecture and sperm parameters when coadministered. Conclusion Mucuna pruriens has beneficial effects against the deleterious effects of sodium arsenite on various tissues. Thus, M. pruriens (530 and 700 mg/kg BW) supplementation would reduce the adverse changes observed with sodium arsenite exposure.
Collapse
Affiliation(s)
- Preethi Lavina Concessao
- Department of Physiology, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India,Corresponding author: Preethi Lavina Concessao, e-mail: Co-authors: KLB: , APR:
| | - Kurady Laxminarayana Bairy
- Department of Pharmacology, RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Archana Parampalli Raghavendra
- Department of Physiology, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
25
|
XueXia L, YaNan L, Zi T, YuSheng Z, ZeLin W, Peng Z, MeiNa X, FuJun L. Di-2-ethylhexyl phthalate (DEHP) exposure induces sperm quality and functional defects in mice. CHEMOSPHERE 2023; 312:137216. [PMID: 36372335 DOI: 10.1016/j.chemosphere.2022.137216] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 06/16/2023]
Abstract
Di-2-ethylhexyl phthalate (DEHP) harms mammalian testis development, yet the specific mechanism of its effect on sperm quality and function is unclear. In this study, male mice were administrated DEHP (200 mg/kg/day) via intragastric (i.g.) injection for 35 days. The sperm quality and function of DEHP-exposed mice were evaluated. DEHP exposure reduced the relative testis weight and serum testosterone levels. In addition, sperm count and motility parameters decreased significantly, which led to reduced sperm fertility characterized by reduced acrosome reaction rate, sperm-egg binding capacity and blastocyte formation. DEHP exposure decreased anti-oxidant indicators and the expressions of Cat, Sod1, Prdx6 and Sirt1 in the testis. DEHP-exposure also resulted in decreased proliferating cell nuclear antigen (PCNA) expression in mice testis, as well as the dose-dependent inhibition of the proliferation of GC-1 and GC-2 cells. These phenotypes may be related to increased cell apoptosis characterized by BAX/BCL2 and P53 up-regulation. DEHP exposure resulted in the down-regulation of SIRT1 and p-AKT in mice testis and decreased levels of GC-1and GC-2 cells. DEHP co-incubation with sperm in vitro resulted in decreased tyrosine phosphorylation and progressive motility, as well as p-AKT expression in capacitated sperm. Differential sperm proteomics identified 495 differentially expressed proteins, including 257 proteins down-regulated in the DEHP-exposure group. Bioinformatics analysis showed that proteins involved in sperm-egg interaction and fertilization processes were significantly down-regulated. Pathway analysis demonstrated that the adhesion pathway was enriched in down-regulated proteins, while the pathway associated with ribosomes was enriched in up-regulated proteins. Conclusively, DEHP exposure impaired male fertility by affecting sperm quality and function, and a pathway mediating the DEHP-induced decline in sperm quality and function was identified. The study provides additional information for understanding the molecular mechanisms of DEHP exposure and its effects on male reproduction.
Collapse
Affiliation(s)
- Liu XueXia
- Shandong Stem Cell Engineering Technology Research Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China; School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Liu YaNan
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Teng Zi
- Shandong Stem Cell Engineering Technology Research Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Zhang YuSheng
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Wang ZeLin
- Shandong Stem Cell Engineering Technology Research Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Zhu Peng
- Shandong Stem Cell Engineering Technology Research Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Xie MeiNa
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Liu FuJun
- Shandong Stem Cell Engineering Technology Research Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China; School of Bioscience and Technology, Weifang Medical University, Weifang, China.
| |
Collapse
|
26
|
Yu Y, Zhang D, Xu J, Zhang D, Yang L, Xia R, Wang SL. Adolescence is a sensitive period for acrylamide-induced sex hormone disruption: Evidence from NHANES populations and experimental mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114413. [PMID: 36516620 DOI: 10.1016/j.ecoenv.2022.114413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 06/17/2023]
Abstract
Acrylamide (AA) is widely contaminated in environment and diet. However, the association of AA and sex hormones has rarely been investigated, especially in adolescents, a period of particular susceptibility to sex hormone disruption. In this study, survey-weighted multivariate linear regression models were conducted to determine the association between AA Hb biomarkers [HbAA and glycidamide (HbGA)] and sex hormones [total testosterone (TT) and estradiol (E2)] in a total of 3268 subjects from National Health and Nutrition Examination Survey (NHANES) 2013-2016 waves. Additionally, adult and pubertal mice were treated with AA to assess the effect of AA on sex hormones and to explore the potential mechanisms. Among all the subjects, significant negative patterns for HbGA and sex hormones were identified only in youths (6-19 years old), with the lowest β being - 0.53 (95% CI: -0.80 to -0.26) for TT in males and - 0.58 (95% CI: -0.93 to -0.23) for E2 in females. Stratified analysis further revealed significant negative associations between HbGA and sex hormones in adolescents, with the lowest β being - 0.58 (95% CI: -1.02 to -0.14) for TT in males and - 0.54 (95% CI: -1.03 to -0.04) for E2 in females, while there were no significant differences between children or late adolescents. In mice, the levels of TT and E2 were dramatically reduced in AA-treated pubertal mice but not in adult mice. AA disturbed the expression of genes in the hypothalamic-pituitary-gonadal (HPG) axis, induced apoptosis of hypothalamus-produced gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus and reduced serum and hypothalamic GnRH levels in pubertal mice. Our study indicates AA could reduce TT and E2 levels by injuring GnRH neurons and disrupting the HPG axis in puberty, which manifested as severe endocrine disruption on adolescents. Our findings reinforce the idea that adolescence is a vulnerable stage in AA-induced sex hormone disruption.
Collapse
Affiliation(s)
- Yongquan Yu
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China; State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Di Zhang
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Jiayi Xu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Daiwei Zhang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Liu Yang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Rong Xia
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Shou-Lin Wang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China.
| |
Collapse
|
27
|
Bhattacharya I, Dey S, Banerjee A. Revisiting the gonadotropic regulation of mammalian spermatogenesis: evolving lessons during the past decade. Front Endocrinol (Lausanne) 2023; 14:1110572. [PMID: 37124741 PMCID: PMC10140312 DOI: 10.3389/fendo.2023.1110572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Spermatogenesis is a multi-step process of male germ cell (Gc) division and differentiation which occurs in the seminiferous tubules of the testes under the regulation of gonadotropins - Follicle Stimulating Hormone (FSH) and Luteinising hormone (LH). It is a highly coordinated event regulated by the surrounding somatic testicular cells such as the Sertoli cells (Sc), Leydig cells (Lc), and Peritubular myoid cells (PTc). FSH targets Sc and supports the expansion and differentiation of pre-meiotic Gc, whereas, LH operates via Lc to produce Testosterone (T), the testicular androgen. T acts on all somatic cells e.g.- Lc, PTc and Sc, and promotes the blood-testis barrier (BTB) formation, completion of Gc meiosis, and spermiation. Studies with hypophysectomised or chemically ablated animal models and hypogonadal (hpg) mice supplemented with gonadotropins to genetically manipulated mouse models have revealed the selective and synergistic role(s) of hormones in regulating male fertility. We here have briefly summarized the present concept of hormonal control of spermatogenesis in rodents and primates. We also have highlighted some of the key critical questions yet to be answered in the field of male reproductive health which might have potential implications for infertility and contraceptive research in the future.
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology, School of Biological Science, Central University of Kerala, Kasaragod, Kerala, India
- *Correspondence: Arnab Banerjee, ; Indrashis Bhattacharya,
| | - Souvik Dey
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Arnab Banerjee
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS) Pilani, Goa, India
- *Correspondence: Arnab Banerjee, ; Indrashis Bhattacharya,
| |
Collapse
|
28
|
Alemany M. The Roles of Androgens in Humans: Biology, Metabolic Regulation and Health. Int J Mol Sci 2022; 23:11952. [PMID: 36233256 PMCID: PMC9569951 DOI: 10.3390/ijms231911952] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Androgens are an important and diverse group of steroid hormone molecular species. They play varied functional roles, such as the control of metabolic energy fate and partition, the maintenance of skeletal and body protein and integrity and the development of brain capabilities and behavioral setup (including those factors defining maleness). In addition, androgens are the precursors of estrogens, with which they share an extensive control of the reproductive mechanisms (in both sexes). In this review, the types of androgens, their functions and signaling are tabulated and described, including some less-known functions. The close interrelationship between corticosteroids and androgens is also analyzed, centered in the adrenal cortex, together with the main feedback control systems of the hypothalamic-hypophysis-gonads axis, and its modulation by the metabolic environment, sex, age and health. Testosterone (T) is singled out because of its high synthesis rate and turnover, but also because age-related hypogonadism is a key signal for the biologically planned early obsolescence of men, and the delayed onset of a faster rate of functional losses in women after menopause. The close collaboration of T with estradiol (E2) active in the maintenance of body metabolic systems is also presented Their parallel insufficiency has been directly related to the ravages of senescence and the metabolic syndrome constellation of disorders. The clinical use of T to correct hypoandrogenism helps maintain the functionality of core metabolism, limiting excess fat deposition, sarcopenia and cognoscitive frailty (part of these effects are due to the E2 generated from T). The effectiveness of using lipophilic T esters for T replacement treatments is analyzed in depth, and the main problems derived from their application are discussed.
Collapse
Affiliation(s)
- Marià Alemany
- Facultat de Biologia, Universitat de Barcelona, Av. Diagonal, 635, 08028 Barcelona, Catalonia, Spain;
- Institut de Biomedicina, Universitat de Barcelona, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
29
|
Androgen Receptor Gene CAG Repeat Length Varies and Affects Semen Quality in an Ethnic-Specific Fashion in Young Men from Russia. Int J Mol Sci 2022; 23:ijms231810594. [PMID: 36142533 PMCID: PMC9505661 DOI: 10.3390/ijms231810594] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Male infertility is a multi-factorial and multi-genetic disorder, and the prevalence of male infertility in the world is estimated at 5–35%. The search for the causes of male infertility allowed for identifying a number of genetic factors including a single X-linked gene of the androgen receptor (AR), and some of its alleles are assumed to negatively affect male fertility. Our aim was (1) to study the variability of the length of CAG repeats of the AR gene and possible associations in the AR CAG genetic variants with semen quality and reproductive hormone levels in a population-based cohort of men and (2) to estimate distributions of AR CAG repeat alleles and associations with semen parameters in different ethnic subgroups. The cohort of 1324 young male volunteers of different ethnicities (median age 23.0 years) was recruited from the general population of five cities of the Russian Federation, regardless of their fertility status. Semen quality (sperm concentration, motility and morphology), reproductive hormone levels (testosterone, estradiol, LH, FSH and inhibin B) and trinucleotide (CAG) n repeat polymorphism in exon 1 of the AR gene were evaluated. The semen samples were analyzed according to the WHO laboratory manual (WHO, 2010), serum hormones were measured by enzyme immunoassay, and the AR CAG repeat length was analyzed by direct sequencing of leukocyte DNA. The median AR CAG repeat length in men of our multi-ethnic population was 23 (range 6–39). In the entire study population, a significant difference (p ≤ 0.05) was found in the frequency distribution and the mean values for the CAG repeat length between the groups with normal (23.2 ± 3.3) and impaired semen quality (23.9 ± 3.2). Additionally, we demonstrated that the total sperm count, sperm concentration, progressive motility and normal morphology were lower in the category of long CAG repeats (CAG ≥ 25) compared with those in the category of short CAG repeats (CAG ≤ 19); however, hormonal parameters did not differ between the long and short CAG categories, with the exception of estradiol. Significant differences were observed in the AR CAG repeat length between the most common ethnic cohorts of Slavs (Caucasians), Buryats (Asians), and Yakuts (Asians). The Buryats and Yakuts had a higher number of CAG repeats than the Slavs (medians: Slavs—23; Buryats—24; Yakuts—25). The range of alleles differed among ethnicities, with the Slavs having the largest range (7–36 repeats, 24 alleles total), the Yakuts having the smallest range (18–32 repeats, 14 alleles total) and the Buryats having the middle range (11–39 repeats, 20 alleles total). The longer CAG repeats were associated with an impaired semen quality within the Slavic (CAG ≥ 25) and Buryat (CAG ≥ 28) groups, but this effect was not found in Yakuts. Hormonal parameters did not differ between the three CAG repeat categories in men of all ethnic groups. This is the largest Russian study of the distribution of AR CAG repeats and the search for association between length of AR CAG repeat tract and impaired spermatogenesis in men from the general population. Our results confirmed the association of longer CAG repeats with a risk of impaired semen quality, but this association can be modified by ethnic origin. Identification of the number of AR CAG repeats can be an effective tool to assess the risk of male subfertility and the control of androgen hormone therapy of reproductive diseases.
Collapse
|
30
|
Wang JM, Li ZF, Yang WX, Tan FQ. Follicle-stimulating hormone signaling in Sertoli cells: a licence to the early stages of spermatogenesis. Reprod Biol Endocrinol 2022; 20:97. [PMID: 35780146 PMCID: PMC9250200 DOI: 10.1186/s12958-022-00971-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/20/2022] [Indexed: 11/10/2022] Open
Abstract
Follicle-stimulating hormone signaling is essential for the initiation and early stages of spermatogenesis. Follicle-stimulating hormone receptor is exclusively expressed in Sertoli cells. As the only type of somatic cell in the seminiferous tubule, Sertoli cells regulate spermatogenesis not only by controlling their own number and function but also through paracrine actions to nourish germ cells surrounded by Sertoli cells. After follicle-stimulating hormone binds to its receptor and activates the follicle-stimulating hormone signaling pathway, follicle-stimulating hormone signaling will establish a normal Sertoli cell number and promote their differentiation. Spermatogonia pool maintenance, spermatogonia differentiation and their entry into meiosis are also positively regulated by follicle-stimulating hormone signaling. In addition, follicle-stimulating hormone signaling regulates germ cell survival and limits their apoptosis. Our review summarizes the aforementioned functions of follicle-stimulating hormone signaling in Sertoli cells. We also describe the clinical potential of follicle-stimulating hormone treatment in male patients with infertility. Furthermore, our review may be helpful for developing better therapies for treating patients with dysfunctional follicle-stimulating hormone signaling in Sertoli cells.
Collapse
Affiliation(s)
- Jia-Ming Wang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhen-Fang Li
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Fu-Qing Tan
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|
31
|
Liu B, Zheng S, Tang H, Liu Q, Li H, Gao B, Zhao X, Sun F. Highly sensitive detection of free testosterone assisted by magnetic nanobeads and gap-enhanced SERS nanotags. Colloids Surf B Biointerfaces 2022; 214:112460. [PMID: 35298951 DOI: 10.1016/j.colsurfb.2022.112460] [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: 09/27/2021] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 01/03/2023]
Abstract
The quantitative determination of trace free testosterone (FT) is of great significance for the diagnosis of androgen-related endocrine diseases. Herein, a fascinating detection protocol was developed for highly sensitive FT analysis through a competitive immunoassay mechanism, which was composed of magnetic nanobeads (MNBs) and gap-enhanced surface enhanced Raman scattering (SERS) nanotags. With the MNBs as detection carriers, trace FT could be enriched by simple magnetic separation. The SERS nanotag constructed with silver-gold core-shell nanoparticle was acted as quantitative label, and Raman indicators were located at the interface between silver core and gold shell. It is demonstrated that the as-proposed protocol achieves high detection sensitivity for FT of 12.11 fg mL-1, and wider linear dynamic detection range (LDR) in the concentration of 100 fg mL-1 to 100 ng mL-1 with R2 value of 0.979, which is due to the enhanced Raman signal of the gap-enhanced SERS nanotag and the high surface-to-volume ratio of the MNB, respectively. Taking advantages of such sensitivity and accuracy approach, the as-developed powerful strategy presents potential applications for rapid disease diagnosis through analyzing trace levels of FT, and can also provide guidance for the exploitation of analysis project of other analytes.
Collapse
Affiliation(s)
- Bing Liu
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong 226001, China.
| | - Shiya Zheng
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Hanyu Tang
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong 226001, China
| | - Qian Liu
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong 226001, China
| | - Haitao Li
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong 226001, China
| | - Bingbing Gao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Xiangwei Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China; Southeast University Shenzhen Research Institute, Shenzhen 518000, China.
| | - Fei Sun
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong 226001, China.
| |
Collapse
|
32
|
The Role of NLRP3 Inflammasome Activation and Oxidative Stress in Varicocele-Mediated Male Hypofertility. Int J Mol Sci 2022; 23:ijms23095233. [PMID: 35563625 PMCID: PMC9102453 DOI: 10.3390/ijms23095233] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022] Open
Abstract
Varicocele (VC) is the most common abnormality identified in men evaluated for hypofertility. Increased levels of reactive oxygen species (ROS) and reduced antioxidants concentrations are key contributors in varicocele-mediated hypofertility. Moreover, inflammation and alterations in testicular immunity negatively impact male fertility. In particular, NLRP3 inflammasome activation was hypothesized to lead to seminal inflammation, in which the levels of specific cytokines, such as IL-1β and IL-18, are overexpressed. In this review, we described the role played by oxidative stress (OS), inflammation, and NLRP3 inflammasome activation in VC disease. The consequences of ROS overproduction in testis, including inflammation, lipid peroxidation, mitochondrial dysfunction, chromatin damage, and sperm DNA fragmentation, leading to abnormal testicular function and failed spermatogenesis, were highlighted. Finally, we described some therapeutic antioxidant strategies, with recognized beneficial effects in counteracting OS and inflammation in testes, as possible therapeutic drugs against varicocele-mediated hypofertility.
Collapse
|
33
|
Molecular characterization and expression patterns of nuclear androgen receptors in the ovoviviparous black rockfish Sebastes schlegelii. AQUACULTURE AND FISHERIES 2022. [DOI: 10.1016/j.aaf.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
34
|
Abstract
Male infertility contributes to 50% of all cases of infertility. The main cause is low quality and quantity of sperm. In humans, spermatogenesis starts at the beginning of puberty and lasts lifelong. It is under the control of FSH and testicular androgens, and mainly testosterone (T), and therefore requires a normal gonadotroph axis, intratesticular T production by Leydig cells and functional androgen receptors (ARs) within testicular Sertoli cells. Various clinical cases illustrate the roles of T in human spermatogenesis. Men with complete congenital hypogonadotropic hypogonadism (HH) are usually azoospermic. Treatment by exogenous testosterone injection and FSH is not able to produce sperm. However, combined treatment with FSH and hCG is effective. This example shows that intratesticular T plays a major role in spermatogenesis. Furthermore, testicular histology of men with LH receptor mutations shows Leydig cell hypoplasia/agenesis/dysplasia with conserved Sertoli cell count. The sperm count is reduced, as in males with partial inactivating mutation of the androgen receptor. Some protocols of hormonal male contraception or exogenous androgen abuse induce negative feedback in the hypothalamic pituitary axis, decreasing FSH, LH and T levels and inducing sperm defects and testicular atrophy. The time to recovery after cessation of drug abuse is around 14 months for sperm output and 38 months for sperm motility. In summary, abnormal androgen production and/or AR signaling impairs spermatogenesis in humans. The minimal level of intratesticular T for normal sperm production is a matter of debate. Interestingly, some animal models showed that completely T-independent spermatogenesis is possible, potentially through strong FSH activation. Finally, recent data suggest important roles of prenatal life and minipuberty in adult spermatogenesis.
Collapse
|
35
|
Serum Nerve Growth Factor Levels as a Predictor of Bull Candidate Semen Quality of Madura Cattle. Vet Med Int 2022; 2022:7128384. [PMID: 35449725 PMCID: PMC9017486 DOI: 10.1155/2022/7128384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/31/2022] [Indexed: 12/05/2022] Open
Abstract
Madura cattle are the germplasm of native cattle on the verge of extinction because of crossbreeding. Therefore, the present study was aimed to determine the serum nerve growth factor (NGF) concentration as a predictor of fresh ejaculate fertility parameters in Madura bull candidates. Eleven Madura bull candidates used for frozen semen production were selected for the study. Blood samples were collected using a vacutainer from the jugular vein for analyzing serum NGF and testosterone levels. Meanwhile, semen collection was conducted using an artificial vagina for sperm motility, viability, and concentration assessment. Data were analyzed to determine the correlation among variables and the linear regression of NGF concentration to other significant variables. The result showed that NGF had a significant correlation (p < 0.05) with testosterone levels, sperm motility, viability, and concentration. A significant correlation was observed between testosterone levels, sperm concentration, and sperm viability. The regression equation among significantly correlated variables was determined. For artificial insemination, suitable bull ejaculates for obtaining frozen semen should reach at least 2.12 ng/mL of NGF levels, with sperm viability, sperm concentration, and testosterone levels of more than 78.63%, 1,462.177 million/mL ejaculate, and 25.67 ng/mL, respectively. This is the first study to identify NGF as a predictor of male fertility in bull candidates of Madura cattle. Therefore, NGF levels could be used as a marker of male fertility in Madura bull cattle candidates. Thus, based on the minimum NGF levels, the ejaculate of Madura bull candidate that meets the requirements for frozen semen production could predict fertility.
Collapse
|
36
|
Aescin Protects against Experimental Benign Prostatic Hyperplasia and Preserves Prostate Histomorphology in Rats via Suppression of Inflammatory Cytokines and COX-2. Pharmaceuticals (Basel) 2022; 15:ph15020130. [PMID: 35215244 PMCID: PMC8880638 DOI: 10.3390/ph15020130] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023] Open
Abstract
Background: Benign prostatic hyperplasia (BPH) is the most common urogenital condition in aging males, while inflammation and tissue proliferation constitute the main pathophysiological factors. The adverse effects of currently available BPH medications limit patient compliance. We tested the protective effect of aescin against the development of BPH in rats. Methods: A total of 18 male Wistar rats were divided into 3 groups: control (sesame oil 1 mL/kg, s.c.); BPH (testosterone oenanthate 3 mg/kg, s.c., in sesame oil), and BPH-aescin rats (testosterone oenanthate 3 mg/kg, s.c. + aescin 10 mg/kg/day, p.o.). All treatments continued for 4 weeks. Serum and prostatic samples were harvested for biochemical and histopathological examination. Results: Induction of BPH by testosterone increased the prostate weight and prostate weight index, serum testosterone, prostate expression of inflammatory (IL-1β, TNF-α, and COX-2), and proliferative markers (PCNA and TGF-β1). Concurrent treatment with aescin decreased the testosterone-induced increase in prostatic IL-1β, TNF-α, and COX-2 expression by 47.9%, 71.2%, and 64.4%, respectively. Moreover, aescin reduced the prostatic proliferation markers TGF-β1 and PCNA by 58.3% and 71.9%, respectively, and normalized the prostate weight. Conclusion: The results of this study showed, for the first time, that aescin protected against the development of experimental BPH in rats via its anti-inflammatory and antiproliferative effects. These findings warrant further studies to clinically repurpose aescin in the management of BPH.
Collapse
|
37
|
Mohsen S, Dorsaf H, Sabrine M, Zaineb B, Olfa T, Khemais B. Reproductive toxicity of Carlina gummifera L. incense inhalation in adult male wistar rats. J Hum Reprod Sci 2022; 15:12-20. [PMID: 35494197 PMCID: PMC9053348 DOI: 10.4103/jhrs.jhrs_149_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 11/28/2022] Open
Abstract
Background: Burning incense of Carlina gummifera L. is a traditional practice in North African countries for religious or ritual intentions. Aim: The aim of this study was to investigate the effects of smoke incense of this plant on the reproductive function in adult male rats. Setting and Design: This study was conducted in the Integrated Physiology Laboratory. Materials and Methods: Plant roots were collected, dried and finely ground in powder form. Adult Wistar rats were randomly assigned to treated groups exposed daily during 60 min for 15 consecutive days to smoke incense at 2, 4 and 6 g and a control group was subjected to the same conditions in the absence of smoke. Statistical Analysis Used: Statistical analysis was performed using one-way analysis of variance followed by Tukey's multiple comparison as the post hoc test. Results: Exposure to the incense of Carlina gummifera L. seriously affected dose dependently the reproductive function in male rats. It was found that in treated groups, the testicle relative weight decreased, while those of seminal vesicles and prostate increased when compared to the untreated group. Carlina gummifera L. incense inhalation reduced the total number, viability and mobility of epididymis spermatozoa compared to control. Furthermore, incense exposure induced various histological changes in the testes, prostate and seminal vesicles, including in particular a decrease in the number of gametes in the seminiferous tubes, the reduction of prostatic secretions and the macrophagic resorption of the seminal secretions. The effect of Carlina incense on the antioxidant system was evaluated by assaying the two antioxidant enzyme activities catalase and superoxide dismutase as well as thiol group levels in the testicles. Our results showed that fumigation affected these parameters, suggesting that the morphological and functional modifications in the male reproductive system induced by Carlina gummifera L. incense may be related, in part, to the alteration of the oxidative balance in the testicle. Conclusion: Smoke incense of Carlina gummifera L. caused marked reproductive toxicity in adult male rats associated with induced oxidative stress.
Collapse
|
38
|
Wang JM, Li ZF, Yang WX. What Does Androgen Receptor Signaling Pathway in Sertoli Cells During Normal Spermatogenesis Tell Us? Front Endocrinol (Lausanne) 2022; 13:838858. [PMID: 35282467 PMCID: PMC8908322 DOI: 10.3389/fendo.2022.838858] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/01/2022] [Indexed: 01/18/2023] Open
Abstract
Androgen receptor signaling pathway is necessary to complete spermatogenesis in testes. Difference between androgen binding location in Sertoli cell classifies androgen receptor signaling pathway into classical signaling pathway and non-classical signaling pathway. As the only somatic cell type in seminiferous tubule, Sertoli cells are under androgen receptor signaling pathway regulation via androgen receptor located in cytoplasm and plasma membrane. Androgen receptor signaling pathway is able to regulate biological processes in Sertoli cells as well as germ cells surrounded between Sertoli cells. Our review will summarize the major discoveries of androgen receptor signaling pathway in Sertoli cells and the paracrine action on germ cells. Androgen receptor signaling pathway regulates Sertoli cell proliferation and maturation, as well as maintain the integrity of blood-testis barrier formed between Sertoli cells. Also, Spermatogonia stem cells achieve a balance between self-renewal and differentiation under androgen receptor signaling regulation. Meiotic and post-meiotic processes including Sertoli cell - Spermatid attachment and Spermatid development are guaranteed by androgen receptor signaling until the final sperm release. This review also includes one disease related to androgen receptor signaling dysfunction named as androgen insensitivity syndrome. As a step further ahead, this review may be conducive to develop therapies which can cure impaired androgen receptor signaling in Sertoli cells.
Collapse
|
39
|
Ruthig VA, Lamb DJ. Updates in Sertoli Cell-Mediated Signaling During Spermatogenesis and Advances in Restoring Sertoli Cell Function. Front Endocrinol (Lausanne) 2022; 13:897196. [PMID: 35600584 PMCID: PMC9114725 DOI: 10.3389/fendo.2022.897196] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/31/2022] [Indexed: 01/16/2023] Open
Abstract
Since their initial description by Enrico Sertoli in 1865, Sertoli cells have continued to enchant testis biologists. Testis size and germ cell carrying capacity are intimately tied to Sertoli cell number and function. One critical Sertoli cell function is signaling from Sertoli cells to germ cells as part of regulation of the spermatogenic cycle. Sertoli cell signals can be endocrine or paracrine in nature. Here we review recent advances in understanding the interplay of Sertoli cell endocrine and paracrine signals that regulate germ cell state. Although these findings have long-term implications for treating male infertility, recent breakthroughs in Sertoli cell transplantation have more immediate implications. We summarize the surge of advances in Sertoli cell ablation and transplantation, both of which are wedded to a growing understanding of the unique Sertoli cell niche in the transitional zone of the testis.
Collapse
Affiliation(s)
- Victor A. Ruthig
- Department of Urology, Weill Cornell Medicine, New York, NY, United States
- Sexual Medicine Lab, Weill Cornell Medicine, New York, NY, United States
| | - Dolores J. Lamb
- Department of Urology, Weill Cornell Medicine, New York, NY, United States
- Center for Reproductive Genomics, Weill Cornell Medicine, New York, NY, United States
- *Correspondence: Dolores J. Lamb,
| |
Collapse
|
40
|
Li X, Yao X, Mu C, Wang Z, Hu R, Chang Y, Wu J. Serum- and Feeder-Free Culture of Juvenile Monkey Female Germline Stem Cells and Testosterone Regulation of their Self-Renewal. Stem Cell Rev Rep 2021; 18:336-345. [PMID: 34642851 DOI: 10.1007/s12015-021-10278-9] [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] [Accepted: 10/01/2021] [Indexed: 10/20/2022]
Abstract
Female germline stem cells (FGSCs) have been found in mouse, rat, pig, sheep and human ovaries. However, there is no information on the isolation or long-term culture of FGSCs from non-human primates. Here, we identified the presence of FGSCs in the ovaries of juvenile (3-4-year-old) cynomolgus monkeys using DDX4 and Ki67 double immunofluorescence. Then, a long-term serum- and cell feeder-free culture system for these FGSCs was used to establish a cell line, and its biological characteristics were analyzed. We found that testosterone promoted self-renewal of the cells. This study confirmed for the first time the presence of FGSCs in the ovary of non-human primates. This culture system and cell line will be of great significance for research in medicine and reproductive biology.
Collapse
Affiliation(s)
- Xiaoyong Li
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoying Yao
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Chunlan Mu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China
| | - Zhaoxia Wang
- Laboratory Animal Center, Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Rong Hu
- Reproductive Medicine Center, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| | - Yan Chang
- Shanghai Innostar biotech Co. Ltd, Shanghai, 200120, China.
| | - Ji Wu
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China. .,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|