1
|
Hafez MH, Gad SB, El-Sayed YS. Quercetin-mediated restoration of high-fat diet-induced male reproductive dysfunction through modifying spermatogenesis and unraveling 3β-HSD, 17β-HSD, and StAR pathways. BMC Pharmacol Toxicol 2025; 26:90. [PMID: 40264244 PMCID: PMC12016373 DOI: 10.1186/s40360-025-00918-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/02/2025] [Indexed: 04/24/2025] Open
Abstract
PURPOSE We explored the astounding potential of quercetin (QRT) to counteract the determinantal impacts of a high-fat diet (HFD) on testicular function in rat model. The goal was to understand how QRT, and its mechanisms of action can protect testicular health from HFD. METHODS Rats were divided into four groups receiving a control diet, QRT supplement (100 mg/kg), HFD, or HFD plus QRT for 8 weeks. Afterward, assessments were conducted, and reproductive organs were analyzed for hormone levels, gene expression, and subjected to biochemical, histopathological, and immunohistochemical analyses. RESULTS The HFD caused substantial declines in testicular weight, accessory sex glands and epididymis. The HFD also negatively impacted sperm characteristics including reduced motility, viability, and count, along with impaired morphology. Additionally, the HFD decreased testosterone levels in the testes and serum, impaired antioxidant enzymes like superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase, and reduced expression of key steroid metabolism enzymes 17β-hydroxysteroid dehydrogenase (17β-HSD), 3β-hydroxysteroid dehydrogenase (3β-HSD), and steroidogenic acute regulatory protein (StAR) involved in testosterone synthesis. These changes were paired with enhanced testicular lipid peroxidation, nitrite, and the inflammatory marker tumor necrosis factor-alpha (TNF-α), reflecting the damaging еffеcts of the HFD. Examination of testicular tissues verified structural damage and significantly fewer proliferating cell nuclear antigen (PCNA)-positive spermatogenic cells in seminiferous tubules of HFD-fed group, confirming HFD's adverse еffеcts. CONCLUSION QRT supplementation was able to curb the harmful impacts of the HFD on testicular spermatogenesis and steroidogenesis through its antioxidant, anti-inflammatory and androgen boosting properties.
Collapse
Affiliation(s)
- Mona H Hafez
- Physiology Department, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 22758, Egypt
| | - Shereen B Gad
- Physiology Department, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 22758, Egypt
| | - Yasser S El-Sayed
- Veterinary Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt.
| |
Collapse
|
2
|
Ruiz-Valderrama L, Mendoza-Sánchez JE, Rodríguez-Tobón E, Arrieta-Cruz I, González-Márquez H, Salame-Méndez PA, Tarragó-Castellanos R, Cortés-Barberena E, Rodríguez-Tobón A, Arenas-Ríos E. High-Fat Diets Disturb Rat Epididymal Sperm Maturation. Int J Mol Sci 2025; 26:1850. [PMID: 40076475 PMCID: PMC11899043 DOI: 10.3390/ijms26051850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Infertility is increasingly recognized as being closely linked to obesity in humans. The successful production of fertile spermatozoa requires adequate spermatogenesis within the testis and proper spermatozoa maturation through the epididymis. This study aimed to evaluate the impact of body adiposity on male fertility, focusing on sperm parameters, epididymal sperm maturation, and sperm capacitation in Wistar rats. Male rats were randomized into three dietary groups over four weeks: a control group receiving less than 4% lard, regular chow, a 10% lard group, and a 60% lard group. Following dietary interventions, fertility tests were conducted across the groups. The epididymis was dissected into caput, corpus, and cauda regions to assess sperm concentration, vitality capacitation, carbohydrate distribution, tyrosine phosphorylation, and phosphatidylserine levels. Additionally, serum testosterone levels were measured to evaluate hormonal influences on fertility. The rats subjected to high-fat diets leading to overweight and obesity exhibited significant alterations in fertility. These changes were characterized by impaired epididymal sperm maturation, as evidenced by lower testosterone levels, decreased sperm viability, and capacitation. Furthermore, increased adiposity was associated with a lack of asymmetry in the plasma membrane, alteration in carbohydrate distribution, and changes in tyrosine phosphorylation. This study underscores the adverse effects of high-fat diets on male fertility, particularly through mechanisms affecting sperm maturation in the epididymis. The evidence suggests that obesity-induced alterations in sperm parameters and hormonal profiles may contribute to reduced fertility in male rats, which could have implications for understanding similar human processes.
Collapse
Affiliation(s)
- Lorena Ruiz-Valderrama
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 09340, Mexico; (L.R.-V.)
| | - José Edwin Mendoza-Sánchez
- Doctorado en Biología Experimental, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 09340, Mexico
| | - Ernesto Rodríguez-Tobón
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 09340, Mexico; (L.R.-V.)
| | - Isabel Arrieta-Cruz
- Departamento de Investigación Básica, Instituto Nacional de Geriatría, Magdalena Contreras, Ciudad de México 10200, Mexico
| | - Humberto González-Márquez
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 09340, Mexico
| | - Pablo Arturo Salame-Méndez
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 09340, Mexico; (L.R.-V.)
| | - Rosario Tarragó-Castellanos
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 09340, Mexico; (L.R.-V.)
| | - Edith Cortés-Barberena
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 09340, Mexico
| | - Ahiezer Rodríguez-Tobón
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 09340, Mexico; (L.R.-V.)
| | - Edith Arenas-Ríos
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 09340, Mexico; (L.R.-V.)
| |
Collapse
|
3
|
Moreira RJ, Oliveira PF, Spadella MA, Ferreira R, Alves MG. Do Lifestyle Interventions Mitigate the Oxidative Damage and Inflammation Induced by Obesity in the Testis? Antioxidants (Basel) 2025; 14:150. [PMID: 40002337 PMCID: PMC11851673 DOI: 10.3390/antiox14020150] [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: 12/27/2024] [Revised: 01/18/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity results from a disproportionate accumulation of fat and has become a global health concern. The increase in adipose tissue is responsible for several systemic and testicular changes including hormone levels (leptin, adiponectin, testosterone, estrogen), inflammatory cytokines (increase in TNF-α and IL-6 and decrease in IL-10), and redox state (increase in reactive oxygen species and reduction in antioxidant enzymes). This results in poor sperm quality and compromised fertility in men with obesity. Lifestyle modifications, particularly diet transition to caloric restriction and physical exercise, are reported to reverse these negative effects. Nevertheless, precise mechanisms mediating these benefits, including how they modulate testicular oxidative stress, inflammation, and metabolism, remain to be fully elucidated. The main pathway described by which these lifestyle interventions reverse obesity-induced oxidative damage is the Nrf2-SIRT1 axis, which modulates the overexpression of antioxidant defenses. Of note, some of the detrimental effects of obesity on the testis are inherited by the descendants of individuals with obesity, and while caloric restriction reverses some of these effects, no significant work has been carried out regarding physical exercise. This review discusses the consequences of obesity-induced testicular oxidative stress on adult and pediatric populations, emphasizing the therapeutic potential of lifestyle to mitigate these detrimental effects.
Collapse
Affiliation(s)
- Ruben J. Moreira
- Institute of Biomedicine, Department of Medical Sciences (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal;
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (P.F.O.); (R.F.)
| | - Pedro F. Oliveira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (P.F.O.); (R.F.)
| | | | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (P.F.O.); (R.F.)
| | - Marco G. Alves
- Institute of Biomedicine, Department of Medical Sciences (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal;
| |
Collapse
|
4
|
Aguilar-Lozano A, Palacios-González B, Guevara-Cruz M, Tovar AR, Palma-Guzman A, Noriega LG. The type of diet consumed during prepuberty modulates plasma cholesterol, hepatic LXRα expression, and DNA methylation and hydroxymethylation during adulthood in male rats. PLoS One 2025; 20:e0315197. [PMID: 39854485 PMCID: PMC11761095 DOI: 10.1371/journal.pone.0315197] [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: 05/12/2024] [Accepted: 11/22/2024] [Indexed: 01/26/2025] Open
Abstract
Childhood obesity increases the risk of developing metabolic diseases in adulthood, since environmental stimuli during critical windows of development can impact on adult metabolic health. Studies demonstrating the effect of prepubertal diet on adult metabolic disease risk are still limited. We hypothesized that a prepubertal control diet (CD) protects the adult metabolic phenotype from diet-induced obesity (DIO), while a high-fat diet (HFD) would predispose to adult metabolic alterations. Sprague-Dawley male rats were fed either a CD or a HFD during the prepubertal period (day 30-40 of age) and subsequently a chronic HFD or CD, respectively, until adulthood (day 220 of age). As controls, rats aged 30 days were exclusively fed a CD or a HFD until adulthood. Body weight and composition, metabolic rate, biochemical and hormonal plasma measurements, hepatic gene expression and methylation and hydroxymethylation levels were analyzed at ages 30, 40 and 220 days. The prepubertal CD prevented fat mass accumulation, lean mass loss and metabolic inflexibility, showed lower insulin, leptin and cholesterol concentrations in adulthood despite the chronic HFD. Notably, the prepubertal CD led to higher hepatic Lxrα expression, lower hepatic global DNA methylation and higher hydroxymethylation in adulthood despite a chronic HFD. Conversely, a prepubertal HFD decreased adult metabolic flexibility, increased serum cholesterol, and decreased Lxrα expression and global DNA hydroxymethylation, while also increasing DNA methylation levels despite a chronic CD. In summary, a prepubertal CD protected the adult metabolic phenotype from high cholesterol concentrations associated with increased hepatic Lxrα expression and lower hepatic global DNA methylation in adulthood, despite exposure to a chronic HFD. Conversely, a prepubertal HFD altered the adult metabolic phenotype.
Collapse
Affiliation(s)
- Ana Aguilar-Lozano
- Department of Nutritional Physiology, National Institute of Medical and Nutritional Sciences “Salvador Zubirán”, Mexico City, Mexico
- Biomedical Sciences PhD program, National Autonomous Mexican University (UNAM), Mexico City, Mexico
| | - Berenice Palacios-González
- Healthy Aging Unit from the National Institute for Genomic Medicine, Aging Research Center, Mexico City, Mexico
| | - Martha Guevara-Cruz
- Department of Nutritional Physiology, National Institute of Medical and Nutritional Sciences “Salvador Zubirán”, Mexico City, Mexico
| | - Armando R. Tovar
- Department of Nutritional Physiology, National Institute of Medical and Nutritional Sciences “Salvador Zubirán”, Mexico City, Mexico
| | - Alam Palma-Guzman
- Histology Laboratory from the XXI Century National Medical Center–Social Security Mexican Institute, Mexico City, Mexico
| | - Lilia G. Noriega
- Department of Nutritional Physiology, National Institute of Medical and Nutritional Sciences “Salvador Zubirán”, Mexico City, Mexico
| |
Collapse
|
5
|
Cannarella R, Curto R, Condorelli RA, Lundy SD, La Vignera S, Calogero AE. Molecular insights into Sertoli cell function: how do metabolic disorders in childhood and adolescence affect spermatogonial fate? Nat Commun 2024; 15:5582. [PMID: 38961093 PMCID: PMC11222552 DOI: 10.1038/s41467-024-49765-1] [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: 08/07/2023] [Accepted: 06/12/2024] [Indexed: 07/05/2024] Open
Abstract
Male infertility is a major public health concern globally with unknown etiology in approximately half of cases. The decline in total sperm count over the past four decades and the parallel increase in childhood obesity may suggest an association between these two conditions. Here, we review the molecular mechanisms through which obesity during childhood and adolescence may impair future testicular function. Several mechanisms occurring in obesity can interfere with the delicate metabolic processes taking place at the testicular level during childhood and adolescence, providing the molecular substrate to hypothesize a causal relationship between childhood obesity and the risk of low sperm counts in adulthood.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Roberto Curto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Scott D Lundy
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
6
|
Zhang Y, Ding R, Zhang Y, Qi J, Cao W, Deng L, Zhou L, Ye Y, Xue Y, Liu E. Dysfunction of DMT1 and miR-135b in the gut-testis axis in high-fat diet male mice. GENES & NUTRITION 2024; 19:1. [PMID: 38243197 PMCID: PMC10797958 DOI: 10.1186/s12263-024-00737-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 01/04/2024] [Indexed: 01/21/2024]
Abstract
BACKGROUND Obese patients have been found to be susceptible to iron deficiency, and malabsorption of dietary iron is the cause of obesity-related iron deficiency (ORID). Divalent metal transporter 1 (DMT1) and ferroportin (FPN), are two transmembrane transporter proteins expressed in the duodenum that are closely associated with iron absorption. However, there have been few studies on the association between these two proteins and the increased susceptibility to iron deficiency in obese patients. Chronic inflammation is also thought to be a cause of obesity-related iron deficiency, and both conditions can have an impact on spermatogenesis and impair male reproductive function. Based on previous studies, transgenerational epigenetic inheritance through gametes was observed in obesity. RESULTS Our results showed that obese mice had decreased blood iron levels (p < 0.01), lower protein and mRNA expression for duodenal DMT1 (p < 0.05), but no statistically significant variation in mRNA expression for duodenal FPN (p > 0.05); there was an increase in sperm miR-135b expression (p < 0.05). Bioinformatics revealed ninety overlapping genes and further analysis showed that they were primarily responsible for epithelial cilium movement, fatty acid beta-oxidation, protein dephosphorylation, fertilization, and glutamine transport, which are closely related to spermatogenesis, sperm development, and sperm viability in mice. CONCLUSIONS In obese mice, we observed downregulation of DMT1 in the duodenum and upregulation of miR-135b in the spermatozoa.
Collapse
Affiliation(s)
- Yanru Zhang
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, 710061, China
| | - Ruike Ding
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, 710061, China
| | - Yulin Zhang
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, 710061, China
| | - Jia Qi
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, 710061, China
| | - Wenbin Cao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, 710061, China
| | - Lijun Deng
- Spring Biological Technology Development Co., Ltd, Fangchenggang, Guangxi, 538000, China
| | - Lin Zhou
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, 710061, China
| | - Yun Ye
- Central Laboratory, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710000, China
| | - Ying Xue
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, 710061, China.
| | - Enqi Liu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, 710049, China.
| |
Collapse
|
7
|
Yu W, Fan S, Wang X, Zhu J, Yuan Z, Han Y, Zhang H, Weng Q. Seasonal change of circulating leptin associated with testicular activities of the wild ground squirrels (Citellus dauricus). Integr Zool 2023; 18:76-92. [PMID: 35841626 DOI: 10.1111/1749-4877.12668] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The purpose of this study was to explore the variations in the circulating leptin concentrations of the wild ground squirrels in relation to seasonal changes in testicular activities. Hematoxylin-eosin staining showed all types of elongated spermatids and spermatogenic cells existed in the testis in April, while the primary spermatocytes and spermatogonia were most advanced stages of germ cells in June. In addition, the primary spermatocytes, secondary spermatocytes, and spermatogonia were most advanced stages of germ cells in September. The highest circulating leptin concentration was consistent with the maximum body weight results from accumulation of adipose tissue in September. The mRNA expression level of leptin receptor (Ob-R) and STAT3 was lowest in June, raised in September, and remained increased in April. Ob-R and STAT3 were stronger staining in the Leydig cells in July. Moreover, the concentrations of testosterone (T) showed the maximum values in April, the minimum values in June, and significant increases in September. Furthermore, it is worth noting that the levels of T increased with the mRNA levels of Ob-R, STAT3, StAR, and testicular steroidogenic enzymes (3β-HSD, P450c17, and P450scc). Moreover, RNA-seq analyses of testis during the different periods showed that a total of 4209 genes were differentially expressed genes (DEGs); further analysis revealed that DEGs related with the Jak/STAT pathways and reproduction were altered. Taken together, the results suggested that the leptin regulated testicular function through the Jak/STAT pathways and testicular steroidogenic factor expressions.
Collapse
Affiliation(s)
- Wenyang Yu
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Sijie Fan
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Xi Wang
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Jueyu Zhu
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Zhengrong Yuan
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Yingying Han
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Haolin Zhang
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Qiang Weng
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| |
Collapse
|
8
|
Suleiman JB, Abu Bakar AB, Noor MM, Nna VU, Othman ZA, Zakaria Z, Eleazu CO, Mohamed M. Bee bread mitigates downregulation of steroidogenic genes, decreased spermatogenesis, and epididymal oxidative stress in male rats fed with high-fat diet. Am J Physiol Endocrinol Metab 2021; 321:E351-E366. [PMID: 34229480 DOI: 10.1152/ajpendo.00093.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023]
Abstract
The pituitary-gonadal axis plays an important role in steroidogenesis and spermatogenesis, and by extension, fertility. The aim of this study was to investigate the protective role of bee bread, a natural bee product, against obesity-induced decreases in steroidogenesis and spermatogenesis. Thirty-two adult male Sprague-Dawley rats weighing between 200 and 300 g were divided into four groups (n = 8/group), namely: normal control (NC), high-fat diet (HFD), HFD plus bee bread administered concurrently for 12 wk (HFD + B), HFD plus orlistat administered concurrently for 12 wk (HFD + O) groups. Bee bread (0.5 g/kg) or orlistat (10 mg/kg/day) was suspended in distilled water and given by oral gavage daily for 12 wk. Levels of follicle-stimulating hormone, luteinizing hormone, testosterone, and adiponectin, as well as sperm count, motility, viability, normal morphology, and epididymal antioxidants decreased, whereas levels of leptin, malondialdehyde, and sperm nDNA fragmentation increased significantly in the HFD group relative to the NC group. There were significant decreases in the testicular mRNA transcript levels of androgen receptor, luteinizing hormone receptor, steroidogenic acute regulatory protein, cytochrome P450 enzyme, 3β-hydroxysteroid dehydrogenase (HSD) and 17β-HSD in the testes of the HFD group. Furthermore, mount, intromission and ejaculatory latencies increased, and penile cGMP level decreased significantly in the HFD group. Supplementation with bee bread significantly reduced leptin level and increased adiponectin level, enhanced sperm parameters and reduced sperm nDNA fragmentation, upregulated the levels of steroidogenic genes and proteins in HFD-induced obese male rats. Bee bread improved steroidogenesis and spermatogenesis by upregulating steroidogenic genes. Therefore, bee bread may be considered as a potential supplementation to protect against infertility in overweight men or men with obesity.NEW & NOTEWORTHY The high-fat diet utilized in the present study induced obesity in the male rats. Bee bread supplementation mitigated impaired steroidogenesis, spermatogenesis, mating behavior, and fertility potential by counteracting the downregulation of steroidogenic genes, thus increasing testosterone levels and suppressing epididymal oxidative stress. These benefits may be due to the abundance of phenolic and flavonoid compounds in bee bread.
Collapse
Affiliation(s)
- Joseph Bagi Suleiman
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Department of Science Laboratory Technology, Akanu Ibiam Federal Polytechnic, Unwana, Afikpo, Nigeria
| | - Ainul Bahiyah Abu Bakar
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Mahanem Mat Noor
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Victor Udo Nna
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Zaidatul Akmal Othman
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Unit of Physiology, Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu, Malaysia
| | - Zaida Zakaria
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Chinedum Ogbonnaya Eleazu
- Department of Chemistry, Biochemistry and Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Mahaneem Mohamed
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Unit of Integrative Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
9
|
Wagner IV, Klöting N, Savchuk I, Eifler L, Kulle A, Kralisch-Jäcklein S, Dötsch J, Hiort O, Svechnikov K, Söder O. Diabetes Type 1 Negatively Influences Leydig Cell Function in Rats, Which is Partially Reversible By Insulin Treatment. Endocrinology 2021; 162:6122542. [PMID: 33507237 DOI: 10.1210/endocr/bqab017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Indexed: 12/29/2022]
Abstract
Type 1 diabetes mellitus (T1DM) is associated with impaired spermatogenesis and lower testosterone levels and epididymal weight. However, the underlying processes in the testis are unknown and remain to be elucidated. Therefore, the present study focused on the effects of T1DM on testicular function in a spontaneously diabetic rat model. BB/OKL rats after diabetes manifestation were divided into 3 groups: those without insulin treatment and insulin treatment for a duration of 2 and of 6 weeks. Anthropometrical data, circulating levels of gonadotrophins, testosterone, and inhibin B were measured. Intratesticular testosterone, oxidative stress, inflammation, and apoptosis were analyzed. Key enzymes of steroidogenesis were evaluated in the testis. Untreated diabetic rats had significantly lower serum follicle-stimulating hormone and luteinizing hormone levels. Serum and intratesticular testosterone levels significantly decreased in untreated diabetic rats compared to healthy controls. Key markers of Leydig cell function were significantly downregulated at the RNA level: insulin-like factor 3 (Insl3) by 53% (P = .006), Star by 51% (P = .004), Cyp11A1 by 80% (P = .003), 3Beta-Hsd2 by 61% (P = .005), and Pbr by 52% (P = .002). In the insulin-treated group, only Cyp11A1 and 3Beta-Hsd2 transcripts were significantly lower. Interestingly, the long-term insulin-treated group showed significant upregulation of most steroidogenic enzymes without affecting testosterone levels. Tumor necrosis factor α and apoptosis were significantly increased in the long-term insulin-treated rats. In conclusion T1DM, with a severe lack of insulin, has an adverse action on Leydig cell function. This is partially reversible with well-compensated blood glucose control. Long-term T1DM adversely affects Leydig cell function because of the process of inflammation and apoptosis.
Collapse
Affiliation(s)
- Isabel Viola Wagner
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatrics, Medical Faculty, University of Cologne, Cologne, Germany
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - Nora Klöting
- Integrated Research and Treatment Center (IFB Adiposity Diseases), Department of Medicine, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig, Leipzig, Germany
| | - Iuliia Savchuk
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Lisa Eifler
- Department of Pediatrics, Medical Faculty, University of Cologne, Cologne, Germany
| | - Alexandra Kulle
- University Hospital Kiel, Hormone Center for Pediatric Endocrinology Laboratory, Kiel, Germany
| | - Susan Kralisch-Jäcklein
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Jörg Dötsch
- Department of Pediatrics, Medical Faculty, University of Cologne, Cologne, Germany
| | - Olaf Hiort
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - Konstantin Svechnikov
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Olle Söder
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
De León-Ramírez YM, Lara-García M, Pacheco P, Lara-García O, Martínez-Gómez M, Cuevas-Romero E, Rodríguez-Antolín J, Nicolás-Toledo L. Histomorphological testicular changes and decrease in the sperm count in pubertal rats induced by a high-sugar diet. Ann Anat 2021; 235:151678. [PMID: 33515690 DOI: 10.1016/j.aanat.2021.151678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/01/2020] [Accepted: 12/29/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND During childhood and adolescence, excessive food consumption stimulates adipose tissue expansion promoting overweight in humans, and mice. A high-sucrose diet is related to obesity and metabolic syndrome. Infertility is commonly related to these pathologies. We aim to evaluate possible histomorphological testicular changes induced by a high-sucrose diet on sperm count during the post-weaning period. METHODS Wistar male rats aged 21 days, weaned, were randomly assigned into two groups: control (fed and hydrated normally) and sugar group (fed normally but hydrated with a solution containing 30% of diluted sucrose during 30 days). At the pubertal age of 51 days, animals were killed and blood samples were taken to measure testosterone and leptin. Testicles were collected and gonadal adipose tissue and semen samples from the epididymis were excised. Testicle samples were used for morphological description using H&E staining, as well as to quantify the triacylglycerol content and the lactate dehydrogenase (LDH) expression. Semen samples were used to assess motility, viability, and sperm count. RESULTS The sugar group presented an increase in the testicular weight, but a reduction in the cross-sectional area of seminiferous tubules. Moreover, disorganization of Sertoli cells and spermatogonia, an increase in the LDH expression within the entire seminiferous tubule, and a reduced sperm count and spermatozoid motility were found. These alterations were accompanied by high serum levels of testosterone and leptin. CONCLUSIONS Our results indicate strong damage of testis by sugar consumption during early life that may lead to the onset of infertility in adulthood.
Collapse
Affiliation(s)
| | | | - Pablo Pacheco
- Instituto de Neuroetología, Universidad Veracruzana, Veracruz, México; Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México
| | - Omar Lara-García
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Margarita Martínez-Gómez
- Instituto de Neuroetología, Universidad Veracruzana, Veracruz, México; Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México
| | - Estela Cuevas-Romero
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Jorge Rodríguez-Antolín
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Leticia Nicolás-Toledo
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, México.
| |
Collapse
|
11
|
Wagner IV, Oliver E, Dötsch J, Söder O. Adverse effects of metabolic disorders in childhood on adult reproductive function and fertility in the male. J Pediatr Endocrinol Metab 2021; 34:13-23. [PMID: 33185575 DOI: 10.1515/jpem-2020-0276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/21/2020] [Indexed: 12/21/2022]
Abstract
Over the last 50 years, there has been a steady decline in fertility rates in humans, which has occurred in parallel with an increasing incidence of obesity and metabolic disorders. The potential impact of these disorders and plausible mechanisms by which they negatively influence male reproduction are only partly understood and published data are often controversial. Obesity is one of the most important health challenges worldwide and is becoming more prevalent in children and adolescents. Obesity, the metabolic syndrome and related co-morbidities can lead to impaired male reproductive function, including adverse effects on spermatogenesis and steroidogenesis as illustrated by reduced sperm number and quality, decreased testosterone levels and elevated inflammatory markers. The incidence of diabetes mellitus type I is also dramatically increasing and may negatively impact spermatogenesis and testicular function, resulting in decreased serum testosterone and epididymal weight. In this review, we summarize and discuss the effects of metabolic diseases that typically develop during childhood and adolescence on later reproductive function and fertility. While impact on reproductive health is likely observed in both sexes, we have chosen to focus on the male in the current review. Specifically, we illustrate adverse effects of obesity, type 1 diabetes, the metabolic syndrome and insulin resistance on sperm function and testosterone metabolism. Identification of pathophysiological mechanisms during childhood may open up new avenues for early prevention and treatment resulting in better reproductive outcomes and improved fertility rates during adulthood.
Collapse
Affiliation(s)
- Isabel Viola Wagner
- Karolinska Institutet, Department of Women's and Children's Health, Pediatric Endocrinology Unit, Stockholm, Sweden.,Department of Pediatrics, Medical Faculty, University of Cologne, Pediatric Endocrinology Unit, Cologne, Germany.,Department of Pediatrics, Medical Faculty, University of Lübeck, Pediatric Endocrinology Unit, Lübeck, Germany
| | - Elizabeth Oliver
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Jörg Dötsch
- Department of Pediatrics, Medical Faculty, University of Cologne, Pediatric Endocrinology Unit, Cologne, Germany
| | - Olle Söder
- Karolinska Institutet, Department of Women's and Children's Health, Pediatric Endocrinology Unit, Stockholm, Sweden.,Department of Pediatrics, Medical Faculty, University of Lübeck, Pediatric Endocrinology Unit, Lübeck, Germany
| |
Collapse
|
12
|
Bunay J, Gallardo LM, Torres-Fuentes JL, Aguirre-Arias MV, Orellana R, Sepúlveda N, Moreno RD. A decrease of docosahexaenoic acid in testes of mice fed a high-fat diet is associated with impaired sperm acrosome reaction and fertility. Asian J Androl 2021; 23:306-313. [PMID: 33269725 PMCID: PMC8152421 DOI: 10.4103/aja.aja_76_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Obesity is a major worldwide health problem that is related to most chronic diseases, including male infertility. Owing to its wide impact on health, mechanisms underlying obesity-related infertility remain unknown. In this study, we report that mice fed a high-fat diet (HFD) for over 2 months showed reduced fertility rates and increased germ cell apoptosis, seminiferous tubule degeneration, and decreased intratesticular estradiol (E2) and E2-to-testosterone ratio. Interestingly, we also detected a decrease in testicular fatty acid levels, behenic acid (C22:0), and docosahexaenoic acid (DHA, 22:6n-3), which may be related to the production of dysfunctional spermatozoa. Overall, we did not detect any changes in the frequency of seminiferous tubule stages, sperm count, or rate of in vitro capacitation. However, there was an increase in spontaneous and progesterone-induced acrosomal exocytosis (acrosome reaction) in spermatozoa from HFD-fed mice. These data suggest that a decrease in E2 and fatty acid levels influences spermatogenesis and some steps of acrosome biogenesis that will have consequences for fertilization. Thus, our results add new evidence about the adverse effect of obesity in male reproduction and suggest that the acrosomal reaction can also be affected under this condition.
Collapse
Affiliation(s)
- Julio Bunay
- Physiology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile (PUC), Santiago 8331150, Chile
| | - Luz-Maria Gallardo
- Physiology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile (PUC), Santiago 8331150, Chile
| | - Jorge Luis Torres-Fuentes
- Physiology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile (PUC), Santiago 8331150, Chile
| | - M Verónica Aguirre-Arias
- Physiology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile (PUC), Santiago 8331150, Chile
| | - Renan Orellana
- Department of Chemistry and Biological Sciences, Health Sciences Faculty, Universidad Bernardo O Higgins, Santiago 8370854, Chile
| | - Néstor Sepúlveda
- Center of Excellence in Biotechnology of Reproduction, Universidad de la Frontera, Temuco 4780000, Chile
| | - Ricardo D Moreno
- Physiology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile (PUC), Santiago 8331150, Chile
| |
Collapse
|
13
|
Pini T, Parks J, Russ J, Dzieciatkowska M, Hansen KC, Schoolcraft WB, Katz-Jaffe M. Obesity significantly alters the human sperm proteome, with potential implications for fertility. J Assist Reprod Genet 2020; 37:777-787. [PMID: 32026202 PMCID: PMC7183029 DOI: 10.1007/s10815-020-01707-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/30/2020] [Indexed: 11/25/2022] Open
Abstract
PURPOSE In men, obesity may lead to poor semen parameters and reduced fertility. However, the causative links between obesity and male infertility are not totally clear, particularly on a molecular level. As such, we investigated how obesity modifies the human sperm proteome, to elucidate any important implications for fertility. METHODS Sperm protein lysates from 5 men per treatment, classified as a healthy weight (body mass index (BMI) ≤ 25 kg/m2) or obese (BMI ≥ 30 kg/m2), were FASP digested, submitted to liquid chromatography tandem mass spectrometry, and compared by label-free quantification. Findings were confirmed for several proteins by qualitative immunofluorescence and a quantitative protein immunoassay. RESULTS A total of 2034 proteins were confidently identified, with 24 proteins being significantly (p < 0.05) less abundant (fold change < 0.05) in the spermatozoa of obese men and 3 being more abundant (fold change > 1.5) compared with healthy weight controls. Proteins with altered abundance were involved in a variety of biological processes, including oxidative stress (GSS, NDUFS2, JAGN1, USP14, ADH5), inflammation (SUGT1, LTA4H), translation (EIF3F, EIF4A2, CSNK1G1), DNA damage repair (UBEA4), and sperm function (NAPA, RNPEP, BANF2). CONCLUSION These results suggest that oxidative stress and inflammation are closely tied to reproductive dysfunction in obese men. These processes likely impact protein translation and folding during spermatogenesis, leading to poor sperm function and subfertility. The observation of these changes in obese men with no overt andrological diagnosis further suggests that traditional clinical semen assessments fail to detect important biochemical changes in spermatozoa which may compromise fertility.
Collapse
Affiliation(s)
- T Pini
- Colorado Center for Reproductive Medicine, Lone Tree, CO, 80124, USA.
| | - J Parks
- Colorado Center for Reproductive Medicine, Lone Tree, CO, 80124, USA
| | - J Russ
- Colorado Center for Reproductive Medicine, Lone Tree, CO, 80124, USA
| | - M Dzieciatkowska
- School of Medicine Biological Mass Spectrometry Facility, University of Colorado, Aurora, CO, 80045, USA
| | - K C Hansen
- School of Medicine Biological Mass Spectrometry Facility, University of Colorado, Aurora, CO, 80045, USA
| | - W B Schoolcraft
- Colorado Center for Reproductive Medicine, Lone Tree, CO, 80124, USA
| | - M Katz-Jaffe
- Colorado Center for Reproductive Medicine, Lone Tree, CO, 80124, USA
| |
Collapse
|
14
|
Braga PC, Pereira SC, Ribeiro JC, Sousa M, Monteiro MP, Oliveira PF, Alves MG. Late-onset hypogonadism and lifestyle-related metabolic disorders. Andrology 2020; 8:1530-1538. [PMID: 31991053 DOI: 10.1111/andr.12765] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/22/2019] [Accepted: 01/23/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Late-onset hypogonadism (LOH) is a condition defined by low levels of testosterone (T), occurring in advanced age. LOH is promoted by senescence, which, in turn, has negative effects on male fertility. Interestingly, the impact of metabolic disorders on the male reproductive system has been the topic of several studies, but the association with LOH is still debatable. OBJECTIVES Herein, we discuss the hypothesis that the prevalence of metabolic abnormalities potentiates the effects of LOH on the male reproductive system, affecting the reproductive potential of those individuals. MATERIAL AND METHODS We analyzed the bibliography available, until June 2019, about LOH in relation to metabolic and hormonal dysregulation, sperm quality profiles and assisted-reproduction treatment outcomes. RESULTS LOH affects the hypothalamic-pituitary testis (HPT) axis. Additionally, metabolic disorders can also induce T deficiency, which is reflected in decreased male fertility, highlighting a possible connection. Indeed, T replacement therapy (TRT) is widely used to restore T levels. Although this therapy is unable to reverse all deleterious effects promoted by LOH on male reproductive function, it can improve metabolic and reproductive health. DISCUSSION AND CONCLUSIONS Emerging new evidence suggests that metabolic disorders may aggravate LOH effects on the fertility potential of males in reproductive age, by enhancing T deficiency. These results clearly show that metabolic disorders, such as obesity and diabetes, have a greater impact on causing hypogonadotropic hypogonadism than tissue senescence. Further, TRT and off-label alternatives capable of restoring T levels appear as suitable to improve LOH, while also counteracting comorbidities related with metabolic diseases.
Collapse
Affiliation(s)
- Patrícia C Braga
- Department of Microscopy, Laboratory of Cell Biology, and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Sara C Pereira
- Department of Microscopy, Laboratory of Cell Biology, and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - João C Ribeiro
- Department of Microscopy, Laboratory of Cell Biology, and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Mário Sousa
- Department of Microscopy, Laboratory of Cell Biology, and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Centre for Reproductive Genetics Professor Alberto Barros, Porto, Portugal
| | - Mariana P Monteiro
- Department of Anatomy, Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Pedro F Oliveira
- Department of Microscopy, Laboratory of Cell Biology, and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Department of Genetics, Faculty of Medicine, University of Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,QOPNA & LAQV, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Marco G Alves
- Department of Microscopy, Laboratory of Cell Biology, and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| |
Collapse
|
15
|
Wagner IV, Yango P, Svechnikov K, Tran ND, Söder O. Adipocytokines may delay pubertal maturation of human Sertoli cells. Reprod Fertil Dev 2019; 31:1395-1400. [PMID: 31056083 DOI: 10.1071/rd18487] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 02/15/2019] [Indexed: 12/19/2022] Open
Abstract
Reproduction is an important target of obesity complications, including adverse effects on spermatogenesis and steroidogenesis. Adipocytokines are key mediators in various complications of obesity. Our aim was to study the potential of adipocytokines to affect Sertoli cell function, which is crucial for spermatogenesis, and possibly link these findings to the observed attenuation of spermatogenesis in obese males. Testicular biopsies were obtained from healthy donors. Highly purified adult human Sertoli cells (HSCs) were isolated by fluorescence-activated cell sorting. Cells were cultured and exposed to different concentrations of adipocytokines (10-1000ngmL-1 ) for 2-7 days. Expression of selected Sertoli cell genes was quantified by quantitative polymerase chain reaction. Long-term treatment (7 days) of HSCs with higher concentrations of chemerin, irisin, nicotinamide phosphoribosyltransferase (Nampt), resistin and progranulin significantly suppressed FSH receptor expression (by 79%, 83%, 64%, 71% and 26% respectively; P P invitro , may negatively affect Sertoli cell maturation and retain these cells in a more prepubertal stage. This could negatively affect testis function and add to fertility problems in obese adults.
Collapse
Affiliation(s)
- I V Wagner
- Karolinska Institutet, Department of Women's and Children's Health, Pediatric Endocrinology Unit, 17176 Stockholm, Sweden; and Department of Pediatrics, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany; and Corresponding author
| | - P Yango
- Center for Reproductive Sciences, University of California, San Francisco, 513 Parnassus Avenue, CA 94143, USA
| | - K Svechnikov
- Karolinska Institutet, Department of Women's and Children's Health, Pediatric Endocrinology Unit, 17176 Stockholm, Sweden
| | - N D Tran
- Center for Reproductive Sciences, University of California, San Francisco, 513 Parnassus Avenue, CA 94143, USA
| | - O Söder
- Karolinska Institutet, Department of Women's and Children's Health, Pediatric Endocrinology Unit, 17176 Stockholm, Sweden
| |
Collapse
|
16
|
Regulation of Leydig cell steroidogenesis: intriguing network of signaling pathways and mitochondrial signalosome. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.coemr.2019.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
17
|
Obesity and Hypogonadism-A Narrative Review Highlighting the Need for High-Quality Data in Adolescents. CHILDREN-BASEL 2019; 6:children6050063. [PMID: 31052376 PMCID: PMC6560454 DOI: 10.3390/children6050063] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 12/30/2022]
Abstract
The prevalence of obesity continues to rise in adult and pediatric populations throughout the world. Obesity has a direct impact on all organ systems, including the reproductive system. This review summarizes current knowledge about the effects of obesity on the male reproductive system across age, highlighting the need for more data in children and adolescents. Male hypogonadism is commonly seen in patients with obesity and affects the onset, duration, and progression of puberty. Different pathophysiologic mechanisms include increased peripheral conversion of testosterone to estrone and increased inflammation due to increased fat, both of which lead to suppression of the hypothalamic-pituitary-gonadotropin (HPG) axis and delayed development of secondary sexual characteristics in adolescent males. Evaluation of the HPG axis in obesity includes a thorough history to exclude other causes of hypogonadism and syndromic associations. Evaluation should also include investigating the complications of low testosterone, including increased visceral fat, decreased bone density, cardiovascular disease risk, and impaired mood and cognition, among others. The mainstay of treatment is weight reduction, but medications such as testosterone and clomiphene citrate used in adults, remain scarcely used in adolescents. Male hypogonadism associated with obesity is common and providers who care for adolescents and young adults with obesity should be aware of its impact and management.
Collapse
|
18
|
Shalitin S, Kiess W. Putative Effects of Obesity on Linear Growth and Puberty
. Horm Res Paediatr 2018; 88:101-110. [PMID: 28183093 DOI: 10.1159/000455968] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/10/2017] [Indexed: 01/02/2023] Open
Abstract
Childhood obesity is a major public health problem that has grown to epidemic proportions throughout the world. Obesity is influenced by genetic and environmental factors. The nutritional status plays an important role in growth and body weight regulation. Excess adiposity during childhood can affect the process of growth and puberty. Obese children are frequently tall for their age, with accelerated epiphyseal growth plate maturation despite low growth hormone levels. Several regulatory hormones may affect the process of linear growth in the constellation of obesity, as high levels of insulin and leptin are observed in obese children. Leptin can act as a skeletal growth factor, with a direct effect on skeletal growth centers. The finding that overweight children, especially girls, tend to mature earlier than lean children has led to the hypothesis that the degree of body fatness may trigger the neuroendocrine events that lead to the onset of puberty. Leptin receptors have been identified in the hypothalamus, as well as in gonadotrope cells, ovarian follicular cells, and Leydig cells. The increased leptin and androgen levels seen in obese children may be implicated in their earlier onset of puberty and accelerated pubertal growth. This review is focused on the interaction between childhood obesity and growth and pubertal processes.
.
Collapse
Affiliation(s)
- Shlomit Shalitin
- The Jesse Z. and Sara Lea Shafer Institute of Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Wieland Kiess
- Department of Women and Child Health, Hospital for Children and Adolescents, University Hospitals, University of Leipzig, Leipzig, Germany
| |
Collapse
|
19
|
Zhao JL, Zhao YY, Zhu WJ. A high-fat, high-protein diet attenuates the negative impact of casein-induced chronic inflammation on testicular steroidogenesis and sperm parameters in adult mice. Gen Comp Endocrinol 2017; 252:48-59. [PMID: 28743557 DOI: 10.1016/j.ygcen.2017.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 06/30/2017] [Accepted: 07/16/2017] [Indexed: 12/16/2022]
Abstract
The interaction between obesity and chronic inflammation has been studied. Diet-induced obesity or chronic inflammation could reduce the testicular functions of males. However, the mechanism underlying the reproductive effects of fattening foods in males with or without chronic inflammation still needs further discussion. This study was aimed to investigate the effects of high-fat, high-protein diet on testicular steroidogenesis and sperm parameters in adult mice under physiological and chronic inflammatory conditions. Because casein can trigger a non-infectious systemic inflammatory response, we used casein injection to induce chronic inflammation in male adult Kunming mice. Twenty-four mice were randomly and equally divided into four groups: (i) normal diet+saline (Control); (ii) normal diet+casein (ND+CS); (iii) high-fat, high-protein diet+saline (HFPD+SI); (iv) high-fat, high-protein diet+casein (HFPD+CS). After 8weeks, there was a significant increase in body weight for groups HFPD+SI and HFPD+CS and a decrease in group ND+CS compared with the control. The serum levels of tumor necrosis factor alpha (TNF-α), interleukin-10 (IL-10) and lipid profiles were increased markedly in groups ND+CS, HFPD+SI and HFPD+CS compared with the control. A remarkable reduction of serum adiponectin level occurred in group HFPD+CS compared with group ND+CS. Sperm parameters (sperm count, viability and abnormality) were also adversely affected in groups ND+CS and HFPD+SI. Groups ND+CS and HFPD+SI showed severe pathological changes in testicular tissues. Semiquantitative RT-PCR, Western blot and immunohistochemical staining also showed significant reductions in both testicular mRNA and protein levels of steroidogenic acute regulatory (StAR) and cytochrome P450scc (CYP11A1) in groups HFPD+SI and HFPD+CS compared with the control, whereas testicular mRNA and protein levels of 3β-hydroxysteroid dehydrogenase (3β-HSD) in groups HFPD+SI and HFPD+CS significantly increased. The mRNA and protein levels of the StAR and 3β-HSD in group HFPD+CS were both higher than those of in group ND+CS. These results indicated that Kunming male mice with high-fat, high-protein diet and casein injection for 8weeks can be used to establish a diet-induced obesity and chronic systemic inflammation. The sperm parameters in groups ND+CS and HFPD+SI decreased accompanied by pathological changes of testicular tissue. This resultant effect of reduced serum testosterone levels was associated with the overproduction of TNF-α and IL-10 and down-regulation of StAR and CYP11A1. Under the same casein-induced chronic inflammation condition, the mice with high-fat, high-protein diet had better testicular steroidogenesis activity and sperm parameters compared with the mice in normal diet, indicating that the mice with casein-induced inflammatory injury consuming a high-fat, high-protein diet gained weight normally, reduced serum adiponectin level and increased testosterone production by an upregulation of 3β-HSD expression. High-fat, high-protein diet attenuated the negative impact of casein-induced chronic inflammation on testicular steroidogenesis and sperm parameters.
Collapse
Affiliation(s)
- Jing-Lu Zhao
- Department of Developmental and Regenerative Biology, College of Life Science and Technology, Jinan University, Guangdong, Guangzhou 510632, China
| | - Yu-Yun Zhao
- College of Chemical and Biological Engineering, Hunan University of Science and Engineering, Hunan, Yongzhou 425199, China
| | - Wei-Jie Zhu
- Department of Developmental and Regenerative Biology, College of Life Science and Technology, Jinan University, Guangdong, Guangzhou 510632, China.
| |
Collapse
|
20
|
Melatonin ameliorates restraint stress-induced oxidative stress and apoptosis in testicular cells via NF-κB/iNOS and Nrf2/ HO-1 signaling pathway. Sci Rep 2017; 7:9599. [PMID: 28851995 PMCID: PMC5575312 DOI: 10.1038/s41598-017-09943-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/01/2017] [Indexed: 01/03/2023] Open
Abstract
Decline in semen quality has become a global public health concern. Psychological stress is common in the current modern society and is associated with semen decline. Increasing evidence demonstrated that melatonin has anti-apoptotic and antioxidant functions. Whether melatonin can ameliorate the damage in testes induced by psychological stress has never been investigated. Here, a mouse model of restraint stress demonstrated that melatonin normalized the sperm density decline, testicular cells apoptosis, and testicular oxidative stress in stressed male mice. Melatonin decreased reactive oxygen species (ROS) level, increased superoxide dismutase (SOD) and glutathione (GSH) activities, and downregulated inducible nitric oxide synthase (iNOS) and tumor necrosis factor-α (TNF-α) activities in stressed mice testes. Furthermore, melatonin reduced the stress-induced activation of the NF-κB signaling pathway by decreasing the phosphorylation of nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IκBα) and p65 nuclear translocation. In addition, melatonin upregulated the expression of anti-oxidant proteins including nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). Meanwhile, in vitro studies also demonstrated melatonin could reduce oxidative apoptosis of testicular cells. Collectively, melatonin mitigated psychological stress-induced spermatogenic damage, which provides evidence for melatonin as a therapy against sperm impairment associated with psychological stress.
Collapse
|
21
|
Chen G, Li H, Zhao Y, Zhu H, Cai E, Gao Y, Liu S, Yang H, Zhang L. Saponins from stems and leaves of Panax ginseng prevent obesity via regulating thermogenesis, lipogenesis and lipolysis in high-fat diet-induced obese C57BL/6 mice. Food Chem Toxicol 2017; 106:393-403. [PMID: 28599882 DOI: 10.1016/j.fct.2017.06.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 06/02/2017] [Accepted: 06/05/2017] [Indexed: 02/06/2023]
Abstract
In this study, high-fat diet (HFD)-induced obesity in mouse model was used to evaluate the dietary effect of saponins from stems and leaves of Panax ginseng (SLG), and to explore its mechanism of action in producing anti-obesity effects. The results indicate that SLG showed significant anti-obesity effects in diet-induced obese mice, represented by decreased serum levels of free fatty acids (FFA), total cholesterol (TC), triglycerides (TG), low-density lipoprotein (LDL)-cholesterol, glucose, leptin and insulin, as well as a reduction in overall body and liver weight, epididymal adipose tissue weight, and food efficiency, and inhibition of abnormal increases in acyl carnitine levels normally caused by an HFD. Additionally, the down-regulated expression of PPARγ, FAS, CD36, FATP2 and up-regulated expression of CPT-1, UCP-2, PPARα, HSL, and ATGL in liver tissue was induced by SLG. In addition, the SLG groups showed decreased PPARγ, aP2 and leptin mRNA levels and increased expression of PPARα, PGC-1α, UCP-1 and UCP-3 genes in adipose tissues, compared with the HFD group. In short, SLG may play a key role in producing anti-obesity effects in mice fed an HFD, and its mechanism may be related to regulation of thermogenesis, lipogenesis and lipolysis.
Collapse
Affiliation(s)
- Guilin Chen
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | - Haijun Li
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yan Zhao
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China.
| | - Hongyan Zhu
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | - Enbo Cai
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | - Yugang Gao
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China.
| | - Shuangli Liu
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | - He Yang
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | - Lianxue Zhang
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| |
Collapse
|
22
|
Oliveira PF, Sousa M, Silva BM, Monteiro MP, Alves MG. Obesity, energy balance and spermatogenesis. Reproduction 2017; 153:R173-R185. [DOI: 10.1530/rep-17-0018] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/04/2017] [Accepted: 03/09/2017] [Indexed: 01/01/2023]
Abstract
Obesity has grown to pandemic proportions. It affects an increasing number of children, adolescents and young adults exposed to the silent comorbidities of this disorder for a longer period. Infertility has arisen as one important comorbidity associated with the energy dysfunction promoted by obesity. Spermatogenesis is a highly regulated process that is determined by specific energetic requirements. The reproductive potential of males relies on hormonal-dependent and -independent stimuli that control sperm quality. There are conflicting data concerning the impact of male overweight and obesity on sperm quality, as well as on the possible paternal-induced epigenetic trait inheritance of obesity. In addition, it remains a matter of debate whether massive weight loss induced by lifestyle interventions, drugs or bariatric surgery may or may not benefit obese men seeking fatherhood. Herein, we propose to discuss how energy balance may modulate hormonal signalling and sperm quality in overweight and obese men. We also discuss some molecular mechanisms that mediate obesity-related dysfunction in male reproductive system and how paternal obesity may lead to trait inheritance. Finally, we will discuss how lifestyle modifications and sustained weight loss, particularly the loss achieved by bariatric surgery, may revert some of the deleterious effects of obesity in men and their offspring.
Collapse
|