1
|
Badejogbin OC, Chijioke-Agu OE, Olubiyi MV, Agunloye MO. Pathogenesis of testicular dysfunction in diabetes: exploring the mechanism and therapeutic interventions. J Assist Reprod Genet 2025; 42:367-379. [PMID: 39625650 PMCID: PMC11871280 DOI: 10.1007/s10815-024-03314-3] [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: 07/23/2024] [Accepted: 11/01/2024] [Indexed: 03/01/2025] Open
Abstract
Diabetes mellitus is a global epidemic contributing to the rising male infertility rates. Addressing testicular dysfunction in diabetic patients requires a multimodal strategy encompassing medication, lifestyle changes, early diagnosis, and innovative treatments targeting specific biochemical pathways. This review explores the mechanisms of diabetes-induced testicular dysfunction and potential intervention targets. A comprehensive literature search was conducted using PubMed, Science Direct, Google Scholar, and Web of Science with keywords related to diabetes and testicular dysfunction. Diabetes leads to reduced testosterone synthesis, decreased spermatogenesis, increased germ cell apoptosis, and damage to Leydig and Sertoli cells. Mechanisms involved in the pathogenesis of diabetes-induced testicular dysfunction include: hyperglycaemia oxidative stress, inflammation, apoptosis and disrupted hormone levels among others. Targeting biomolecular regulators involved in the pathogenic pathways offers a promising therapeutic avenue. Additionally, exploring plant-based therapies as alternative treatments shows potential in alleviating testicular dysfunction in diabetes. Implementing a comprehensive approach combining diagnostics, pharmacological interventions, and lifestyle modifications is crucial in managing testicular dysfunction in diabetic individuals. Future research directions suggest the need for large-scale clinical trials, personalized medicine strategies, and innovative technologies to address and mitigate testicular dysfunction in diabetic populations effectively.
Collapse
Affiliation(s)
- Olabimpe Caroline Badejogbin
- Department of Physiology, School of Basic Medical Sciences, Babcock University, Ilishan-Remo, Ogun State, Nigeria
| | | | | | - Mary Olaoluwa Agunloye
- Department of Physiology, Kampala International University, Western Campus, Ishaka, Uganda.
| |
Collapse
|
2
|
Papadimitriou K, Mousiolis AC, Mintziori G, Tarenidou C, Polyzos SA, Goulis DG. Hypogonadism and nonalcoholic fatty liver disease. Endocrine 2024; 86:28-47. [PMID: 38771482 DOI: 10.1007/s12020-024-03878-1] [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: 01/15/2024] [Accepted: 05/12/2024] [Indexed: 05/22/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD), recently proposed to be renamed to metabolic dysfunction-associated steatotic liver disease (MASLD), is a major global public health concern, affecting approximately 25-30% of the adult population and possibly leading to cirrhosis, hepatocellular carcinoma, and liver transplantation. The liver is involved in the actions of sex steroids via their hepatic metabolism and production of the sex hormone-binding globulin (SHBG). Liver disease, including NAFLD, is associated with reproductive dysfunction in men and women, and the prevalence of NAFLD in patients with hypogonadism is considerable. A wide spectrum of possible pathophysiological mechanisms linking NAFLD and male/female hypogonadism has been investigated. As therapies targeting NAFLD may impact hypogonadism in men and women, and vice versa, treatments of the latter may affect NAFLD, and an insight into their pathophysiological pathways is imperative. This paper aims to elucidate the complex association between NAFLD and hypogonadism in men and women and discuss the therapeutic options and their impact on both conditions.
Collapse
Affiliation(s)
- Kasiani Papadimitriou
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Athanasios C Mousiolis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gesthimani Mintziori
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
3
|
Ansari M. Recent strategies to mitigate reproductive aging in male broiler breeders: A review. Anim Reprod Sci 2024; 268:107570. [PMID: 39068813 DOI: 10.1016/j.anireprosci.2024.107570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
The continued improvement of genetics, nutrition, and management has resulted in rapid growth, better feed efficiency, and higher meat yield with competitive prices in the broiler industry. Nowadays, however, it is well-documented that productive traits and fertility are negatively correlated, and male broiler breeders are exposed to a fertility decline after 45 wk of age. Considering a low male-to-female ratio in breeder flocks, roosters have a prominent impact on flock fertility. Consequently, strategies to maintain the fertility of male broiler breeders could guarantee the reproductive performance of commercial herds. Understanding reproductive aging demands deep insights into its molecular and physiological mechanisms. Over-weighting, Sertoli and Leydig cell dysfunctions, compromised antioxidant capacity, imbalance in sexual hormones, and epididymal lithiasis are among candidate culprits associated with reproductive aging in roosters. Nutritional and managing strategies have been successfully applied to modulate body weight, improve sperm fatty acid profile and antioxidant status, and boost spermatogenic and steroidogenic pathways. The current review characterizes the physiology and biochemistry of reproductive aging in male broiler breeders and then highlights strategies and their underlying mechanisms to mitigate this failure. In summary, applying one or more of the abovementioned strategies might result in consistent post-peak reproduction and benefit producers in the poultry industry.
Collapse
Affiliation(s)
- Mahdi Ansari
- Department of Animal and Poultry Physiology, Faculty of Animal Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan 49138-15739, Iran.
| |
Collapse
|
4
|
Jing J, Ouyang L, Zhang H, Liang K, Ma R, Ge X, Tang T, Zhao S, Xue T, Shen J, Ma J, Li Z, Wu J, Yang Y, Zhao W, Zheng L, Qian Z, Sun S, Ge Y, Chen L, Li C, Yao B. Omega-3 polyunsaturated fatty acids and its metabolite 12-HEPE rescue busulfan disrupted spermatogenesis via target to GPR120. Cell Prolif 2024; 57:e13551. [PMID: 37743695 PMCID: PMC10849791 DOI: 10.1111/cpr.13551] [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: 11/25/2022] [Revised: 08/26/2023] [Accepted: 09/08/2023] [Indexed: 09/26/2023] Open
Abstract
Busulfan is an antineoplastic, which is always accompanied with the abnormal of spermatogonia self-renewal and differentiation. It has been demonstrated that the omega-3 polyunsaturated fatty acids (PUFAs) benefits mature spermatozoa. However, whether omega-3 can protect endogenous spermatogonia and the detailed mechanisms are still unclear. Evaluate of spermatogenesis function (in vivo) were examined by histopathological analysis, immunofluorescence staining, and western blotting. The levels of lipid metabolites in testicular tissue were determined via liquid chromatography. We investigated the effect of lipid metabolites on Sertoli cells provided paracrine factors to regulate spermatogonia proliferation and differentiation using co-culture system. In our study, we showed that omega-3 PUFAs significantly improved the process of sperm production and elevated the quantity of both undifferentiated Lin28+ spermatogonia and differentiated c-kit+ spermatogonia in a mouse model where spermatogenic function was disrupted by busulfan. Mass spectrometry revealed an increase in the levels of several omega-3 metabolites in the testes of mice fed with omega-3 PUFAs. The eicosapentaenoic acid metabolite 12-hydroxyeicosapentaenoic acid (12-HEPE) up-regulated bone morphogenic protein 4 (BMP4) expression through GPR120-ERK1/2 pathway activation in Sertoli cells and restored spermatogonia proliferation and differentiation. Our study provides evidence that omega-3 PUFAs metabolite 12-HEPE effectively protects spermatogonia and reveals that GPR120 might be a tractable pharmacological target for fertility in men received chemotherapy or severe spermatogenesis dysfunction.
Collapse
Affiliation(s)
- Jun Jing
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Lei Ouyang
- Department of Reproductive Medicine, Affiliated Jinling Hospital, The First School of Clinical MedicineSouthern Medical UniversityNanjingChina
| | - Hong Zhang
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Kuan Liang
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, The First School of Clinical MedicineSouthern Medical UniversityNanjingChina
| | - Rujun Ma
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Xie Ge
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Ting Tang
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Shanmeizi Zhao
- School of Life ScienceNanjing Normal UniversityNanjingChina
| | - Tongmin Xue
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Reproductive Medical Center, Clinical Medical College (Northern Jiangsu People's Hospital)Yangzhou UniversityYangzhouChina
| | - Jiaming Shen
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Jinzhao Ma
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Zhou Li
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Jing Wu
- Core Laboratory, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Yang Yang
- Basic Medical Laboratory, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Wei Zhao
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Lu Zheng
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Zhang Qian
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Shanshan Sun
- School of Life ScienceNanjing Normal UniversityNanjingChina
| | - Yifeng Ge
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Li Chen
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Chaojun Li
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
| | - Bing Yao
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, The First School of Clinical MedicineSouthern Medical UniversityNanjingChina
- School of Life ScienceNanjing Normal UniversityNanjingChina
| |
Collapse
|
5
|
Huang R, Chen J, Guo B, Jiang C, Sun W. Diabetes-induced male infertility: potential mechanisms and treatment options. Mol Med 2024; 30:11. [PMID: 38225568 PMCID: PMC10790413 DOI: 10.1186/s10020-023-00771-x] [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: 08/12/2023] [Accepted: 12/14/2023] [Indexed: 01/17/2024] Open
Abstract
Male infertility is a physiological phenomenon in which a man is unable to impregnate a fertile woman during a 12-month period of continuous, unprotected sexual intercourse. A growing body of clinical and epidemiological evidence indicates that the increasing incidence of male reproductive problems, especially infertility, shows a very similar trend to the incidence of diabetes within the same age range. In addition, a large number of previous in vivo and in vitro experiments have also suggested that the complex pathophysiological changes caused by diabetes may induce male infertility in multiple aspects, including hypothalamic-pituitary-gonadal axis dysfunction, spermatogenesis and maturation disorders, testicular interstitial cell damage erectile dysfunction. Based on the above related mechanisms, a large number of studies have focused on the potential therapeutic association between diabetes progression and infertility in patients with diabetes and infertility, providing important clues for the treatment of this population. In this paper, we summarized the research results of the effects of diabetes on male reproductive function in recent 5 years, elaborated the potential pathophysiological mechanisms of male infertility induced by diabetes, and reviewed and prospected the therapeutic measures.
Collapse
Affiliation(s)
- Runchun Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000
| | - Jiawang Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000
| | - Buyu Guo
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000
| | - Chenjun Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000
| | - Weiming Sun
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000.
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
6
|
El Helew EA, Hamed WS, Moustafa AM, Sakkara ZA. Structural changes in testes of Streptozotocin induced diabetic rats and possible protective effect of royal jelly: light and electron microscopic study. Ultrastruct Pathol 2024; 48:1-15. [PMID: 37927047 DOI: 10.1080/01913123.2023.2277170] [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: 09/14/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023]
Abstract
Diabetes mellitus (DM) is one of the most common metabolic diseases causing damage in many organs in the body including the testes. Royal Jelly (RJ) is one of the honey bee products that has antioxidant, anti-inflammatory and antidiabetic properties. This study was performed to evaluate the changes in the microscopic structure of the testes in Streptozotocin (STZ)-induced diabetic rats, and the possible protective role of RJ. 60 adult male albino rats were divided into three groups. Group I Control group, Group II STZ group, and Group III STZ+RJ group. Group II received a single dose of STZ (50 mg/kg) by intraperitoneal injection. Group III received a single dose of STZ as in the second group then received RJ orally by intragastric tube in dose of (100 mg/kg/day) for 4 weeks after confirmation of diabetes. Light and electron microscopic studies were performed. Group II revealed marked structural changes affecting seminiferous tubules with sever reduction in germinal epithelium and loss of mature spermatozoa in their lumina. The interstitial tissue revealed degenerated Leydig cells and congested blood vessels. Mallory trichrome stained section of group II revealed marked increase in the amount of collagen fibers. Group III revealed highly preserved testicular architecture almost near to that appeared in the control group except few tubules that were damaged. In conclusion, RJ protected the testicular structure from the damaging effect of diabetic oxidative stress through its antioxidant effect thus preserving male fertility.
Collapse
Affiliation(s)
- Eman A El Helew
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Wafaa S Hamed
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amal M Moustafa
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Zeinab A Sakkara
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
7
|
Emad NA, Sultana Y, Aqil M, Saleh A, Al kamaly O, Nasr FA. Omega-3 fatty acid-based self-microemulsifying drug delivery system (SMEDDS) of pioglitazone: Optimization, in vitro and in vivo studies. Saudi J Biol Sci 2023; 30:103778. [PMID: 37663396 PMCID: PMC10470285 DOI: 10.1016/j.sjbs.2023.103778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 09/05/2023] Open
Abstract
Pioglitazone (PGL) is an effective insulin sensitizer, however, side effects such as accumulation of subcutaneous fat, edema, and weight gain as well as poor oral bioavailability limit its therapeutic potential for oral delivery. Recent studies have shown that combination of both, PGL and fish oil significantly reduce fasting plasma glucose, improve insulin resistance, and mitigate pioglitazone-induced subcutaneous fat accumulation and weight gain. Nevertheless, developing an effective oral drug delivery system for administration of both medications have not been explored yet. Thus, this study aimed to develop a self-micro emulsifying drug delivery system (SMEDDS) for the simultaneous oral administration of PGL and fish oil. SMEDDS was developed using concentrated fish oil,Tween® 80, and Transcutol HP and optimized by central composite design (CCD). The reconstituted, optimized PGL-SMEDDS exhibited a globule size of 142 nm, a PDI of 0.232, and a zeta potential of -20.9 mV. The in-vitro drug release study of the PGL-SMEDDS showed a first-order model kinetic release and demonstrated remarkable 15-fold enhancement compared to PGL suspension. Additionally, following oral administration in fasting albino Wistar rats, PGL-SMEDDS exhibited 3.4-fold and 1.4-fold enhancements in the AUC0-24h compared to PGL suspension and PGL marketed product. The accelerated stability testing showed that the optimized SMEDDS formulation was stable over a three-month storage period. Taken together, our findings demonstrate that the developed fish oil-based SMEDDS for PGL could serve as effective nanoplatforms for the oral delivery of PGL, warranting future studies to explore its synergistic therapeutic potential in rats.
Collapse
Affiliation(s)
- Nasr A. Emad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed University), M. B. Road, New Delhi 110062, India
| | - Yasmin Sultana
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed University), M. B. Road, New Delhi 110062, India
| | - Mohd Aqil
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed University), M. B. Road, New Delhi 110062, India
| | - Asmaa Saleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O Box 84428, Riyadh 11671, Saudi Arabia
| | - Omkulthom Al kamaly
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O Box 84428, Riyadh 11671, Saudi Arabia
| | - Fahd A Nasr
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Tsampoukas G, Tharakan T, Narayan Y, Khan F, Cayetano A, Papatsoris A, Buchholz N, Minhas S. Investigating the therapeutic options for diabetes-associated male infertility as illustrated in animal experimental models. Andrologia 2022; 54:e14521. [PMID: 35934995 DOI: 10.1111/and.14521] [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: 03/31/2022] [Revised: 05/15/2022] [Accepted: 05/25/2022] [Indexed: 11/30/2022] Open
Abstract
Diabetes is a rising global health concern and an increasingly common cause of male infertility. Although the definitive pathophysiological mechanisms underpinning the association between diabetes and infertility is unclear, there are several animal studies showing diabetes to be a detrimental factor on reproductive health through apoptosis, oxidative stress and impairment of steroidogenesis. Furthermore, as reflected in animal models, antidiabetic strategies and relevant treatments are beneficial in the management of infertile men with diabetes as the recovery of euglycemic status affects positively the spermatogenesis. However, the available data are still evolving and specific conclusion in human populations are not possible yet. In this review, we are discussing the current literature concerning the association of diabetes and male infertility, focusing on the therapeutic approach as illustrated in animals' models.
Collapse
Affiliation(s)
- Georgios Tsampoukas
- U-merge Ltd. (Urology for emerging countries), London, UK.,Department of Urology, Great Western Hospital NHS Trust, Swindon, UK
| | - Tharu Tharakan
- Department of Urology, Imperial Healthcare NHS Trust, Charing Cross Hospital, London, UK.,Section of Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Yash Narayan
- Department of Surgery, Cairns Hospital, Cairns North, Queensland, Australia
| | - Faisal Khan
- Department of Urology, North Devon Hospital, Barnstaple, UK
| | - Axel Cayetano
- Department of Urology, Imperial Healthcare NHS Trust, Charing Cross Hospital, London, UK
| | - Athanasios Papatsoris
- U-merge Ltd. (Urology for emerging countries), London, UK.,Department of Urology, Sismanoglio University Hospital of Athens, Athens, Greece
| | - Noor Buchholz
- U-merge Ltd. (Urology for emerging countries), London, UK
| | - Suks Minhas
- Department of Urology, Imperial Healthcare NHS Trust, Charing Cross Hospital, London, UK
| |
Collapse
|
9
|
Luo M, Liao B, Ma D, Wang J, Wang J, Liu J, Lei X, Cai Y, Tang L, Zhao L, Long S, Yang F, Lei X. Dendrobium nobile-derived polysaccharides ameliorate spermatogenic disorders in mice with streptozotocin-induced diabetes through regulation of the glycolytic pathway. Int J Biol Macromol 2022; 216:203-212. [DOI: 10.1016/j.ijbiomac.2022.06.193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/01/2022] [Accepted: 06/28/2022] [Indexed: 01/17/2023]
|
10
|
Therapeutic Potential of Mesenchymal Stem Cells versus Omega n − 3 Polyunsaturated Fatty Acids on Gentamicin-Induced Cardiac Degeneration. Pharmaceutics 2022; 14:pharmaceutics14071322. [PMID: 35890218 PMCID: PMC9319609 DOI: 10.3390/pharmaceutics14071322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/28/2022] [Accepted: 06/17/2022] [Indexed: 01/27/2023] Open
Abstract
This study compared the cardioprotective action of mesenchymal stem cells (MSCs) and PUFAs in a rat model of gentamicin (GM)-induced cardiac degeneration. Male Wistar albino rats were randomized into four groups of eight rats each: group I (control group), group II (gentamicin-treated rats receiving gentamicin intraperitoneally (IP) at dose of 100 mg/kg/day for 10 consecutive days), group III (gentamicin and PUFA group receiving gentamicin IP at dose of 100 mg/kg/day for 10 consecutive days followed by PUFAs at a dose of 100 mg/kg/day for 4 weeks), and group IV (gentamicin and MSC group receiving gentamicin IP at dose of 100 mg/kg/day followed by a single dose of MSCs (1 × 106)/rat IP). Cardiac histopathology was evaluated via light and electron microscopy. Immunohistochemical detection of proliferating cell nuclear antigen (PCNA), caspase-3 (apoptosis), Bcl2, and Bax expression was performed. Moreover, cardiac malonaldehyde (MDA) content, catalase activity, and oxidative stress parameters were biochemically evaluated. Light and electron microscopy showed that both MSCs and PUFAs had ameliorative effects. Their actions were mediated by upregulating PCNA expression, downregulating caspase-3 expression, mitigating cardiac MDA content, catalase activity, and oxidative stress parameters. MSCs and PUFAs had ameliorative effects against gentamicin-induced cardiac degeneration, with MSCs showing higher efficacy compared to PUFAs.
Collapse
|
11
|
Alfarhan MW, Al-Hussaini H, Kilarkaje N. Role of PPAR-γ in diabetes-induced testicular dysfunction, oxidative DNA damage and repair in leptin receptor-deficient obese type 2 diabetic mice. Chem Biol Interact 2022; 361:109958. [PMID: 35472412 DOI: 10.1016/j.cbi.2022.109958] [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: 12/21/2021] [Revised: 04/06/2022] [Accepted: 04/20/2022] [Indexed: 11/03/2022]
Abstract
The testis expresses peroxisome proliferator-activated receptor-γ (PPAR-γ), but its involvement in regulating diabetes-induced testicular dysfunction and DNA damage repair is not known. Pioglitazone-induced activation of PPAR-γ for 12 weeks in db/db obese diabetic mice increases bodyweights and reduces blood glucose levels, but PPAR-γ inhibition by 2-chloro-5-nitro-N-phenylbenzamide does not alter these parameters; instead, improves testis and epididymis weights and sperm count. Neither activation nor inhibition of PPAR-γ normalizes the diabetes-induced seminiferous epithelial degeneration. The PPAR-γ activation normalizes testicular lipid peroxidation, but its inhibition reduces lipid peroxidation and oxidative DNA damage (8-oxo-dG) in diabetic mice. As a response to diabetes-induced oxidative DNA damage, the base-excision repair (BER) mechanism proteins- 8-oxoguanine DNA glycosylases (OGG1/2) and X-ray repair cross-complementing protein-1 (XRCC1) increase, whereas the redox-factor-1 (REF1), DNA polymerase (pol) δ and poly (ADP-ribose) polymerase-1 (PARP1) show a tendency to increase suggesting an attempt to repair the oxidative DNA damage. The PPAR-γ stimulation inhibits OGG2, DNA pol δ, and XRCC1 in diabetic mice testes, but PPAR-γ inhibition reduces oxidative DNA damage and normalizes BER protein levels. In conclusion, type 2 diabetes negatively affects testicular structure and function and increases oxidative DNA damage and BER protein levels due to increased DNA damage. The PPAR-γ modulation does not significantly affect the structural changes in the testis. The PPAR-γ stimulation aggravates diabetes-induced effects on testis, including oxidative DNA damage and BER proteins, but PPAR-γ inhibition marginally recovers these diabetic effects indicating the involvement of the receptor in the reproductive effects of diabetes.
Collapse
Affiliation(s)
| | - Heba Al-Hussaini
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| | | |
Collapse
|
12
|
Jiang Q, Linn T, Drlica K, Shi L. Diabetes as a potential compounding factor in COVID-19-mediated male subfertility. Cell Biosci 2022; 12:35. [PMID: 35307018 PMCID: PMC8934536 DOI: 10.1186/s13578-022-00766-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 02/21/2022] [Indexed: 01/09/2023] Open
Abstract
Recent work indicates that male fertility is compromised by SARS-CoV-2 infection. Direct effects derive from the presence of viral entry receptors (ACE2 and/or CD147) on the surface of testicular cells, such as spermatocytes, Sertoli cells, and Leydig cells. Indirect effects on testis and concentrations of male reproductive hormones derive from (1) virus-stimulated inflammation; (2) viral-induced diabetes, and (3) an interaction between diabetes and inflammation that exacerbates the deleterious effect of each perturbation. Reproductive hormones affected include testosterone, luteinizing hormone, and follicle-stimulating hormone. Reduction of male fertility is also observed with other viral infections, but the global pandemic of COVID-19 makes demographic and public health implications of reduced male fertility of major concern, especially if it occurs in the absence of serious symptoms that would otherwise encourage vaccination. Clinical documentation of COVID-19-associated male subfertility is now warranted to obtain quantitative relationships between infection severity and subfertility; mechanistic studies using animal models may reveal ways to mitigate the problem. In the meantime, the possibility of subfertility due to COVID-19 should enter considerations of vaccine hesitancy by reproductive-age males.
Collapse
Affiliation(s)
- Qingkui Jiang
- grid.430387.b0000 0004 1936 8796Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers The State University of New Jersey, Newark, NJ USA
| | - Thomas Linn
- grid.8664.c0000 0001 2165 8627Clinical Research Unit, Centre of Internal Medicine, Justus-Liebig-University (JLU), Giessen, Germany
| | - Karl Drlica
- grid.430387.b0000 0004 1936 8796Public Health Research Institute and Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers The State University of New Jersey, Newark, NJ USA
| | - Lanbo Shi
- grid.430387.b0000 0004 1936 8796Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers The State University of New Jersey, Newark, NJ USA
| |
Collapse
|
13
|
Pourazadi L, Sharafi M, Torshizi MAK, Shahverdi A, Alizadeh A. Modulatory effects of pioglitazone as a ligand for the peroxisome proliferator-activated receptor on semen quality and fertility potential of broiler breeder roosters. Poult Sci 2022; 101:101795. [PMID: 35349953 PMCID: PMC8965139 DOI: 10.1016/j.psj.2022.101795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/23/2022] [Accepted: 02/13/2022] [Indexed: 11/25/2022] Open
Abstract
Fertility potential in roosters is a crucial topic in broiler breeder reproduction which is thought to be associated with age. This study aims to investigate effects of 2 levels of pioglitazone (PIO) supplementation on peroxisome proliferator-activated receptor gamma (PPAR-γ) expression, semen quality, and fertility parameters of aged broiler breeder roosters. The efficacy of PIO was divided into 2 sections: receptor-dependent and receptor-independent. Expression of PPAR-γ mRNA and protein was assessed in sperm to monitor receptor-dependent actions. Sperm motility, velocity parameters, viability, mitochondrial activity, and apoptosis were assessed for the receptor-independent actions. Broiler breeder roosters were randomly assigned to 3 groups: 1) control received a basal diet (CTRL); 2) PIO-5 received a basal diet supplemented with 5 mg PIO/bird/day, and 3) PIO-10 received a basal diet supplemented with 10 mg PIO/bird/day. In addition, semen samples were collected from 24 Ross broiler breeder roosters at 30, 43, and 53 wk of age. Effects of PIO were significant in terms of total motility, straight-line velocity, mitochondrial activity, and apoptosis (P ≤ 0.05). Total motility, straight-line velocity and mitochondrial activity improved in both PIO groups (P ≤ 0.05) along with a significant reduction in early and late apoptosis in the PIO groups (P ≤ 0.05). Pioglitazone addition affected total motility, mitochondrial activity, early apoptosis and late apoptosis in a linearly and quadratically manner (P < 0.05). PPAR-γ mRNA and protein expression were not significantly upregulated by the different doses of PIO (P > 0.05). Similarly, fertility performance was not significantly changed in the PIO groups (P > 0.05). Moreover, PIO improved mitochondrial activity and decreased the apoptosis rate in the sperm of aged broiler breeder roosters. These improvements were associated with the receptor-independent actions of PIO and the mechanism of action of PIO did not appear to be affected by the PPAR-γ receptor in broiler breeder roosters.
Collapse
Affiliation(s)
- Laya Pourazadi
- Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, Tehran 14115-336, Iran
| | - Mohsen Sharafi
- Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, Tehran 14115-336, Iran.
| | | | - Abdolhossein Shahverdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran 16635-148, Iran
| | - AliReza Alizadeh
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran 16635-148, Iran
| |
Collapse
|
14
|
Khazaei MR, Gravandi E, Ghanbari E, Niromand E, Khazaei M. Trifolium pratense extract increases testosterone and improves sperm characteristics and antioxidant status in diabetic rats. Biotech Histochem 2022; 97:576-583. [PMID: 35164615 DOI: 10.1080/10520295.2022.2039766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Male reproductive dysfunction is a common complication of diabetes mellitus. Trifolium pratense exhibits antioxidant and antidiabetic effects. We investigated the effects of an extract of T. pratense on serum antioxidant status, sperm characteristics, testicular tissue changes and testosterone level in diabetic rats. Male Wistar rats were divided into six groups: 1, untreated control; 2, diabetic; 3 and 4, 100 or 200 mg/kg T. pratense extract treated, respectively; 5 and 6, diabetic 100 or 200 mg/kg T. pratense extract treated, respectively. Diabetes was induced by intraperitoneal injection of streptozotocin. After 3 weeks, serum glucose, testosterone and nitric oxide (NO); sperm parameters; testicular histology and total antioxidant capacity (TAC) were evaluated. In diabetic rats treated with T. pratense extract, sperm motility, count and viability, as well as TAC and testosterone were increased significantly compared to untreated diabetic rats, while serum NO and bcl-2 and p53 expression was decreased significantly compared to untreated diabetic rats. T. pratense extract reduced testicular tissue destruction caused by diabetes.
Collapse
Affiliation(s)
- Mohammad Rasool Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Gravandi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Ghanbari
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Niromand
- Department of Internal Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
15
|
El-Sayed K, Ali DA, Maher SA, Ghareeb D, Selim S, Albogami S, Fayad E, Kolieb E. Prophylactic and Ameliorative Effects of PPAR-γ Agonist Pioglitazone in Improving Oxidative Stress, Germ Cell Apoptosis and Inflammation in Gentamycin-Induced Testicular Damage in Adult Male Albino Rats. Antioxidants (Basel) 2022; 11:antiox11020191. [PMID: 35204074 PMCID: PMC8868260 DOI: 10.3390/antiox11020191] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 02/07/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPAR-γ) is ubiquitously expressed in testicular tissue and plays a crucial role in regulating various physiological processes. Pioglitazone (PIO) is one of the PPAR-γ agonists, having anti-oxidant and anti-inflammatory effects. Patients on gentamycin treatment may undergo serious side effects such as testicular damage. To the best of our knowledge, this was the first study to investigate the possible protective anti-inflammatory and anti-apoptotic effects of PIO on gentamycin-induced testicular damage. Fifty adult male Wistar albino rats included in the study as the control group (CTL) received normal saline; a gentamycin-induced testicular damage group (GM) received gentamycin (100 mg/kg); PIO5, PIO10, PIO20 groups received PIO at a dose of 5, 10, and 20 mg/ kg, respectively, for 21 days, and gentamycin was started at day 15 of the experiment for 6 days. The parameters of spermatozoa and histopathological alterations in the testes were significantly improved in the PIO20 group. Moreover, MDA levels, inflammatory mediators, and apoptotic Bax expression were decreased. The activity of glutathione peroxidase, catalase, total antioxidant capacity, and anti-apoptotic Bcl-2 genes expression were increased. It was concluded that PIO20 could protect against gentamycin-induced testicular damage in Wistar rats through its anti-oxidant, anti-inflammatory, and antiapoptotic effects.
Collapse
Affiliation(s)
- Karima El-Sayed
- Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Dina A. Ali
- Clinical Pharmacology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Shymaa Ahmed Maher
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
- Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Dalia Ghareeb
- Clinical Pathology Department, Faculty of Medicine, Suez University, Suez 41522, Egypt;
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72341, Saudi Arabia;
| | - Sarah Albogami
- Department of Biotechnology, Faculty of Sciences, Taif University, Taif 21944, Saudi Arabia; (S.A.); (E.F.)
| | - Eman Fayad
- Department of Biotechnology, Faculty of Sciences, Taif University, Taif 21944, Saudi Arabia; (S.A.); (E.F.)
| | - Eman Kolieb
- Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
- Correspondence: ; Tel.: +20-1006738513
| |
Collapse
|
16
|
Akarca Dizakar SÖ, Saribas GS, Tekcan A. Effects of ellagic acid in the testes of streptozotocin induced diabetic rats. Drug Chem Toxicol 2021; 45:2123-2130. [PMID: 33832387 DOI: 10.1080/01480545.2021.1908714] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Diabetes mellitus (DM) is a serious and common in the world health problem that leads to different complications. Changes in oxidative stress and antioxidant capacity play an important role in the pathogenesis of DM. The purpose of this study was to investigate ellagic acid (EA) treatment in diabetes induced testicular damage. In our study, 24 male Sprague Dawley rats were divided into four groups. Group 1: Control (n = 6), Group 2: EA (n = 6), Group 3: Diabet (n = 6), Group 4: Diabet + EA (n = 6). Diabetes was induced by intraperitoneal injection of streptozocin (STZ) (55 mg/kg) to group 3 and 4. EA was given 100 mg/kg/day group 2 and 4 for 35 days by oral gavage. We used that Hematoxylen-Eosin (H&E) and Johnsen's scoring to determine histological change. The terminal-deoxynucleoitidyl-transferase mediated nick end-labeling assay (TUNEL) was used for apoptosis. Oxidative stress markers were determined by qRT-PCR and immunexpression of Nrf2 was evaluated in testicular tissue. In conclusion, EA administration on the diabetes model has changed the histopathological features, apopotosis and oxidative stress marker genes in the testis and may have an effect on the reduction of diabetes induced testicular damage.
Collapse
Affiliation(s)
| | - Gulistan Sanem Saribas
- Department of Histology and Embryology, Faculty of Medicine, Ahi Evran University, Kirsehir, turkey
| | - Akın Tekcan
- Department of Medical Biology Faculty of Medicine, Amasya University, Amasya, Turkey
| |
Collapse
|
17
|
Antonuccio P, Marini HR, Micali A, Romeo C, Granese R, Retto A, Martino A, Benvenga S, Cuzzocrea S, Impellizzeri D, Di Paola R, Fusco R, Cervellione RM, Minutoli L. The Nutraceutical N-Palmitoylethanolamide (PEA) Reveals Widespread Molecular Effects Unmasking New Therapeutic Targets in Murine Varicocele. Nutrients 2021; 13:nu13030734. [PMID: 33668991 PMCID: PMC7996616 DOI: 10.3390/nu13030734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/26/2022] Open
Abstract
Varicocele is an age-related disease with no current medical treatments positively impacting infertility. Toll-like receptor 4 (TLR4) expression is present in normal testis with an involvement in the immunological reactions. The role of peroxisome proliferator-activated receptor-α (PPAR-α), a nuclear receptor, in fertility is still unclear. N-Palmitoylethanolamide (PEA), an emerging nutraceutical compound present in plants and animal foods, is an endogenous PPAR-α agonist with well-demonstrated anti-inflammatory and analgesics characteristics. In this model of mice varicocele, PPAR-α and TLR4 receptors’ roles were investigated through the administration of ultra-micronized PEA (PEA-um). Male wild-type (WT), PPAR-α knockout (KO), and TLR4 KO mice were used. A group underwent sham operation and administration of vehicle or PEA-um (10 mg/kg i.p.) for 21 days. Another group (WT, PPAR-α KO, and TLR4 KO) underwent surgical varicocele and was treated with vehicle or PEA-um (10 mg/kg i.p.) for 21 days. At the end of treatments, all animals were euthanized. Both operated and contralateral testes were processed for histological and morphometric assessment, for PPAR-α, TLR4, occludin, and claudin-11 immunohistochemistry and for PPAR-α, TLR4, transforming growth factor-beta3 (TGF-β3), phospho-extracellular signal-Regulated-Kinase (p-ERK) 1/2, and nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) Western blot analysis. Collectively, our data showed that administration of PEA-um revealed a key role of PPAR-α and TLR4 in varicocele pathophysiology, unmasking new nutraceutical therapeutic targets for future varicocele research and supporting surgical management of male infertility.
Collapse
Affiliation(s)
- Pietro Antonuccio
- Department of Human Pathology of Adult and Childhood, University of Messina, 98125 Messina, Italy; (P.A.); (C.R.); (R.G.); (A.R.); (A.M.)
| | - Herbert Ryan Marini
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (S.B.); (L.M.)
| | - Antonio Micali
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy;
| | - Carmelo Romeo
- Department of Human Pathology of Adult and Childhood, University of Messina, 98125 Messina, Italy; (P.A.); (C.R.); (R.G.); (A.R.); (A.M.)
| | - Roberta Granese
- Department of Human Pathology of Adult and Childhood, University of Messina, 98125 Messina, Italy; (P.A.); (C.R.); (R.G.); (A.R.); (A.M.)
| | - Annalisa Retto
- Department of Human Pathology of Adult and Childhood, University of Messina, 98125 Messina, Italy; (P.A.); (C.R.); (R.G.); (A.R.); (A.M.)
| | - Antonia Martino
- Department of Human Pathology of Adult and Childhood, University of Messina, 98125 Messina, Italy; (P.A.); (C.R.); (R.G.); (A.R.); (A.M.)
| | - Salvatore Benvenga
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (S.B.); (L.M.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (S.C.); (D.I.); (R.F.)
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (S.C.); (D.I.); (R.F.)
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (S.C.); (D.I.); (R.F.)
- Correspondence: ; Tel.: +39-090-2213655; Fax: +39-090-2213300
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (S.C.); (D.I.); (R.F.)
| | | | - Letteria Minutoli
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (S.B.); (L.M.)
| |
Collapse
|
18
|
Derkach KV, Bakhtyukov AA, Bayunova LV, Zorina II, Shpakov AO. Normalization of Testicular Steroidogenesis and Spermatogenesis in Male Rats with Type 2 Diabetes Mellitus under the Conditions of Metformin Therapy. DOKLADY BIOLOGICAL SCIENCES : PROCEEDINGS OF THE ACADEMY OF SCIENCES OF THE USSR, BIOLOGICAL SCIENCES SECTIONS 2020; 493:110-113. [PMID: 32894423 DOI: 10.1134/s0012496620040031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 12/19/2022]
Abstract
One of the complications of type 2 diabetes mellitus in men is steroidogenic and spermatogenic dysfunctions. There is evidence of a restoring effect of the antidiabetic drug metformin on them. We studied the effect of MF therapy (4 weeks, 200 mg/kg/day) on the hormonal parameters of the gonad axis and on the morphological characteristics of epididymal spermatozoa in male rats with a severe form of T2DM caused by a high-fat diet and a low-dose streptozotocin. It has been shown that MF therapy, along with the restoration of the metabolic parameters, normalizes the plasma levels of testosterone and leptin and the content of testosterone, its precursors, leptin and its receptors in the testes, and also increases sperm motility, which is reduced in T2DM. This is the result of both the systemic action of MF and its direct effect on testicular cells.
Collapse
Affiliation(s)
- K V Derkach
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223, St. Petersburg, Russia
| | - A A Bakhtyukov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223, St. Petersburg, Russia
| | - L V Bayunova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223, St. Petersburg, Russia
| | - I I Zorina
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223, St. Petersburg, Russia
| | - A O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223, St. Petersburg, Russia.
| |
Collapse
|
19
|
Santoro M, De Amicis F, Aquila S, Bonofiglio D. Peroxisome proliferator-activated receptor gamma expression along the male genital system and its role in male fertility. Hum Reprod 2020; 35:2072-2085. [PMID: 32766764 DOI: 10.1093/humrep/deaa153] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) acts as a ligand activated transcription factor and regulates processes, such as energy homeostasis, cell proliferation and differentiation. PPARγ binds to DNA as a heterodimer with retinoid X receptor and it is activated by polyunsaturated fatty acids and fatty acid derivatives, such as prostaglandins. In addition, the insulin-sensitizing thiazolidinediones, such as rosiglitazone, are potent and specific activators of PPARγ. PPARγ is present along the hypothalamic-pituitary-testis axis and in the testis, where low levels in Leydig cells and higher levels in Sertoli cells as well as in germ cells have been found. High amounts of PPARγ were reported in the normal epididymis and in the prostate, but the receptor was almost undetectable in the seminal vesicles. Interestingly, in the human and in pig, PPARγ protein is highly expressed in ejaculated spermatozoa, suggesting a possible role of PPARγ signaling in the regulation of sperm biology. This implies that both natural and synthetic PPARγ ligands may act directly on sperm improving its performance. Given the close link between energy balance and reproduction, activation of PPARγ may have promising metabolic implications in male reproductive functions. In this review, we first describe PPARγ expression in different compartments of the male reproductive axis. Subsequently, we discuss the role of PPARγ in both physiological and several pathological conditions related to the male fertility.
Collapse
Affiliation(s)
- Marta Santoro
- Department of Pharmacy, Health and Nutritional Sciences (Department of Excellence, Italian Law 232/2016), Arcavacata di Rende, Cosenza 87036, Italy.,Centro Sanitario, University of Calabria, Arcavacata di Rende, Cosenza 87036, Italy
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences (Department of Excellence, Italian Law 232/2016), Arcavacata di Rende, Cosenza 87036, Italy
| | - Saveria Aquila
- Department of Pharmacy, Health and Nutritional Sciences (Department of Excellence, Italian Law 232/2016), Arcavacata di Rende, Cosenza 87036, Italy.,Centro Sanitario, University of Calabria, Arcavacata di Rende, Cosenza 87036, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences (Department of Excellence, Italian Law 232/2016), Arcavacata di Rende, Cosenza 87036, Italy.,Centro Sanitario, University of Calabria, Arcavacata di Rende, Cosenza 87036, Italy
| |
Collapse
|
20
|
Abdel-Aziz AM, Abozaid SMM, Yousef RKM, Mohammed MM, Khalaf HM. Fenofibrate ameliorates testicular damage in rats with streptozotocin-induced type 1 diabetes: role of HO-1 and p38 MAPK. Pharmacol Rep 2020; 72:1645-1656. [PMID: 32515004 DOI: 10.1007/s43440-020-00096-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/05/2020] [Accepted: 03/14/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Since diabetes mellitus type-1 (DM-1) induces testicular oxidative and inflammatory damage with finally an ultimate male infertility, and as fenofibrate (FEN) plays an important antioxidant and anti-inflammatory role, the aim of the present study was to investigate the effects of FEN on diabetes-induced reproductive damage and clarifying the underlying related mechanisms. METHODS DM-1 was induced in male Wistar rats by a single intraperitoneal injection of streptozotocin (50 mg/kg). FEN (100 mg/kg/day, orally) was administrated to diabetic rats for 4 weeks. Testicular damage was detected by estimation of both testicular and body weights, assessment of serum testosterone, testicular oxidative stress parameters (malondialdehyde and nitric oxide levels) and testicular oxidant defenses (reduced glutathione, superoxide dismutase and hemeoxygenase-1). Expressions of the inflammatory markers (inducible nitric oxide synthase, p38 mitogen-activated protein kinase (MAPK), tumor necrosis factor alpha, interleukin-6 and apoptotic marker (caspase-3) were evaluated in testicular tissue. Our results were confirmed by histopathological examination of testicular tissues. RESULTS Diabetic testicular damage was proved by both biochemical and histopathological examinations. FEN treatment reversed diabetic testicular damage; normalized the serum testosterone level, improved anti-oxidative capacity, ameliorated the pro-inflammatory cytokine expression in testicular tissue with the down regulation of p38 MAPK mediated-testicular apoptosis. CONCLUSION FEN treatment exerted a protective effect against streptozotocin-induced diabetic reproductive dysfunction not only through its powerful antioxidant and hypoglycemic effects, but also through its anti-inflammatory and anti-apoptotic effect via down-regulation of testicular p38 MAPK expression in diabetic rats.
Collapse
Affiliation(s)
| | | | | | | | - Hanaa Mohamed Khalaf
- Department of Pharmacology, Faculty of Medicine, Minia University, Minia, 61511, Egypt.
| |
Collapse
|