Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Biol Rep ; 51(1): 631, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38722405

ABSTRACT

Adipokines are now well-known to regulate reproduction. Visfatin is an adipokine expressed in the hypothalamus, pituitary, ovary, uterus, and placenta of different species, and since it has been found to modulate the endocrine secretion of the hypothalamus, pituitary gland and ovary, it may be considered a novel regulator of female reproduction. Although the majority of the literature explored its role in ovarian regulation, visfatin has also been shown to regulate uterine remodeling, endometrial receptivity and embryo development, and its expression in the uterus is steroid dependent. Like other adipokines, visfatin expression and levels are deregulated in pathological conditions including polycystic ovary syndrome. Thus, the present mini-review focuses on the role of visfatin in female reproduction under both physiological and pathological conditions.


Subject(s)
Nicotinamide Phosphoribosyltransferase , Polycystic Ovary Syndrome , Reproduction , Female , Humans , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Reproduction/physiology , Reproduction/genetics , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Animals , Ovary/metabolism , Uterus/metabolism , Cytokines/metabolism , Pregnancy , Adipokines/metabolism
2.
J Biochem Mol Toxicol ; 37(9): e23400, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37335250

ABSTRACT

Cadmium (Cd) is one of the heavy metal pollutants present in the environment due to human intervention. It is well known that Cd causes toxicological effects on various organs, including the testes. Morin hydrate is a plant-derived bioflavonoid with antioxidant, anti-inflammatory, and anti-stress properties. Thus, the question can be raised as to whether Morin has an effect on Cd-intoxication-induced testicular impairment. Therefore, the aim of this study was to investigate the role of Morin on Cd-mediated disruption of testicular activity. Mice were divided into three groups: group 1 served as the control group, group 2 was given Cd (10 mg/kg) orally for 35 days, and group 3 was given Cd and Morin hydrate (100 mg/kg) for 35 days. To validate the in vivo findings, an in vitro study on testicular explants was also performed. The results of the in vivo study showed that Cd-intoxicated mice had testicular disorganization, reduced circulating testosterone levels, decreased sperm density, and elevated oxidative stress and sperm abnormality. The expression of the germ cell proliferation marker, germ cell nuclear acidic protein (GCNA), and adipocytokine visfatin were also downregulated. It was observed that Morin hydrate upregulated testicular visfatin and GCNA expression in Cd-intoxicated mice, along with improvement in circulating testosterone, testicular histology, and sperm parameters. Furthermore, the in vitro study showed that Cd-mediated downregulation of testicular visfatin and GCNA expression, along with the suppressed secretion of testosterone from testicular explants, was normalized by Morin treatment, whereas visfatin expression was not. Overall, these data indicate that environmental cadmium exposure impairs testicular activity through downregulation of visfatin and GCNA expression, and Morin might play a protective role against Cd-induced testicular toxicity.


Subject(s)
Cadmium Poisoning , Testis , Humans , Male , Mice , Animals , Testis/metabolism , Cadmium/toxicity , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide Phosphoribosyltransferase/pharmacology , Semen/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Oxidative Stress , Testosterone/pharmacology , Flavonoids/pharmacology , Flavonoids/metabolism , Cadmium Poisoning/metabolism , Cell Proliferation
3.
Cytokine ; 144: 155554, 2021 08.
Article in English | MEDLINE | ID: mdl-33962842

ABSTRACT

Type 1 diabetes mellitus (T1DM) is a metabolic disorder with severe hyperglycemia, one of the complications of which is testicular dysfunctions, androgen deficiency and decreased male fertility. In the diabetic testes, the expression and signaling pathways of leptin and a number of other adipokines are significantly changed. However, there is no information on the localization and expression of adipokine, apelin and its receptor (APJ) in the diabetic testes, although there is information on the involvement of apelin in the regulation of reproductive functions. The aim of this study was to investigate the expression and localization of apelin and APJ in the testes of mice with streptozotocin-induced T1DM and to estimate the effects of agonist (apelin-13) and antagonist (ML221) of APJ on the testosterone production by diabetic testis explants in the in vitro conditions. We first detected the expression of apelin and its receptor in the mouse testes, and showed an increased intratesticular expression of apelin and APJ along with the reduced testosterone secretion in T1DM. Using imunohistochemical approach, we showed that apelin and APJ are localized in the Leydig and germ cells, and in diabetes, the amount of these proteins was significantly higher than in the control mice. The diabetic testes had a decrease in germ cell proliferation (the reduced PCNA and GCNA levels) and an increase in apoptosis (the increased active caspase-3 and decreased BCL2 levels). These results suggest an involvement of apelin and APJ in T1DM-induced testicular pathogenesis. Treatment of the cultured testis explants with ML221 significantly increased the testosterone secretion, whereas apelin-13 was ineffective. Thus, hyperapelinemia in the testes can significantly contribute to testicular pathogenesis in T1DM, and pharmacological inhibition of apelin receptors can improve testicular steroidogenesis.


Subject(s)
Apelin/metabolism , Diabetes Mellitus, Experimental/metabolism , Steroids/metabolism , Testis/metabolism , Adipokines/metabolism , Animals , Apelin Receptors/metabolism , Apoptosis/physiology , Cell Proliferation/physiology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/metabolism , Germ Cells/metabolism , Leydig Cells/metabolism , Male , Mice , Nitrobenzoates/pharmacology , Pyrans/pharmacology , Signal Transduction/physiology , Streptozocin/pharmacology , Testosterone/metabolism
4.
Life Sci ; 276: 119409, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33781825

ABSTRACT

Polycystic ovary syndrome is a common reproductive disorder in the female of reproductive age, which is characterized by hyperandrogenism, insulin resistance, cystic ovary and infertility. The level of pro-inflammatory adipokine, visfatin is elevated in PCOS conditions in human and animal. In this study, letrozole induced hyperandrogenised PCOS mice model have been used to unravel the effects of visfatin inhibition. The results showed that letrozole induced hyperandrogenisation significantly (p < 0.05) elevates ovarian visfatin concentration from 66.03 ± 1.77 to 112.08 ± 3.7 ng/ml, and visfatin expression to 2.5 fold (p < 0.05) compared to control. Visfatin inhibition in PCOS by FK866 has significantly (p < 0.05) suppressed the secretion of androgens, androstenedione (from 0.329 ± 0.07 to 0.097 ± 0.01 ng/ml) and testosterone levels (from 0.045 ± 0.003 to 0.014 ± 0.0009 ng/ml). Ovarian histology showed that visfatin inhibition suppressed cyst formation and promotes corpus luteum formation. Visfatin inhibition has suppressed apoptosis and increases the expression of BCL2 along with increase in the proliferation (GCNA expression elevated). Visfatin inhibition has increased ovarian glucose content (from 167.05 ± 8.5 to 210 ± 7 mg/dl), along with increase in ovarian GLUT8 expression. In vitro study has also supported the in vivo findings where FK866 treatment significantly (p < 0.05) suppressed testosterone (control-3.84 ± 0.44 ng/ml, 1 nM FK866-2.02 ± 0.048 ng/ml, 10 nM FK866-1.74 ± 0.20 ng/ml) and androstenedione (control-4.68 ± 0.91 ng/ml, 1 nM FK866-3.38 ± 0.27 ng/ml, 10 nM FK866-4.55 ± 0.83 ng/ml) production from PCOS ovary. In conclusion, this is first report, which showed that visfatin inhibition by FK866 in hyperandrogenised mice ameliorates pathogenesis of PCOS. Thus, it may be suggested that visfatin inhibition could have a therapeutic potential in PCOS management along with other intervention.


Subject(s)
Acrylamides/pharmacology , Cytokines/antagonists & inhibitors , Disease Models, Animal , Hyperandrogenism/drug therapy , Letrozole/toxicity , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Piperidines/pharmacology , Polycystic Ovary Syndrome/drug therapy , Androgens/metabolism , Animals , Blood Glucose/metabolism , Female , Hyperandrogenism/chemically induced , Hyperandrogenism/metabolism , Hyperandrogenism/pathology , Insulin Resistance , Mice , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology
5.
Cytokine ; 140: 155422, 2021 04.
Article in English | MEDLINE | ID: mdl-33476980

ABSTRACT

Visfatin is an important adipokines, which are expressed in different tissues including ovary of mammals. The postnatal ovary in rodents undergoes dramatic changes of intra-ovarian factors in relation to proliferation and apoptosis. There are studies which showed that gonadal visfatin changes in postnatal life. However, role of visfatin in the early postnatal period i.e. infantile period has not been studied. Therefore, the present study was aimed to explore the role of visfatin in the early postnatal ovarian functions. Furthermore, to explore the role of visfatin, the endogenous visfatin was inhibited from PND14-PND21 by FK866 with dose of 1.5 mg/kg. Our results showed gain in body weight and ovarian weight after visfatin inhibition. The inhibition of visfatin increased the ovarian proliferation (increase in PCNA, GCNA expression and BrdU incorporation) and apoptosis (increase in BAX and active caspase3 expression). Moreover, visfatin inhibition decreased the expression of antiapoptotic/survival protein, BCL2 in the ovary. These findings suggest that visfatin in the infantile ovary may suppress the proliferation and apoptosis by up-regulating BCL2 expression. An interesting finding has been observed that circulating estrogen and progesterone remain unaffected, although visfatin inhibition up-regulated ER-ß and down-regulated ER-α. It may also be suggested that visfatin could regulates proliferation and apoptosis via modulating estrogen signaling. In conclusion, visfatin inhibits the proliferation and apoptosis without modulating the ovarian steroid biosynthesis and visfatin mediated BCL2 expression could also be mechanism to preserve the good quality follicle in early postnatal period.


Subject(s)
Apoptosis/physiology , Cell Proliferation/physiology , Nicotinamide Phosphoribosyltransferase/metabolism , Ovary/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Down-Regulation/physiology , Estrogens/metabolism , Female , Mice , Progesterone/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Up-Regulation/physiology
6.
Acta Histochem ; 122(8): 151630, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32992122

ABSTRACT

Visfatin is a crucial adipokine, which also regulates ovarian functions in many animals. Mice estrous cycle is characterized by a dynamic complex physiological process in the reproductive system. Expression of various factors changes during the estrous cycle in the ovary. To the best of our knowledge, no previous study has been conducted on the expression of visfatin in mice ovaries during the estrous cycle. Therefore, we investigated the localization and expression of visfatin protein in the ovary of mice during the estrous cycle. Western blot analysis showed the elevated expression of visfatin in proestrus and lowest in diestrus. Immunohistochemical localization of visfatin showed intense staining in the corpus luteum of proestrus and diestrus ovaries. Thecal cells, granulosa cells, and oocytes also showed the presence of visfatin. Expression of ovarian visfatin was correlated to BCL2 and active caspase3 expression and exhibited a significant positive correlation. Furthermore, in vivo inhibition of visfatin by FK866 in the proestrus ovary down-regulated active caspase3 and PCNA expression, and up-regulated the BCL2 expression. These results suggest the role of visfatin in the proliferation and apoptosis of the follicles and specific localization of visfatin in the corpus luteum also indicate its role in corpus luteum function, which may be in progesterone biosynthesis and regression of old corpus luteum. However, further study is required to support these findings. In conclusion, visfatin may also be regulating follicular growth during the estrous cycle by regulating proliferation and apoptosis.


Subject(s)
Corpus Luteum/metabolism , Cytokines/genetics , Granulosa Cells/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Oocytes/metabolism , Theca Cells/metabolism , Acrylamides/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Caspase 3/genetics , Caspase 3/metabolism , Cell Proliferation/drug effects , Corpus Luteum/cytology , Corpus Luteum/drug effects , Corpus Luteum/growth & development , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Diestrus/drug effects , Diestrus/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Developmental , Granulosa Cells/cytology , Granulosa Cells/drug effects , Immunohistochemistry , Mice , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Oocytes/cytology , Oocytes/drug effects , Piperidines/pharmacology , Proestrus/drug effects , Proestrus/genetics , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Theca Cells/cytology , Theca Cells/drug effects
7.
J Steroid Biochem Mol Biol ; 204: 105763, 2020 11.
Article in English | MEDLINE | ID: mdl-32987128

ABSTRACT

Pubertal ovarian function might be dependent on the factors present in the pre-pubertal stages. Visfatin regulates ovarian steroidogenesis in adult. To date, no study has investigated the role of visfatin either in pre-pubertal or pubertal mice ovary. Thus, we investigated the role of visfatin in pre-pubertal mice ovary in relation to steroidogenesis and proliferation and apoptosis in vitro by inhibiting the endogenous visfatin by a specific inhibitor, FK866. Inhibition of visfatin increased the estrogen secretion and also up-regulated the expression of CYP11A1, 17ßHSD and CYP19A1 in mice ovary. Furthermore, active caspase3 was up-regulated along with the down-regulation of BAX and BCL2 in the pre-pubertal ovary after visfatin inhibition. The expression of GCNA, PCNA, and BrdU labeling was also decreased by FK866 treatment. These results suggest that visfatin inhibits steroidogenesis, increases proliferation, and suppresses apoptosis in the pre-pubertal mice ovary. So, visfatin is a new regulator of ovary function in pre-pubertal mice.


Subject(s)
Nicotinamide Phosphoribosyltransferase/metabolism , Ovary/metabolism , 17-Hydroxysteroid Dehydrogenases/metabolism , Acrylamides/pharmacology , Animals , Apoptosis , Aromatase/metabolism , Cell Proliferation , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Female , Mice , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Ovary/drug effects , Piperidines/pharmacology , Sexual Maturation
8.
Mol Reprod Dev ; 87(5): 620-629, 2020 05.
Article in English | MEDLINE | ID: mdl-32249489

ABSTRACT

Diabetes mellitus impairs testicular activity and leads to infertility. Leptin is one of the endogenous regulators of the male reproductive functions, but the role of leptin and its receptor (LEPR/Ob-R) in the control of testosterone production and testicular proliferation has not been investigated so far, especially in the Type 1 diabetes mellitus (DM1). Metformin is an anti-hyperglycemic drug which is beneficial for treating the both DM2 and DM1. The aim of this work was to study the possible role of leptin and Ob-R in the regulation of steroidogenesis and proliferation in the testes of mice with streptozotocin-induced DM1 (75 mg/kg/day, 4 days) and to estimate the restoring effect of metformin treatment (500 mg/kg, 2 weeks) on the diabetic testes. In the diabetic testes, the plasma and intratesticular leptin levels and plasma testosterone levels were reduced and completely restored by metformin treatment. Metformin also restored the expression of the steroidogenic transport protein steroidogenic acute regulatory protein reduced in DM1. In the diabetic testes, the expression of Ob-R was downregulated and the immunolocalization of Ob-R showed weak staining in the Leydig cells, the primary spermatocytes and the round spermatids. The germ cell proliferation was also reduced in DM1, as noticed with proliferating cell nuclear antigen (PCNA) expression. Metformin increased the Ob-R expression and immunostaining in the different cell types and improved the PCNA expression. Thus, DM1 impairs the testicular steroidogenesis and proliferation by inhibiting the leptin signaling, causing a decrease in leptin levels and Ob-R expression in the testes of diabetic mice, while metformin improves the leptin signaling and restores testosterone production and testicular proliferation.


Subject(s)
Diabetes Mellitus, Experimental , Leptin/metabolism , Metformin/pharmacology , Receptors, Leptin , Testis/drug effects , Animals , Cell Proliferation/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Gene Expression/drug effects , Leptin/blood , Leydig Cells/drug effects , Leydig Cells/physiology , Male , Metformin/therapeutic use , Mice , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Streptozocin , Testis/cytology , Testis/metabolism , Testosterone/blood , Tissue Distribution/drug effects
9.
J Steroid Biochem Mol Biol ; 185: 225-236, 2019 01.
Article in English | MEDLINE | ID: mdl-30227242

ABSTRACT

Visfatin is an adipokine which has an endocrine effect on reproductive functions and regulates ovarian steroidogenesis. There is scant information about the expression, regulation, and functions of visfatin in the mammalian uterus. The present study examined expression and localization of visfatin in the mouse uterus at various stages of the natural estrous cycle, effects of estrogen and progesterone on localization and expression of visfatin in the ovariectomised mouse uterus and effect of visfatin inhibition by a specific inhibitor, FK866 on proliferation and apoptosis in the uterus. Western blot analysis of visfatin showed high expression in proestrus and metestrus while it declined in estrus and diestrus. Immulocalization study also showed strong immunostaining in the cells of endometrium, myometrium, luminal and glandular epithelium during proestrus and metestrus that estrus and diestrus. The uterine visfatin expression closely related to the increased estrogen levels in proestrus and suppressed when progesterone rose to a high level in diestrus. The treatment with estrogen to ovariectomised mice up-regulates visfatin, PCNA, and active caspase3 whereas progesterone up-regulates PCNA and down-regulates visfatin and active caspase3 expression in mouse uterus. The co-treatment with estrogen and progesterone up-regulates visfatin and down-regulates PCNA and active caspase3. In vitro study showed endogenous visfatin inhibition by FK866 increased expression of PCNA and BCL2 increased catalase activity while FK866 treatment decreased expression of active caspase3 and BAX with decreased SOD and GPx activity. BrdU labeling showed that inhibition of visfatin modulates the uterine proliferation. This study showed that expression of visfatin protein is steroid dependent in mouse uterus which is involved in the regulation of proliferation and apoptosis via modulating antioxidant system in the uterus of mice during the reproductive cycle.


Subject(s)
Apoptosis/physiology , Cell Proliferation/physiology , Endometrium/metabolism , Estrogens/metabolism , Estrous Cycle/metabolism , Myometrium/metabolism , Nicotinamide Phosphoribosyltransferase/biosynthesis , Progesterone/metabolism , Acrylamides/pharmacology , Animals , Caspase 3/biosynthesis , Catalase/biosynthesis , Diestrus/metabolism , Estrus/metabolism , Female , Glutathione Peroxidase/biosynthesis , Mice , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Piperidines/pharmacology , Proestrus/metabolism , Proliferating Cell Nuclear Antigen/biosynthesis , Superoxide Dismutase/biosynthesis , bcl-2-Associated X Protein/biosynthesis
10.
Acta Histochem ; 121(2): 182-188, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30579591

ABSTRACT

Dexamethasone, a synthetic glucocorticoid has been used as an immunosuppressive and anti-inflammatory and affects reproduction. It has been suggested that testicular steroidogenesis involves PGC-1α and visfatin as key regulators. Previous studies have shown that dexamethasone down-regulates PGC-1α and visfatin expression in muscle and mammary epithelial cells respectively. However, the effect of dexamethasone on testicular visfatin and PGC-1α expressions has not been investigated. The aims of the present study were to investigate the effect of dexamethasone, on the expression of PGC-1α, visfatin and antioxidant enzymes activities in mouse testis. The results of the present study showed that dexamethasone treatment significantly decreased the expression of visfatin and PGC-1α in mice testis, along with significant decreased in testicular antioxidant enzymes activates. Further, dexamethasone treatment also significantly increased the testicular lipid peroxidation and decreased testosterone synthesis. The dexamethasone induced changes in PGC-1α and visfatin in the testis were significantly correlated with changes in serum testosterone concentrations and antioxidant enzymes activities. Thus, dexamethasone induced testicular toxicity may involve the PGC-1α and visfatin as important molecules to exhibit its effects.


Subject(s)
Dexamethasone/pharmacology , Down-Regulation , Lipid Peroxidation/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Testosterone/metabolism , Animals , Antioxidants/pharmacology , Glucocorticoids/metabolism , Male , Mice , Nicotinamide Phosphoribosyltransferase/drug effects , Nicotinamide Phosphoribosyltransferase/metabolism , Testis/drug effects , Transcription Factors/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...