Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Endocrinol Diabetes Metab ; 6(6): e450, 2023 11.
Article in English | MEDLINE | ID: mdl-37723884

ABSTRACT

INTRODUCTION: Prenatal programming with dexamethasone increases the risk of the development of hyperglycaemia and insulin resistance, leading to diabetes in adulthood. Dexamethasone also causes a decline in renal glomerular filtration in the adult offspring. Sodium-glucose cotransporter-2 (SGLT2) plays a significant role in regulating blood glucose and renal haemodynamics in diabetic patients. However, the role of SGLT2 in dexamethasone-induced programming and the putative sex-dependent effects on the changes named earlier is unknown. Therefore, this study aimed to investigate the impact of maternal dexamethasone treatment on glucose tolerance, insulin sensitivity, renal perfusion and renal function in adult male and female offspring and the possible contribution of SGLT2 to these changes. METHODS AND RESULTS: Pregnant Sprague Dawley rats (F0 ) were treated with either vehicle or dexamethasone (0.2 mg/kg ip) from gestation Day 15 to 20. F1 males and F1 females were randomly selected from each mother at 4 months of age. There was no change in serum Na+ , Na+ excretion rate, glucose tolerance or insulin sensitivity in F1 male or female rats. However, dexamethasone caused significant glomerular hypertrophy and decreases in CSinistrin and CPAH indicating decreased glomerular filtration rate and renal plasma flow, respectively, in dexamethasone-treated F1 male but not female rats. Dexamethasone did not affect SGLT2 mRNA or protein expression in F1 males or females. CONCLUSION: We conclude that dexamethasone-mediated prenatal programming of glomerular volume, renal function and haemodynamics is sex-dependent, occurring only in adult male offspring.


Subject(s)
Insulin Resistance , Prenatal Exposure Delayed Effects , Humans , Pregnancy , Rats , Animals , Male , Female , Sodium-Glucose Transporter 2/pharmacology , Insulin Resistance/physiology , Rats, Sprague-Dawley , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , Kidney/metabolism , Blood Glucose , Hemodynamics , Dexamethasone/adverse effects
2.
Pharmaceuticals (Basel) ; 16(2)2023 Feb 03.
Article in English | MEDLINE | ID: mdl-37259385

ABSTRACT

BACKGROUND: Diabetes mellitus (DM) is a risk factor for cardiovascular diseases, specifically, the ischemic heart diseases (IHD). The renin-angiotensin system (RAS) affects the heart directly and indirectly. However, its role in the protection of the heart against I/R injury is not completely understood. The aim of the current study was to evaluate the efficacy of the angiotensin-converting enzyme (ACE) inhibitor and Angiotensin II receptor (AT1R) blocker or a combination thereof in protection of the heart from I/R injury. METHODS: Hearts isolated from adult male Wistar rats (n = 8) were subjected to high glucose levels; acute hyperglycemia or streptozotocin (STZ)-induced diabetes were used in this study. Hearts were subjected to I/R injury, treated with Captopril, an ACE inhibitor; Losartan, an AT1R antagonist; or a combination thereof. Hemodynamics data were measured using a suitable software for that purpose. Additionally, infarct size was evaluated using 2,3,5-Triphenyltetrazolium chloride (TTC) staining. The levels of apoptosis markers (caspase-3 and -8), antioxidant enzymes, superoxide dismutase (SOD) and catalase (CAT), nitric oxide synthase (eNOS), and glucose transporter type 4 (GLUT-4) protein levels were evaluated by Western blotting. Pro-inflammatory and anti-inflammatory cytokines levels were evaluated by enzyme-linked immunosorbent assay (ELISA). RESULTS: Captopril and Losartan alone or in combination abolished the effect of I/R injury in hearts subjected to acute hyperglycemia or STZ-induced diabetes. There was a significant (p < 0.05) recovery in hemodynamics, infarct size, and apoptosis markers following the treatment with Captopril, Losartan, or their combination. Treatment with Captopril, Losartan, or their combination significantly (p < 0.05) reduced pro-inflammatory cytokines and increased GLUT-4 protein levels. CONCLUSIONS: The blockade of the RAS system protected the diabetic heart from I/R injury. This protection followed a pathway that utilizes GLUT-4 to decrease the apoptosis markers, pro-inflammatory cytokines, and to increase the anti-inflammatory cytokines. This protection seems to employ a pathway which is not involving ERK1/2 and eNOS.

3.
Pharmaceuticals (Basel) ; 16(4)2023 Mar 27.
Article in English | MEDLINE | ID: mdl-37111249

ABSTRACT

Ischemia and perfusion (I/R) induce inflammation and oxidative stress, which play a notable role in tissue damage. The aim of this study was to investigate the role of an NADPH oxidase inhibitor (apocynin) in the protection of the heart from I/R injury. Hearts isolated from Wistar rats (n = 8 per group) were perfused with a modified Langendorff preparation. Left ventricular (LV) contractility and cardiovascular hemodynamics were evaluated by a data acquisition program, and infarct size was evaluated by 2,3,5-Triphenyl-2H-tetrazolium chloride (TTC) staining. Furthermore, the effect of apocynin on the pro-inflammatory cytokines (IL-1ß, IL-6 and TNF-α) and anti-inflammatory cytokine (IL-10) was evaluated using an enzyme linked immunosorbent assay (ELISA). Hearts were subjected to 30 min of regional ischemia, produced by ligation of the left anterior descending (LAD) coronary artery, followed by 30 min of reperfusion. Hearts were infused with apocynin before ischemia, during ischemia or at reperfusion. To understand the potential pathways of apocynin protection of the heart, a nitric oxide donor (S-nitroso-N-acetylpenicillamine, SNAP), nitric oxide blocker (N (gamma)-nitro-L-arginine methyl ester, L-Name), nicotinic acid adenine dinucleotide phosphate (NAADP) inhibiter (Ned-K), cyclic adenosine diphosphate ribose (cADPR) agonist, or CD38 blocker (Thiazoloquin (az)olin (on)e compound, 78c) was infused with apocynin. Antioxidants were evaluated by measuring superoxide dismutase (SOD) and catalase (CAT) activity. Apocynin infusion before ischemia or at reperfusion protected the heart by normalizing cardiac hemodynamics and decreasing the infarct size. Apocynin treatment resulted in a significant (p < 0.05) decrease in pro-inflammatory cytokine levels and a significant increase (p < 0.05) in anti-inflammatory and antioxidant levels. Apocynin infusion protected the heart by improving LV hemodynamics and coronary vascular dynamics. This treatment decreased the infarct size and inflammatory cytokine levels and increased anti-inflammatory cytokine and antioxidant levels. This protection follows a pathway involving CD38, nitric oxide and acidic stores.

4.
Front Oncol ; 13: 1080008, 2023.
Article in English | MEDLINE | ID: mdl-37114127

ABSTRACT

Intoduction: Identification of molecular alterations associated with tumor behavior is necessary to guide clinical management. The 2022 WHO classification has organized the thyroid follicular cell-derived neoplasms into benign, low-risk and high-risk neoplasms, and emphasized the value of biomarkers that may provide differential diagnostic and prognostic information to avoid overtreatment of low risk neoplasms. This work aims to study the epidermal growth factor receptor (EGFR) expression, functional and spatial dynamics in relation to specific miRNAs alterations in papillary thyroid cancer (PTC) and in non-invasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP) considered as models of high-risk and low-risk thyroid tumors respectively. Methods: Primary thyroid cultured cells were used for miRNA gain/loss of function and luciferase reporter assays. Paraffin embedded tissues were used for real time PCR, immuno-fluorescence stain and confocal microscopy experiments. Results: Our results showed that in PTC, EGFR mRNA is reduced as an effect of miR-146b-5p upregulation. The EGF expression is low and the ERK pathway is inhibited. The EGFR protein high cytoplasmic expression and colocalization with the endosomal/exosomal markers, ALIX and CD63, suggest the occurrence of stress-induced EGFR internalization, accumulation in endosomal vesicles and secretion via exosomes. In NIFTP EGFR transcription is increased in association with downregulation of miR-7-5p and the EGFR/ERK pathway is active indicating dependence on the canonical EGFR pathway for growth. Conclusion: Downregulation of transcript level along with cytoplasmic accumulation of undegraded protein is a new pattern of EGFR regulation associated with malignancy in thyroid. Further research is needed to elucidate the intracellular trafficking defects responsible for this specific EGFR dynamic in PTC.

5.
Biol Reprod ; 108(1): 133-149, 2023 01 14.
Article in English | MEDLINE | ID: mdl-36322157

ABSTRACT

BACKGROUND: Intrauterine growth restriction (IUGR) is manifested by lower maternal progesterone levels, smaller placental size, and decreased placental vascularity indicated by lower expression of vascular endothelial growth factor (VEGF). Studies showed that progesterone increases angiogenesis and induces VEGF expression in different tissues. Therefore, the aim of the present study is to evaluate the effect of progesterone on placental vascular bed and VEGF expression and the modulation of nuclear and membranous progesterone receptors (PR) in dexamethasone-induced rat IUGR model. METHODS: Pregnant Sprague-Dawley rats were allocated into four groups and given intraperitoneal injections of either saline, dexamethasone, dexamethasone, and progesterone or progesterone. Injections started on gestation day (DG) 15 and lasted until the days of euthanization (19 and 21 DG). Enzyme-linked immunosorbent assay was used to evaluate plasma progesterone levels. Real-time PCR and western blotting were used to evaluate gene and protein expressions of VEGF, and PR in labyrinth and basal placental zones. Immunohistochemistry was used to locate VEGF and different PRs in placental cells. Immunofluorescence was used to monitor the expression of blood vessel marker (αSMA). RESULTS: Dexamethasone decreased the vascular bed fraction and the expression of VEGF in both placental zones. Progesterone co-treatment with dexamethasone prevented this reduction. Nuclear and membrane PRs showed tissue-specific expression in different placental zones and responded differently to both dexamethasone and progesterone. CONCLUSIONS: Progesterone treatment improves the outcomes in IUGR pregnancy. Progesterone alleviated DEX-induced IUGR probably by promoting placental VEGF and angiogenesis.


Subject(s)
Placenta , Progesterone , Receptors, Progesterone , Animals , Female , Humans , Pregnancy , Rats , Dexamethasone/pharmacology , Fetal Growth Retardation/metabolism , Placenta/drug effects , Placenta/metabolism , Progesterone/pharmacology , Rats, Sprague-Dawley , Receptors, Progesterone/drug effects , Receptors, Progesterone/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
6.
Mol Neurobiol ; 59(6): 3767-3777, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35396693

ABSTRACT

Prenatal exposure to dexamethasone (DEX) results in long-lasting effects on cognitive functions such as learning and memory impairment. However, the mechanisms underlying these DEX-induced deleterious effects are not well known. Here, we assessed whether cyclooxygenase-2 (COX-2) is involved in the impact of prenatal exposure to DEX on learning and memory during adulthood. Pregnant Sprague-Dawley rats received daily injections of either DEX (0.2 mg/kg; i.p.) or saline from gestation day (GD) 14 until GD21. Gene and protein expression of COX-2, as well as presynaptic (synaptophysin) and postsynaptic (postsynaptic density protein-95) proteins, were monitored in the dorsal and ventral hippocampi of adult male and female offspring. A different cohort of adult male and female rat offspring was given daily injections of either vehicle or a specific COX-2 inhibitor (celecoxib 10 mg/kg, i.p.) for 5 consecutive days and was subsequently subjected to Morris water maze memory test. Prenatal DEX enhanced the expression of COX-2 protein and cox-2 mRNA in the dorsal hippocampus of adult female but not male rats. This enhanced COX-2 expression was associated with reduced expression in pre- and postsynaptic proteins and altered memory acquisition and retention. Administration of COX-2-specific inhibitor alleviated prenatal DEX-induced memory impairment in adult female rats. This study suggests that prenatal activation of glucocorticoid receptors stimulates COX-2 gene and protein expression and impairs hippocampal-dependent spatial memory in female but not male rat offspring. Furthermore, COX-2 selective inhibitors can be used to alleviate the long-lasting deleterious effects of corticosteroid medication during pregnancy.


Subject(s)
Cyclooxygenase 2 , Dexamethasone , Prenatal Exposure Delayed Effects , Receptors, Glucocorticoid , Animals , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dexamethasone/adverse effects , Female , Hippocampus/metabolism , Male , Maze Learning , Memory Disorders/chemically induced , Memory Disorders/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/metabolism
7.
Reprod Biomed Online ; 44(4): 595-607, 2022 04.
Article in English | MEDLINE | ID: mdl-35232674

ABSTRACT

RESEARCH QUESTION: How does progesterone improve fetal outcome and change the expression of placental glucose transporters (GLUT) in dexamethasone-induced intrauterine growth restriction (IUGR)? DESIGN: A total of 64 rats were divided randomly into four different treatment groups based on daily i.p. injections of either saline or dexamethasone in the presence or absence of progesterone. Injections started on the 15th day of gestation (15dg) and lasted until the day of sacrifice at 19dg or 21dg. Maternal plasma progesterone concentrations were measured by enzyme-linked immunosorbent assay. The gene and protein expression of placental GLUT1 and GLUT3 were evaluated in the placental labyrinth and basal zones by real-time polymerase chain reaction and Western blotting, respectively. The localization of GLUT1 and GLUT3 was evaluated by immunohistochemistry. RESULTS: Dexamethasone induced significant decreases in maternal serum progesterone concentrations (P = 0.029) and placental (P < 0.001) and fetal body (P = 0.009) weights. Dexamethasone also reduced the expression of GLUT1 in the labyrinth zone (P = 0.028) and GLUT3 in both the labyrinth (P = 0.002) and basal zones (P = 0.026). Coadministration of dexamethasone and progesterone prevented the reduction in fetal body weight, placental weight and placental GLUT expression compared with that seen in dexamethasone-treated groups. CONCLUSION: These results suggest that progesterone prevents the significant reduction in fetal and placental weights in dexamethasone-induced IUGR, possibly through improving the expression of placental GLUT.


Subject(s)
Fetal Growth Retardation , Placenta , Animals , Female , Pregnancy , Rats , Dexamethasone/adverse effects , Dexamethasone/metabolism , Fetal Growth Retardation/chemically induced , Glucose/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Placenta/metabolism , Progesterone/metabolism
8.
Toxicol Appl Pharmacol ; 418: 115510, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33775663

ABSTRACT

Prevention or repair of DNA damage is critical to inhibit carcinogenesis in living organisms. Using quantitative RT2 Profiler™ PCR array, we investigated if trans-resveratrol could modulate the transcription of DNA damage sensing/repair pathway genes in euglycemic and non-obese type 2 diabetic Goto-Kakizaki rat testis. Trans-resveratrol imparted disparate effects on gene expressions. In euglycemic rats, it downregulated 79% and upregulated 2% of genes. However, in diabetic rats, it upregulated only 2% and downregulated 4% of genes. As such, diabetes upregulated 16% and downregulated 4% of genes. Trans-resveratrol normalized the expression of 9 (60%) out of 15 upregulated genes in diabetic rats. In euglycemic rats, trans-resveratrol inhibited ATM/ATR, DNA damage repair, pro-cell cycle progression, and apoptosis signaling genes. However, it increased Cdkn1a and Sumo1, indicating cell cycle arrest, apoptosis, and cytostasis in conjunction with increased DNA double-strand breaks and apoptosis. Diabetes increased DNA damage and apoptosis but did not affect ATM/ATR and double-strand break repair genes, although it increased few single-strand repair genes. Diabetes increased Abl1 and Sirt1, which may be related to apoptosis, but their increase may well suggest the enhanced cell cycle progression and putative carcinogenicity. The transcription of Rad17 and Smc1a increased in diabetic rats indicating G2 phase arrest and increases in a few DNA single-strand breaks repair genes suggesting DNA damage repair. Trans-resveratrol inhibits the cell cycle and causes cell death in euglycemic rat testis but normalizes diabetes-induced genes related to DNA damage and cell cycle control, suggesting its usefulness in maintaining DNA integrity in diabetes.


Subject(s)
Blood Glucose/metabolism , DNA Damage , DNA Repair , Diabetes Mellitus, Type 2/blood , Resveratrol/toxicity , Testis/drug effects , Transcription, Genetic/drug effects , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Biomarkers/blood , Cell Cycle Checkpoints , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Gene Expression Regulation , Male , Rats, Wistar , Testis/metabolism , Testis/pathology
10.
Int J Endocrinol ; 2019: 6794837, 2019.
Article in English | MEDLINE | ID: mdl-31341474

ABSTRACT

Many Arab women in the Gulf region cover their bodies for cultural and religious reasons, limiting the skin's exposure to sunlight and therefore its ability to synthesize vitamin D. The aim of this study is to determine whether the clothing style of Kuwaiti premenopausal women affects their vitamin D status, bone marker expression, and bone density. Three groups of healthy unmarried single Kuwaiti females (20-35 years old; n=30 per group) were recruited randomly from the general community: a control group who wear Western-style clothing (unveiled group), a group who wear a hejab that covers the whole body except for the face and hands (hejab group), and a group who wear a black veil with the entire body covered (veiled group). Bone mineral density (BMD), bone markers (procollagen type 1 N-terminal propeptide [P1NP], osteocalcin, and ß-CrossLaps), 25-hydroxy vitamin D, intact parathyroid hormone [iPTH], and calcitonin were measured. The bone marker osteocalcin was significantly higher in the hejab group compared to the control group, whereas P1NP and ß-CrossLaps were significantly higher in the veiled group compared to the control group. 25-hydroxy vitamin D, iPTH, calcitonin, and BMD were not significantly different across the three groups despite the observed elevation in bone turnover markers. The majority of participants in all three groups exhibited vitamin D deficiency; however, the lowest vitamin D levels were observed among the hejab and veiled participants. These findings suggest that clothing style may contribute to vitamin D deficiency in young Kuwaiti women.

11.
Brain Res ; 1707: 117-123, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30476470

ABSTRACT

Fetal exposure to dexamethasone (DEX) alters brain plasticity and cognitive functions during adulthood in a sex-dependent manner. The mechanisms underlying such long-lasting sex-dependent change of prenatal DEX is not well understood. The p73 gene plays an important role in brain development. It encodes for two protein variants; the neural cell death protein (TAp73) and the anti-neural cell death protein (ΔNp73). Therefore, we sought to determine how prenatal exposure to DEX alters the expression of these p73 gene variants in the brain of male and female fetuses. Pregnant dams received daily injections of either DEX (0.4 mg/kg, i.p.) or saline from gestation day (GD) 14 until GD21. On GD21, body and brain weights were monitored and mRNA and protein levels of TAp73 and ΔNp73 were measured in male and female fetal brains using RT-PCR, Western blot, and immunohistochemistry. Prenatal exposure to DEX significantly reduced the body and brain weights of both male and female fetuses, although reduction in brain weight was less severe than that of the body weight. Administration of DEX to pregnant dams led to enhanced expression of both TAp73 and ΔNp73 gene/protein variants in the brain of male but not in that of female fetuses. Dexamethasone induced a sex-dependent effect on the expression of p73 gene variants. DEX-induced growth restriction in the brain of female fetuses is independent of p73 gene. This study strongly suggests that survival/death programs operate differently during the development of male and female brains.


Subject(s)
Brain/drug effects , Brain/embryology , Dexamethasone/pharmacology , Prenatal Exposure Delayed Effects , Tumor Protein p73/genetics , Animals , Brain/metabolism , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Male , Nuclear Proteins/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Sex Factors , Tumor Protein p73/biosynthesis , Tumor Suppressor Proteins/metabolism
12.
Reprod Biol Endocrinol ; 15(1): 97, 2017 Dec 29.
Article in English | MEDLINE | ID: mdl-29321064

ABSTRACT

BACKGROUND: Plasma and urine levels of the potent vasodilator Ang-(1-7) are elevated in mid and late pregnancy and are correlated with elevated placental angiogenesis, fetal blood flow, and rapid fetal growth. We hypothesized that Ang-(1-7), its receptor (Mas1) and the enzymes involved in Ang-(1-7) production (ACE2 and Membrane metallo-endopeptidase; MME) are down regulated in response to glucocorticoid administration contributing to IUGR. METHODS: Pregnant female Sprague-Dawley rats were injected with dexamethasone (DEX; 0.4 mg/kg/day) starting from 14 day gestation (dg) till sacrifice at 19 or 21 dg while control groups were injected with saline (n = 6/group). The gene and protein expression of ACE2, MME, Ang-(1-7) and Mas1 receptor in the placental labyrinth (LZ) and basal zones (BZ) were studied. RESULTS: DEX administration caused a reduction in LZ weight at 19 and 21 dg (p < 0.001). IUGR, as shown by decreased fetal weights, was evident in DEX treated rats at 21 dg (p < 0.01). ACE2 gene expression was elevated in the LZ of control placentas at 21 dg (p < 0.01) compared to 19 dg and DEX prevented this rise at both gene (p < 0.01) and protein levels (p < 0.05). In addition, Ang-(1-7) protein expression in LZ was significantly reduced in DEX treated rats at 21 dg (p < 0.05). On the other hand, Mas1 and MME were upregulated in LZ at 21 dg in both groups (p < 0.05 and p < 0.001, respectively). CONCLUSION: The results of this study indicate that a reduced expression of ACE2 and Ang-(1-7) in the placenta by DEX treatment may be responsible for IUGR and consequent disease programming later in life.


Subject(s)
Angiotensin I/metabolism , Dexamethasone/adverse effects , Fetal Growth Retardation/metabolism , Glucocorticoids/adverse effects , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Angiotensin-Converting Enzyme 2 , Animals , Female , Fetal Growth Retardation/chemically induced , Placenta/drug effects , Placenta/metabolism , Pregnancy , Proto-Oncogene Mas , Rats , Rats, Sprague-Dawley
13.
Reprod Sci ; 24(8): 1164-1175, 2017 08.
Article in English | MEDLINE | ID: mdl-27932593

ABSTRACT

Molecular mechanisms affecting placental formation in intrauterine growth-restricted (IUGR) pregnancies are not clearly understood. Since metastasis tumor antigens (MTAs) MTA1 and MTA2 promote cell proliferation and MTA3 suppresses it, we hypothesized that IUGR alters cell survival/cell death programs driven by placental MTAs. To induce IUGR, pregnant Sprague Dawley rats were given daily intraperitoneal injections of either saline or dexamethasone (0.4 mg/kg) starting from 14 days of gestation (dg) to either 19 dg or 21 dg. Gene and protein expressions of MTA1-3 in the placental basal and labyrinth zones were investigated by real-time polymerase chain reaction, Western blotting, and immunohistochemistry. We also explored the expressions of proliferating cell nuclear antigen (PCNA), caspase-3, p53, p21, and ß-catenin. Dexamethasone-induced IUGR resulted in decreased expression of MTA1 in the nuclei of cells in the basal zone. The expression of p21 was increased and that of PCNA was reduced in both placental zones of IUGR rats. Cytoplasmic expression of MTA1 and p53 increased in the labyrinth zone of IUGR placentas in association with an increase in cell death as indicated by an increased caspase-3 expression. The labyrinth zone of IUGR placentas showed a significant reduction in MTA2-MTA3 gene expression and an increase in p53 protein levels. Total MTA3 level increased and ß-catenin level decreased in the labyrinth zone of IUGR placentas associated with a reduction in cell proliferation. Taken together, these results strongly suggest that dexamethasone-induced IUGR is associated with changes in MTA expression, decreased cell proliferation, and increased cell death in placentas.


Subject(s)
Cell Proliferation/drug effects , Dexamethasone/pharmacology , Fetal Growth Retardation/metabolism , Placenta/metabolism , Proteins/genetics , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/genetics , Caspase 3/metabolism , Cell Death/drug effects , Cell Death/physiology , Cell Proliferation/physiology , Female , Fetal Growth Retardation/chemically induced , Pregnancy , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proteins/metabolism , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , beta Catenin/genetics , beta Catenin/metabolism
14.
Data Brief ; 10: 230-237, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27995160

ABSTRACT

This article contains data related to the article "Effects of Trans-Resveratrol on hyperglycemia-induced abnormal spermatogenesis, DNA damage and alterations in poly (ADP-ribose) polymerase signaling in rat testis" (A. Abdelali, M. Al-Bader, N. Kilarkaje, 2016) [1]. The data are related to Resveratrol on diabetes-induced changes in blood glucose levels, body weights of rats, sperm count and motility, expression of poly (ADP-ribose) polymerase-1 (PARP1) in Leydig cells and in intratesticular blood vessels, and stage-dependent expression of PARP1 and Sirtuin 1 (SirT1) in the rat testis. In this experiment, the data were obtained from control, Resveratrol-treated, diabetic and Resveratrol-treated diabetic rats on day 42 after the induction of diabetes. Resveratrol treatment for a group each of normal and diabetic rats started on day 22 and extended up to day 42. The sperm parameters were conducted in samples obtained from the epididymis. The expression of proteins was evaluated by immunohistochemistry by using specific primary antibodies. The data are presented in the form of figures and significance of them has been given in the research article [1].

15.
Eur J Pharmacol ; 791: 675-685, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27693517

ABSTRACT

Gestational carbenoxolone exposure inhibits placental 11ß-hydroxysteroid dehydrogenase (11ß-HSD), the physiological barrier for glucocorticoids, which increases fetal exposure to glucocorticoids and induces intrauterine growth restriction (IUGR). We hypothesized that carbenoxolone exposure influences the expression of placental estrogen receptors-α and ß (ERα & ERß) and p53 leading to inhibited fetal and placental growth. Pregnant Sprague-Dawley rats were injected twice daily with either carbenoxolone (10mg/kg; s.c.) or vehicle (control group) from gestational days (dg) 12 onwards. Maternal blood and placentas were collected on 16 dg, 19 dg and 21 dg. The expression of ERα, ERß and p53 were studied in placental basal and labyrinth zones by RT-PCR, Western blotting and immunohistochemistry. Carbenoxolone did not affect placental and fetal body weights, but ELISA showed decreased estradiol levels on 19 dg and 21 dg, and increased maternal luteinizing hormone levels on all dg. The follicle stimulating hormone levels decreased on 16 dg and 19 dg, and increased on 21 dg. Carbenoxolone decreased ERα mRNA levels on 16 dg in both zones and its protein level on 19 dg in the labyrinth zone. However, carbenoxolone increased ERß mRNA levels on 19 dg and 21 dg and protein levels on 16 dg and 19 dg in the labyrinth zone. The p53 mRNA levels increased on all dg, but its protein levels increased on 21 dg in both zones. In conclusion, carbenoxolone exposure changes placental p53, ERα, ERß expression in favor of cell death but these changes do not induce IUGR in rats.


Subject(s)
Carbenoxolone/adverse effects , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Gene Expression Regulation/drug effects , Maternal Exposure/adverse effects , Placenta/drug effects , Tumor Suppressor Protein p53/metabolism , Animals , Body Weight/drug effects , Cell Proliferation/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Female , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Mothers , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Placenta/cytology , Placenta/metabolism , Pregnancy , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Time Factors , Tumor Suppressor Protein p53/genetics
16.
Toxicol Appl Pharmacol ; 311: 61-73, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27687054

ABSTRACT

Diabetes induces oxidative stress, DNA damage and alters several intracellular signaling pathways in organ systems. This study investigated modulatory effects of Trans-Resveratrol on type 1 diabetes mellitus (T1DM)-induced abnormal spermatogenesis, DNA damage and alterations in poly (ADP-ribose) polymerase (PARP) signaling in rat testis. Trans-Resveratrol administration (5mg/kg/day, ip) to Streptozotocin-induced T1DM adult male Wistar rats from day 22-42 resulted in recovery of induced oxidative stress, abnormal spermatogenesis and inhibited DNA synthesis, and led to mitigation of 8-hydroxy-2'-deoxyguanosine formation in the testis and spermatozoa, and DNA double-strand breaks in the testis. Trans-Resveratrol aggravated T1DM-induced up-regulation of aminoacyl tRNA synthetase complex-interacting multifunctional protein 2 expression; however, it did not modify the up-regulated total PARP and down-regulated PARP1 expressions, but recovered the decreased SirT1 (Sirtuin 1) levels in T1DM rat testis. Trans-Resveratrol, when given alone, reduced the poly (ADP-ribosyl)ation (pADPr) process in the testis due to an increase in PAR glycohydrolase activity, but when given to T1DM rats it did not affect the pADPr levels. T1DM with or without Trans-Resveratrol did not induce nuclear translocation of apoptosis-inducing factor and the formation of 50 kb DNA breaks, suggesting to the lack of caspase-3-independent cell death called parthanatos. T1DM with or without Trans-Resveratrol did not increase necrotic cell death in the testis. Primary spermatocytes, Sertoli cells, Leydig cells and intra-testicular vessels showed the expression of PARP pathway related proteins. In conclusion, Trans-Resveratrol mitigates T1DM-induced sperm abnormality and DNA damage, but does not significantly modulate PARP signaling pathway, except the SirT1 expression, in the rat testis.


Subject(s)
DNA Damage/drug effects , Hyperglycemia/physiopathology , Poly(ADP-ribose) Polymerases/metabolism , Signal Transduction/drug effects , Spermatogenesis/drug effects , Stilbenes/pharmacology , Testis/drug effects , Animals , Diabetes Mellitus, Type 1/physiopathology , Male , Oxidation-Reduction , Oxidative Stress , Rats , Rats, Wistar , Resveratrol , Spermatozoa/drug effects , Spermatozoa/metabolism , Testis/enzymology
17.
Toxicol Ind Health ; 32(1): 89-97, 2016 Jan.
Article in English | MEDLINE | ID: mdl-24021429

ABSTRACT

Metastasis-associated protein 1 (MTA1) is involved in tumor growth and metastasis of cancers. Being a component of nucleosome remodeling and histone deacetylase complex, the protein is also associated with DNA damage response pathway. Since the protein is involved in cancer pathology, we first investigated the effects of bleomycin, etoposide, and cisplatin (BEP) on MTA1 signaling in the testis. Second, since the antioxidants (AOs) have protective effects, we further investigated whether or not an AO cocktail modulates the effects of the drugs. Adult male Sprague Dawley rats (N = 4) were treated either with saline, or AO (α-tocopherol, l-ascorbic acid, zinc, and selenium), or therapeutic dose levels of etoposide (15 mg/kg) and cisplatin (3 mg/kg) from day 1-4 of the week and B (1.5 mg/kg) on the second day of the week, or BEP + AO. The real-time polymerase chain reaction showed that MTA1 and MTA1s (short form) gene expression was downregulated in AO (100% and 100%), BEP (86% and 71%), and BEP + AO (97% and 93%) groups. Western blotting and immunohistochemistry results showed that unnormalized MTA1 protein expression was upregulated in AO (38%) and BEP + AO (34%) groups; however, the MTA1/ß-actin ratio was upregulated in all treated groups (21, 19, and 15%, respectively). In conclusion, the results indicate that both BEP and AO suppress MTA1 and MTA1s transcription, which may render the germ cells to be more prone to apoptosis. However, upregulation of MTA1 protein expression may be related to induced DNA damage. Modulation of MTA1 signaling is a novel mechanism of action of BEP and AO, which may be useful in developing newer anticancer drugs.


Subject(s)
Antineoplastic Agents/adverse effects , Antioxidants/pharmacology , Proteins/metabolism , Testicular Neoplasms/drug therapy , Testis/drug effects , Actins/genetics , Actins/metabolism , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Ascorbic Acid/pharmacology , Bleomycin/administration & dosage , Bleomycin/adverse effects , Cisplatin/administration & dosage , Cisplatin/adverse effects , DNA Damage/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Etoposide/administration & dosage , Etoposide/adverse effects , Histone Deacetylases/metabolism , Male , Proteins/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Selenium/pharmacology , Signal Transduction , Testis/metabolism , Up-Regulation , alpha-Tocopherol/pharmacology
18.
Reprod Sci ; 22(4): 484-94, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25217305

ABSTRACT

Metastasis-associated protein 1 (MTA1) and its short form (MTA1s) regulate the function of estrogen receptors (ERs). Estrogens, via ERs, affect placental growth and fetal development, a process that may involve MTA1 signaling. Expression of MTA1, MTA1s, ERα, and ERß genes and proteins in rat placentas was studied on 16, 19, and 21 days of gestation (dg). The ERß messenger RNA decreased significantly toward the end of gestation, whereas its protein level increased in the nuclear fraction on 21 dg. Both MTA1 and MTA1s increased with gestation. Decidual, trophoblast giant, glycogen, and villous trophoblast cells expressed MTA1, ERα, and ERß proteins on all dg with colocalization of MTA1 with ERα and ERß in the nucleus and cytoplasm. Expression of MTA1 suggests a possible role in regulating placental functions; considering the repressive function of MTA1 on ERs, the expression of MTA1 suggests that placental cells may be less sensitive to estrogens during late pregnancy.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Proteins/metabolism , Animals , Estradiol/blood , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Female , Gene Expression Regulation, Developmental , Gestational Age , Placenta/cytology , Placenta/metabolism , Pregnancy , Proteins/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley
19.
Eur J Pharmacol ; 747: 150-9, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25523482

ABSTRACT

Cytotoxic anticancer chemotherapy affects pituitary-testicular hormonal axis in humans and in animals. This study investigated the effects on Leydig cells of three cycles of bleomycin, etoposide and cisplatin (0.75, 7.5, and 1.5mg/kg, respectively; BEP) chemotherapy in rat testis. The chemotherapy has induced hyperplasia of and degenerative changes in Leydig cells at the end of BEP exposure, which remained so even after a recovery time of 63 days. The increased testicular oxidative stress at the end of the chemotherapy returned to normal level after the recovery time. The chemotherapy has stimulated the transcription of scavenger receptor class type-B1 (SCARB1), steroidogenic acute-regulatory protein (StAR), cytochrome P450 cholesterol side-chain cleavage (CYP11A1), CYP17A1, and inhibited that of 17ß-hydroxysteroid dehydrogenase (HSD17B6) and CYP19A1 in association with increased cholesterol and decreased testosterone levels. Even after the recovery time, the chemotherapy still had inhibitory effects on the transcription of all of the above genes in addition to luteinizing hormone receptor and HSD3B1, but not on the StAR gene. The cholesterol and testosterone levels also did not show any significant differences with the control group. The decreased testosterone level at the end of chemotherapy was probably due to inhibition of HSD3B1 and HSD17B6 genes. In conclusion, clinically relevant dose-levels and treatment protocols of BEP chemotherapy adversely affect Leydig cell function. The BEP chemotherapy inhibits the transcription of steroidogenic enzymes and that these effects sustain over an extended period of time without returning to normal levels.


Subject(s)
Bleomycin/pharmacology , Cisplatin/pharmacology , Etoposide/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Leydig Cells/drug effects , Testosterone/biosynthesis , Transcription, Genetic/drug effects , Animals , Cholesterol/metabolism , Dose-Response Relationship, Drug , Leydig Cells/metabolism , Luteinizing Hormone/metabolism , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Testosterone/metabolism , Time Factors
20.
Reprod Sci ; 22(1): 102-12, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24828139

ABSTRACT

Diabetes is increasingly becoming a major cause of large-scale morbidity and mortality. Diabetes-induced oxidative stress alters numerous intracellular signaling pathways. Although testicular dysfunction is a major concern in diabetic men, the mechanistic alterations in the testes that lead to hypogonadism are not yet clear. Oxidative mitochondrial DNA damage, as indicated by 7,8-dihydro-8-oxo-2'-deoxyguanosine, and phosphorylation of p53 at ser315 residue (p-p53ser315) increased in a stage- and cell-specific manner in the testes of rats that were diabetic for 1 month (DM1). Prolongation of diabetes for 3 months (DM3) led to an increase in nuclear oxidative DNA damage in conjunction with a decrease in the expression of p-p53ser315. The nuclei of pachytene and preleptotene spermatocytes, steps 1, 11, and 12 spermatids, secondary spermatocytes and the Sertoli cells, and the meiotic figures showed an increase in the expression of p-p53ser315. An increase in the expression of a downstream target of p53 and protein 21(cyclin-dependent kinase interacting protein 1/wild-type p53-activated factor 1) (p21(CIP1/Waf1)) in both diabetic groups did not show any time-dependent effects but occurred concurrent with an upregulation of p-p53ser315 in DM1 and a downregulation of the protein in DM3. In diabetic groups, the expression of p21(CIP1/Waf1) was mainly cytoplasmic but also perinuclear in pachytene spermatocytes and round spermatids. The cytoplasmic localization of p21(CIP1/Waf1) may be suggestive of an antiapoptotic role for the protein. The perinuclear localization is probably related to the cell cycle arrest meant for DNA damage repair. Diabetes upregulates p21(CIP1/Waf1) signaling in testicular germ cells in association with alteration in p-p53ser315 expression, probably to counteract DNA damage-induced cell death.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , DNA, Mitochondrial/metabolism , Diabetes Mellitus, Experimental/metabolism , Oxidative Stress , Signal Transduction , Spermatozoa/metabolism , Testis/metabolism , Tumor Suppressor Protein p53/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Animals , Apoptosis , Cell Cycle , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Diabetes Mellitus, Experimental/pathology , Male , Phosphorylation , Rats, Sprague-Dawley , Spermatozoa/pathology , Testis/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL