Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Poult Sci ; 95(11): 2624-2632, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27143762

ABSTRACT

This study examined the ability of flax-based ingredients to attenuate nonalcoholic fatty liver disease ( NAFLD: ) in aged laying hens-a novel and more physiologically relevant model of human disease. Our results showed only hens supplemented with whole flaxseed ( WFX: ) reduced steatosis and hepatocellular ballooning. Serum AST was also reduced in hens provided WFX and defatted flaxseed meal ( DFM: ). Hepatic ω-3 PUFA enrichment was improved with supplementation of WFX, DFM, and flaxseed oil ( FXO: ). However, this effect was more evident in the WFX group. In contrast, transcript abundance of genes linked to NAFLD were predominantly modified with FXO supplementation. Taken together, our data indicate a potential synergistic relationship between the fatty acid and lignan content in flaxseed which attenuated the progression of NAFLD in aged laying hens. Although more research is necessary, these findings demonstrate the potential use of whole flaxseed for the treatment and prevention of NAFLD in humans.


Subject(s)
Diet , Flax/metabolism , Non-alcoholic Fatty Liver Disease/diet therapy , Animal Feed/analysis , Animals , Chickens/physiology , Disease Models, Animal , Female , Flax/chemistry , Non-alcoholic Fatty Liver Disease/etiology , Random Allocation , Seeds/chemistry
2.
Reproduction ; 151(3): 271-83, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26647418

ABSTRACT

Luteinized unruptured follicle (LUF) syndrome is a recurrent anovulatory dysfunction that affects up to 23% of women with normal menstrual cycles and up to 73% with endometriosis. Mechanisms underlying the development of LUF syndrome in mares were studied to provide a potential model for human anovulation. The effect of extended increase in circulating LH achieved by administration of recombinant equine LH (reLH) or a short surge of LH and decrease in progesterone induced by prostaglandin F2α (PGF2α) on LUF formation (Experiment 1), identification of an optimal dose of COX-2 inhibitor (flunixin meglumine, FM; to block the effect of prostaglandins) for inducing LUFs (Experiment 2), and evaluation of intrafollicular endocrine milieu in LUFs (Experiment 3) were investigated. In Experiment 1, mares were treated with reLH from Day 7 to Day 15 (Day 0=ovulation), PGF2α on Day 7, or in combination. In Experiment 2, FM at doses of 2.0 or 3.0 mg/kg every 12 h and human chorionic gonadotropin (hCG) (1500 IU) were administered after a follicle ≥32 mm was detected. In Experiment 3, FM at a dose of 2.0 mg/kg every 12 h plus hCG was used to induce LUFs and investigate the intrafollicular endocrine milieu. No LUFs were induced by reLH or PGF2α treatment; however, LUFs were induced in 100% of mares using FM. Intrafollicular PGF2α metabolite, PGF2α, and PGE2 were lower and the ratio of PGE2:PGF2α was higher in the induced LUF group. Higher levels of intrafollicular E2 and total primary sex steroids were observed in the induced LUF group along with a tendency for higher levels of GH, cortisol, and T; however, LH, PRL, VEGF-A, and NO did not differ between groups. In conclusion, this study reveals part of the intrafollicular endocrine milieu and the association of prostaglandins in LUF formation, and indicates that the mare might be an appropriate model for studying the poorly understood LUF syndrome.


Subject(s)
Anovulation/etiology , Dinoprost/physiology , Disease Models, Animal , Horses , Luteinizing Hormone/physiology , Animals , Clonixin/analogs & derivatives , Female
3.
Oncogene ; 33(28): 3619-33, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-23934194

ABSTRACT

Epithelial ovarian cancer (OvCa) is associated with high mortality and, as the majority (>75%) of women with OvCa have metastatic disease at the time of diagnosis, rates of survival have not changed appreciably over 30 years. A mechanistic understanding of OvCa initiation and progression is hindered by the complexity of genetic and/or environmental initiating events and lack of clarity regarding the cell(s) or tissue(s) of origin. Metastasis of OvCa involves direct extension or exfoliation of cells and cellular aggregates into the peritoneal cavity, survival of matrix-detached cells in a complex ascites fluid phase and subsequent adhesion to the mesothelium lining covering abdominal organs to establish secondary lesions containing host stromal and inflammatory components. Development of experimental models to recapitulate this unique mechanism of metastasis presents a remarkable scientific challenge, and many approaches used to study other solid tumors (for example, lung, colon and breast) are not transferable to OvCa research given the distinct metastasis pattern and unique tumor microenvironment (TME). This review will discuss recent progress in the development and refinement of experimental models to study OvCa. Novel cellular, three-dimensional organotypic, and ex vivo models are considered and the current in vivo models summarized. The review critically evaluates currently available genetic mouse models of OvCa, the emergence of xenopatients and the utility of the hen model to study OvCa prevention, tumorigenesis, metastasis and chemoresistance. As these new approaches more accurately recapitulate the complex TME, it is predicted that new opportunities for enhanced understanding of disease progression, metastasis and therapeutic response will emerge.


Subject(s)
Models, Biological , Neoplasms, Glandular and Epithelial , Ovarian Neoplasms , Animals , Animals, Genetically Modified , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Transformation, Neoplastic , Humans , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Tumor Microenvironment
4.
J Androl ; 32(3): 307-23, 2011.
Article in English | MEDLINE | ID: mdl-20966422

ABSTRACT

Hypoxia-inducible factors (HIF) are transcription factors that serve essential regulatory roles in cellular and molecular responses to oxygen debt. HIFs are composed of hypoxia-dependent α subunits (1α, 2α, 3α) and an oxygen-independent ß subunit. Previously we demonstrated that HIF-1, the master regulator of hypoxic responses, is expressed in the adult rat testis. We hypothesized that HIF-1 is involved in regulating responses to oxygen tension in the testis. Goals of this study were to determine if HIF-2α and HIF-3α are expressed in rat testis, identify testis cell types that express HIF-1α, and examine patterns of testicular HIF-1α protein expression under conditions of ischemia and hypoxia in vivo and in vitro. Reverse transcriptase polymerase chain reaction revealed that mRNA for Hif-1α, Hif-2α, and Hif-3α is expressed in the testis. The HIF-1α protein is the predominant subunit in testis. HIF-1α protein was abundant in normoxic testis, and its levels remained unchanged following ischemia created by surgically induced testicular torsion and reperfusion. Immunoblot and immunocytochemical experiments demonstrated that Leydig cells are the major source of HIF-1α in normoxic and hypoxic testes. To examine potential mechanisms of testicular HIF-1 stabilization, nuclear proteins from Leydig cells cultured in 5% or 21% oxygen, or cells cultured with H2O2, were analyzed by immunoblotting. Levels of HIF-1α were significantly diminished in 5% or 21% oxygen cultures compared with freshly isolated cells. Treating Leydig cells with H2O2 as a source of reactive oxygen species did not affect HIF-1α levels. High levels of constitutively expressed HIF-1α in normoxic Leydig cells suggest potentially unique roles for HIF-1 in Leydig cell responsiveness to oxygen.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Leydig Cells/metabolism , Animals , Base Sequence , Blotting, Western , DNA Primers , Electrophoresis, Polyacrylamide Gel , Hypoxia-Inducible Factor 1/genetics , Immunoprecipitation , In Vitro Techniques , Male , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
5.
Andrologia ; 35(1): 55-63, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12558529

ABSTRACT

Inflammatory disease is known to affect male reproductive function and fertility. Male accessory gland infections (MAGI) account for almost 15% of all cases of male infertility seen in infertility clinics. Infections of the male accessory glands are associated with increased counts of white blood cells in semen and elevated levels of pro-inflammatory cytokines in the semen and the testis. Numerous studies have underscored the importance of cytokines in the regulation of testicular and glandular function during pathophysiological events as well as under normal physiological conditions when cytokines act as growth and differentiation factors. The purpose of this paper is to particularly review the role of cytokines in the regulation of Leydig cell function in the testis primarily under pathophysiological conditions, and also considers clinical investigations that help to improve the evaluation and treatment of male infertility.


Subject(s)
Cytokines/physiology , Hormones/physiology , Immunity , Leydig Cells/physiology , Animals , Genital Diseases, Male , Humans , Inflammation , Interferons/physiology , Interleukins/physiology , Lipopolysaccharides , Male , Spermatogenesis , Steroids/biosynthesis , Tumor Necrosis Factor-alpha/physiology
6.
Eur J Endocrinol ; 147(3): 387-95, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12213677

ABSTRACT

OBJECTIVE: The aim of the present work was to study whether immunocytochemical parameters present in the normal ovary were altered after tumor development under high gonadotropin levels. METHODS: Ovarian tumors (luteoma): castrated female rats had an ovary grafted into the spleen; tumors were left to develop for 1, 2, 3 or 7 months. The presence of apoptotic cells (TUNEL method) and the expression of proliferating cell nuclear antigen (PCNA), gap junction protein (Cx43), steroidogenic acute regulatory protein (StAR), aromatase and synaptosome-associated protein of 25 kDa (SNAP-25) were determined by immunocytochemistry. Some of these findings were confirmed by RT-PCR (Cx43, StAR, SNAP-25). Inhibin subunit mRNAs were investigated by Northern blot. RESULTS: PCNA staining of tumors was mainly found in granulosa cells of transforming follicles and was absent from luteinized follicles. A nearly complete absence of apoptosis was observed. Cx43 was mainly found in follicles, while it was very weakly expressed or absent in luteinized follicles. StAR protein expression, indicating active steroidogenesis, was demonstrated only in luteinized follicles and in thecal cells, but was absent from granulosa cells. Aromatase immunoreactivity was very intense in granulosa and also present in luteal cells. Membrane-associated and cytoplasmic SNAP-25 immunostaining was determined in patches of endocrine cells in the follicles, as well as in the luteinized follicles. The expression of mRNAs for Cx43, StAR and SNAP-25 (RT-PCR) and inhibin subunits (Northern blots) were confirmed in 1-, 3- and 7-month-old tumors. CONCLUSIONS: These results indicated that luteoma most likely develop from unruptured follicles by hypertrophy and proliferation of follicular cells. Circulating gonadotropins seem to play a fundamental role in maintaining the expression of proteins typically expressed in normal ovary, while avoiding apoptosis in this tissue.


Subject(s)
Disease Models, Animal , Immunohistochemistry , Ovarian Neoplasms/chemistry , Animals , Apoptosis , Aromatase/analysis , Blotting, Northern , Cell Division , Connexin 43/analysis , Connexin 43/genetics , Female , In Situ Nick-End Labeling , Luteoma/chemistry , Membrane Proteins/analysis , Membrane Proteins/genetics , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Ovariectomy , Ovary/transplantation , Phosphoproteins/analysis , Phosphoproteins/genetics , Proliferating Cell Nuclear Antigen/analysis , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spleen/chemistry , Synaptosomal-Associated Protein 25 , Time Factors , Tyrosine 3-Monooxygenase/analysis
7.
J Biol Chem ; 276(49): 46583-96, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11579102

ABSTRACT

The metabolism of cholesterol by cytochrome P450 side chain cleavage enzyme is hormonally regulated in steroidogenic tissues via intramitochondrial cholesterol transport. The mediating steroidogenic acute regulatory protein (StAR) is synthesized as a 37-kDa (p37) precursor that is phosphorylated by protein kinase A and cleaved within the mitochondria to generate 30-kDa forms (p30, pp30). The effectiveness of modified recombinant StAR forms in COS-1 cells without mitochondrial import has led to a prevailing view that cholesterol transport is mediated by p37 StAR via activity on the outer mitochondrial membrane. The present study of the activation of cholesterol metabolism by bromo-cAMP in adrenal cells in relation to (35)S-StAR turnover indicates that targeting of pp30 to the inner membrane provides the dominant cholesterol transport mechanism. We show that 1) only newly synthesized StAR is functional, 2) phosphorylation and processing of p37 to pp30 occurs rapidly and stoichiometrically, 3) both steps are necessary for optimum transport, and 4) newly synthesized pp30 exhibits very high activity (400 molecules of cholesterol/StAR/min). Segregation of cAMP activation and synthesis of StAR from cholesterol metabolism showed that very low levels of newly synthesized StAR (1 fmol/min/10(6) cells) sustained activated cholesterol metabolism (0.4 pmol/min/10(6) cells, t(1/2) = 70 min) long after complete removal of p37 (t(1/2) = 5 min). This activity was highly sensitive to inhibition of processing by CCCP only until sufficient pp30 was formed. Maximum activation preceded bromo-cAMP-induced StAR expression, indicating other limiting steps in cholesterol metabolism.


Subject(s)
Adrenal Cortex/metabolism , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cholesterol/metabolism , Mitochondria/metabolism , Phosphoproteins/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adrenal Cortex/cytology , Adrenal Cortex/enzymology , Animals , Biological Transport , Female , Hydrolysis , Kinetics , Phenanthrolines/pharmacology , Phosphorylation , Rats , Rats, Sprague-Dawley
8.
Proc Natl Acad Sci U S A ; 98(14): 7988-93, 2001 Jul 03.
Article in English | MEDLINE | ID: mdl-11427738

ABSTRACT

DAX-1 [dosage-sensitive sex reversal, adrenal hypoplasia congenita (AHC) critical region on the X chromosome, gene 1] is an orphan nuclear receptor that represses transcription by steroidogenic factor-1 (SF-1), a factor that regulates expression of multiple steroidogenic enzymes and other genes involved in reproduction. Mutations in the human DAX1 gene (also known as AHC) cause the X-linked syndrome AHC, a disorder that is associated with hypogonadotropic hypogonadism also. Characterization of Dax1-deficient male mice revealed primary testicular defects that included Leydig cell hyperplasia (LCH) and progressive degeneration of the germinal epithelium, leading to infertility. In this study, we investigated the effect of Dax1 disruption on the expression profile of various steroidogenic enzyme genes in Leydig cells isolated from Dax1-deficient male mice. Expression of the aromatase (Cyp19) gene, which encodes the enzyme that converts testosterone to estradiol, was increased significantly in the Leydig cells isolated from mutant mice, whereas the expression of other proteins (e.g., StAR and Cyp11a) was not altered. In in vitro transfection studies, DAX-1 repressed the SF-1-mediated transactivation of the Cyp19 promoter but did not inhibit the StAR or Cyp11a promoters. Elevated Cyp19 expression was accompanied by increased intratesticular levels of estradiol. Administration of tamoxifen, a selective estrogen-receptor modulator, restored fertility to the Dax1-deficient male mice and partially corrected LCH, suggesting that estrogen excess contributes to LCH and infertility. Based on these in vivo and in vitro analyses, aromatase seems to be a physiologic target of Dax-1 in Leydig cells, and increased Cyp19 expression may account, in part, for the infertility and LCH in Dax1-deficient mice.


Subject(s)
Aromatase/genetics , DNA-Binding Proteins/genetics , Receptors, Retinoic Acid/genetics , Repressor Proteins , Transcription Factors/genetics , Animals , DAX-1 Orphan Nuclear Receptor , Infertility, Male/genetics , Leydig Cells/physiology , Male , Mice , Steroidogenic Factor 1 , Up-Regulation
10.
Mol Cell Endocrinol ; 171(1-2): 49-51, 2001 Jan 22.
Article in English | MEDLINE | ID: mdl-11165010

ABSTRACT

Cultured human luteinized granulosa cells (GC) are a unique model to study regulation of the human corpus luteum, e.g. by local factors, including acetylcholine (ACh). GC express functional ACh-receptors of the M1-type (M1R). Priming with the cholinergic agonist carbachol for 24 h, followed by a 24 h stimulation with human chorionic gonadotrophin (hCG), significantly increased progesterone levels (up to 2-fold), compared with hCG stimulation alone. This is likely due to the effect of carbachol, observed after 24 h, to increase the levels of steroid acute regulatory (StAR) protein, as found in Western blots. StAR protein was increased (2-10-fold), while P450 side chain cleavage (SCC) enzyme was unaffected. Thus, one role of muscarinic receptor activation in human GC is regulation of StAR protein and, subsequently, progesterone production.


Subject(s)
Corpus Luteum/physiology , Granulosa Cells/metabolism , Phosphoproteins/metabolism , Receptors, Muscarinic/physiology , Carbachol/pharmacology , Cells, Cultured , Cholinergic Agonists/pharmacology , Chorionic Gonadotropin/pharmacology , Female , Humans , Progesterone/biosynthesis , Receptors, Muscarinic/drug effects
11.
Endocrinology ; 141(11): 4000-12, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11089530

ABSTRACT

Immune activation results in the activation of adrenal steroidogenesis and inhibition of gonadal steroidogenesis. Previous studies indicated that these effects were caused primarily by activation and suppression of the secretion of ACTH and LH, respectively. However, other evidence indicated a direct effect of the immune system on the gonads. In this study, serum testosterone, quantitated by RIA after lipopolysaccharide injection, showed a significant decrease within 2 h. Parallel measurement of serum LH showed no change. There were no differences in LH receptor or cAMP produced in Leydig cells between vehicle- and lipopolysaccharide-injected mice. The 30-kDa form of the steroidogenic acute regulatory (StAR) protein was quantitated, by Western blot, in Leydig cells and was found to decrease in a time-dependent manner. No change in StAR protein messenger RNA (mRNA) was detected by Northern analysis during this time, nor were any changes found in the levels of mRNA for the steroidogenic enzymes P450scc, 3beta-hydroxysteroid dehydrogenase delta4-delta5-isomerase, or P450c17. In the adrenal, StAR protein was increased, as was StAR protein mRNA. No changes were observed in the levels of mRNA for P450scc, 3beta-hydroxysteroid dehydrogenase delta4-delta5-isomerase, or P450c21. Thus, although the mechanisms of regulation differ, changes in the levels of StAR protein are a sensitive indicator of the steroidogenic capacity of these two tissues.


Subject(s)
Adrenal Glands/metabolism , Leydig Cells/metabolism , Lipopolysaccharides/pharmacology , Phosphoproteins/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , Animals , Blotting, Northern , Cholesterol Side-Chain Cleavage Enzyme/genetics , Escherichia coli , Male , Mice , Phosphoproteins/genetics , RNA, Messenger/metabolism , Steroid 21-Hydroxylase/genetics
12.
Curr Opin Urol ; 10(1): 39-44, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10650514

ABSTRACT

Male accessory sex gland infections are considered to be hazards to male fertility. Various pathophysiologic concepts have evolved from experimental and clinical studies that begin to explain the effects of bacteria and immunologic events on spermatozoa. Recent studies have identified and evaluated mediators that are responsible for specific molecular processes in infections that particularly affect the function of spermatozoa.


Subject(s)
Bacterial Infections/complications , Female Urogenital Diseases/complications , Infertility, Male/etiology , Male Urogenital Diseases , Sperm Motility , Spermatozoa/physiology , Cytokines/pharmacology , Female , Female Urogenital Diseases/microbiology , Genitalia, Male/microbiology , Humans , Infertility, Male/microbiology , Male , Sperm-Ovum Interactions
13.
J Steroid Biochem Mol Biol ; 75(2-3): 177-86, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11226834

ABSTRACT

In peripheral steroidogenic tissues, dehydroepiandrosterone (D) is formed from pregnenolone (P) by the microsomal cytochrome P450c17 enzyme. Although some steroidogenic P450s have been found in brain tissue, no enzyme has been shown to possess P450c17 activity. We recently demonstrated the presence of an alternative, Fe(2+)-dependent pathway responsible for D formation from alternative precursors in rat glioma cells. We and others could not find P450c17 mRNA and protein in rat brain, but demonstrate herein the presence of Fe(2+)-dependent alternative pathway for D formation in rat brain cortex microsomes. Using primary cultures of differentiating rat glial cells, we observed that P450c17 mRNA and protein were present in O-2A oligodendrocyte precursors and mature oligodendrocytes. In the presence of P, O-2A and mature oligodendrocytes formed D. Addition of Fe(2+) together with submaximal concentrations of P increased D formation by these cells. Treatment of oligodendrocytes with the P450c17 inhibitor SU 10603 in the presence or absence of P failed to inhibit D production. These data suggest that D formation in oligodendrocytes occurs independently of the P450c17 protein present in the cells. In isolated type I astrocytes we did not find neither P450c17 mRNA nor protein. These cells responded to Fe(2+) by producing D and addition of P together with Fe(2+) further increased D synthesis. SU 10603 failed to inhibit D formation by astrocytes. Taken together these results suggest that in differentiating rat brain oligodendrocytes and astrocytes D is formed via a P450c17-independent and oxidative stress-dependent alternative pathway.


Subject(s)
Brain/metabolism , Cerebral Cortex/physiology , Dehydroepiandrosterone/biosynthesis , Neuroglia/metabolism , Steroid Hydroxylases/metabolism , Animals , Brain/drug effects , Cell Differentiation , Cells, Cultured , Dehydroepiandrosterone/metabolism , Ferrous Compounds/pharmacology , Immunohistochemistry , Microsomes/metabolism , Neuroglia/drug effects , Pregnenolone/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Steroid 17-alpha-Hydroxylase/biosynthesis , Steroid Hydroxylases/genetics
14.
Endocrinology ; 140(11): 5154-64, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10537144

ABSTRACT

We have studied the in vivo expression of steroidogenic acute regulatory protein (StAR) in adrenals of control, ACTH-treated, and Na+-restricted rats. Indirect immunofluorescence by microscopy revealed the presence of StAR in the zonae glomerulosa (ZG) and fasciculata-reticularis (ZFR). An increased signal was observed in the ZG and zona fasciculata, 5 h after ACTH injection; a few cells of the medulla were also positive. Immunogold electron microscopy showed that StAR was mainly located over mitochondria (MT). By immunoblotting, a major 29-kDa and other minor StAR bands migrating between 30 and 39 kDa were increased 5 h after ACTH treatment but remained unchanged after 1 h. By two-dimensional-PAGE, four StAR species were revealed in homogenates of control ZG, and their intensity was increased 5 h after ACTH treatment but not after 1 h. Also, additional acidic species were seen 5 h after treatment. Other bands with basic isoelectric point were revealed between 29 and 37 kDa. Analyses on whole gland MT and supernatant (SN) revealed four bands in the control SN and five in ACTH SN; the intensity of one band was increased, and that of another one was decreased, in SN of treated rats. ACTH treatment resulted in the localization of many low-isoelectric point StARs in MT. After two-dimensional-PAGE, differences were found in the mobility of some StAR species in the ZG between controls and Na+-restricted rats. In MT, four bands were revealed in the ZG preparations of Na+-restricted and two bands in controls. Four bands were revealed in the ZG SNs of control and Na+-restricted rats; an additional band was observed only in the SN of treated animals, whereas the intensity of another band decreased. Na+ restriction did not affect StAR in the ZFR. In conclusion, StAR was present in the rat adrenal cortex ZG and ZFR and was mainly located in MT. StAR expression was inducible in the ZG and the ZF by ACTH, resulting in the formation of many StAR acidic species; interestingly, such changes were detectable 5 h, but not 1 h, after ACTH administration, suggesting that steroidogenesis stimulation by StAR might occur mainly outside MT. Although less spectacular than for ACTH, Na+ restriction also affected StAR expression in the ZG but not in the ZFR, by increasing two mitochondrial and one SN species, implying that StAR is involved in the mechanism of action of Na+ restriction in promoting aldosterone formation. These results suggest that differential processing and/or changes in phosphorylation may occur in vivo upon ACTH treatment and Na+ restriction. We hypothesize that modification of a relatively small quantity of StAR, mainly located outside MT, is necessary to increase adrenal steroidogenesis challenged either by ACTH or Na+ restriction.


Subject(s)
Adrenal Glands/metabolism , Adrenocorticotropic Hormone/pharmacology , Diet, Sodium-Restricted , Phosphoproteins/biosynthesis , Adrenal Glands/chemistry , Adrenal Glands/drug effects , Angiotensin II/pharmacology , Animals , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Fluorescent Antibody Technique, Indirect , Isoelectric Point , Male , Microscopy, Electron , Mitochondria/chemistry , Mitochondria/metabolism , Phosphoproteins/analysis , Rats , Tissue Distribution , Zona Fasciculata/chemistry , Zona Fasciculata/metabolism , Zona Glomerulosa/chemistry , Zona Glomerulosa/metabolism
15.
Endocrine ; 10(3): 201-17, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10484284

ABSTRACT

Inflammatory disease has been established to affect male reproductive function and fertility. Relevant inflammatory diseases include general and chronic infectious diseases as well as localized acute or chronic infections of the male genitourinary tract. Male accessory gland infections account for almost 15% of all cases of male infertility seen in infertility clinics while fertility usually is not a clinical objective among patients with acute systemic infections such as Gram-negative sepsis. Infections of the male accessory glands frequently are associated with increased counts of white blood cells in semen and elevated levels of proinflammatory cytokines in semen and the testis. There is a mounting body of evidence that demonstrates the importance of cytokines and chemokines in the regulation of testicular and glandular function during pathophysiological states as well as under normal physiological conditions when cytokines act as growth and differentiation factors. The purpose of this review is to examine the role of cytokines in the regulation of steroidogenesis and spermatogenesis in the testis under physiological and pathophysiological conditions and considers clinical investigations that help to improve the evaluation and treatment of male infertility.


Subject(s)
Cytokines/physiology , Testis/physiopathology , Animals , Humans , Immunity , Inflammation/physiopathology , Leydig Cells/immunology , Leydig Cells/physiology , Male , Steroids/biosynthesis , Testicular Diseases/immunology , Testicular Diseases/physiopathology , Testis/immunology
16.
Biol Reprod ; 61(3): 643-50, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10456840

ABSTRACT

With interest in steroidogenic acute regulatory protein (StAR) involvement in the luteolytic process, we studied changes in serum progesterone levels and the concomitant expression of StAR mRNA and protein (37-, 32-, and 30-kDa forms) in postovulatory Day 7 corpora lutea (CL) isolated from rats 1 h after injection with prostaglandin F(2alpha) (PGF(2alpha), n = 6) or saline (n = 6). Serum progesterone levels were determined by RIA, StAR and beta-actin mRNA expression by Northern analysis, and StAR and beta-actin protein expression by Western analysis. Adrenal, brain, and spleen from control animals were used as positive and negative controls for StAR expression. Scanning optical densitometry measurements were standardized by dividing the signal strength from each StAR autoradiogram lane by that from the corresponding beta-actin autoradiogram lane. ANOVA was used for significance testing, with alpha set at 0.05. The 37-, 32-, and 30-kDa forms of StAR protein were expressed in all adrenal samples, whereas only the 37- and 30-kDa forms were found in CL. Serum progesterone levels and expression of the 30-kDa and 37-kDa forms of the StAR protein in CL were all found to be significantly lower in the PGF(2alpha)-treated than the saline-treated group. StAR mRNA expression was not significantly different in the saline- and PGF(2alpha)-treated rats. The rapid decline in StAR protein expression that accompanies PGF(2alpha) induced luteolysis, therefore, does not result from significant decline in mRNA expression.


Subject(s)
Corpus Luteum/metabolism , Dinoprost/pharmacology , Gene Expression/drug effects , Phosphoproteins/genetics , Progesterone/biosynthesis , RNA, Messenger/metabolism , Actins/genetics , Animals , Autoradiography , Blotting, Northern , Female , Progesterone/blood , Rats , Rats, Sprague-Dawley
17.
Shock ; 11(4): 298-301, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10220309

ABSTRACT

The hypothesis that induction of chronic peritoneal sepsis would produce depression of serum testosterone due to a decrease in Leydig cell steroidogenic acute regulatory (StAR) protein or P450c17 steroidogenic enzyme was tested. Male Sprague-Dawley rats (350-400 g) were randomized to septic and nonseptic groups. Sepsis was induced with a cecal slurry (200 mg/kg in 5 mL of 5% dextrose in water (D5W); intraperitoneal) while nonseptic rats received only sterile D5W. Animals (n = 6, in each group) were killed by CO2 asphyxiation and blood samples were collected by direct cardiac puncture at 24 h after induction of sepsis/sham sepsis. The serum concentration of corticosterone, progesterone, estradiol, and testosterone was determined using radioimmunoassay. Western blot analysis was utilized to quantify Leydig cell StAR protein and P450c17 enzyme. Sepsis produced a significant decrease in the serum concentration of testosterone, a down-regulation of StAR protein, and an increase in serum estradiol 24 h after induction of sepsis (as compared with the nonseptic group). Protein levels of P450c17 in Leydig cells and serum concentrations of progesterone and corticosterone 24 h after induction of sham sepsis or sepsis were not different. It is concluded that the decreases in serum testosterone after 24 h of chronic peritoneal sepsis correlated with reductions in StAR protein.


Subject(s)
Phosphoproteins/metabolism , Sepsis/metabolism , Testosterone/blood , Animals , Leydig Cells/metabolism , Male , Peritonitis/metabolism , Rats , Rats, Sprague-Dawley , Steroid 17-alpha-Hydroxylase/metabolism
18.
Biol Reprod ; 60(2): 527-33, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9916024

ABSTRACT

Exposure to disease or injury often results in impaired reproductive activity accompanied by decreased testosterone levels. After immune activation, the cytokine interleukin 1-beta (IL-1beta) circulates in high concentrations, and its exogenous administration evokes many of the sequelae of immune activation. Previously, we have shown that the administration of this cytokine into the cerebral ventricles blunts hCG-stimulated testosterone secretion. This effect, though time-dependent, occurs before significant elevation of interleukin 6 in the peripheral bloodstream, does not depend on adrenal activation, and/or changes in LH concentrations, leading us to hypothesize a direct connection between the brain and testis. To explore this mechanism further, we isolated testicular tissue from rats treated intracerebroventricularly (icv) with vehicle or IL-1beta 30 or 90 min before they were killed. We found that in vivo cytokine treatment blunted ex vivo testosterone secretion in response to hCG, showing that the mechanism is independent of circulating cytokines. Though hCG binding was moderately reduced by icv IL-1beta in these preparations, the extent of this inhibition did not explain our observations. As the first acutely and hormonally regulated step in the biosynthesis of testosterone is the transfer of cholesterol into the inner mitochondrial membrane, which is mediated by steroidogenic acute regulatory (StAR) protein, we hypothesized that the rapid effects of icv IL-1beta on testicular responsiveness to hCG might be due to reduced levels of StAR. We report here that StAR protein was indeed reduced in Leydig cells isolated from rats treated in vivo with IL-1beta. Furthermore, treatment with a water-permeable form of cholesterol that bypasses the requirement for StAR partially restored hCG-stimulated testosterone secretion from testes isolated from rats treated icv with IL-1beta. Taken together, our data indicate that StAR plays a role in the suppression of testicular function evoked by central administration of IL-1beta.


Subject(s)
Interleukin-1/administration & dosage , Phosphoproteins/physiology , Testis/metabolism , Testosterone/metabolism , Animals , Chorionic Gonadotropin/metabolism , Hydroxycholesterols/pharmacology , Injections, Intraventricular , Interleukin-1/pharmacology , Leydig Cells/physiology , Luteinizing Hormone/metabolism , Male , Rats , Rats, Sprague-Dawley , Steroid 17-alpha-Hydroxylase/metabolism
19.
Alcohol Clin Exp Res ; 22(8): 1754-62, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9835291

ABSTRACT

Ethanol (EtOH) exerts deleterious actions on reproductive function at all three levels: the hypothalamus, pituitary, and gonad (HPG). Nitric oxide (NO), a newly identified messenger molecular in a variety of biological systems, has been suggested as playing a role in HPG hormone regulation. NO stimulates luteinizing hormone releasing hormone secretion from the hypothalamus and has variable effects on luteinizing hormone release from the pituitary. NO is inhibitory to testosterone production, and it may also directly inhibit some steroidogenic enzymes. Related studies in the accompanying paper have demonstrated that inhibiting NO synthase (NOS) using various NOS inhibitors can prevent the EtOH-induced suppression of testosterone on the male HPG axis, and this action is mainly, although not entirely, due to a direct gonadal effect. To further investigate the role of NO in the HPG axis, we assessed the HPG NO-NOS system by determining NOS mRNA levels, protein levels, and enzyme activity in the presence and absence of EtOH. At the testicular level, EtOH's action did not appear to be mediated by increasing NO content. However, EtOH was able to potentiate NO's suppressive effect on the testicular synthesis system. One locus where EtOH and NO interacted was at the steroidogenic enzyme level. N(G)-nitro-L-arginine methyl ester, a NOS inhibitor, was found to antagonize the EtOH-induced fall on P-450 17alpha-hydroxylase/C17-20 lyase mRNA levels when administered along with EtOH. EtOH had no apparent effect on the pituitary NO-NOS system and its effects on the hypothalamic NO-NOS system do not explain its ability to reduce luteinizing hormone releasing hormone secretion.


Subject(s)
Ethanol/toxicity , Hypothalamo-Hypophyseal System/drug effects , Nitric Oxide/physiology , Testis/drug effects , Animals , Brain Mapping , Gonadotropin-Releasing Hormone/blood , Hypothalamo-Hypophyseal System/physiology , Male , Nitric Oxide Synthase/physiology , Rats , Testis/physiology , Testosterone/blood
20.
Endocrinology ; 139(12): 4820-31, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9832418

ABSTRACT

Steroidogenic acute regulatory (StAR) protein is synthesized in response to tropic hormones to facilitate cholesterol transport to the inner mitochondrial membrane-bound P450 side-chain cleavage enzyme (P450scc), the first enzymatic step in the steroid hormone biosynthetic pathway. Gonadotropins activate expression of their target genes via the cAMP second messenger system. We have demonstrated that cAMP administration to rat luteal cells stimulates expression of both StAR messenger RNA and protein. Because cholesterol delivery is the first regulated step in steroidogenesis, and because StAR messenger RNA levels are increased in response to tropic hormone and cAMP stimulation, the mechanism by which tropic hormones/cAMP stimulate transcription needs to be elucidated. To this end, approximately 2.7 kb of the rat StAR promoter was isolated and sequenced. Sequence analysis revealed the presence of a TATA-like element as well as multiple regulatory motifs including steroidogenic factor 1 (SF-1) binding sites, an estrogen receptor half-site, and two AP-1 sites within the promoter region. 5'-RACE experiments determined the transcription start site to be located 82 bp upstream of the ATG translation start codon. Electrophoretic mobility shift assays and supershift analysis demonstrated SF-1 binding to three SF-1 binding sites in the rat StAR promoter with high affinity and two SF-1 binding sites with low affinity. Transfection of mouse Y1 adrenal tumor cells and human bladder carcinoma cells (HTB9s) with the rat StAR promoter demonstrated that SF-1 was able to activate transcription of the luciferase reporter gene and that the rat StAR promoter was responsive to cAMP. Nested deletions of the rat StAR promoter (1.9 kb) identified a region between -1413 and -998 that is essential for maximal activation of the rat StAR gene in HTB9 cells; however, deletion of this region does not affect responsiveness to cAMP. 5'-Deletion and site-directed mutagenesis experiments demonstrated that the SF-1 motifs identified within the rat StAR promoter (located at positions -764, -455, and -106) were sufficient to activate transcription as well as confer cAMP responsiveness to the rat StAR gene. Site-directed mutagenesis studies using the smallest promoter fragment demonstrated that the two proximal SF-1 binding sites are crucial for StAR gene transcription, both at a basal level and in response to cAMP stimulation. These studies provide novel insights into the regulation of the rat StAR gene at the transcriptional level by SF-1.


Subject(s)
DNA-Binding Proteins/physiology , Phosphoproteins/genetics , Transcription Factors/physiology , Transcription, Genetic/physiology , Animals , Base Sequence , Cyclic AMP/physiology , Female , Fushi Tarazu Transcription Factors , Gene Deletion , Homeodomain Proteins , Humans , Mice , Mutagenesis, Site-Directed , Phosphoproteins/isolation & purification , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear , Steroidogenic Factor 1 , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...