Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
J Cell Physiol ; 226(12): 3403-12, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21344398

ABSTRACT

The mechanisms by which varicocele affects fertility remain undetermined. Estrogens play a key role in the human male reproduction and human sperm expresses the estrogen receptors (ERs) and aromatase. In this study, by Western blotting we evidenced the ERs content concomitantly in healthy sperm and in oligoastenoteratozoospermic (OAT) samples without and with varicocele. In varicocele a strong reduction of the ERß was observed, while the ERα was almost absent. Besides, transmission electron microscopy (TEM) confirmed the reduction of ERs expression in "varicocele" sperm, indicating that varicocele has a detrimental effect on sperm structure at molecular level. To further define the estrogen significance in male gamete and the pathophysiology of varicocele we investigated both the expression of ERα and ERß in normal and pathologic sperm samples as well as we evaluated estradiol (E2) action on lipid and glucose sperm metabolism. Responses to E2 treatments on cholesterol efflux, protein tyrosine phosphorylations, motility, and acrosin activity in varicocele sperm were reduced or absent. The evaluation of the triglycerides content, lipase and acyl-CoA dehydrogenase activities, suggest that E2 exerts a lipolytic effect on human sperm metabolism. Concerning glucose metabolism, it appears that E2 induces G6PDH activity concomitantly to the insulin secretion. In "varicocele" sperm, the E2 did not induce energy expenditure. OAT sperm had E2-responsiveness but in a lesser extent with respect healthy sperm. This study discovered a novel role for E2/ERs in human sperm physiology, since they modulate sperm metabolism and new detrimental effects related to the pathophysiology of the varicocele condition.


Subject(s)
Energy Metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Infertility, Male/etiology , Spermatozoa/metabolism , Varicocele/complications , Acrosin/metabolism , Acyl-CoA Dehydrogenase/metabolism , Blotting, Western , Case-Control Studies , Cholesterol/metabolism , Estradiol/metabolism , Glucose/metabolism , Glucosephosphate Dehydrogenase/metabolism , Humans , Infertility, Male/metabolism , Infertility, Male/pathology , Infertility, Male/physiopathology , Insulin/metabolism , Italy , Lipase/metabolism , Male , Microscopy, Electron, Transmission , Phosphorylation , Sperm Capacitation , Sperm Motility , Spermatozoa/ultrastructure , Triglycerides/metabolism , Varicocele/metabolism , Varicocele/pathology , Varicocele/physiopathology
2.
Breast Cancer Res Treat ; 124(1): 63-77, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20052536

ABSTRACT

In breast tumors the expression of estrogen receptor alpha (ERα) is known to be associated with a more favorable prognosis. ERα expression has been reported to reduce the metastatic potential of breast cancer cells. Recently, we have observed that extracellular matrix proteins activate ERα and that both liganded and unliganded receptor modulate cell invasiveness acting at nuclear level. To explain the mechanisms by which ERα regulates cell adhesion, we have evaluated the expression of α(5)ß(1) integrin, prevalently expressed in stationary cells, in response to 17ß-estradiol (E2). Here we show that E2/ERα increases the expression of integrin α(5)ß(1) through Sp1-mediated binding to a GC-rich region located upstream of an ERE half-site in the 5' flanking region of the α(5) gene forming a ternary ERα-Sp1-DNA complex. Estrogen responsiveness of the α(5) gene promoter, as observed in HeLa cells, underlies a general mechanism of regulation which is not strictly linked to the cell type. Our data reveal novel insight into the molecular mechanisms sustaining the reduced invasiveness of ERα expressing cells demonstrating that α(5)ß(1) integrin expression is related to the maintenance of the stationary status of the cells, counteracting E2/ERα capability to enhance breast cancer cell migration and invasion.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Estrogen Receptor alpha/metabolism , Integrin alpha5/metabolism , Sp1 Transcription Factor/metabolism , 5' Flanking Region , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Integrin alpha5/genetics , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Neoplasm Invasiveness , RNA Interference , RNA, Messenger/metabolism , Response Elements , Time Factors , Transcription, Genetic , Transfection , Up-Regulation
3.
Reprod Biol Endocrinol ; 7: 140, 2009 Nov 30.
Article in English | MEDLINE | ID: mdl-19948036

ABSTRACT

BACKGROUND: A wider biological role of 1alpha,25-Dihydroxyvitamin D3 (1,25(OH)2D3), the active metabolite of vitamin D3, in tissues not primarily related to mineral metabolism was suggested. Recently, we evidenced the ultrastructural localization the 1,25(OH)2D3 receptor in the human sperm. However, the 1,25(OH)2D3 action in human male reproduction has not yet been clarified. METHODS AND RESULTS: By RT-PCR, Western blot and Immunofluorescence techniques, we demonstrated that human sperm expresses the 1,25(OH)2D3 receptor (VDR). Besides, 25(OH)D3-1 alpha-hydroxylase, evidenced by Western blot analysis, indicated that in sperm 1,25(OH)2D3 is locally produced, highlighting the potential for autocrine-paracrine responses. 1,25(OH)2D3 through VDR, increased intracellular Ca2+ levels, motility and acrosin activity revealing an unexpected significance of this hormone in the acquisition of fertilizing ability. In sperm, 1,25(OH)2D3 through VDR, reduces triglycerides content concomitantly to the increase of lipase activity. Rapid responses stimulated by 1,25(OH)2D3 have been observed on Akt, MAPK and GSK3 implying that this secosteroid is involved in different sperm signalling pathways. CONCLUSION: Our data extended the role of 1,25(OH)2D3 beyond its conventional physiological actions, paving the way for novel therapeutic opportunities in the treatment of the male reproduction disorders.


Subject(s)
Calcitriol/physiology , Germ Cells/physiology , Receptors, Calcitriol/physiology , Spermatozoa/physiology , Acrosin/metabolism , Acyl-CoA Dehydrogenase/metabolism , Adult , Blotting, Western , Calcium/metabolism , Cell Separation , Enzyme Activation/drug effects , Fluorescent Antibody Technique , Germ Cells/metabolism , Glucosephosphate Dehydrogenase/metabolism , Humans , In Vitro Techniques , Lipase/metabolism , Male , RNA/biosynthesis , RNA/isolation & purification , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Sperm Motility/physiology , Spermatozoa/enzymology , Spermatozoa/metabolism , Triglycerides/metabolism
4.
Hum Reprod ; 22(10): 2594-605, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17656415

ABSTRACT

BACKGROUND: Results from mice lacking the androgen receptor (AR) showed that it is critical for the proper development and function of the testes. The aim of this study was to investigate whether a functional AR is present in human sperm. METHODS: The expression of AR and its effects on sperm were evaluated by RT-PCR, Western Blot, Immunocytochemistry, PI3Kinase and DNA laddering assays. RESULTS: We showed in human sperm that AR is located at the head region. Dihydrotestosterone (DHT), in a dose-dependent manner, leads to the rapid phosphorylation of the AR on tyrosine, serine and threonine residues and this effect was reduced by the AR antagonist hydroxyflutamide (OH-Flut). The effects of AR were evaluated on the phosphoinositide-3 kinase/protein kinase B (PI3K/AKT) pathway. Specifically, 0.1 and 1 nM DHT stimulated PI3K activity, whereas 10 nM DHT decreased PI3K activity and levels of p-AKT S473 and p-AKT T308, p-BCL2, and enhanced phosphatase and tensin homologue (PTEN) phosphorylation. In addition, 10 nM DHT was able to induce the cleavage of caspases 8, 9 and 3 and cause DNA laddering, and these effects were reversed either by casodex or OHFlut. By using wortmannin, a specific PI3K inhibitor, the cleavage of caspase 3 was reproduced, confirming that in sperm the PI3K/AKT pathway is involved in caspase activation. CONCLUSIONS: Human sperm express a functional AR that have the ability to modulate the PI3K/AKT pathway, on the basis of androgen concentration.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/biosynthesis , Spermatozoa/metabolism , Androgens/pharmacology , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Survival , Dihydrotestosterone/pharmacology , Fluorescent Antibody Technique , Humans , Male , PTEN Phosphohydrolase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Testosterone/pharmacology , src-Family Kinases/metabolism
5.
J Cell Physiol ; 211(2): 448-56, 2007 May.
Article in English | MEDLINE | ID: mdl-17167783

ABSTRACT

The testis is an immunologically privileged site of the body where Sertoli cells work on to favor local immune tolerance by testicular autoantigens segregation and immunosuppressive factors secretion. Fas/Fas Ligand (FasL) system, expressed prevalently in Sertoli cells, has been considered to be one of the central mechanisms in testis immunological homeostasis. In different cell lines it has been reported that the proapoptotic protein FasL is regulated by 17-beta estradiol (E2). Thus, using as experimental model mouse Sertoli cells TM4, which conserve a large spectrum of functional features present in native Sertoli cells, like aromatase activity, we investigated if estradiol "in situ" production may influence FasL expression. Our results demonstrate that an aromatizable androgen like androst-4-ene-3,17-dione (Delta4) enhanced FasL mRNA, protein content and promoter activity in TM4 cells. The treatment with N(6),2'-O-dibutyryladenosine-3'-5'-cyclic monophosphate [(Bu)(2)cAMP] (simulating FSH action), that is well known to stimulate aromatase activity in Sertoli cells, amplified Delta4 induced FasL expression. Functional studies of mutagenesis, electrophoretic mobility shift (EMSA) and chromatin immunoprecipitation (ChIP) assays revealed that the Sp-1 motif on FasL promoter was required for E2 enhanced FasL expression in TM4 cells. These data let us to recruit FasL among those genes whose expression is up-regulated by E2 through a direct interaction of ERalpha with Sp-1 protein. Finally, evidence that an aromatizable androgen is able to increase FasL expression suggests that E2 production by aromatase activity may contribute to maintain the immunoprivilege status of Sertoli cells.


Subject(s)
Aromatase/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Fas Ligand Protein/biosynthesis , Sertoli Cells/metabolism , Sp1 Transcription Factor/metabolism , Androstenedione/metabolism , Animals , Aromatase Inhibitors/pharmacology , Bucladesine/pharmacology , Chromatin Immunoprecipitation , DNA/metabolism , Electrophoretic Mobility Shift Assay , Estradiol/pharmacology , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Follicle Stimulating Hormone/metabolism , Genes, Reporter , HeLa Cells , Humans , Immune Tolerance , Letrozole , Luciferases , Male , Mice , Mice, Inbred BALB C , Mutation , Nitriles/pharmacology , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , Sertoli Cells/drug effects , Sertoli Cells/immunology , Transcription, Genetic , Transfection , Triazoles/pharmacology , Up-Regulation
6.
J Clin Pathol ; 60(6): 633-41, 2007 Jun.
Article in English | MEDLINE | ID: mdl-16882697

ABSTRACT

BACKGROUND: Insulin receptor substrate 1 (IRS-1), a cytoplasmic protein transmitting signals from the insulin and insulin-like growth factor 1 receptors, has been implicated in breast cancer. Previously, it was reported that IRS-1 can be translocated to the nucleus and modulate oestrogen receptor alpha (ERalpha) activity in vitro. However, the expression of nuclear IRS-1 in breast cancer biopsy specimens has never been examined. AIMS: To assess whether nuclear IRS-1 is present in breast cancer and non-cancer mammary epithelium, and whether it correlates with other markers, especially ERalpha. Parallel studies were carried out for the expression of cytoplasmatic IRS-1. METHODS: IRS-1 and ERalpha expression was assessed by immunohistochemical analysis. Data were evaluated using Pearson's correlation, linear regression and receiver operating characteristic analysis. RESULTS: Median nuclear IRS-1 expression was found to be low in normal mammary epithelial cells (1.6%) and high in benign tumours (20.5%), ductal grade 2 carcinoma (11.0%) and lobular carcinoma (approximately 30%). Median ERalpha expression in normal epithelium, benign tumours, ductal cancer grade 2 and 3, and lobular cancer grade 2 and 3 were 10.5, 20.5, 65.0, 0.0, 80 and 15%, respectively. Nuclear IRS-1 and ERalpha positively correlated in ductal cancer (p<0.001) and benign tumours (p<0.01), but were not associated in lobular cancer and normal mammary epithelium. In ductal carcinoma, both nuclear IRS-1 and ERalpha negatively correlated with tumour grade, size, mitotic index and lymph node involvement. Cytoplasmic IRS-1 was expressed in all specimens and positively correlated with ERalpha in ductal cancer. CONCLUSIONS: A positive association between nuclear IRS-1 and ERalpha is a characteristic for ductal breast cancer and marks a more differentiated, non-metastatic phenotype.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cell Nucleus/metabolism , Phosphoproteins/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Cytoplasm/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Immunoenzyme Techniques , Insulin Receptor Substrate Proteins , Mammary Glands, Human/metabolism , Microscopy, Confocal , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/metabolism
7.
J Cell Physiol ; 209(3): 977-86, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16972250

ABSTRACT

The peroxisome proliferation-activated receptor gamma (PPARgamma) is mainly expressed in the adipose tissue and integrates the control of energy, lipid, and glucose homeostasis. The present study, by means of RT-PCR, Western blot, and immunofluorescence techniques, demonstrates that human sperm express the PPARgamma. The functionality of the receptor was evidenced by 15-deoxy-12,14-prostaglandin J(2) (PGJ2) and rosiglitazone (BRL) PPARgamma-agonists that were tested on capacitation, acrosome reaction, and motility. Both treatments also increase AKT phosphorylations and influence glucose and lipid metabolism in sperm. The specificity of PGJ2 and BRL effects through PPARgamma on human sperm was confirmed by an irreversible PPARgamma antagonist, GW9662. Our findings provide evidence that human sperm express a functional PPARgamma whose activation influences sperm physiology. In conclusion, the presence of PPARgamma in male gamete broadens the field of action of this nuclear receptor, bringing us to look towards sperm as an endocrine mobile unit independent of the systemic regulation.


Subject(s)
PPAR gamma/metabolism , Spermatozoa/metabolism , Spermatozoa/physiology , Acrosome Reaction/physiology , Cell Movement/physiology , Glucose/metabolism , Homeostasis , Humans , Lipid Metabolism , Male , PPAR gamma/agonists , PPAR gamma/genetics , Pentose Phosphate Pathway/physiology , Phosphatidylinositol 3-Kinases/metabolism , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Sperm Capacitation/physiology , Spermatozoa/cytology
8.
Mol Endocrinol ; 20(12): 3083-92, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16887883

ABSTRACT

The aim of the present study was to provide new mechanistic insight into the growth arrest and apoptosis elicited by peroxisome proliferator-activated receptor (PPAR)gamma in breast cancer cells. We ascertained that PPARgamma mediates the inhibition of cycle progression in MCF7 cells exerted by the specific PPARgamma agonist rosiglitazone [BRL4653 (BRL)], because this response was no longer notable in the presence of the receptor antagonist GW9662. We also provided evidence that BRL is able to up-regulate mRNA and protein levels of the tumor suppressor gene p53 and its effector p21(WAF1/Cip1) in a time- and dose-dependent manner. Moreover, in transfection experiments with deletion mutants of the p53 gene promoter, we documented that the nuclear factor-kappaB sequence is required for the transcriptional response to BRL. Interestingly, EMSA showed that PPARgamma binds directly to the nuclear factor-kappaB site located in the promoter region of p53, and chromatin immunoprecipitation experiments demonstrated that BRL increases the recruitment of PPARgamma on the p53 promoter sequence. Next, both PPARgamma and p53 were involved in the cleavage of caspases-9 and DNA fragmentation induced by BRL, given that GW9662 and an expression vector for p53 antisense blunted these effects. Our findings provide evidence that the PPARgamma agonist BRL promotes the growth arrest and apoptosis in MCF7 cells, at least in part, through a cross talk between p53 and PPARgamma, which may be considered an additional target for novel therapeutic interventions in breast cancer patients.


Subject(s)
Breast Neoplasms/metabolism , Genes, p53 , NF-kappa B/metabolism , PPAR gamma/metabolism , Tumor Suppressor Protein p53/metabolism , Base Pairing , Breast Neoplasms/genetics , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Fragmentation , G1 Phase/drug effects , Humans , PPAR gamma/agonists , Promoter Regions, Genetic , Resting Phase, Cell Cycle/drug effects , Rosiglitazone , Thiazolidinediones/pharmacology , Tumor Suppressor Protein p53/genetics
9.
Clin Cancer Res ; 11(17): 6139-47, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16144913

ABSTRACT

PURPOSE: The molecular mechanisms involved in the repressive effects exerted by estrogen receptors (ER) on peroxisome proliferator-activated receptor (PPAR) gamma-mediated transcriptional activity remain to be elucidated. The aim of the present study was to provide new insight into the crosstalk between ERalpha and PPARgamma pathways in breast cancer cells. EXPERIMENTAL DESIGN: Using MCF7 and HeLa cells as model systems, we did transient transfections and electrophoretic mobility shift assay and chromatin immunoprecipitation studies to evaluate the ability of ERalpha to influence PPAR response element-mediated transcription. A possible direct interaction between ERalpha and PPARgamma was ascertained by co-immunoprecipitation assay, whereas their modulatory role in the phosphatidylinositol 3-kinase (PI3K)/AKT pathway was evaluated by determining PI3K activity and AKT phosphorylation. As a biological counterpart, we investigated the growth response to the cognate ligands of both receptors in hormone-dependent MCF7 breast cancer cells. RESULTS: Our data show for the first time that ERalpha binds to PPAR response element and represses its transactivation. Moreover, we have documented the physical and functional interactions of ERalpha and PPARgamma, which also involve the p85 regulatory subunit of PI3K. Interestingly, ERalpha and PPARgamma pathways have an opposite effect on the regulation of the PI3K/AKT transduction cascade, explaining, at least in part, the divergent response exerted by the cognate ligands 17beta-estradiol and BRL49653 on MCF7 cell proliferation. CONCLUSION: ERalpha physically associates with PPARgamma and functionally interferes with PPARgamma signaling. This crosstalk could be taken into account in setting new pharmacologic strategies for breast cancer disease.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , PPAR gamma/metabolism , Response Elements/genetics , Signal Transduction , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Estradiol/pharmacology , HeLa Cells , Humans , Ligands , PPAR gamma/genetics , PTEN Phosphohydrolase , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/drug effects , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Thiazolidinediones/pharmacology , Thymidine/metabolism , Transcription, Genetic , Transcriptional Activation , Tumor Suppressor Proteins/drug effects , Tumor Suppressor Proteins/metabolism , Vasodilator Agents/pharmacology
10.
J Clin Endocrinol Metab ; 90(8): 4753-61, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15944217

ABSTRACT

INTRODUCTION: Leptin action is a dynamic area of investigation that continues to broaden beyond the basic lipostatic model originally envisaged. Here, we show that leptin is expressed in and secreted from human ejaculated spermatozoa. METHODS: By RT-PCR, Western blot, and immunofluorescence techniques, we have demonstrated that human sperm express leptin. RIA method evidenced leptin secretion. Phosphatidyl-inositol-kinase-3 (PI3K)/Akt pathway was examined by PI3K activity assay and Western blot. Leptin and insulin regulation of glycogen synthesis was evaluated by glycogen synthase activity (GSA). RESULTS: The large differences of leptin secretion between uncapacitated and capacitated sperm suggest a functional role for leptin in capacitation. Indeed, in uncapacitated sperm, leptin enhances both cholesterol efflux and protein tyrosine phosphorylation. In uncapacitated sperm, both insulin and leptin increased PI3K activity, Akt S473, and glycogen synthase kinase-3 S9 phosphorylation. Interestingly, during capacitation, concomitantly to the massive release of both hormones, we observed a strong reduction in the phosphorylation of glycogen synthase kinase-3 S9, kinase downstream of Akt that regulates the glycogen synthase. Our results from GSA showed that the enzymatic activity was significantly higher in uncapacitated than in capacitated sperm. Particularly, in uncapacitated sperm, GSA appeared to depend on the hormones concentration, because the enzymatic activity was stimulated at low doses, whereas it was inhibited at high doses. Moreover, both leptin and insulin regulate in autocrine fashion sperm glycogen synthesis. CONCLUSION: Leptin secretion by sperm suggests that the male gamete may be able to modulate its metabolism independently by systemic leptin. These data open new considerations about leptin significance in male fertility.


Subject(s)
Leptin/genetics , Leptin/metabolism , Spermatozoa/metabolism , Spermatozoa/physiology , Blotting, Southern , Blotting, Western , Cholesterol/metabolism , Ejaculation , Humans , Insulin/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Tyrosine/metabolism
11.
Endocrinology ; 146(2): 552-7, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15550513

ABSTRACT

A striking feature of insulin expression is its almost complete restriction to beta-cells of the pancreatic islet in normal mammals. Here we show that insulin is expressed in and secreted from human ejaculated spermatozoa. Both insulin transcript and protein were detected. In addition, the large differences in insulin secretion, assessed by RIA, between noncapacitated and capacitated sperm suggest a role for insulin in capacitation. Insulin had an oscillatory secretory pattern involving glucose dose-dependent increases and significant decreases during the blockage of an insulin autocrine effect. It appears that the effect of glucose on the fertilizing ability of sperm is mediated by glucose metabolism through the pentose phosphate pathway. Then we evaluated the autocrine effect of sperm insulin on glucose metabolism by studying the activity of glucose-6-phosphate dehydrogenase, the key rate-limiting enzyme in the pentose phosphate pathway. The simultaneous decrease in both insulin release and glucose-6-phosphate dehydrogenase activity induced by blocking the autocrine insulin effect with three different procedures (blockage of insulin release by nifedipine, immune neutralization of the released insulin by antiinsulin serum, and blockage of an insulin intracellular effector such as phosphotidylinositol 3-kinase by wortmannin) strongly suggests a physiological role of sperm insulin on these two events. Insulin secretion by spermatozoa may provide an autocrine regulation of glucose metabolism based on their energetic needs independent of systemic insulin. In conclusion, these data open a new area of study in male reproduction.


Subject(s)
Insulin/genetics , Insulin/metabolism , Spermatozoa/metabolism , Spermatozoa/physiology , Autocrine Communication , Blotting, Western , Ejaculation , Gene Expression/physiology , Glucosephosphate Dehydrogenase/metabolism , Humans , Insulin Secretion , Male , Reverse Transcriptase Polymerase Chain Reaction
12.
Oncogene ; 23(55): 8920-30, 2004 Nov 25.
Article in English | MEDLINE | ID: mdl-15467744

ABSTRACT

The expression of estrogen receptor alpha (ERalpha) is generally associated with a less invasive and aggressive phenotype in breast carcinoma. In an attempt to understand the role of ERalpha in regulating breast cancer cells invasiveness, we have demonstrated that cell adhesion on fibronectin (Fn) and type IV Collagen (Col) induces ERalpha-mediated transcription and reduces cell migration in MCF-7 and in MDA-MB-231 cell lines expressing ERalpha. Analysis of deleted mutants of ERalpha indicates that the transcriptional activation function (AF)-1 is required for ERalpha-mediated transcription as well as for the inhibition of cell migration induced by cell adhesion on extracellular matrix (ECM) proteins. In addition, the nuclear localization signal region and some serine residues in the AF-1 of the ERalpha are both required for the regulation of cell invasiveness as we have observed in HeLa cells. It is worth noting that c-Src activation is coincident with adhesion of cells to ECM proteins and that the inhibition of c-Src activity by PP2 or the expression of a dominant-negative c-Src abolishes ERalpha-mediated transcription and partially reverts the inhibition of cell invasiveness in ERalpha-positive cancer cells. These findings address the integrated role of ECM proteins and ERalpha in influencing breast cancer cell motility through a mechanism that involves c-Src and seems not to be related to a specific cell type.


Subject(s)
Breast Neoplasms/metabolism , Collagen Type IV/chemistry , Collagen Type IV/physiology , Estrogen Receptor alpha/chemistry , Fibronectins/chemistry , Fibronectins/physiology , Phosphotransferases/metabolism , Proto-Oncogene Proteins/metabolism , Active Transport, Cell Nucleus , Blotting, Western , CSK Tyrosine-Protein Kinase , Cathepsin D/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , Cytosol/metabolism , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , HeLa Cells , Humans , Immunohistochemistry , Immunoprecipitation , Ligands , Luciferases/metabolism , Plasmids/metabolism , Protein Structure, Tertiary , Protein-Tyrosine Kinases , RNA, Messenger/metabolism , Response Elements , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection , Up-Regulation , src-Family Kinases
13.
J Clin Endocrinol Metab ; 89(3): 1443-51, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15001646

ABSTRACT

Human and animal models have evidenced how estrogen insufficiency is associated with abnormal spermatogenesis and male infertility. We previously demonstrated that estradiol is able to influence both capacitation and acrosome reaction in human ejaculated spermatozoa. It remains to be elucidated whether the biochemical changes induced by estradiol, in a rapid nongenomic way, are mediated by a single estrogen receptor (ER) or by the two ER subtypes, ER alpha and ER beta. In the present study, we have first demonstrated the concomitant expression of ER beta and ER alpha in human ejaculated spermatozoa. By RT-PCR and Southern blot, transcripts of both ERs were detected. Western blot analysis showed ER alpha and ER beta proteins at the same size as the "classical" ERs. The localization of ER alpha and ER beta with the immunocytochemistry shows a differential distribution of the two ER subtypes, the former being prevalently located in the midpiece, but the latter being in the tail. Estradiol has been associated with sperm longevity; however, the mechanism through which estradiol acts in sperm survival was never investigated. Upon estradiol exposure, we observed an enhanced phosphorylation of the proteins involved in the phosphatidylinositol-3-OH kinase (PI3K)/Akt pathway like PDK1, Akt, GSK-3, Bcl-2, together with ERK1/2, which was also involved in cell survival signals. Moreover, such phosphorylations were reduced in the presence of ICI 182, 780, addressing the role of estradiol and ERs in sperm survival. For instance we have provided, for the first time, a different interaction of the two ERs with the PI3K/Akt pathway, because ER alpha interacts with the p55 regulatory subunit of PI3K, whereas ER beta interacts with Akt1. However, it still remains to be elucidated whether the functional role of each of the ER subtypes in sperm survival signaling is redundant or distinct.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Spermatozoa/metabolism , Blotting, Southern , Blotting, Western , Cell Survival/physiology , Ejaculation , Estradiol/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Gene Expression/physiology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spermatozoa/cytology
14.
Hum Reprod ; 18(8): 1650-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12871877

ABSTRACT

BACKGROUND: Advances in the definition of the function and the mechanism of estrogen action in different tissues have come from human and animal models of estrogen insufficiency. Recently we have demonstrated that aromatase is present and biologically active in human ejaculated sperm, suggesting that autonomous estradiol sperm production may influence sperm functions. In the present study we investigate a possible physiological role for enzymatically active P450 aromatase in human ejaculated sperm. METHODS AND RESULTS: To confirm the presence of mRNA coding for P450 aromatase, total RNA isolated from human sperm underwent RT-PCR and then Southern blot analysis. In non-capacitating medium, we observed that only estradiol and aromatizable steroids were able to increase sperm motility/migration; concomitantly they enhanced protein tyrosine phosphorylation and increased p-44/42 extracellular signal-regulated kinase activity. When we tested acrosin activity, it emerged that estradiol and aromatizable androgens were also able to induce the acrosome reaction evaluated by two different cytological staining techniques (triple-stain and fluorescein isothiocyanate-Pisum sativum agglutinin). All these events were enhanced by the 2'-O-dibutyryladenosine-3',5'-cyclic monophosphate and inhibited in the presence of the specific aromatase inhibitor, letrozole. CONCLUSIONS: From this study, it appears that a link exists between the locally produced estradiol (from ejaculated sperm), sperm capacitation and the acrosome reaction. The induction of both events by aromatizable androgens in the absence of exogenous mediators suggests that estrogen biosynthesis in ejaculated sperm is a process that may influence the intrinsic sperm fertilizing capability.


Subject(s)
Aromatase/physiology , Spermatozoa/enzymology , Acrosin/metabolism , Acrosome Reaction/physiology , Aromatase/genetics , Base Sequence , DNA, Complementary/genetics , Ejaculation , Estradiol/pharmacology , Estradiol/physiology , Humans , In Vitro Techniques , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sperm Capacitation/physiology , Sperm Motility/drug effects , Sperm Motility/physiology , Staining and Labeling , Substrate Specificity
15.
J Clin Endocrinol Metab ; 87(7): 3385-90, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12107254

ABSTRACT

The generation of cytochrome P450 aromatase (P450arom) and estrogen receptor (ER) knockout mice has raised new interest in the physiological role of estrogens in male reproduction. Testicular expression of P450arom, the enzyme that converts androgens into estrogens, has been shown in both somatic and germ cell types in several species, whereas in humans, testicular expression is confined to the somatic cells. The aim of this study was to determine whether P450arom is present in human ejaculated spermatozoa. Using RT-PCR and specific primers, we amplified the highly conserved helical, aromatic, and heme-binding sequences of the conventional human P450arom from RNA isolated from human spermatozoa. Employing a rabbit polyclonal antiserum directed against human placental P450arom, immunoblotting analysis demonstrated aromatase protein expression, which was localized primarily to the tail and midpiece of spermatozoa. Measurement of enzymatic activity using a sensitive (3)H(2)O aromatase assay revealed that activity was enhanced by the 2'-O-dibutyryl cAMP and completely inhibited in the presence of the specific aromatase inhibitor, letrozole. These results represent the first demonstration that human spermatozoa are a potential site of estrogen biosynthesis. The physiological relevance of estrogen synthesis in spermatozoa remains to be elucidated and opens a new area of investigation in male fertility.


Subject(s)
Aromatase/metabolism , Ejaculation , Spermatozoa/enzymology , Aromatase/genetics , Enzyme Activation , Humans , Male , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL