Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biology (Basel) ; 11(5)2022 Apr 26.
Article in English | MEDLINE | ID: mdl-35625396

ABSTRACT

The blood-testis barrier (BTB) is formed from tight junctions (TJs) between Sertoli cells. This dynamic structure, which establishes an immune-privileged environment protecting haploid germ cells formed in puberty from cells of the innate immune system, protects male fertility. Testosterone produced in Leydig cells is one of the main regulators of TJ protein expression and BTB dynamics. Nevertheless, although it has been assumed that testosterone effects on TJs and BTB are mediated through the classical androgen receptor (AR), newer results call the importance of this receptor into question. ZIP9, a recently identified androgen receptor of plasma membranes, mediates testosterone effects that promote the expression of TJ proteins and TJ formation in a rat Sertoli cell line that lacks the classical AR. Although these findings suggest that ZIP9 mediates these testosterone effects, participation of the classical AR in these events cannot be excluded. Here we used immortalized adult rat Sertoli cells that express both ZIP9 and AR and addressed the involvement of these receptors in the stimulation of TJ protein expression and TJ formation in response to testosterone and to the androgenic peptide IAPG that acts via ZIP9. We find that both testosterone and IAPG trigger the so-called non-classical signaling pathway of testosterone and stimulate the expression of TJ-associated proteins and TJ formation. Silencing classical AR expression had no effect on the responses, whereas silencing of ZIP9 expression completely blocked them. Our results demonstrate that ZIP9 is the sole androgen receptor involved in the regulation of TJ protein expression and TJ formation at the BTB.

2.
Biology (Basel) ; 11(1)2021 Dec 23.
Article in English | MEDLINE | ID: mdl-35053017

ABSTRACT

ZIP9 is a recently identified membrane-bound androgen receptor of physiological significance that may mediate certain physiological responses to androgens. Using in silico methods, six tetrapeptides with the best docking properties at the testosterone binding site of ZIP9 were synthesized and further investigated. All tetrapeptides displaced T-BSA-FITC, a membrane-impermeable testosterone analog, from the surface of mouse myogenic L6 cells that express ZIP9 but not the classical androgen receptor (AR). Silencing the expression of ZIP9 with siRNA prevented this labeling. All tetrapeptides were found to be pro-androgenic; in L6 cells they stimulated the expression of myogenin, triggered activation of focal adhesion kinase, and prompted the fusion of L6 myocytes to syncytial myotubes. In human osteoblastic SAOS-2 cells that express AR and ZIP9, they reduced the expression of alkaline phosphatase and stimulated mineralization. These latter effects were prevented by silencing ZIP9 expression, indicating that the osteoblast/osteocyte conversion is exclusively mediated through ZIP9. Our results demonstrate that the synthetic tetrapeptides, by acting as ZIP9-specific androgens, have the potential to replace testosterone or testosterone analogs in the treatment of bone- or muscle-related disorders by circumventing the undesirable effects mediated through the classical AR.

3.
Biology (Basel) ; 11(1)2021 Dec 31.
Article in English | MEDLINE | ID: mdl-35053053

ABSTRACT

Androgens stimulate the expression of tight junction (TJ) proteins and the formation of the blood-testis barrier (BTB). Interactions of testosterone with the zinc transporter ZIP9 stimulate the expression of TJ-forming proteins and promote TJ formation in Sertoli cells. In order to investigate androgenic effects mediated by ZIP9 but not by the nuclear androgen receptor (AR), the effects of three tetrapeptides fitting the androgen binding site of ZIP9 were compared with those induced by testosterone in a Sertoli cell line expressing ZIP9 but not the AR. Three tetrapeptides and testosterone displaced testosterone-BSA-FITC from the surface of 93RS2 cells and stimulated the non-classical testosterone signaling pathway that includes the activation of Erk1/2 kinases and transcription factors CREB and ATF-1. The expression of the TJ-associated proteins ZO-1 and claudin-5 was triggered as was the re-distribution of claudin-1 from the cytosol to the membrane and nucleus. Furthermore, TJ formation was stimulated, indicated by increased transepithelial electrical resistance. Silencing ZIP9 expression by siRNA prevented all of these responses. These results are consistent with an alternative pathway for testosterone action at the BTB that does not involve the nuclear AR and highlight the significant role of ZIP9 as a cell-surface androgen receptor that stimulates TJ formation.

4.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118600, 2020 02.
Article in English | MEDLINE | ID: mdl-31760088

ABSTRACT

It is generally assumed that circulating dehydroepiandrosterone sulfate (DHEAS) can be desulfated and further metabolized to estrogen, which is of concern for all patients with estrogen-responsive breast cancer. We addressed this issue by comparing the effects of DHEAS, its desulfated form DHEA, and 17ß-estradiol on human metastatic, estrogen-responsive MCF-7 breast cancer cells. Physiological concentrations of DHEAS promoted phosphorylation of Erk1/2, whereas DHEA and 17ß-estradiol failed to stimulate Erk1/2 phosphorylation, indicating that the sulfated steroid acts as an autonomous hormone. Exposure of MCF-7 cells to 17ß-estradiol stimulated cell proliferation and the expression of pro-metastatic and pro-invasive elements such as claudin-1, matrix metalloproteinase 9 (MMP9), and the CC chemokine ligand 2 (CCL2). In contrast, treatment with DHEAS did not stimulate these responses but prevented all of the actions of 17ß-estradiol, and as a consequence cell migration and invasion were completely inhibited. The results of this study not only challenge the assumption that DHEAS poses a danger as an endogenous source of estrogen, they rather favor the idea that keeping DHEAS levels within a physiological range might be supportive in treating estrogen-responsive breast cancer.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Estradiol/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemokine CCL2/metabolism , Claudin-1/metabolism , Female , Humans , MCF-7 Cells , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects
5.
J Steroid Biochem Mol Biol ; 191: 105372, 2019 07.
Article in English | MEDLINE | ID: mdl-31042565

ABSTRACT

Cardiotonic steroids such as ouabain are potent inhibitors of the sodium pump and have been widely used for centuries in the treatment of congestive heart failure. In recent decades, however, they have also been identified as hormone-like molecules that trigger signaling cascades of physiological relevance by using the various sodium pump α subunit isoforms as receptors. The murine Leydig cell line MLTC-1 expresses both the ubiquitous, relatively ouabain-insensitive α1 isoform of the sodium pump and the ouabain-sensitive α3 isoform that is normally found in neuronal cells. The physiological relevance of the simultaneous presence of the two isoforms in Leydig cells has not been previously addressed. MLTC-1 Leydig cells contain lipid droplets (LDs) and are capable of progesterone biosynthesis when stimulated by luteinizing hormone (LH). When exposed to low nanomolar concentrations of ouabain, they respond with stimulation of Erk1/2, CREB, and ATF-1 phosphorylation, LD enlargement, and perilipin2 mobilization to the LDs. As a result, progesterone biosynthesis is augmented. Abrogation of α3 isoform expression by siRNA prevents all of the above responses, indicating that it is the hormone/receptor-like interaction of ouabain exclusively with this isoform that triggers the signaling events that normally occur when LH binds to its receptor. Considering that ouabain is produced endogenously and is found in seminal fluid, one can speculate that effects of this substance on germ and somatic cells of the testis might play a role in male reproductive physiology.


Subject(s)
Cardiotonic Agents/pharmacology , Leydig Cells/drug effects , Ouabain/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Steroids/metabolism , Animals , Biosynthetic Pathways/drug effects , Cell Line , Leydig Cells/metabolism , Male , Mice , Progesterone/metabolism , Protein Isoforms/metabolism
6.
Biochim Biophys Acta Mol Cell Res ; 1865(12): 1857-1868, 2018 12.
Article in English | MEDLINE | ID: mdl-30262433

ABSTRACT

LNCaP cells are derived from a metastatic lesion of human prostate adenocarcinoma. They express the classical androgen receptor (AR) and ZIP9, a Zn2+ transporter that also binds testosterone and mediates signaling by interacting with G-proteins. Our results show that LNCaP cells respond to testosterone by mobilizing their migratory machinery. Their exposure to testosterone triggers the formation of lamellipodia, reorganization of the actin cytoskeleton, phosphorylation of focal adhesion kinase (FAK) at Tyr925 and of paxillin at Tyr118, expression of matrix metalloproteinase 2 (MMP-2), and cell migration. Silencing ZIP9 expression by means of siRNA does not affect the responsiveness of the classical AR to testosterone; however, it prevents all of the testosterone effects described above: formation of lamellipodia cannot be induced, stimulation of FAK or paxillin phosphorylation or MMP-2 expression is prevented, and cell migration does not take place in the absence of ZIP9. The data presented show that testosterone/ZIP9 interactions might have not only physiological but also pathophysiological relevance. The fact that the migratory machinery of a metastatic prostate cancer cell line is activated exclusively through testosterone/ZIP9 and not through testosterone/AR interactions suggests that targeting specific inhibition of testosterone/ZIP9-mediated events might help in developing new therapeutic strategies against androgen-induced progression of prostate cancer.


Subject(s)
Cation Transport Proteins/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Testosterone/pharmacology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Focal Adhesion Kinase 1/metabolism , Humans , Male , Matrix Metalloproteinase 2/metabolism , Paxillin/metabolism , Phosphorylation , Prostatic Neoplasms/genetics , Signal Transduction/drug effects
7.
J Steroid Biochem Mol Biol ; 182: 50-61, 2018 09.
Article in English | MEDLINE | ID: mdl-29684479

ABSTRACT

Although dehydroepiandrosterone sulfate (DHEAS) constitutes the most abundant steroid in humans, in-depth investigations of its effects are rather scarce. We address here DHEAS effects on the estrogen receptor-positive metastatic human breast cancer cell line MCF-7. We focus on DHEAS-mediated signaling that might influence expression of claudin-1 and matrix metalloproteinase-9 (MMP-9), both known to be critical factors for migration and invasiveness of various cancers, including breast cancer cells. Physiological concentrations of DHEAS trigger persistent phosphorylation of Erk1/2 in MCF-7 cells. Exposure of these cells for 24 h to 1 µM DHEAS also leads to a significant reduction of claudin-1 expression that cannot be prevented by high concentrations of the steroid sulfatase inhibitor STX64, indicating that desulfation and further conversion of DHEAS to some other steroid hormone is not required for this action. In addition, exposure of MCF-7 cells to the same concentration of DHEAS completely abolishes MMP-9 expression and considerably impairs cell migratory behavior. Abrogation of Gnα11 expression by siRNA prevents the stimulatory effect of DHEAS on Erk1/2 phosphorylation, consistent with a G-protein-coupled receptor being involved in the DHEAS-induced signaling. Nevertheless, Gnα11 also has direct effects that do not depend on DHEAS; thus, when Gnα11 expression is suppressed, expression of claudin-1 and MMP-9 as well as cell migration are significantly reduced. This is the first report demonstrating direct involvement of DHEAS and Gnα11 in the regulation of claudin-1 and MMP-9 expression and migration of MCF-7 cells.


Subject(s)
Breast Neoplasms/pathology , Cell Movement/drug effects , Claudin-1/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , GTP-Binding Protein alpha Subunits/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Matrix Metalloproteinase 9/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Claudin-1/genetics , Female , GTP-Binding Protein alpha Subunits/antagonists & inhibitors , GTP-Binding Protein alpha Subunits/genetics , Humans , MCF-7 Cells , Matrix Metalloproteinase 9/genetics , Phosphorylation , RNA, Small Interfering/genetics , Signal Transduction
8.
Biochim Biophys Acta Mol Cell Res ; 1864(12): 2402-2414, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28943399

ABSTRACT

ZIP9 is a Zn2+ transporter, testosterone receptor, and mediator of signaling events through G-proteins. Despite these pivotal properties, however, its physiological and pathophysiological significance has not yet been comprehensively addressed. Using a cell line that lacks the classical androgen receptor we show that ZIP9-mediated phosphorylation of Erk1/2, CREB, or ATF-1 and expression of claudin-5 and zonula occludens-1 by testosterone can be completely antagonized by bicalutamide (Casodex), an anti-androgen of significant clinical impact. Computational modeling and docking experiments with ZIP9 reveal typical characteristics of ZIP transporters and an extracellular binding site for testosterone capable of accommodating bicalutamide. The presence of this site is verified by our demonstration that the membrane-impermeable testosterone analogue T-BSA-FITC labels the membrane only when ZIP9 is expressed and that this labeling is completely prevented by bicalutamide. The study connects structural features of ZIP9 to its functions and indicates a possible relevance of ZIP9 as a pharmacological target.


Subject(s)
Androgens/chemistry , Apoptosis/drug effects , Cation Transport Proteins/chemistry , Receptors, Androgen/genetics , Androgens/genetics , Androgens/metabolism , Anilides/antagonists & inhibitors , Anilides/chemistry , Binding Sites/drug effects , Cation Transport Proteins/genetics , Humans , Male , Molecular Docking Simulation , Nitriles/antagonists & inhibitors , Nitriles/chemistry , Phosphorylation/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Receptors, Androgen/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship , Testosterone/antagonists & inhibitors , Testosterone/chemistry , Tosyl Compounds/antagonists & inhibitors , Tosyl Compounds/chemistry
9.
Mol Reprod Dev ; 83(12): 1092-1101, 2016 12.
Article in English | MEDLINE | ID: mdl-27764533

ABSTRACT

A gonadotropin-releasing hormone agonist (GnRH-A) implant induces hormonal castration in dogs that is associated with reduced prostate and testes size. We address the molecular events associated with hormonal castration by examining GnRH-A effects on expression and phosphorylation of a number of key signaling proteins. Male beagles were treated for 5 months with a GnRH-A implant, and then surgically castrated at 0, 3, 6, 12, and, 24 weeks after implant removal; untreated animals served as controls. GnRH-A treatment led to activation of c-Raf, Erk1/2, and, p53 in the testes. Phosphorylation of p53 occurred at Ser15, consistent with activation of the c-Raf-Erk1/2-p53 signaling cascade that triggers growth arrest or apoptosis. GnRH-A also suppressed the anti-apoptotic protein Bcl-xL; reduced phosphorylation of the transcription factors CREB and ATF1; and down-regulated expression of StAR and P450scc, proteins involved in steroidogenesis. Although androgen receptor expression was little affected by GnRH-A treatment, levels of ZIP9, a membrane-bound Zn2+ transporter that mediates non-classical signaling of testosterone, were abrogated. All of these effects were reversed within 24 weeks after implant removal. Thus, molecular signatures of implant-dependent hormonal castration include reversible cell cycle arrest and apoptosis, loss of steroidogenesis, and reduced transcriptional activity. Mol. Reprod. Dev. 83: 1092-1101, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Apoptosis , Gonadotropin-Releasing Hormone/agonists , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 3/metabolism , Orchiectomy , Tumor Suppressor Protein p53/metabolism , Animals , Dogs , Male , Sterilization, Reproductive
10.
Cell Signal ; 28(8): 1075-85, 2016 08.
Article in English | MEDLINE | ID: mdl-27164415

ABSTRACT

In the classical signaling pathway, testosterone regulates gene expression by activating the cytosolic/nuclear androgen receptor. In the non-classical pathway, testosterone activates cytosolic signaling cascades that are normally triggered by growth factors. The nature of the receptor involved in this signaling pathway is a source of controversy. In the Sertoli cell line 93RS2, which lacks the classical AR, we determined that testosterone stimulates the non-classical signaling pathway, characterized by the phosphorylation of Erk1/2 and transcription factors CREB and ATF-1. We also demonstrated that testosterone increases the expression of the tight junction (TJ) proteins claudin-1 and claudin-5. Both of these proteins are known to be essential constituents of TJs between Sertoli cells, and as a consequence of their increased expression transepithelial resistance across Sertoli cell monolayers is increased. ZIP9 is a Zn(2+)transporter that was recently shown to be a membrane-bound testosterone receptor. Silencing its expression in 93RS2 Sertoli cells by siRNA completely prevents Erk1/2, CREB, and ATF-1 phosphorylation as well the stimulation of claudin-1 and -5 expression and TJ formation between neighboring cells. The study presented here demonstrates for the first time that in Sertoli cells testosterone acts through the receptor ZIP9 to trigger the non-classical signaling cascade, resulting in increased claudin expression and TJ formation. Since TJ formation is a prerequisite for the maintenance of the blood-testis barrier, the testosterone/ZIP9 effects might be significant for male physiology. Further assessment of these interactions will help to supplement our knowledge concerning the mechanism by which testosterone plays a role in male fertility.


Subject(s)
Cation Transport Proteins/metabolism , Claudin-1/metabolism , Claudin-5/metabolism , Sertoli Cells/metabolism , Signal Transduction , Testosterone/metabolism , Tight Junctions/metabolism , Activating Transcription Factor 1/metabolism , Animals , Blotting, Western , Cyclic AMP Response Element-Binding Protein/metabolism , Electric Impedance , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorescent Antibody Technique , Gene Silencing/drug effects , Male , Phosphorylation/drug effects , RNA, Small Interfering/metabolism , Rats , Sertoli Cells/drug effects , Signal Transduction/drug effects , Testosterone/pharmacology , Tight Junctions/drug effects
11.
Biochim Biophys Acta ; 1843(6): 1172-81, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24631506

ABSTRACT

Testosterone is known to mediate its effects by two different mechanisms of action. In the so-called "classical" pathway testosterone binds to cytosolic androgen receptors (AR), which essentially function as ligand-activated transcription factors. Once activated, these receptors bind to DNA and activate the expression of target genes. In the "non-classical" pathway, the steroid hormone binds to receptors associated with the plasma membrane and induces signaling cascades mediated through activation of Erk1/2. The precise nature of the membrane-associated AR, however, remains controversial. Although some assume that the membrane and cytosolic AR are identical, others propose that the AR of the membrane is a G-protein-coupled receptor (GPCR). To evaluate these two possibilities we first searched for testosterone-induced signaling cascades in the spermatogenic cell line GC-2. Testosterone was found to cause phosphorylation (activation) of Erk1/2, CREB, and ATF-1, consistent with its non-classical mechanism of action. Silencing of AR expression by means of siRNA did not influence testosterone-induced activation of Erk1/2, CREB, or ATF-1, indicating that this pathway is not activated by the classical cytosolic/nuclear AR. In contrast, when the expression of the G-protein Gnα11 is suppressed, the activation of these signaling molecules is abolished, suggesting that these responses are elicited through a membrane-bound GPCR. The results presented here and the identification of the testosterone-specific GPCR in future investigations will help to reveal and characterize new testosterone-mediated mechanisms associated not only with fertility and reproduction but perhaps also with other physiological processes.


Subject(s)
Androgens/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptors, Androgen/metabolism , Receptors, G-Protein-Coupled/metabolism , Spermatocytes/metabolism , Testosterone/pharmacology , Activating Transcription Factor 1/genetics , Activating Transcription Factor 1/metabolism , Animals , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Fluorescent Antibody Technique , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Male , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Spermatocytes/cytology , Spermatocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...