Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Intervalo de año de publicación
1.
Andrology ; 4(1): 95-110, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26588606

RESUMEN

Testicular germ cell tumors (TGCTs) are the most common type of cancer in young men and their incidence has been steadily increasing for the past decades. TGCTs and their precursor carcinoma in situ (CIS) are thought to arise from the deficient differentiation of gonocytes, precursors of spermatogonial stem cells. However, the mechanisms relating failed gonocyte differentiation to CIS formation remain unknown. The goal of this study was to uncover genes regulated during gonocyte development that would show abnormal patterns of expression in testicular tumors, as prospective links between failed gonocyte development and TGCT. To identify common gene and protein signatures between gonocytes and seminomas, we first performed gene expression analyses of transitional rat gonocytes, spermatogonia, human normal testicular, and TGCT specimens. Gene expression arrays, pathway analysis, and quantitative real-time PCR analysis identified cell adhesion molecules as a functional gene category including genes downregulated during gonocyte differentiation and highly expressed in seminomas. In particular, the mRNA and protein expressions of claudins 6 and 7 were found to decrease during gonocyte transition to spermatogonia, and to be abnormally elevated in seminomas. The dynamic changes in these genes suggest that they may play important physiological roles during gonocyte development. Moreover, our findings support the idea that TGCTs arise from a disruption of gonocyte differentiation, and position claudins as interesting genes to further study in relation to testicular cancer.


Asunto(s)
Diferenciación Celular/fisiología , Claudinas/biosíntesis , Neoplasias de Células Germinales y Embrionarias/metabolismo , Seminoma/metabolismo , Espermatogonias/citología , Células Madre/citología , Neoplasias Testiculares/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Perfilación de la Expresión Génica , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
2.
Curr Mol Med ; 12(4): 467-75, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22348614

RESUMEN

Translocator protein (TSPO) is a high affinity 18 kDa drug- and cholesterol-binding protein strongly expressed in steroidogenic tissues where it mediates cholesterol transport into mitochondria and steroid formation. Testosterone formation by Leydig cells in the testis is critical for the regulation of spermatogenesis and male fertility. Male germ cell development comprises two main phases, the pre-spermatogenesis phase occurring from fetal life to infancy and leading to spermatogonial stem cell (SSC) formation, and spermatogenesis, which consists of repetitive cycles of germ cell mitosis, meiosis and differentiation, starting with SSC differentiation and ending with spermiogenesis and spermatozoa formation. Little is known about the molecular mechanisms controlling the progression from one germ cell phenotype to the next. Here, we report that testicular germ cells express TSPO from neonatal to adult phases, although at lower levels than Leydig cells. TSPO mRNA and protein were found at specific steps of germ cell development. In fetal and neonatal gonocytes, the precursors of SSCs, TSPO appears to be mainly nuclear. In the prepubertal testis, TSPO is present in pachytene spermatocytes and dividing spermatogonia. In adult testes, it is found in a stage-dependent manner in pachytene spermatocyte and round spermatid nuclei, and in mitotic spermatogonia. In search of TSPO function, the TSPO drug ligand PK 11195 was added to isolated gonocytes with or without the proliferative factors PDGF and 17ß-estradiol, and was found to have no effect on gonocyte proliferation. However, TSPO strong expression in dividing spermatogonia suggests that it might play a role in spermatogonial mitosis. Taken together, these results suggest that TSPO plays a role in specific phases of germ cell development.


Asunto(s)
Proteínas Portadoras/metabolismo , Receptores de GABA-A/metabolismo , Espermatocitos/metabolismo , Espermatogonias/metabolismo , Testículo/citología , Animales , Proteínas Portadoras/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Isoquinolinas/farmacología , Células Intersticiales del Testículo/metabolismo , Masculino , Naftoles , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/genética , Testículo/embriología , Testículo/crecimiento & desarrollo , Transcripción Genética , Triazinas
3.
Endocr Res ; 28(4): 395-401, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12530641

RESUMEN

Acute stimulation of cholesterol transport into mitochondria involves the cAMP-dependent protein kinase (PKA), peripheral-type benzodiazepine receptor (PBR), and the steroidogenesis acute regulatory (StAR) proteins. We investigated the respective role of these proteins in hormone-induced steroidogenesis. Oligonucleotides antisense, but not sense, to PBR and StAR reduced their respective levels in steroidogenic cells and inhibited hormone-stimulated steroid formation in MA-10 mouse Leydig tumor cells. In search of the proteins regulating PBR we identified a protein, PAP7, which interacts with PBR and the PKA regulatory subunit RIalpha, is present in adrenal and gonadal cells and is found in mitochondria. Overexpression of the full length PAP7 increased the hormone-induced steroid production. However, inhibition of PAP7 expression reduced the gonadotropin-induced steroid formation. In search of a PBR functional antagonist that would facilitate the studies on the biological function of PBR, we screened a phage display library. A 7-mer competitive PBR peptide antagonist was identified, which when transduced into Leydig cells inhibited the benzodiazepine and hormone-stimulated steroid production suggesting that the endogenous PBR agonist/receptor interaction is critical for the hormone-dependent steroidogenesis. These data indicate that hormone-induced cholesterol transport and the subsequent steroid formation is a dynamic multistep process involving protein-protein interactions.


Asunto(s)
Colesterol/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfoproteínas/metabolismo , Receptores de GABA-A/metabolismo , Esteroides/biosíntesis , Animales , Transporte Biológico/fisiología , Ratones , Células Tumorales Cultivadas
4.
Anticancer Res ; 20(5A): 2835-47, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11062691

RESUMEN

The peripheral-type benzodiazepine receptor (PBR) expression and localization correlate with human breast cancer cell proliferation and aggressive phenotype expression. The standardized extract of Ginkgo biloba leaves (EGb 761) and isolated ginkgolide B (GKB) were shown to decrease PBR mRNA expression in adrenal cells. We examined the effect of EGb 761 and GKB on PBR expression and cell proliferation in human breast cancer cells. EGb 761 and GKB decreased in a time- and dose-dependent manner PBR expression and cell proliferation in the highly aggressive, rich in PBR, human breast cancer cell line MDA-231 whereas they did not affect the proliferation of the non-aggressive human breast cancer cell line MCF-7, which contains very low PBR levels. This effect was reversible and not due to the antioxidant properties of the compounds tested. Using a human cDNA expression array we determined that EGb 761 treatment altered, in addition to PBR, the expression of 36 gene products involved in various pathways regulating cell proliferation. These in vitro data were further validated in an in vivo model where EGb 761 and GKB significantly inhibited the nuclear PBR expression and growth of MDA-231 cell xenografts in nude mice. Taken together, these data suggest that the manipulation of PBR expression could be used to control tumor growth and that EGb 761 and GKB, under the conditions used, exert cytostatic properties.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Antioxidantes/farmacología , Diterpenos , Flavonoides/farmacología , Lactonas/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Receptores de GABA-A/biosíntesis , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Antioxidantes/uso terapéutico , División Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Flavonoides/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Ginkgo biloba , Ginkgólidos , Humanos , Lactonas/uso terapéutico , Ligandos , Ratones , Ratones Desnudos , Fitoterapia , Extractos Vegetales , Plantas Medicinales , ARN Mensajero , Receptores de GABA-A/genética , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas
5.
Endocrinology ; 141(9): 3137-48, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10965884

RESUMEN

The peroxisome proliferator perfluordecanoic acid (PFDA) has been shown to exert an antiandrogenic effect in vivo by acting directly on the interstitial Leydig cells of the testis. The objective of this study was to examine the in vitro effects of PFDA and identify its site of action in steroidogenesis using as model systems the mouse tumor MA-10 and isolated rat Leydig cells. PFDA inhibited in a time- and dose-dependent manner the hCG-stimulated Leydig cell steroidogenesis. This effect was localized at the level of cholesterol transport into the mitochondria. PFDA did not affect either the total cell protein synthesis or the mitochondrial integrity. Moreover, it did not induce any DNA damage. Morphological studies indicated that PFDA induced lipid accumulation in the cells, probably due to the fact that cholesterol mobilized by hCG did not enter the mitochondria to be used for steroidogenesis. In search of the target of PFDA, we examined its effect on key regulatory mechanisms of steroidogenesis. PFDA did not affect the hCG-induced steroidogenic acute regulatory protein (StAR) levels. However, it was found to inhibit the mitochondrial peripheral-type benzodiazepine receptor (PBR) ligand binding capacity, 18-kDa protein, and messenger RNA (mRNA) levels. Further studies indicated that PFDA did not affect PBR transcription, but it rather accelerated PBR mRNA decay. Taken together, these data suggest that PFDA inhibits the Leydig cell steroidogenesis by affecting PBR mRNA stability, thus inhibiting PBR expression, cholesterol transport into the mitochondria, and the subsequent steroid formation. Moreover, this action of PFDA on PBR mRNA stability indicates a new mechanism of action of peroxisome proliferators distinct from the classic transcription-mediated regulation of target genes.


Asunto(s)
Colesterol/metabolismo , Ácidos Decanoicos/farmacología , Fluorocarburos/farmacología , Células Intersticiales del Testículo/metabolismo , Mitocondrias/metabolismo , Proliferadores de Peroxisomas/farmacología , Receptores de GABA-A/biosíntesis , Esteroides/biosíntesis , Animales , Northern Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Daño del ADN/efectos de los fármacos , Electroforesis en Gel de Poliacrilamida , Antagonistas de Receptores de GABA-A , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/ultraestructura , Masculino , Ratones , Microscopía Electrónica , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Biosíntesis de Proteínas , Radioinmunoensayo , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Transfección/genética
6.
Biochem Pharmacol ; 58(9): 1389-93, 1999 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-10513982

RESUMEN

We have demonstrated that continuous administration of a gonadotropin-releasing hormone agonist (GnRH-Ag) decreases the expression of the mitochondrial peripheral-type benzodiazepine receptor (PBR) and increases the rate of DNA degradation in a time-dependent manner in the corpora lutea of pregnant rats. In the present study, we show in situ the GnRH-Ag-induced DNA fragmentation and correlate the increase of the rate of DNA degradation with the decrease in mitochondrial PBR ligand binding (r = 0.89). The GnRH-Ag-induced decrease in the 18-kDa PBR protein also correlated with the reduction in the Bcl-X(L), but not Bcl-2 (cell survival), gene product levels and the increase in the Bax (cell death) gene product expression in the luteal mitochondrial preparations. Considering the function of PBR in cholesterol uptake and intramitochondrial movement, we propose that decreased PBR expression may lead to reduced levels of mitochondrial membrane cholesterol, which, together with the ability of Bcl-X(L) and Bax to form ion channels, produces breaks in the outer membranes allowing the exit of cytochrome c, thus triggering apoptosis. Alternatively, PBR may exert an as yet unidentified anti-apoptotic function.


Asunto(s)
Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/agonistas , Mitocondrias/efectos de los fármacos , Receptores de GABA-A/biosíntesis , Animales , Apoptosis , Cuerpo Lúteo/efectos de los fármacos , Cuerpo Lúteo/metabolismo , Daño del ADN , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína X Asociada a bcl-2 , Proteína bcl-X
7.
Cancer Res ; 59(4): 831-42, 1999 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10029072

RESUMEN

Aberrant cell proliferation and increased invasive and metastatic behavior are hallmarks of the advancement of breast cancer. Numerous studies implicate a role for cholesterol in the mechanisms underlying cell proliferation and cancer progression. The peripheral-type benzodiazepine receptor (PBR) is an Mr 18,000 protein primarily localized to the mitochondria. PBR mediates cholesterol transport across the mitochondrial membranes in steroidogenic cells. A role for PBR in the regulation of tumor cell proliferation has also been shown. In this study, we examined the expression, characteristics, localization, and function of PBR in a battery of human breast cancer cell lines differing in their invasive and chemotactic potential as well as in several human tissue biopsies. Expression of PBR ligand binding and mRNA was dramatically increased in the highly aggressive cell lines, such as MDA-231, relative to nonaggressive cell lines, such as MCF-7. PBR was also found to be expressed at high levels in aggressive metastatic human breast tumor biopsies compared with normal breast tissues. Subcellular localization with both antibodies and a fluorescent PBR drug ligand revealed that PBR from the MDA-231 cell line as well as from aggressive metastatic human breast tumor biopsies localized primarily in and around the nucleus. This localization is in direct contrast to the largely cytoplasmic localization seen in MCF-7 cells, normal breast tissue, and to the typical mitochondrial localization seen in mouse tumor Leydig cells. Pharmacological characterization of the receptor and partial nucleotide sequencing of PBR cDNA revealed that the MDA-231 PBR is similar, although not identical, to previously described PBR. Addition of high affinity PBR drug ligands to MDA-231 cells increased the incorporation of bromodeoxyuridine into the cells in a dose-dependent manner, suggesting a role for PBR in the regulation of MDA-231 cell proliferation. Cholesterol uptake into isolated MDA-231 nuclei was found to be 30% greater than into MCF-7 nuclei. High-affinity PBR drug ligands regulated the levels of cholesterol present in MDA-231 nuclei but not in MCF-7. In addition, the PBR-dependent MDA-231 cell proliferation was found to highly correlate (r = -0.99) with the PBR-mediated changes in nuclear membrane cholesterol levels. In conclusion, these data suggest that PBR expression, nuclear localization, and PBR-mediated cholesterol transport into the nucleus are involved in human breast cancer cell proliferation and aggressive phenotype expression, thus participating in the advancement of the disease.


Asunto(s)
Neoplasias de la Mama/química , Núcleo Celular/química , Colesterol/metabolismo , Receptores de GABA-A/análisis , Secuencia de Aminoácidos , Animales , Transporte Biológico , Neoplasias de la Mama/patología , Proteínas Portadoras/análisis , División Celular , Núcleo Celular/metabolismo , Inhibidor de la Unión a Diazepam , Femenino , Humanos , Ratones , Datos de Secuencia Molecular , Fenotipo , ARN Mensajero/análisis , Receptores de GABA-A/genética , Receptores de GABA-A/fisiología
8.
J Mol Endocrinol ; 22(1): 45-54, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9924179

RESUMEN

We have demonstrated that continuous administration of a gonadotropin-releasing hormone agonist (GnRH-Ag) suppresses luteal steroidogenesis in the pregnant rat. We further demonstrated that the peripheral-type benzodiazepine receptor (PBR) and the steroidogenic acute regulatory protein (StAR) play key roles in cholesterol transport leading to steroidogenesis. The purpose of this study was to understand the cellular and molecular mechanisms involved in the suppression of luteal steroidogenesis leading to a fall in serum progesterone levels in GnRH-Ag-treated rats during early pregnancy. Pregnant rats were treated individually starting on day 8 of pregnancy with 5 microgram/day GnRH-Ag using an osmotic minipump. Sham-operated control rats received no treatment. At 0, 4, 8 and 24 h after initiation of the treatment, rats were killed and corpora lutea (CL) were removed for PBR mRNA, protein and radioligand binding analyses, immunoblot 1-D gel analysis of StAR, P450 scc and 3beta-hydroxysteroid dehydrogenase as well as 2-D gel analysis of StAR. The treatment decreased the luteal PBR mRNA expression at all time periods starting at 4 h compared with that in corresponding sham controls. GnRH-Ag also reduced, in the CL, the PBR protein/ligand binding, the StAR protein and P450 scc protein and its activity as early as 8 h after the treatment and they remained low compared with those in corresponding sham controls. The data from 2-D gel studies suggest that the majority of the decrease in StAR protein appears to be in the phosphorylated forms of StAR. Thus, we have demonstrated, for the first time, the presence of PBR and StAR in the pregnant rat CL and that the coordinated suppression of these proteins involved in the mitochondrial cholesterol transport along with P450 scc by GnRH-Ag leads to reduced ovarian steroidogenesis.


Asunto(s)
Cuerpo Lúteo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/agonistas , Fosfoproteínas/biosíntesis , Preñez/metabolismo , Progesterona/biosíntesis , Receptores de GABA-A/biosíntesis , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Colesterol/metabolismo , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Cuerpo Lúteo/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosfoproteínas/genética , Fosforilación , Embarazo , Progesterona/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/genética
9.
Endocr Res ; 24(3-4): 479-87, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9888528

RESUMEN

In various steroidogenic cell models, mitochondrial preparations and submitochondrial fractions, the expression of the mitochondrial 18 kDa peripheral-type benzodiazepine receptor (PBR) protein confers the ability to take up and release, upon ligand activation, cholesterol. Thus, cholesterol becomes available to P450scc on the inner mitochondrial membrane. These in vitro studies were validated by in vivo experiments. Treatment of rats with ginkgolide B (GKB), specifically reduced the ligand binding capacity, protein, and mRNA expression of the adrenocortical PBR and circulating glucocorticoid levels. Treatment with GKB also resulted in inhibition of PBR protein synthesis and corticosterone production by isolated adrenocortical cells in response to ACTH. The ontogeny of both PBR binding capacity and protein directly paralleled that of ACTH-inducible steroidogenesis in rat adrenal cells and in rats injected with ACTH. In addition, the previously described suppression of luteal progesterone synthesis in the pregnant rat by continuous in vivo administration of a gonadotropin-releasing hormone agonist may be due to decreased luteal PBR ligand binding and mRNA. These results suggest that (i) PBR is an absolute prerequisite for adrenocortical and luteal steroidogenesis, (ii) regulation of adrenal PBR expression may be used as a tool to control circulating glucocorticoid levels and (iii) the stress hypo-responsive period of neonatal rats may result from decreased adrenal cortical PBR expression.


Asunto(s)
Receptores de GABA-A/fisiología , Esteroides/biosíntesis , Corticoesteroides/biosíntesis , Glándulas Suprarrenales/metabolismo , Animales , Animales Recién Nacidos/metabolismo , Femenino , Flavonoides/farmacología , Ginkgo biloba/química , Glucocorticoides/biosíntesis , Hormona Liberadora de Gonadotropina/agonistas , Fase Luteínica , Ratones , Ratones Noqueados/genética , Extractos Vegetales/farmacología , Plantas Medicinales , Embarazo , Progesterona/biosíntesis , Ratas , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo
10.
Endocrinology ; 138(3): 1289-98, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9048638

RESUMEN

To determine what factors regulate gonocyte proliferation in newborn rats, we first examined the expression of several signal transduction molecules by immunocytochemistry in 3-day-old rat testis sections. We found that gonocytes specifically expressed the iota and zeta isoforms of protein kinase (PK) C (PKC) and the phosphatidylinositol 3-kinase (PI3-K). Because both the zeta PKC and PI 3-K have been shown to play a role in platelet-derived growth factor (PDGF)-induced cell proliferation, we examined the effects of PDGF on gonocytes. For this, we developed a method to obtain highly purified and viable gonocytes in culture. After enzymatic digestion, differential adhesion, and two successive gradient fractionations, the gonocyte suspension obtained was over 90% pure, as assessed by light microscopy. The viability of cultured gonocytes exceeded 90% after 48 h in the presence of 2.5% FBS used as a survival factor. Immunodetection studies showed that isolated gonocytes expressed zeta PKC, PI 3-K, and the PDGF receptor. Treatment with 10 ng/ml PDGF induced a 4-fold increase of bromodeoxyuridine incorporation into gonocytes (from 5% proliferative gonocytes under basal conditions to 20% in the presence of PDGF). Because neonatal Sertoli cells secrete high levels of the growth promoting steroid, 17 beta-estradiol, we also tested its effect and found that it induced gonocyte proliferation at a level comparable with that of PDGF and that this effect was blocked by the estrogen receptor antagonist, ICI 164384. The combination of PDGF and estradiol, however, was not additive, suggesting that their effects were mediated by common molecular target(s). These results demonstrate that PDGF and estradiol activate gonocyte proliferation in vitro, suggesting that they may act as the physiological regulators of gonocyte development in vivo.


Asunto(s)
Estradiol/farmacología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Transducción de Señal , Testículo/citología , Testículo/efectos de los fármacos , Animales , División Celular/efectos de los fármacos , Supervivencia Celular , Células Cultivadas , Inmunohistoquímica , Isoenzimas/metabolismo , Masculino , Fosfatidilinositol 3-Quinasas , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Fosfotirosina/metabolismo , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo
11.
Steroids ; 62(1): 21-8, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9029710

RESUMEN

Steroidogenesis begins with the metabolism of cholesterol to pregnenolone by the inner mitochondrial membrane cytochrome P450 side-chain cleavage (P450scc) enzyme. The rate of steroid formation, however, depends on the rate of cholesterol transport from intracellular stores to the inner mitochondrial membrane and loading of P450scc with cholesterol. In previous in vitro studies, we demonstrated that a key element in the regulation of cholesterol transport is the mitochondrial peripheral-type benzodiazepine receptor (PBR). We also showed that the polypeptide diazepam binding inhibitor (DBI), an endogenous PBR ligand, stimulates cholesterol transport and promotes loading of cholesterol to P450scc in vitro, and that its presence is vital for hCG-induced steroidogenesis by Leydig cells. Based on these data and the observations that i) the mitochondrial PBR binding and topography are regulated by hormones; ii) the 18-kDa PBR protein is functionally coupled to the mitochondrial contact site voltage-dependent anion channel protein; iii) the 18-kDa PBR protein is a channel for cholesterol, as shown by molecular modeling and in vitro reconstitution studies; iv) targeted disruption of the PBR gene in steroidogenic cells dramatically reduces the ability of the cells to transport cholesterol in the mitochondria and produce steroids; v) endocrine disruptors, with known anisteroidogenic effect, inhibit PBR ligand binding; and vi) in vivo reduction of adrenal PBR expression results in reduced circulating glucocorticoid levels, we conclude that PBR is an indispensable element of the steroidogenic machinery.


Asunto(s)
Colesterol/metabolismo , Receptores de GABA-A/química , Receptores de GABA-A/fisiología , Esteroides/metabolismo , Animales , Sitios de Unión , Transporte Biológico , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/metabolismo , Proteínas Portadoras/farmacología , Membrana Celular/metabolismo , Gonadotropina Coriónica/farmacología , AMP Cíclico/farmacología , Inhibidor de la Unión a Diazepam , Flunitrazepam/farmacología , Humanos , Ratones , Mitocondrias/metabolismo , Modelos Biológicos , Modelos Moleculares , Mutación , Conformación Proteica , Receptores de GABA-A/efectos de los fármacos
12.
Int J Cancer ; 63(6): 815-22, 1995 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-8847140

RESUMEN

Infection with erbB-2 (E) of Ha-ras (H) oncogene-transfected cells has been previously shown to cooperatively induce anchorage-independent growth of the MCF10A human mammary epithelial cell line in vitro, but not to induce nude mouse tumorigenicity. Here we show that oncogene-transformed MCF10A are able to halt in the lungs of nude mice, a sign of organ colonization potential. We have therefore studied the transformants for in vitro migratory and invasive properties known to correlate with the metastatic potential of human mammary carcinoma cells in nude mice. MCF10A transfected with Ha-ras, infected with a recombinant retroviral vector containing the human c-erB-2 proto-oncogene (MCF10A-HE cells), show a higher invasive index than either the single transfectant (MCF10A-H) or MCF10A-erB-2(MCF10A-E) cells in the Boyden chamber chemotaxis and chemoinvasion assays. The MCF10A-HE cells also adopted an invasive stellate growth pattern when plated or embedded in Matrigel, in contrast to the spherical colonies formed by the single transformants MCF10A-H, MCF10A-E, and the parental cells. Dot-blot analysis of gelatinase A and TIMP-2 mRNA levels revealed increasing gelatinase A mRNA levels (HE > E > H > MCF10A) and reduced TIMP-2 expression in both single and double transformants. Furthermore, MCF10A-HE cells show more MMP-2 activity than parental MCF10A cells or the single transformants. CD44 analysis revealed differential isoform banding for the MCF10A-HE cells compared to parental cells, MCF10A-H and MCF10A-E, accompanied by increased binding of hyaluronan by the double transformants. Our results indicate that erB-2 and Ha-ras co-expression can induce a more aggressive phenotype in vitro, representative of the malignancy of mammary carcinomas.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias Pulmonares/metabolismo , Invasividad Neoplásica , Proteína Oncogénica p21(ras)/biosíntesis , Receptor ErbB-2/biosíntesis , Animales , Neoplasias de la Mama/patología , Línea Celular Transformada , Femenino , Gelatinasas/biosíntesis , Técnicas de Transferencia de Gen , Humanos , Receptores de Hialuranos/biosíntesis , Neoplasias Pulmonares/patología , Metaloproteinasa 2 de la Matriz , Metaloendopeptidasas/biosíntesis , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Proteína Oncogénica p21(ras)/genética , Biosíntesis de Proteínas , Proto-Oncogenes Mas , Receptor ErbB-2/genética , Inhibidor Tisular de Metaloproteinasa-2
13.
J Leukoc Biol ; 56(5): 605-11, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7525818

RESUMEN

CD44 glycoproteins are present on the surfaces of many hematopoietic cells and in some cases can bind hyaluronan, a major component of the extracellular matrix. In the present study, we have found that newly explanted human peripheral blood monocytes (PBMs) exhibit a major CD44 band of 85 kDa, whereas autologous alveolar macrophages (AM phi) express multiple isoforms ranging from 85 to 200 kDa. Within 4 h in culture, PBMs began expressing new CD44 isoforms of 120, 150, and 180 kDa. Newly explanted AM phi specifically bound [3H]hyaluronan (135 cpm/microgram protein), but newly explanted PBMs did not. However, in vitro cultured PBM progressively acquired the ability to bind [3H]hyaluronan and exhibited specific binding of hyaluronan similar to that of AM phi (113 cpm/microgram protein) after 4 days in culture. In both case, the binding of [3H]hyaluronan was specifically inhibited by the addition of monoclonal antibody directed against CD44. AM phi readily degraded [3H]hyaluronan and reached a plateau after 4 days in culture (115 cpm/microgram protein). Newly explanted PBM exhibit no hyaluronan degradation and only a small degradative activity after 4 days in culture (6 to 11 cpm/microgram protein). Thus, CD44 expression and function appear to change as PBM mature in vitro resembling more that found in AM phi.


Asunto(s)
Proteínas Portadoras/fisiología , Macrófagos Alveolares/química , Monocitos/química , Receptores de Superficie Celular/fisiología , Receptores Mensajeros de Linfocitos/fisiología , Adulto , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Células Cultivadas , Femenino , Humanos , Receptores de Hialuranos , Ácido Hialurónico/metabolismo , Linfocitos/metabolismo , Masculino , Peso Molecular , Receptores de Superficie Celular/química , Receptores de Superficie Celular/metabolismo , Receptores Mensajeros de Linfocitos/química , Receptores Mensajeros de Linfocitos/metabolismo
14.
J Cell Physiol ; 160(2): 275-86, 1994 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7518822

RESUMEN

In the present study, we examined a panel of human breast cancer cell lines with regard to their expression of CD44 and ability to bind and degrade hyaluronan. The cell lines expressed varying amounts of different molecular weight forms of CD44 (85-200 kDa) and, in general, those that expressed the greatest amounts of CD44 were the most invasive as judged by in vitro assays. In addition, the ability to bind and degrade hyaluronan was restricted to the cell lines expressing high levels of CD44, and both these functions were blocked by an antibody to CD44 (Hermes-1). Moreover, the rate of [3H]hyaluronan degradation was highly correlated with the amount of CD44 (r = 0.951, P < 0.0001), as well as with the invasive potential of the cells. Scatchard analysis of the [3H]hyaluronan binding of these cells revealed the existence of significant differences in both their binding capacity and their dissociation constant. To determine the source of this deviation, the different molecular weight forms of CD44 were partially separated by gel filtration chromatography. In all cell lines, the 85 kDa form was able to bind hyaluronan, although with different affinities. In contrast, not all of the high molecular weight forms of CD44 had this ability. These results illustrate the diversity of CD44 molecules in invasive tumor cells, and suggest that one of their major functions is to degrade hyaluronan.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Portadoras/metabolismo , Ácido Hialurónico/metabolismo , Invasividad Neoplásica , Receptores de Superficie Celular/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Western Blotting , Neoplasias de la Mama/patología , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/química , Cromatografía en Gel , Humanos , Receptores de Hialuranos , Peso Molecular , Unión Proteica , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/química , Receptores Mensajeros de Linfocitos/biosíntesis , Receptores Mensajeros de Linfocitos/química , Células Tumorales Cultivadas
15.
In Vitro Cell Dev Biol Anim ; 29A(12): 943-9, 1993 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8167918

RESUMEN

Primary cultures of immature rat Sertoli cells in plastic dishes are highly responsive to follicle stimulating hormone (FSH) and its second messenger, cAMP, in metabolizing testosterone to estradiol, thus indicating the presence of an active, hormone-regulated aromatase cytochrome P450 (P450arom). However, in vivo studies indicated that P450arom is FSH-responsive only in very young animals, where the cells have not yet differentiated, but they lose this ability later on in development. Sertoli cells grown on Matrigel (a reconstituted basement membrane), laminin (a basement membrane component), or in bicameral chambers coated with Matrigel, assume structural and functional characteristics more similar to that of in vivo differentiated Sertoli cells. When the cells were cultured on laminin or Matrigel, the FSH- and cAMP-induced estradiol production was greatly reduced by 30 and 60%, respectively. When Sertoli cells were cultured in bicameral chambers coated with Matrigel, no induction of testosterone aromatization by FSH or cAMP was observed. However, FSH-induced cAMP formation was greater when the cells were cultured on basement membrane or in the chambers than on plastic dishes. These results suggest that culture conditions favoring the assumption by Sertoli cells of a phenotype closer that of the differentiated cells in vivo (tall columnar and highly polarized) suppress the induction of P450arom by FSH and cAMP. We then examined the mechanism(s) by which cell phenotype affects p450arom activity. Northern blot analyses of Sertoli cell RNA revealed one major band of 1.9 Kb and two minor bands of 3.3 and 5.2 Kb. However, there were no changes at the level of the expression of P450arom messenger RNA under the different culture conditions.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Aromatasa/metabolismo , Hormona Folículo Estimulante/farmacología , Células de Sertoli/enzimología , Animales , Bucladesina/farmacología , Diferenciación Celular/efectos de los fármacos , Colágeno , ADN/análisis , Combinación de Medicamentos , Estradiol/metabolismo , Laminina , Masculino , Plásticos , Proteoglicanos , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Células de Sertoli/citología , Células de Sertoli/efectos de los fármacos , Testosterona/metabolismo
16.
J Cell Biol ; 116(4): 1055-62, 1992 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-1370836

RESUMEN

The hyaluronan receptor belongs to the polymorphic family of CD44 glycoproteins, which have been implicated in a variety of cellular functions including adhesion to hyaluronan and collagen, the binding of lymphocytes to high endothelial cells during extravasation, and conferring metastatic potential to carcinoma cells. Here, we demonstrate that the receptor also participates in the uptake and degradation of hyaluronan by both transformed fibroblasts (SV-3T3 cells) and alveolar macrophages. These cells were incubated with isotopically labeled hyaluronan for various periods of time, and the extent of degradation was determined by either molecular-sieve chromatography or centrifugation through Centricon 30 microconcentrators. The macrophages degraded the hyaluronan at a faster rate than the SV-3T3 cells, which may reflect the fact that they contained a greater number of receptors. More importantly, in both cell types, the degradation of hyaluronan was specifically blocked by antibodies directed against the receptor. However, the receptor by itself did not have the ability to degrade hyaluronan, since preparations of SV-3T3 membranes containing the receptor did not break down hyaluronan. Subsequent experiments revealed that macrophages can internalize fluorescein-tagged hyaluronan, and this process was blocked by antibodies against the receptor. Furthermore, the subsequent degradation of hyaluronan was inhibited by agents that block the acidification of lysosomes (chloroquine and NH4Cl). Thus, the most likely explanation for these results is that the receptor mediates the uptake of hyaluronan into the cell where it can be degraded by acid hydrolases in lysosomes. The ability of cells expressing the receptor to degrade hyaluronan may be important during tissue morphogenesis and cell migration.


Asunto(s)
Ácido Hialurónico/metabolismo , Macrófagos Alveolares/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Cloruro de Amonio/farmacología , Animales , Anticuerpos Monoclonales , Línea Celular Transformada , Cloroquina/farmacología , Fluoresceína , Fluoresceínas , Receptores de Hialuranos , Hialuronoglucosaminidasa/metabolismo , Lisosomas/enzimología , Ratones , Receptores de Superficie Celular/inmunología
17.
Biochem J ; 243(2): 457-65, 1987 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-2820381

RESUMEN

Rabbit platelets were labelled with [3H]inositol and a membrane fraction was isolated in the presence of ATP, MgCl2 and EGTA. Incubation of samples for 10 min with 0.1 microM-Ca2+free released [3H]inositol phosphates equivalent to about 2.0% of the membrane [3H]phosphoinositides. Addition of 10 microM-guanosine 5'-[gamma-thio]triphosphate (GTP[S]) caused an additional formation of [3H]inositol phosphates equivalent to 6.6% of the [3H]phosphoinositides. A half-maximal effect was observed with 0.4 microM-GTP[S]. The [3H]inositol phosphates that accumulated consisted of 10% [3H]inositol monophosphate, 88% [3H]inositol bisphosphate ([3H]IP2) and 2% [3H]inositol trisphosphate ([3H]IP3). Omission of ATP and MgCl2 led to depletion of membrane [3H]polyphosphoinositides and marked decreases in the formation of [3H]inositol phosphates. Thrombin (2 units/ml) or GTP (4-100 microM) alone weakly stimulated [3H]IP2 formation, but together they acted synergistically to exert an effect comparable with that of 10 microM-GTP[S]. The action of thrombin was also potentiated by 0.1 microM-GTP[S]. Guanosine 5'-[beta-thio]diphosphate not only inhibited the effects of GTP[S], GTP and GTP with thrombin, but also blocked the action of thrombin alone, suggesting that this depended on residual GTP. Incubation with either GTP[S] or thrombin and GTP decreased membrane [3H]phosphatidylinositol 4-phosphate ([H]PIP) and prevented an increase in [3H]phosphatidylinositol 4,5-bisphosphate ([3H]PIP2) observed in controls. Addition of unlabelled IP3 to trap [3H]IP3 before it was degraded to [3H]IP2 showed that only about 20% of the additional [3H]inositol phosphates that accumulated with GTP[S] or thrombin and GTP were derived from the action of phospholipase C on [3H]PIP2. The results provide further evidence that guanine-nucleotide-binding protein mediates signal transduction between the thrombin receptor and phospholipase C, and suggest that PIP may be a major substrate of this enzyme in the platelet.


Asunto(s)
Plaquetas/enzimología , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/farmacología , Tionucleótidos/farmacología , Trombina/farmacología , Fosfolipasas de Tipo C/sangre , Adenosina Trifosfato/farmacología , Animales , Plaquetas/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Guanosina 5'-O-(3-Tiotrifosfato) , Fosfatos de Inositol/sangre , Fosfatidilinositoles/sangre , Conejos
18.
Biochem Biophys Res Commun ; 124(2): 393-9, 1984 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-6093798

RESUMEN

Addition of angiotensin II (0.3 microM) to bovine adrenal fasciculata cell suspensions prelabeled with [32P] induced a rapid (15 seconds) and marked decrease of the radioactivity from phosphatidylinositol 4,5-biphosphate (62%) and phosphatidylinositol 4-monophosphate (35%). This effect was concentration-dependent and specifically inhibited in the presence of (Sar1-Ala8)-angiotensin II; it was also completely prevented in the absence of extracellular calcium. The present data appear to illustrate the earliest biological response detectable in bovine fasciculata cells under angiotensin II challenge.


Asunto(s)
Corteza Suprarrenal/metabolismo , Angiotensina II/farmacología , Fosfatidilinositoles/metabolismo , Corteza Suprarrenal/citología , Corteza Suprarrenal/efectos de los fármacos , Animales , Bovinos , Técnicas In Vitro , Cinética , Fosfatos de Fosfatidilinositol , Radioisótopos de Fósforo
19.
Biochem Biophys Res Commun ; 123(1): 33-40, 1984 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-6089796

RESUMEN

Phospholipase C (Bacillus cereus) added to the incubation medium stimulated the steroidogenic activity of bovine adrenal zona fasciculata cell suspensions to a level similar to that induced by optimal concentration of ACTH. This effect was not related to an increase of cyclic AMP; it was calcium-dependent and was also induced by an other bacterial phospholipase C (from Clostridium perfringens) whereas phospholipases A2 and D were ineffective. Phospholipid metabolism was examined in these cells after radiolabeling with [14C]-glycerol or [32P]orthophosphate. Phospholipase C induced a very fast (5 seconds) increase in cellular [14C]-1,2-diacylglycerol followed by [32P] labeling of phosphatidic acid and phosphatidylinositol. These events preceded the stimulation of steroidogenesis which was detectable after 2 minutes of incubation. These observations suggest that activation of an endogenous phospholipase C activity may be considered as an early event in the response of bovine adrenocortical cells to steroidogenic effectors such as angiotensin II and acetylcholine.


Asunto(s)
Corteza Suprarrenal/metabolismo , Hidrocortisona/biosíntesis , Fosfolipasas/farmacología , Fosfolípidos/biosíntesis , Fosfolipasas de Tipo C/farmacología , Corteza Suprarrenal/efectos de los fármacos , Animales , Bacillus cereus/enzimología , Calcio/farmacología , Bovinos , AMP Cíclico/metabolismo , Diglicéridos/metabolismo , Cinética , Fosfatos/metabolismo , Fosfatidilinositoles/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA