Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Front Neuroendocrinol ; 36: 108-29, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25223867

RESUMEN

Sex steroids play critical roles in the regulation of the brain and many other organs. Traditionally, researchers have focused on sex steroid signaling that involves travel from the gonads via the circulation to intracellular receptors in target tissues. This classic concept has been challenged, however, by the growing number of cases in which steroids are synthesized locally and act locally within diverse tissues. For example, the brain and prostate carcinoma were previously considered targets of gonadal sex steroids, but under certain circumstances, these tissues can upregulate their steroidogenic potential, particularly when circulating sex steroid concentrations are low. We review some of the similarities and differences between local sex steroid synthesis in the brain and prostate cancer. We also share five lessons that we have learned during the course of our interdisciplinary collaboration, which brought together neuroendocrinologists and cancer biologists. These lessons have important implications for future research in both fields.


Asunto(s)
Encéfalo/metabolismo , Hormonas Esteroides Gonadales/biosíntesis , Neoplasias de la Próstata/metabolismo , Conducta Cooperativa , Humanos , Masculino
2.
Cancer Cell Int ; 13(1): 93, 2013 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-24059654

RESUMEN

BACKGROUND: Somatostatin receptors (SSTRs) and opioid receptors (ORs) belong to the superfamily of G-protein coupled receptors and function as negative regulators of cell proliferation in breast cancer. In the present study, we determined the changes in SSTR subtype 2 (SSTR2) and µ, δ and κ-ORs expression, signaling cascades and apoptosis in three different breast cancer cells namely MCF-7, MDA-MB231 and T47D. METHODS: Immunocytochemistry and western blot analysis were employed to study the colocalization and changes in MAPKs (ERK1/2 and p38), cell survival pathway (PI3K/AKT) and tumor suppressor proteins (PTEN and p53) in breast cancer cell lines. The nature of cell death upon activation of SSTR2 or OR was analysed using flow cytometry analysis. RESULTS: The activation of SSTR2 and ORs modulate MAPKs (ERK1/2 and p38) in cell dependent and possibly estrogen receptor (ER) dependent manner. The activation of tumor suppressor proteins phosphatase and tensin homolog (PTEN) and p53 antagonized the PI3K/AKT cell survival pathway. Flow cytometry analyses reveal increased necrosis as opposed to apoptosis in MCF-7 and T47D cells when compared to ER negative MDA-MB231 cells. Furthermore, receptor and agonist dependent expression of ORs in SSTR2 immunoprecipitate suggest that SSTR2 and ORs might interact as heterodimers and inhibit epidermal growth factor receptor phosphorylation. CONCLUSION: Taken together, findings indicate a new role for SSTR2/ORs in modulation of signaling pathways involved in cancer progression and provide novel therapeutic approaches in breast cancer treatment.

3.
Biochim Biophys Acta ; 1813(6): 1172-89, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21419811

RESUMEN

Epidermal growth factor (EGF) regulates normal and tumor cell proliferation via epidermal growth factor receptor (EGFR) phosphorylation, homo- or heterodimerization and activation of mitogen-activated protein kinases (MAPKs) and PI3K/AKT cell survival pathways. In contrast, SST via activation of five different receptor subtypes inhibits cell proliferation and has been potential target in tumor treatment. To gain further insight for the effect of SSTRs on EGFR activated signaling, we determine the role of SSTR1 and SSTR1/5 in human embryonic kidney (HEK) 293 cells. We here demonstrate that cells transfected with SSTR1 or SSTR1/5 negatively regulates EGF mediated effects attributed to the inhibition of EGFR phosphorylation, MAPKs as well as the cell survival signaling. Furthermore, SSTR effects were significantly enhanced in cells when EGFR was knock down using siRNA or treated with selective antagonist (AG1478). Most importantly, the presence of SSTR in addition to modulating signaling pathways leads to the dissociation of the constitutive and EGF induced heteromeric complex of EGFR/ErbB2. Furthermore, cells cotransfected with SSTR1/5 display pronounced effect of SST on the signaling and dissociation of the EGFR/ErbB2 heteromeric complex than the cells expressing SSTR1 alone. Taken together this study provides the first evidence that the presence of SSTR controls EGF mediated cell survival pathway via dissociation of ErbB heteromeric complex. We propose that the activation of SSTR and blockade of EGFR might serve novel therapeutic approach in inhibition of tumor proliferation.


Asunto(s)
Receptores ErbB/metabolismo , Receptores de Somatostatina/metabolismo , Transducción de Señal/fisiología , Western Blotting , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Células HEK293 , Humanos , Inmunoprecipitación , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica , Multimerización de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas , Interferencia de ARN , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Somatostatina/genética , Transducción de Señal/efectos de los fármacos , Somatostatina/farmacología , Tirfostinos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Int Immunopharmacol ; 7(3): 277-86, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17276885

RESUMEN

Signal Transducer and Activator of Transcription (STAT)-6 is a transcriptional factor activated mainly through the cytokines IL-4 and IL-13 leading to the Th2 cell differentiation. Th2 cells play a role in the etiology and pathogenesis of allergic disease. Histamine alters the Th1/Th2 cytokine balance towards the Th2 cytokine profile and consequently plays a role in allergic diseases and asthma. This study was designed to investigate the effects of histamine on the STAT6 phosphorylation. C57/BL6 splenocytes were pretreated with different concentrations of histamine (10(-)(4) M to 10(-)(13) M) followed by stimulation with PMA+ionomycin or IL-4. The phosphorylated and total basal STAT6 levels were assessed by employing the immunoblotting technique. Histamine caused the hyper-phosphorylation of STAT6. H1 receptor antagonist pyrilamine reversed the effect of histamine on STAT6 phosphorylation. However, H2 receptor antagonist ranitidine and H3/H4 receptor antagonist thioperamide did not affect the histamine mediated hyper-phosphorylation of STAT6. Furthermore, H1 receptor agonist betahistine enhanced the phosphorylation of STAT6 whereas H2 receptor agonist amthamine did not affect the phosphorylation STAT6. Furthermore, tyrosine kinase inhibitor, tyrphostin, inhibited the histamine mediated phosphorylation of STAT6 when stimulated with PMA+ionomycin. The effects of histamine on the STAT6 phosphorylation were indirect since they were blocked either by the antibodies to IL-4 and IL-13 or in IL-4 knock out mice in the presence of IL-13 antibody. These observations suggest that histamine indirectly affected the STAT6 phosphorylation via its effects on the secretion of cytokines (IL-4) and H1 receptor played a role in this process.


Asunto(s)
Histamina/farmacología , Interleucina-4/biosíntesis , Receptores Histamínicos H1/fisiología , Factor de Transcripción STAT6/metabolismo , Animales , Femenino , Interleucina-13/fisiología , Ionomicina/farmacología , Ratones , Ratones Endogámicos C57BL , Fosforilación , Receptores Histamínicos/fisiología , Acetato de Tetradecanoilforbol/farmacología
5.
Int Immunopharmacol ; 6(3): 485-93, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16428084

RESUMEN

Histamine shifts TH1/TH2 cytokine balance from TH1 to TH2 cytokines and regulates the function of lymphocytes after binding to histamine receptors. The phosphorylation of STAT factors and the translocation to the nucleus are important steps in the regulation of TH1/TH2 cytokine balance. This study was designed to investigate the effects of histamine on the phosphorylation of STAT4. C57BL/6 splenocytes were isolated and treated with histamine (10(-4) to 10(-9) M) after activation with either PMA (phorbol 12 myristate 13-acetate) plus ionomycin or IL-12. The phosphorylated STAT4 levels were analyzed by Western Blot Analysis. Unstimulated splenocytes expressed both STAT4 and phosphorylated STAT4. However, phosphorylated STAT4 gradually declined within 24 h. Histamine increased the phosphorylation of STAT4 at lower concentrations (10(-6) to 10(-9) M), and had no effect at higher concentrations (10(-4) and 10(-5) M) after the cells were stimulated with PMA + ionomycin. Histamine did not affect IL-12-induced phosphorylation of STAT4. To characterize the histamine receptor subtypes involved in the up-regulation of STAT4 phosphorylation, various H1, H2 and H3/H4 receptor antagonists and/or agonists were employed. H1 receptor agonist (betahistine), but not H2 receptor agonist (amthamine), induced phosphorylation of STAT4. H1 receptor antagonist (pyrilamine) inhibited histamine-mediated phosphorylation of STAT4. However, H2 receptor antagonist (ranitidine) and H3/H4 receptor antagonist (thioperamide) did not alter this effect. Tyrosine kinase inhibitor (tyrphostin) failed to block histamine-mediated phosphorylation of STAT4. These observations suggest that histamine up-regulated the phosphorylation of STAT4 via H1 receptors, and that the Ca2+-PKC pathway, but not the tyrosine kinase pathway, was involved in this effect.


Asunto(s)
Histamina/fisiología , Receptores Histamínicos H1/fisiología , Factor de Transcripción STAT4/metabolismo , Regulación hacia Arriba/fisiología , Animales , Carcinógenos/farmacología , Células Cultivadas , Femenino , Agonistas de los Receptores Histamínicos/farmacología , Antagonistas de los Receptores Histamínicos H1/farmacología , Interleucina-12/fisiología , Ionomicina/farmacología , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo , Acetato de Tetradecanoilforbol/farmacología
6.
PLoS One ; 11(5): e0154967, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27152724

RESUMEN

Exosomes proteins and microRNAs have gained much attention as diagnostic tools and biomarker potential in various malignancies including prostate cancer (PCa). However, the role of exosomes and membrane-associated receptors, particularly epidermal growth factor receptor (EGFR) as mediators of cell proliferation and invasion in PCa progression remains unexplored. EGFR is frequently overexpressed and has been associated with aggressive forms of PCa. While PCa cells and tissues express EGFR, it is unknown whether exosomes derived from PCa cells or PCa patient serum contains EGFR. The aim of this study was to detect and characterize EGFR in exosomes derived from PCa cells, LNCaP xenograft and PCa patient serum. Exosomes were isolated from conditioned media of different PCa cell lines; LNCaP xenograft serum as well as patient plasma/serum by differential centrifugation and ultracentrifugation on a sucrose density gradient. Exosomes were confirmed by electron microscopy, expression of exosomal markers and NanoSight™ analysis. EGFR expression was determined by western blot analysis and ELISA. This study demonstrates that exosomes may easily be derived from PCa cell lines, serum obtained from PCa xenograft bearing mice and clinical samples derived from PCa patients. Presence of exosomal EGFR in PCa patient exosomes may present a novel approach for measuring of the disease state. Our work will allow to build on this finding for future understanding of PCa exosomes and their potential role in PCa progression and as minimal invasive biomarkers for PCa.


Asunto(s)
Receptores ErbB/metabolismo , Exosomas/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Biomarcadores de Tumor/sangre , Western Blotting , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Xenoinjertos , Humanos , Masculino , Ratones , Microscopía Electrónica de Transmisión , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/patología
7.
PLoS One ; 11(6): e0157392, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27295211

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0154967.].

8.
Oncotarget ; 7(12): 14639-58, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-26840259

RESUMEN

Prostate cancer (PCa) is the most frequently diagnosed cancer in men. Current research on tumour-related extracellular vesicles (EVs) suggests that exosomes play a significant role in paracrine signaling pathways, thus potentially influencing cancer progression via multiple mechanisms. In fact, during the last decade numerous studies have revealed the role of EVs in the progression of various pathological conditions including cancer. Moreover, differences in the proteomic, lipidomic, and cholesterol content of exosomes derived from PCa cell lines versus benign prostate cell lines confirm that exosomes could be excellent biomarker candidates. As such, as part of an extensive proteomic analysis using LCMS we previously described a potential role of exosomes as biomarkers for PCa. Current evidence suggests that uptake of EV's into the local tumour microenvironment encouraging us to further examine the role of these vesicles in distinct mechanisms involved in the progression of PCa and castration resistant PCa. For the purpose of this study, we hypothesized that exosomes play a pivotal role in cell-cell communication in the local tumour microenvironment, conferring activation of numerous survival mechanisms during PCa progression and development of therapeutic resistance. Our in vitro results demonstrate that PCa derived exosomes significantly reduce apoptosis, increase cancer cell proliferation and induce cell migration in LNCaP and RWPE-1 cells. In conjunction with our in vitro findings, we have also demonstrated that exosomes increased tumor volume and serum PSA levels in vivo when xenograft bearing mice were administered DU145 cell derived exosomes intravenously. This research suggests that, regardless of androgen receptor phenotype, exosomes derived from PCa cells significantly enhance multiple mechanisms that contribute to PCa progression.


Asunto(s)
Comunicación Celular , Proliferación Celular , Exosomas/patología , Neoplasias de la Próstata/patología , Microambiente Tumoral , Animales , Apoptosis , Movimiento Celular , Progresión de la Enfermedad , Exosomas/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Fenotipo , Neoplasias de la Próstata/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Oncotarget ; 6(13): 11327-41, 2015 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-25857301

RESUMEN

Large oncosomes (LO) are atypically large (1-10 µm diameter) cancer-derived extracellular vesicles (EVs), originating from the shedding of membrane blebs and associated with advanced disease. We report that 25% of the proteins, identified by a quantitative proteomics analysis, are differentially represented in large and nano-sized EVs from prostate cancer cells. Proteins enriched in large EVs included enzymes involved in glucose, glutamine and amino acid metabolism, all metabolic processes relevant to cancer. Glutamine metabolism was altered in cancer cells exposed to large EVs, an effect that was not observed upon treatment with exosomes. Large EVs exhibited discrete buoyant densities in iodixanol (OptiPrep(TM)) gradients. Fluorescent microscopy of large EVs revealed an appearance consistent with LO morphology, indicating that these structures can be categorized as LO. Among the proteins enriched in LO, cytokeratin 18 (CK18) was one of the most abundant (within the top 5th percentile) and was used to develop an assay to detect LO in the circulation and tissues of mice and patients with prostate cancer. These observations indicate that LO represent a discrete EV type that may play a distinct role in tumor progression and that may be a source of cancer-specific markers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Micropartículas Derivadas de Células/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Micropartículas Derivadas de Células/patología , Centrifugación , Cromatografía Líquida de Alta Presión , Cromatografía de Fase Inversa , Forminas , Glutamina/metabolismo , Humanos , Queratina-18/metabolismo , Masculino , Ratones , Proteínas de Neoplasias/sangre , Proteínas de Neoplasias/genética , Tamaño de los Orgánulos , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Transporte de Proteínas , Proteómica/métodos , Interferencia de ARN , Espectrometría de Masas en Tándem , Transfección
10.
Clin Biochem ; 47(13-14): 1286-92, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24956264

RESUMEN

OBJECTIVES: Exosomes are emerging as a source of biomarkers with putative prognostic and diagnostic value. However, little is known about the efficiency, reproducibility and reliability of the protocols routinely used to quantify exosomes in the human serum. DESIGN AND METHODS: We used increasing amounts of the same serum sample to isolate exosomes using two different methods: ultracentrifugation onto a sucrose cushion and ExoQuick™. Quantitative analysis of serum-derived exosomes was performed by determining protein concentration (BCA assay) and the number of nanoparticles (Nanosight™ technology). Exosome quality was assessed by Coomassie staining and Western blotting for CD9, LAMP2 exosomal markers and a negative marker Grp94. RESULTS: Correlation between serum volume and the number of isolated exosomes is significant for both methods when exosomes are quantified using protein concentration. However, when the number of nanoparticles is used to quantify exosomes, ExoQuick™ is the only reproducible and efficient method. CD9, LAMP2 and Grp94 exosomal markers are equivalently expressed in both methods. However, exosomes isolated using ultracentrifuge method are strongly contaminated with albumin and IgG. CONCLUSION: ExoQuick™ is an efficient and reproducible method to isolate exosomes for quantitative studies, whereas ultracentrifugation is not. Moreover, high albumin contamination of ultracentrifuged-derived exosomes impairs the use of protein concentration as a mean to quantify serum-derived exosomes.


Asunto(s)
Biomarcadores/sangre , Exosomas/química , Exosomas/metabolismo , Suero/química , Albúminas/metabolismo , Humanos , Inmunoglobulina G/sangre , Proteína 2 de la Membrana Asociada a los Lisosomas/sangre , Glicoproteínas de Membrana/sangre , Nanopartículas/química , Reproducibilidad de los Resultados , Tetraspanina 29/sangre , Ultracentrifugación/métodos
11.
J Steroid Biochem Mol Biol ; 143: 19-28, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24565566

RESUMEN

Castration-resistant prostate cancer (CRPC) remains largely dependent on androgen receptor (AR). Residual tissue androgens are consistently detected within CRPC tumors and play a critical role in facilitating AR-mediated signaling pathways which lead to disease progression. Testosterone and dihydrotestosterone (DHT) are the major androgens detected in tumors. They are produced through three biosynthesis pathways: Δ(4), Δ(5), and backdoor pathways. Both androgens bind to and stimulate AR activation. The current study investigates the effects of pomegranate extracts (POM) and their ability to inhibit androgen biosynthesis using PCa cell lines (22RV1 and LNCaP) in vitro as well as the PTEN knockout mouse model representing prostate cancer. Steroids were extracted using ethyl acetate or solid phase extraction, and then analyzed by UPLC/MS/MS. The results showed that POM (0-12µg/mL) reduced the production of testosterone, DHT, DHEA, androstenedione, androsterone, and pregnenolone in both cell lines. In addition our in vivo data supports this observation with a reduction in serum steroids determined after 20 weeks of POM treatment (0.17 g/L in drinking water). In accordance with these results, Western blotting of cell lysates and tPSA analysis determined that PSA was significantly decreased by the treatment of POM. Interestingly, AKR1C3 and AR levels were shown to be increased in both cell lines, perhaps as a negative feedback effect in response to steroid inhibition. Overall, these results provide mechanistic evidence to support the rationale for recent clinical reports describing efficacy of POM in CRPC patients.


Asunto(s)
Andrógenos/biosíntesis , Lythraceae/química , Fosfohidrolasa PTEN/fisiología , Extractos Vegetales/farmacología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata/tratamiento farmacológico , Androstenodiona/metabolismo , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Dihidrotestosterona/metabolismo , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Noqueados , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/metabolismo , Testosterona/metabolismo , Células Tumorales Cultivadas
12.
J Mol Neurosci ; 48(3): 696-705, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22116741

RESUMEN

Somatostatin (SST)-positive medium-sized aspiny interneurons are selectively spared in excitotoxicity. The biological effects of SST are mediated via five different receptors, namely somatostatin receptor (SSTR)1-5; however, SSTR subtype spared in excitotoxicity and involved in neuroprotection is not known. Dopamine- and cAMP-regulated phosphoprotein (DARPP-32) is predominantly expressed in medium-sized projection neurons that are most vulnerable in excitotoxicity. In the present study, we determined the colocalization of SST and SSTRs with DARPP-32 in rat brain cortical and striatal regions using immunofluorescence immunohistochemistry. We also determined the expression of DARPP-32 in SSTR1-5 immunoprecipitate prepared from cortex and striatum. SST-positive neurons in cortex and striatum are devoid of colocalization with DARPP-32. However, in cortical and striatal brain regions, three different neuronal populations either expressing SSTRs and DARPP-32 alone or displaying colocalization were identified. Quantitative analysis reveals that in cortex and striatum, SSTR1 and 5 are most predominant receptor subtypes colocalized with DARPP-32 followed by SSTR4, 2, and 3 in cortex whereas SSTR2, 4, and 3 in striatum. Importantly, DARPP-32 is expressed in SSTR1-5 immunoprecipitate prepared from cortex and striatum. Taken together, these results provide the first evidence that the SSTR-positive neurons lacking colocalization with DARPP-32 might be spared in excitotoxicity.


Asunto(s)
Corteza Cerebral/química , Cuerpo Estriado/química , Fosfoproteína 32 Regulada por Dopamina y AMPc/análisis , Neuronas/química , Receptores de Somatostatina/análisis , Animales , Corteza Cerebral/ultraestructura , Cuerpo Estriado/ultraestructura , Técnica del Anticuerpo Fluorescente Indirecta , Inmunoprecipitación , Masculino , Neurotoxinas/toxicidad , Especificidad de Órganos , Ratas , Ratas Sprague-Dawley , Receptores de Somatostatina/clasificación , Somatostatina/análisis
13.
PLoS One ; 6(9): e24467, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21912697

RESUMEN

BACKGROUND: Selective degeneration of medium spiny neurons and preservation of medium sized aspiny interneurons in striatum has been implicated in excitotoxicity and pathophysiology of Huntington's disease (HD). However, the molecular mechanism for the selective sparing of medium sized aspiny neurons and vulnerability of projection neurons is still elusive. The pathological characteristic of HD is an extensive reduction of the striatal mass, affecting caudate putamen. Somatostatin (SST) positive neurons are selectively spared in HD and Quinolinic acid/N-methyl-D-aspartic acid induced excitotoxicity, mimic the model of HD. SST plays neuroprotective role in excitotoxicity and the biological effects of SST are mediated by five somatostatin receptor subtypes (SSTR1-5). METHODS AND FINDINGS: To delineate subtype selective biological responses we have here investigated changes in SSTR1 and 5 double knockout mice brain and compared with HD transgenic mouse model (R6/2). Our study revealed significant loss of dopamine and cAMP regulated phosphoprotein of 32 kDa (DARPP-32) and comparable changes in SST, N-methyl-D-aspartic acid receptors subtypes, calbindin and brain nitric oxide synthase expression as well as in key signaling proteins including calpain, phospho-extracellular-signal-regulated kinases1/2, synapsin-IIa, protein kinase C-α and calcineurin in SSTR1/5(-/-) and R6/2 mice. Conversely, the expression of somatostatin receptor subtypes, enkephalin and phosphatidylinositol 3-kinases were strain specific. SSTR1/5 appears to be important in regulating NMDARs, DARPP-32 and signaling molecules in similar fashion as seen in HD transgenic mice. CONCLUSIONS: This is the first comprehensive description of disease related changes upon ablation of G- protein coupled receptor gene. Our results indicate that SST and SSTRs might play an important role in regulation of neurodegeneration and targeting this pathway can provide a novel insight in understanding the pathophysiology of Huntington's disease.


Asunto(s)
Encéfalo/metabolismo , Técnicas de Inactivación de Genes , Enfermedad de Huntington/metabolismo , Receptores de Somatostatina/deficiencia , Receptores de Somatostatina/genética , Animales , Encéfalo/patología , Calbindinas , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Regulación de la Expresión Génica , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Transgénicos , Neostriado/metabolismo , Neuronas/metabolismo , Neuronas/patología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Transducción de Señal/genética
14.
Endocrinology ; 152(3): 931-45, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21190959

RESUMEN

Epidermal growth factor through the stimulation of epidermal growth factor receptor (EGFR) plays a critical role in the activation of MAPKs and phosphatidylinositol-3-protein kinase/AKT cell survival pathways attributed in many pathological conditions. At the cellular level, such functions involve EGFR overactivation and phosphorylation. In the present study, we describe that human embryonic kidney-293 cells transfected with somatostatin (SST) receptor 5 (SSTR5) exhibit inhibition of EGFR phosphorylation and modulate MAPK and phosphatidylinositol-3-protein kinase/AKT cell survival signaling. Furthermore, suppression of EGFR by using small interference RNA and an antagonist (AG1478) potentiates the SST effect via activation of SSTR5 on signaling molecules. In wild-type human embryonic kidney-293 cells, EGFR/ErbB2 exists as constitutive heterodimers. The presence of SSTR5 leads to the dissociation of the heteromeric complex of EGFR/ErbB2 and display preferential heterodimerization between SSTR5 and EGFR in an agonist-dependent manner. These findings highlight a new undiscovered mechanism and potential role of SSTR5 to attenuate the EGFR-mediated signaling pathways involved in tumorigenesis. Our data indicate that the activation and/or overexpression of SST receptors along with the inhibition of EGFR will serve as an important therapeutic approach in the treatment of ErbB-positive tumors.


Asunto(s)
Receptores ErbB/metabolismo , Receptor ErbB-2/metabolismo , Receptores de Somatostatina/metabolismo , Transducción de Señal/fisiología , Apoptosis , Línea Celular , Proliferación Celular , Receptores ErbB/química , Receptores ErbB/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-2/química , Receptor ErbB-2/genética , Receptores de Somatostatina/genética
15.
Cell Signal ; 21(3): 428-39, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19070659

RESUMEN

The role of somatostatin (SST) and epidermal growth factor (EGF) in breast cancer is undisputed; however, the molecular mechanisms underlying their antiproliferative or proliferative effects are not well understood. We initially confirmed that breast tumour tissues express all five somatostatin receptors (SSTR1-5) and four epidermal growth factor receptors (ErbB1-4). Subsequently, to gain insight into the function of SSTRs and ErbBs in oestrogen receptor (ER)-positive (MCF-7) or ERalpha-negative (MDA-MB-231) breast cancer cells, we defined SSTR1, SSTR5 and ErbB1 mRNA and protein expression in these two tumour cell lines. Consistent with previous studies showing SSTR1/SSTR5 heterodimerization and having seen cell-specific and ligand-selective alterations in receptor expression, we next elucidated whether SSTR1 and SSTR5 functionally interact with ErbB1 using pbFRET analysis. We subsequently determined the effects of SST and EGF either alone, or in combination, on selected downstream signalling molecules such as erk1/2, p38 and JNK. Here, we showed that both SST and EGF influenced erk1/2 phosphorylation and that SST modulated the effects of EGF in a cell-specific manner. We also demonstrated agonist-, time and cell-dependent regulation of p38 phosphorylation. We further investigated modulation of Grb2, SOS, Shc, SH-PTP1 and SH-PTP2. ErbB1 adaptor proteins known to play a role in MAPK activation, Shc, Grb2 and SOS, changed in an agonist- and cell-specific manner whereas, SH-PTP1 and SH-PTP2, adaptor proteins reported to interact with SSTRs, translocated from the cytosol to membrane in a cell-specific manner following SST and/or EGF treatment. Although several previous studies have shown crosstalk between RTKs and GPCRs, there are no reports describing SSTR (GPCR) modulation of ErbBs (RTK) in breast cancer. To the best of our knowledge, this is the first report describing crosstalk/interactions between SSTRs and ErbBs.


Asunto(s)
Receptores ErbB/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Receptores de Somatostatina/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Receptores ErbB/efectos de los fármacos , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Plaquinas/efectos de los fármacos , Plaquinas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Receptor Cross-Talk/efectos de los fármacos , Receptor Cross-Talk/fisiología , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Somatostatina/efectos de los fármacos
16.
Cell Signal ; 21(9): 1396-414, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19426801

RESUMEN

Somatostatin receptors show great diversity in response to agonist mediated receptor-specific homo- and heterodimerizations. Here, using photobleaching-fluorescence resonance energy transfer, immunocytochemistry, western blot and co-immunoprecipitation, we investigated dimerization, trafficking, coupling to adenylyl cyclase and signaling of human somatostatin receptor-4 (hSSTR4) in HEK-293 cells. We also determined the role of the C-tail of hSSTR4 on physiological responses of the cells. wt-hSSTR4 exogenously expressed in HEK-293 cells exhibits constitutive dimerization, inhibits forskolin-stimulated cAMP, and displays agonist dependent changes in pERK1/2 and pERK5 expressions. Upon C-tail deletion, the receptor loses membrane expression and ability to dimerize and inhibition of cAMP and pERK5 however, displays several-fold increases in the expression of pERK1/2. Chimeric hSSTR4 with the C-tail of hSSTR5 functions like wt-hSSTR4, in contrast, with the C-tail of hSSTR1 functions like C-tail deleted hSSTR4. hSSTR4 dimerization and signaling are associated with increased cyclin-dependent-kinase p27(kip1) expression and inhibition of the cell proliferation. We also report heterodimerization between hSSTR4/hSSTR5, but not between hSSTR4/hSSTR1, with significant changes in receptor functions. Taken together, these data define a novel mechanism for the role of hSSTR4 in cell proliferation and modulation of signaling pathways.


Asunto(s)
Receptores de Somatostatina/metabolismo , Transducción de Señal , Línea Celular , Proliferación Celular , Dimerización , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Humanos , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Estructura Terciaria de Proteína , Receptores de Somatostatina/química , Receptores de Somatostatina/fisiología
17.
Neurosci Res ; 65(1): 53-63, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19465068

RESUMEN

In the present study using indirect immunofluorescence immunohistochemistry, co-immunoprecipitation and western blot analysis we determined the colocalization of dopamine receptors 1-5 and dopamine and cAMP-regulated phosphoprotein (DARPP-32) in rat brain cortex and striatum. All five DR subtypes and DARPP-32 were expressed in rat brain cortex and striatum. DARPP-32 positive neurons displayed comparative colocalization with DR1-5. In cingulate cortex, the colocalization of DR subtypes was greatly different from frontal or temporal cortex. D1R is one of the most predominant subtypes which colocalized with DARPP-32 in cortex as well as striatum and followed by D2R, D3R, D4R and D5R. Amongst all DR subtypes D5R was coexpressed the least with DARPP-32 positive neurons. Consistent with immunohistochemical data, western blot analysis also reveals comparable distribution of DR subtypes and DARPP-32 in cortex and striatum. Colocalization studies were also supported by using co-immunoprecipitate assay displaying DARPP-32 expression in DR immunoprecipitate from tissue lysate prepared from cortex and striatum. Taken together our data support receptor specific association of DARPP-32 with DR subtypes that might shed new information in drugs of abuse and pathophysiology of neurodegenerative diseases as well as neuropsychiatric disorders such as schizophrenia.


Asunto(s)
Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Receptores Dopaminérgicos/metabolismo , Animales , Western Blotting , Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Técnica del Anticuerpo Fluorescente , Giro del Cíngulo/metabolismo , Masculino , Microscopía Confocal , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
18.
J Chem Neuroanat ; 38(1): 20-33, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19465111

RESUMEN

Apolipoprotein D (ApoD) is widely distributed in central and peripheral nervous system. ApoD expression has been shown to increase in several neurodegenerative and neuropsychiatric disorders, as well as during regeneration in the nervous system. Like ApoD, in the central nervous system somatostatin (SST) is widely present and functions as neurotransmitter and neuromodulator. The biological effects of SST are mediated via binding to five high-affinity G-protein coupled receptors termed SSTR1-5. Mice lacking ApoD exhibit reduced SST labeling in cortex and hippocampus and increased expression in striatum and amygdala without any noticeable changes in substantia nigra. Changes in SSTRs expressions have been described in several neurodegenerative disorders including Alzheimer's, Parkinson's and Huntington's diseases. In the present study, using SSTR1-5 receptor-specific antibodies, we mapped their distribution in wild type (wt) and ApoD knockout (ApoD(-/-)) mouse brain. SSTR1-5 expression was observed both as membrane and cytoplasmic protein and display regions and receptor specific differences between wt and ApoD(-/-) mice brains. In cortex and hippocampus, SSTR subtypes like immunoreactivity are decreased in ApoD(-/-) mice brain. Unlike cortex and hippocampus, in the striatum of ApoD(-/-) mice, projection neurons showed increased SSTR immunoreactivity, as compared to wt. Higher SSTR subtypes immunoreactivity is seen in substantia nigra pars compacta (SNpc) whereas lower in substantia nigra pars reticulata (SNpr) of ApoD(-/-) mice brains as compared to wt. Whereas, amygdala displayed SSTR subtypes changes in different nuclei of ApoD(-/-) mice in comparison to wt mice brain. Taken together, our results describe receptor and region specific changes in SST and SSTR subtypes expression in ApoD(-/-) mice brain, which may be linked to specific neurological disorders.


Asunto(s)
Apolipoproteínas D/genética , Química Encefálica/fisiología , Encéfalo/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/ultraestructura , Animales , Encéfalo/citología , Mapeo Encefálico , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Corteza Cerebral/metabolismo , Corteza Cerebral/ultraestructura , Cuerpo Estriado/metabolismo , Cuerpo Estriado/ultraestructura , Citoplasma/metabolismo , Citoplasma/ultraestructura , Regulación hacia Abajo/fisiología , Hipocampo/metabolismo , Hipocampo/ultraestructura , Inmunohistoquímica , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/ultraestructura , Sustancia Negra/metabolismo , Sustancia Negra/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA