Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Oral Pathol Med ; 48(5): 389-399, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30825343

RESUMEN

BACKGROUND: Oral squamous cell carcinoma (OSCC) is a deadly disease with a mere 40% five-year survival rate for patients with advanced disease. Previously, we discovered that capsazepine (CPZ), a transient receptor potential channel, Vanilloid subtype 1 (TRPV1) antagonist, has significant anti-tumor effects against OSCC via a unique mechanism-of-action that is independent of TRPV1. Thus, we developed novel CPZ analogs with more potent anti-proliferative effects (CIDD-24, CIDD-99, and CIDD-111). METHODS: Using OSCC xenograft models, we determined the efficacy of these analogs in vivo. TRPV1 interactions were evaluated using calcium imaging and a rat model of orofacial pain. Anti-cancer mechanism(s)-of-action were assessed by cell cycle analysis and mitochondrial depolarization assays. RESULTS: CIDD-99 was the most potent analog demonstrating significant anti-tumor effects in vivo (P < 0.001). CIDD-24 was equipotent to the parent compound CPZ, but less potent than CIDD-99. CIDD-111 was the least efficacious analog. Calcium imaging studies confirmed that CIDD-99 neither activates nor inhibits TRPV1 confirming that TRPV1 activity is not involved in its anti-cancer effects. All analogs induced an S-phase block, dose-dependent mitochondrial depolarization, and apoptosis. Histological analyses revealed increased apoptosis and reduced cell proliferation in tumors treated with these analogs. Importantly, CIDD-99 had the most dramatic anti-tumor effects with 85% of tumors resolving leaving only minute traces of viable tissue. Additionally, CIDD-99 was non-noxious and demonstrated no observable adverse reactions CONCLUSION: This study describes a novel, highly efficacious, CPZ analog, CIDD-99, with dramatic anti-tumor effects against OSCC that may be efficacious as a lone therapy or in combination with standard therapies.


Asunto(s)
Apoptosis , Capsaicina/análogos & derivados , Carcinoma de Células Escamosas/tratamiento farmacológico , Estrés del Retículo Endoplásmico , Isoquinolinas/farmacología , Mitocondrias/efectos de los fármacos , Neoplasias de la Boca/tratamiento farmacológico , Canales Catiónicos TRPV/antagonistas & inhibidores , Animales , Capsaicina/farmacología , Línea Celular Tumoral , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Mitocondrias/patología , Ratas , Ratas Sprague-Dawley , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Am Soc Nephrol ; 27(11): 3356-3367, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27000065

RESUMEN

During recovery by regeneration after AKI, proximal tubule cells can fail to redifferentiate, undergo premature growth arrest, and become atrophic. The atrophic tubules display pathologically persistent signaling increases that trigger production of profibrotic peptides, proliferation of interstitial fibroblasts, and fibrosis. We studied proximal tubules after ischemia-reperfusion injury (IRI) to characterize possible mitochondrial pathologies and alterations of critical enzymes that govern energy metabolism. In rat kidneys, tubules undergoing atrophy late after IRI but not normally recovering tubules showed greatly reduced mitochondrial number, with rounded profiles, and large autophagolysosomes. Studies after IRI of kidneys in mice, done in parallel, showed large scale loss of the oxidant-sensitive mitochondrial protein Mpv17L. Renal expression of hypoxia markers also increased after IRI. During early and late reperfusion after IRI, kidneys exhibited increased lactate and pyruvate content and hexokinase activity, which are indicators of glycolysis. Furthermore, normally regenerating tubules as well as tubules undergoing atrophy exhibited increased glycolytic enzyme expression and inhibitory phosphorylation of pyruvate dehydrogenase. TGF-ß antagonism prevented these effects. Our data show that the metabolic switch occurred early during regeneration after injury and was reversed during normal tubule recovery but persisted and became progressively more severe in tubule cells that failed to redifferentiate. In conclusion, irreversibility of the metabolic switch, taking place in the context of hypoxia, high TGF-ß signaling and depletion of mitochondria characterizes the development of atrophy in proximal tubule cells and may contribute to the renal pathology after AKI.


Asunto(s)
Lesión Renal Aguda/complicaciones , Glucólisis , Isquemia/complicaciones , Túbulos Renales Proximales/patología , Riñón/irrigación sanguínea , Mitocondrias/metabolismo , Enfermedades Mitocondriales/etiología , Animales , Atrofia/etiología , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley
3.
Am J Pathol ; 181(4): 1236-49, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22885106

RESUMEN

After ischemia-reperfusion injury (IRI), kidney tubules show activated transforming growth factor ß (TGF-ß) signaling and increased expression of profibrotic peptides, platelet-derived growth factor-B (PDGF-B) and connective tissue growth factor (CTGF). If tubule repair after IRI is incomplete, sustained paracrine activity of these peptides can activate interstitial fibroblast progenitors and cause fibrosis. We show that lysophosphatidic acid (LPA), a ubiquitous phospholipid that is increased at sites of injury and inflammation, signals through LPA2 receptors and Gαq proteins of cultured proximal tubule cells to transactivate latent TGF-ß in a Rho/Rho-kinase and αvß6 integrin-dependent manner. Active TGF-ß peptide then initiates signaling to increase the production and secretion of PDGF-B and CTGF. In a rat model of IRI, increased TGF-ß signaling that was initiated early during reperfusion did not subside during recovery, but progressively increased, causing tubulointerstitial fibrosis. This was accompanied by correspondingly increased LPA2 and ß6 integrin proteins and elevated tubule expression of TGF-ß1, together with PDGF-B and CTGF. Treatment with a pharmacological TGF-ß type I receptor antagonist suppressed TGF-ß signaling, decreased the expression of ß6 integrin, PDGF-B, and CTGF, and ameliorated fibrosis. We suggest that LPA-initiated autocrine signaling is a potentially important mechanism that gives rise to paracrine profibrotic signaling in injured kidney tubule cells.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Citocinas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Integrinas/metabolismo , Túbulos Renales Proximales/metabolismo , Lisofosfolípidos/farmacología , Receptores del Ácido Lisofosfatídico/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Citocinas/genética , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Lípidos/sangre , Masculino , Ratones , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Regeneración/efectos de los fármacos , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta/metabolismo
4.
Am J Physiol Renal Physiol ; 302(9): F1210-23, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22301622

RESUMEN

We investigated the signaling basis for tubule pathology during fibrosis after renal injury. Numerous signaling pathways are activated physiologically to direct tubule regeneration after acute kidney injury (AKI) but several persist pathologically after repair. Among these, transforming growth factor (TGF)-ß is particularly important because it controls epithelial differentiation and profibrotic cytokine production. We found that increased TGF-ß signaling after AKI is accompanied by PTEN loss from proximal tubules (PT). With time, subpopulations of regenerating PT with persistent loss of PTEN (phosphate and tension homolog) failed to differentiate, became growth arrested, expressed vimentin, displayed profibrotic JNK activation, and produced PDGF-B. These tubules were surrounded by fibrosis. In contrast, PTEN recovery was associated with epithelial differentiation, normal tubule repair, and less fibrosis. This beneficial outcome was promoted by TGF-ß antagonism. Tubule-specific induction of TGF-ß led to PTEN loss, JNK activation, and fibrosis even without prior AKI. In PT culture, high TGF-ß depleted PTEN, inhibited differentiation, and activated JNK. Conversely, TGF-ß antagonism increased PTEN, promoted differentiation, and decreased JNK activity. Cre-Lox PTEN deletion suppressed differentiation, induced growth arrest, and activated JNK. The low-PTEN state with JNK signaling and fibrosis was ameliorated by contralateral nephrectomy done 2 wk after unilateral ischemia, suggesting reversibility of the low-PTEN dysfunctional tubule phenotype. Vimentin-expressing tubules with low-PTEN and JNK activation were associated with fibrosis also after tubule-selective AKI, and with human chronic kidney diseases of diverse etiology. By preventing tubule differentiation, the low-PTEN state may provide a platform for signals initiated physiologically to persist pathologically and cause fibrosis after injury.


Asunto(s)
Diferenciación Celular , Túbulos Renales Proximales/patología , MAP Quinasa Quinasa 4/fisiología , Fosfohidrolasa PTEN/deficiencia , Fenotipo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Células Cultivadas , Enfermedad Crónica , Fibrosis , Humanos , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Túbulos Renales Proximales/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Modelos Animales , Ratas , Ratas Sprague-Dawley , Regeneración/fisiología , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología
5.
Biochem Biophys Res Commun ; 399(4): 548-54, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20678484

RESUMEN

We showed earlier that 15 deoxy Delta(12,14) prostaglandin J2 (15d-PGJ2) inactivates Drp1 and induces mitochondrial fusion [1]. However, prolonged incubation of cells with 15d-PGJ2 resulted in remodeling of fused mitochondria into large swollen mitochondria with irregular cristae structure. While initial fusion of mitochondria by 15d-PGJ2 required the presence of both outer (Mfn1 and Mfn2) and inner (OPA1) mitochondrial membrane fusion proteins, later mitochondrial changes involved increased degradation of the fusion protein OPA1 and ubiquitination of newly synthesized OPA1 along with decreased expression of Mfn1 and Mfn2, which likely contributed to the loss of tubular rigidity, disorganization of cristae, and formation of large swollen degenerated dysfunctional mitochondria. Similar to inhibition of Drp1 by 15d-PGJ2, decreased expression of fission protein Drp1 by siRNA also resulted in the loss of fusion proteins. Prevention of 15d-PGJ2 induced mitochondrial elongation by thiol antioxidants prevented not only loss of OPA1 isoforms but also its ubiquitination. These findings provide novel insights into unforeseen complexity of molecular events that modulate mitochondrial plasticity.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Mitocondrias/efectos de los fármacos , Dilatación Mitocondrial/efectos de los fármacos , Prostaglandina D2/análogos & derivados , Animales , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Proteínas Quinasas Asociadas a Muerte Celular , GTP Fosfohidrolasas/genética , Ratones , Ratones Noqueados , Mitocondrias/enzimología , Mitocondrias/genética , Prostaglandina D2/farmacología , Ratas , Ubiquitinación
6.
Biochem Biophys Res Commun ; 395(1): 17-24, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20307494

RESUMEN

Arachidonic acid derived endogenous electrophile 15d-PGJ2 has gained much attention in recent years due to its potent anti-proliferative and anti-inflammatory actions mediated through thiol modification of cysteine residues in its target proteins. Here, we show that 15d-PGJ2 at 1 microM concentration converts normal mitochondria into large elongated and interconnected mitochondria through direct binding to mitochondrial fission protein Drp1 and partial inhibition of its GTPase activity. Mitochondrial elongation induced by 15d-PGJ2 is accompanied by increased assembly of Drp1 into large oligomeric complexes through plausible intermolecular interactions. The role of decreased GTPase activity of Drp1 in the formation of large oligomeric complexes is evident when Drp1 is incubated with a non-cleavable GTP analog, GTPgammaS or by a mutation that inactivated GTPase activity of Drp1 (K38A). The mutation of cysteine residue (Cys644) in the GTPase effector domain, a reported target for modification by reactive electrophiles, to alanine mimicked K38A mutation induced Drp1 oligomerization and mitochondrial elongation, suggesting the importance of cysteine in GED to regulate the GTPase activity and mitochondrial morphology. Interestingly, treatment of K38A and C644A mutants with 15d-PGJ2 resulted in super oligomerization of both mutant Drp1s indicating that 15d-PGJ2 may further stabilize Drp1 oligomers formed by loss of GTPase activity through covalent modification of middle domain cysteine residues. The present study documents for the first time the regulation of a mitochondrial fission activity by a prostaglandin, which will provide clues for understanding the pathological and physiological consequences of accumulation of reactive electrophiles during oxidative stress, inflammation and degeneration.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Prostaglandina D2/análogos & derivados , Animales , Línea Celular , Cisteína/genética , Dinaminas/genética , Dinaminas/metabolismo , GTP Fosfohidrolasas/antagonistas & inhibidores , GTP Fosfohidrolasas/genética , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias/fisiología , Proteínas Mitocondriales/genética , Mutación , Prostaglandina D2/farmacología , Estructura Terciaria de Proteína/genética , Ratas
7.
Genes Cancer ; 10(5-6): 134-149, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31798766

RESUMEN

Screening of several TNBC cell lines showed altered Smad2 and Smad3 protein levels compared to normal mammary epithelial cells, suggesting the possibility that it could play an important role in the escape of cancer cells from TGF-ß mediated growth inhibition. To assess the functional relevance of these endogenous molecules, Smad2 or Smad3 expression was knocked down individually and assessed their effects on pro-oncogenic properties of TGF-ß. Smad3 deficiency reduced growth and invasion capacity of breast cancer cells in comparison to Smad2 which had no effect. Smad3 deficiency was also found to be associated with a reduction in the expressions of TMEPAI/PMEPA1 and EMT inducing transcription factors, E-Cadherin and increased expression of cell cycle inhibitors and Vimentin. On the other hand, Smad2 deficiency had opposite effect on these regulators. Interestingly, the decreased growth, invasion and associated gene expressions were largely reversed by overexpressing TMEPAI in Smad3 knockdown cells, suggesting that Smad3-TMEPAI axis may be involved in subverting growth suppressive effects of TGF-ß into growth promotion. Similarly, altered levels of Smad proteins and TMEPAI were also noted in primary TNBC tumor tissues. Analysis of the existing databases provided additional support in terms of TMEPAI and Smad2 expression impacting the survival of TNBC patients. Taken together, our data demonstrate a novel role for Smad3 in cancer transformation and cancer progression through TMEPAI and further suggest that selective targeting of TGF-ß-Smad3-TMEPAI axis may be beneficial in triple negative breast cancer therapy and prevention.

8.
J Ethnopharmacol ; 111(1): 13-21, 2007 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-17127022

RESUMEN

To find out the active principles against ethanol-induced toxicity in mice, Andrographis paniculata Nees. (Ap) was chosen and isolated andrographolide (ANDRO) and arabinogalactan proteins (AGPs). ANDRO was detected by HPTLC, FTIR and quantified by HPLC (10mg/g of Ap powder). AGPs was detected by beta-glucosyl Yariv staining of SDS-PAGE gel, FTIR and quantified by single radial gel diffusion assay with beta-glucosyl Yariv reagent (0.5mg/g Ap powder). The mice are pretreated intra-peritoneally (i.p.) with different doses (62.5, 125, 250, and 500mg/kg) of body weight of mice] of ANDRO and AGPs for 7 days and then ethanol (7.5g/kg of body weight) was injected, i.p. Besides, silymarin was used as standard hepatoprotective agent for comparative study with ANDRO and AGPs. The ameliorative activity of ANDRO and AGP against hepatic renal alcohol toxicity was measured by assessing GOT, GPT, ACP, ALP and LP levels in liver and kidney. It has been observed that pretreatment of mice with ANDRO and AGPs at 500mg/kg of body weight and 125mg/kg of body weight respectively could able to minimize the toxicity in compare to ethanol treated group as revealed by the different enzymatic assay in liver and kidney tissues and the results were comparable with silymarin. Hence, out of several ill-defined compounds present in Ap, ANDRO and AGPs are the potential bioactive compounds responsible for protection against ethanol-induced toxicity.


Asunto(s)
Andrographis , Diterpenos/farmacología , Galactanos/farmacología , Enfermedades Renales/prevención & control , Hepatopatías Alcohólicas/prevención & control , Sustancias Protectoras/farmacología , Fosfatasa Ácida/metabolismo , Fosfatasa Alcalina/metabolismo , Andrographis/química , Animales , Aspartato Aminotransferasas/metabolismo , Depresores del Sistema Nervioso Central , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Diterpenos/aislamiento & purificación , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Etanol , Galactanos/aislamiento & purificación , Glucósidos , India , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Hepatopatías Alcohólicas/etiología , Hepatopatías Alcohólicas/metabolismo , Masculino , Ratones , Floroglucinol/análogos & derivados , Sustancias Protectoras/aislamiento & purificación , Silimarina/farmacología , Espectroscopía Infrarroja por Transformada de Fourier
9.
Oral Oncol ; 59: 12-19, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27424178

RESUMEN

Squamous cell carcinoma (SCC) comprises 90% of all head and neck cancers and has a poor survival rate due to late-stage disease that is refractive to traditional therapies. Epidermal growth factor receptor (EGFR) is over-expressed in greater than 80% of head and neck SCC (HNSCC). However, EGFR targeted therapies yielded little to no efficacy in clinical trials. This study investigated the efficacy of co-targeting EGFR and the anaplastic lymphoma kinase (ALK) whose promoter is hypomethylated in late-stage oral SCC (OSCC). We observed increased ALK activity in late-stage human OSCC tumors and invasive OSCC cell lines. We also found that while ALK inhibition alone had little effect on proliferation, co-targeting ALK and EGFR significantly reduced OSCC cell proliferation in vitro. Further analysis showed significant efficacy of combined treatment in HSC3-derived xenografts resulting in a 30% decrease in tumor volumes by 14days (p<0.001). Western blot analysis showed that co-targeting ALK and EGFR significantly reduced EGFR phosphorylation (Y1148) in HSC3 cells but not Cal27 cells. ALK and EGFR downstream signaling interactions are also demonstrated by Western blot analysis in which lone EGFR and ALK inhibitors attenuated AKT activity whereas co-targeting ALK and EGFR completely abolished AKT activation. No effects were observed on ERK1/2 activation. STAT3 activity was significantly induced by lone ALK inhibition in HSC3 cells and to a lower extent in Cal27 cells. Together, these data illustrate that ALK inhibitors enhance anti-tumor activity of EGFR inhibitors in susceptible tumors that display increased ALK expression, most likely through abolition of AKT activation.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Receptores ErbB/metabolismo , Neoplasias de la Boca/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Quinasa de Linfoma Anaplásico , Animales , Línea Celular Tumoral , Femenino , Gefitinib , Humanos , Ratones Desnudos , Quinazolinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Fitoterapia ; 74(7-8): 692-4, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14630176

RESUMEN

The antimicrobial activity of aqueous extract, andrographolides and arabinogalactan proteins from Andrographis paniculata were evaluated. The aqueous extract showed significant antimicrobial activity, which may be due to the combined effect of the isolated arabinogalactan proteins and andrographolides.


Asunto(s)
Andrographis , Antiinfecciosos/farmacología , Bacterias/efectos de los fármacos , Fitoterapia , Extractos Vegetales/farmacología , Antiinfecciosos/administración & dosificación , Antiinfecciosos/uso terapéutico , Candida albicans/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Extractos Vegetales/administración & dosificación , Extractos Vegetales/uso terapéutico , Proteínas de Plantas
11.
Genes Cancer ; 5(9-10): 320-36, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25352949

RESUMEN

TMEPAI (transmembrane prostate androgen-induced) is amplified at genomic, transcript and protein levels in triple-negative breast cancers and promotes TGF-ß dependent growth, motility and invasion. Tumor promotion by TMEPAI depends on two different but related actions on TGF-ß signaling. Firstly, TMEPAI binds and sequesters regulatory Smads2/3 and thereby decreases growth suppressive signaling by TGF-ß. Secondly, increased expression of TMEPAI decreases PTEN (phosphatase and tensin homolog) abundance, and thereby increases TGF-ß dependent tumor promotive PI3K/Akt signaling. These actions of TMEPAI give rise to increased cell proliferation and motility. Moreover, signaling alterations produced by high TMEPAI were associated with oncogenic Snail expression and lung metastases. Finally, an inverse correlation between TMEPAI and PTEN levels was confirmed in triple negative breast cancer tumor samples. Together, our findings suggest that TMEPAI has dually critical roles to promote TGF-ß dependent cancer cell growth and metastasis. Thus, redirected TGF-ß signaling through TMEPAI may play a pivotal role in TGF-ß mediated tumor promotion.

12.
Cancer Res ; 70(15): 6377-83, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20610632

RESUMEN

TMEPAI is a transforming growth factor-beta (TGF-beta)-induced transmembrane protein that is overexpressed in several cancers. How TMEPAI expression relates to malignancy is unknown. Here, we report high expression of TMEPAI in estrogen receptor/progesterone receptor-negative and human epidermal growth factor receptor-2-negative breast cancer cell lines and primary breast cancers that was further increased by TGF-beta treatment. Basal and TGF-beta-induced expression of TMEPAI were inhibited by the TGF-beta receptor antagonist SB431542 and overexpression of Smad7 or a dominant-negative mutant of Alk-5. TMEPAI knockdown attenuated TGF-beta-induced growth and motility in breast cancer cells, suggesting a role for TMEPAI in growth promotion and invasiveness. Further, TMEPAI knockdown decreased breast tumor mass in a mouse xenograft model in a manner associated with increased expression of phosphatase and tensin homologue (PTEN) and diminished phosphorylation of Akt. Consistent with the effects through the phosphatidylinositol 3-kinase pathway, tumors with TMEPAI knockdown exhibited elevated levels of the cell cycle inhibitor p27kip1 and attenuated levels of DNA replication and expression of hypoxia-inducible fator 1alpha and vascular endothelial growth factor. Together, these results suggest that TMEPAI functions in breast cancer as a molecular switch that converts TGF-beta from a tumor suppressor to a tumor promoter.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Proteínas de la Membrana/biosíntesis , Factor de Crecimiento Transformador beta/farmacología , Animales , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Procesos de Crecimiento Celular/genética , Movimiento Celular/genética , Femenino , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , ARN Interferente Pequeño/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
13.
J Biol Chem ; 281(29): 20160-70, 2006 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-16707491

RESUMEN

Bone remodeling depends upon proper osteoblast and osteoclast function. Bone morphogenetic protein-2 (BMP-2) stimulates differentiation of osteoblasts from pluripotent precursors. Osteoclast formation depends on the concerted action of osteoblast-derived receptor activator of NF-kappaB ligand and colony-stimulating factor-1 (CSF-1). BMP-2 stimulates receptor activator of NF-kappaB ligand expression. However, the effect of BMP-2 on CSF-1 expression has not been studied. We investigated the role of BMP-2 in CSF-1 expression in osteogenic C2C12 cells. Incubation of C2C12 cells with BMP-2 supported osteoclastogenesis of spleen cells with a concomitant increase in expression of CSF-1 mRNA and protein. To determine the mechanism, we identified a BMP-responsive element between -627 bp and -509 bp in the CSF-1 promoter. DNase I footprint analysis revealed the presence of consensus Smad binding motif in this region. Electrophoretic mobility shift assay showed BMP-2-stimulated binding of proteins to this motif. Mutation of core sequence as well as its 5'- and 3'-flanking sequences abolished the DNA-protein interaction resulting in inhibition of CSF-1 transcription. Supershift analysis detects the presence of Smads 1, 5, and 4 and the transcriptional coactivator CREB-binding protein in the BMP-responsive element-protein complex. In addition, Smads 1 and 5 alone or in combination with Smad 4 increased CSF-1 transcription. Furthermore, CREB-binding protein markedly increased transcription of CSF-1. These data represent the first evidence that BMP-2 increases the osteoclastogenic CSF-1 expression by a transcriptional mechanism using the canonical Smad pathway and provide a mechanism for BMP-2-induced osteoclast differentiation.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Proteína de Unión a CREB/metabolismo , Factor Estimulante de Colonias de Macrófagos/genética , Osteoclastos/citología , Osteoclastos/fisiología , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , Proteína Morfogenética Ósea 2 , Diferenciación Celular/efectos de los fármacos , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Células Musculares/efectos de los fármacos , Células Musculares/fisiología , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Osteoclastos/efectos de los fármacos , Regiones Promotoras Genéticas , Proteínas Recombinantes/farmacología , Transducción de Señal , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA