Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Ann Oncol ; 28(8): 1988-1995, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28595336

RESUMEN

BACKGROUND: Surrogate biomarkers of efficacy are needed for anti-PD1/PD-L1 therapy, given the existence of delayed responses and pseudo-progressions. We evaluated changes in serum IL-8 levels as a biomarker of response to anti-PD-1 blockade in melanoma and non-small-cell lung cancer (NSCLC) patients. PATIENTS AND METHODS: Metastatic melanoma and NSCLC patients treated with nivolumab or pembrolizumab alone or nivolumab plus ipilimumab were studied. Serum was collected at baseline; at 2-4 weeks after the first dose; and at the time-points of response evaluation. Serum IL-8 levels were determined by sandwich ELISA. Changes in serum IL-8 levels were compared with the Wilcoxon test and their strength of association with response was assessed with the Mann-Whitney test. Accuracy of changes in IL-8 levels to predict response was estimated using receiver operation characteristics curves. RESULTS: Twenty-nine melanoma patients treated with nivolumab or pembrolizumab were studied. In responding patients, serum IL-8 levels significantly decreased between baseline and best response (P <0.001), and significantly increased upon progression (P =  0.004). In non-responders, IL-8 levels significantly increased between baseline and progression (P =  0.013). Early changes in serum IL-8 levels (2-4 weeks after treatment initiation) were strongly associated with response (P <0.001). These observations were validated in 19 NSCLC patients treated with nivolumab or pembrolizumab (P =  0.001), and in 15 melanoma patients treated with nivolumab plus ipilimumab (P <0.001). Early decreases in serum IL-8 levels were associated with longer overall survival in melanoma (P =  0.001) and NSCLC (P =  0.015) patients. Serum IL-8 levels also correctly reflected true response in three cancer patients presenting pseudoprogression. CONCLUSIONS: Changes in serum IL-8 levels could be used to monitor and predict clinical benefit from immune checkpoint blockade in melanoma and NSCLC patients.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Interleucina-8/sangre , Neoplasias Pulmonares/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Adulto , Antineoplásicos Inmunológicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/sangre , Estudios de Cohortes , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Neoplasias Pulmonares/sangre , Masculino , Melanoma/sangre , Persona de Mediana Edad , Neoplasias Cutáneas/sangre , Análisis de Supervivencia
2.
Oncogene ; 35(40): 5295-5303, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27041569

RESUMEN

Targeted inhibitors of oncogenic Ras (rat sarcoma viral oncogene)-Raf signaling have shown great promise in the clinic, but resistance remains a major challenge: 30% of tumors with pathway mutations do not respond to targeted inhibitors, and of the 70% that do respond, all eventually develop resistance. Before cancer cells acquire resistance, they respond to initial drug treatment either by undergoing apoptosis ('addiction') or by surviving treatment albeit with reduced growth ('tolerance'). As these drug-tolerant cells serve as a reservoir from which resistant cells eventually emerge, inhibiting the pathways that confer tolerance could potentially delay or even prevent recurrence. Here, we show that melanomas and other cancers acquire tolerance to Ras-Raf pathway inhibitors by activating autophagy, which is mediated by the cellular energy sensor AMP-activated protein kinase (AMPK). Blocking this AMPK-mediated autophagy sensitizes drug-tolerant melanomas to Ras-Raf pathway inhibitors. Conversely, activating AMPK signaling and autophagy enables melanomas that would otherwise be addicted to the Ras-Raf pathway to instead tolerate pathway inhibition. These findings identify a key mechanism of tolerance to Ras-Raf pathway inhibitors and suggest that blocking either AMPK or autophagy in combination with these targeted inhibitors could increase tumor regression and decrease the likelihood of eventual recurrence.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Melanoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas ras/genética , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Autofagia/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Tolerancia a Medicamentos/genética , Humanos , Células MCF-7 , Melanoma/genética , Melanoma/patología , Transducción de Señal/efectos de los fármacos
3.
Oncogene ; 30(13): 1542-50, 2011 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-21119596

RESUMEN

MicroRNAs (miRNAs) are small ∼22nt single stranded RNAs that negatively regulate protein expression by binding to partially complementary sequences in the 3' untranslated region (3' UTRs) of target gene messenger RNAs (mRNA). Recently, mutations have been identified in both miRNAs and target genes that disrupt regulatory relationships, contribute to oncogenesis and serve as biomarkers for cancer risk. KIT, an established oncogene with a multifaceted role in melanogenesis and melanoma pathogenesis, has recently been shown to be upregulated in some melanomas, and is also a target of the miRNA miR-221. Here, we describe a genetic variant in the 3' UTR of the KIT oncogene that correlates with a greater than fourfold increased risk of acral melanoma. This KIT variant results in a mismatch in the seed region of a miR-221 complementary site and reporter data suggests that this mismatch can result in increased expression of the KIT oncogene. Consistent with the hypothesis that this is a functional variant, KIT mRNA and protein levels are both increased in the majority of samples harboring the KIT variant. This work identifies a novel genetic marker for increased heritable risk of melanoma.


Asunto(s)
Regiones no Traducidas 3'/genética , Melanoma/genética , MicroARNs/fisiología , Oncogenes , Proteínas Proto-Oncogénicas c-kit/genética , Neoplasias Cutáneas/genética , Estudios de Casos y Controles , Humanos , Melanoma/etiología , Biosíntesis de Proteínas , ARN Mensajero/análisis , Riesgo , Neoplasias Cutáneas/etiología
4.
J Biol Chem ; 276(15): 11933-8, 2001 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-11124258

RESUMEN

Tyrosinase is essential for pigmentation and is a source of tumor-derived antigenic peptides and cellular immune response. Wild type tyrosinase in melanoma cells and certain albino mutants in untransformed melanocytes are targeted to proteolytic degradation by the 26 S proteasome due to retention of the misfolded protein in the endoplasmic reticulum and its subsequent retranslocation to the cytosol. Here, we demonstrate that the substrates DOPA and tyrosine induced in melanoma cells a transition of misfolded wild type tyrosinase to the native form that is resistant to proteolysis, competent to exit the endoplasmic reticulum, and able to produce melanin. Because the enzymatic activity of tyrosinase is induced by DOPA, we propose that proper folding of the wild type protein, just like mutant forms, is tightly linked to its catalytic state. Loss of pigmentation, therefore, in tyrosinase-positive melanoma cells is a consequence of tumor-induced metabolic changes that suppress tyrosinase activity and DOPA production within these cells.


Asunto(s)
Dihidroxifenilalanina/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Monofenol Monooxigenasa/metabolismo , Tirosina/metabolismo , Células Cultivadas , Humanos , Microscopía Fluorescente , Pliegue de Proteína , Transporte de Proteínas
5.
J Biol Chem ; 276(8): 5924-31, 2001 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-11069924

RESUMEN

Tyrosinase is a type I membrane glycoprotein essential for melanin synthesis. Mutations in tyrosinase lead to albinism due, at least in part, to aberrant retention of the protein in the endoplasmic reticulum and subsequent degradation by the cytosolic ubiquitin-proteasomal pathway. A similar premature degradative fate for wild type tyrosinase also occurs in amelanotic melanoma cells. To understand critical cotranslational events, the glycosylation and rate of translation of tyrosinase was studied in normal melanocytes, melanoma cells, an in vitro cell-free system, and semi-permeabilized cells. Site-directed mutagenesis revealed that all seven N-linked consensus sites are utilized in human tyrosinase. However, glycosylation at Asn-290 (Asn-Gly-Thr-Pro) was suppressed, particularly when translation proceeded rapidly, producing a protein doublet with six or seven N-linked core glycans. The inefficient glycosylation of Asn-290, due to the presence of a proximal Pro, was enhanced in melanoma cells possessing 2-3-fold faster (7.7-10.0 amino acids/s) protein translation rates compared with normal melanocytes (3.5 amino acids/s). Slowing the translation rate with the protein synthesis inhibitor cycloheximide increased the glycosylation efficiency in live cells and in the cell-free system. Therefore, the rate of protein translation can regulate the level of tyrosinase N-linked glycosylation, as well as other potential cotranslational maturation events.


Asunto(s)
Retículo Endoplásmico/metabolismo , Glicoproteínas de Membrana/biosíntesis , Monofenol Monooxigenasa/biosíntesis , Biosíntesis de Proteínas , Procesamiento Proteico-Postraduccional , Secuencia de Aminoácidos , Animales , Sistema Libre de Células , Células Cultivadas , Secuencia de Consenso , Glicosilación , Humanos , Melanocitos/metabolismo , Melanoma Experimental/metabolismo , Ratones , Transporte de Proteínas
6.
Proc Natl Acad Sci U S A ; 97(11): 5889-94, 2000 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-10823941

RESUMEN

Tyrosinase is a melanocyte-specific enzyme critical for the synthesis of melanin, a process normally restricted to a post-Golgi compartment termed the melanosome. Loss-of-function mutations in tyrosinase are the cause of oculocutaneous albinism, demonstrating the importance of the enzyme in pigmentation. In the present study, we explored the possibility that trafficking of albino tyrosinase from the endoplasmic reticulum (ER) to the Golgi apparatus and beyond is disrupted. Toward this end, we analyzed the common albino mouse mutation Tyr(C85S), the frequent human albino substitution TYR(T373K), and the temperature-sensitive tyrosinase TYR(R402Q)/Tyr(H402A) found in humans and mice, respectively. Intracellular localization was monitored in albino melanocytes carrying the native mutation, as well as in melanocytes ectopically expressing green fluorescent protein-tagged tyrosinase. Enzymatic characterization of complex glycans and immunofluorescence colocalization with organelle-specific resident proteins established that all four mutations produced defective proteins that were retained in the ER. TYR(R402Q)/Tyr(H402A) Golgi processing and transport to melanosomes were promoted at the permissive temperature of 32 degrees C, but not at the nonpermissive 37 degrees C temperature. Furthermore, evidence of protein misfolding was demonstrated by the prolonged association of tyrosinase mutants with calnexin and calreticulin, known ER chaperones that play a key role in the quality-control processes of the secretory pathway. From these results we concluded that albinism, at least in part, is an ER retention disease.


Asunto(s)
Albinismo Oculocutáneo/metabolismo , Retículo Endoplásmico/metabolismo , Monofenol Monooxigenasa/deficiencia , Sustitución de Aminoácidos , Animales , Proteínas de Unión al Calcio/metabolismo , Calnexina , Calreticulina , Células Cultivadas , Aparato de Golgi/metabolismo , Humanos , Melanocitos/metabolismo , Melanosomas/metabolismo , Ratones , Ratones Mutantes , Microscopía Fluorescente , Monofenol Monooxigenasa/química , Monofenol Monooxigenasa/genética , Monofenol Monooxigenasa/metabolismo , Mutación Puntual , Unión Proteica , Pliegue de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Ribonucleoproteínas/metabolismo , Transfección
7.
Pigment Cell Res ; 13(1): 4-14, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10761990

RESUMEN

In vitro, normal human melanocytes require synergistic mitogens, in addition to the common growth factors present in serum, in order to proliferate. The peptide growth factors that confer stimulation are fibroblast growth factors (such as bFGF/FGF2), hepatocyte growth factor/scatter factor (HGF/SF), mast/stem cell factor (M/SCF), endothelins (such as ET-1) and melanotropin (MSH). The proper function of these factors and their cognate receptors is likely to be important in vivo, as all five ligands are produced in the skin, and disruption of their normal function, by elimination due to deletions or mutations, or overproduction due to ectopic expression, disrupts the normal distribution of melanocytes. The synergistic growth factors activate intracellular signal transduction cascades and maintain the intermediate effectors at optimal levels and duration required for nuclear translocation and modification of transcription factors. The consequent induction of immediate-early response genes, such as cyclins, and subsequent activation of cyclin-dependent kinases (CDK4, CDK6 and CDK2) inactivates the retinoblastoma family of proteins (pRb, p107 and p130, together termed pocket proteins), and releases their suppressive association with E2F transcription factors. Molecular events that disrupt this tight control of pocket proteins and cause their inactivation, increase E2F transcriptional activity and confer autonomous growth on melanocytes.


Asunto(s)
Sustancias de Crecimiento/farmacología , Melanocitos/citología , Transducción de Señal/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Células Cultivadas , Humanos , Transducción de Señal/efectos de los fármacos
8.
J Exp Med ; 191(6): 1005-16, 2000 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-10727462

RESUMEN

Inactivation of the retinoblastoma tumor suppressor protein (pRb) has been implicated in melanoma cells, but the molecular basis for this phenotype has not yet been elucidated, and the status of additional family members (p107 and p130, together termed pocket proteins) or the consequences on downstream targets such as E2F transcription factors are not known. Because cell cycle progression is dependent on the transcriptional activity of E2F family members (E2F1-E2F6), most of them regulated by suppressive association with pocket proteins, we characterized E2F-pocket protein DNA binding activity in normal versus malignant human melanocytes. By gel shift analysis, we show that in mitogen-dependent normal melanocytes, external growth factors tightly controlled the levels of growth-promoting free E2F DNA binding activity, composed largely of E2F2 and E2F4, and the growth-suppressive E2F4-p130 complexes. In contrast, in melanoma cells, free E2F DNA binding activity (E2F2 and E2F4, to a lesser extent E2F1, E2F3, and occasionally E2F5), was constitutively maintained at high levels independently of external melanocyte mitogens. E2F1 was the only family member more abundant in the melanoma cells compared with normal melanocytes, and the approximately fivefold increase in DNA binding activity could be accounted for mostly by a similar increase in the levels of the dimerization partner DP1. The continuous high expression of cyclin D1, A2, and E, the persistent cyclin-dependent kinase 4 (CDK4) and CDK2 activities, and the presence of hyperphosphorylated forms of pRb, p107, and p130, suggest that melanoma cells acquired the capacity for autonomous growth through inactivation of all three pocket proteins and release of E2F activity, otherwise tightly regulated in normal melanocytes by external growth factors.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Proteínas de Drosophila , Melanoma/metabolismo , Melanoma/patología , Factores de Transcripción/metabolismo , División Celular/efectos de los fármacos , Células Cultivadas , Senescencia Celular , Medios de Cultivo Condicionados/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/aislamiento & purificación , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Factor de Transcripción E2F2 , Factor de Transcripción E2F3 , Factor de Transcripción E2F4 , Factor de Transcripción E2F5 , Factor de Transcripción E2F6 , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Humanos , Melanocitos/metabolismo , Melanocitos/fisiología , Melanoma/enzimología , Fosforilación , Piperidinas/farmacología , Unión Proteica , Proteína 1 de Unión a Retinoblastoma , Transactivadores/biosíntesis , Transactivadores/metabolismo , Factor de Transcripción DP1 , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/biosíntesis , Factores de Transcripción/fisiología , Células Tumorales Cultivadas
9.
Cancer Metastasis Rev ; 18(3): 333-43, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10721488

RESUMEN

Transformation of normal melanocytes to metastatic melanoma cells is characterized by loss of dependency on external growth factors required for the viability and proliferation of normal melanocytes. The molecular events that lead to melanoma cell autonomous growth are not well defined, but are likely to include sustained activity of cyclin-dependent kinases (CDK2, CDK4 and CDK6) as a result of loss of CDK inhibitors (such as p16INK4a and possibly p27KIP1), and persistent upregulation of several cyclins (cyclin D1, cyclin A and cyclin E), the positive regulators of CDKs. CDKs phosphorylate, and consequently, inactivate the retinoblastoma family of tumor suppressor proteins (pRb, p107 and p130), termed pocket proteins. The inactivation of pocket proteins liberates E2F transcription factors from suppressive complexes ('free' E2F) that, in turn, induces the continuous expression of target genes whose products promote cell cycle progression. In normal melanocytes, external growth factors suppress the activity of all three pocket proteins, allowing E2F activity to accumulate and sustain transcription of target genes required for cell proliferation. In contrast, in melanoma cells from advanced lesions, all three pocket proteins are highly phosphorylated and inactive, even in the absence of environmental mitogens, and free E2F activity is constitutively high. Manipulations of normal mouse melanocytes in vitro, and in vivo in transgenic mouse expressing ectopic genes, further support the notion that growth rate, and release from dependency on external mitogens, positively correlate with inactivation of pocket proteins. The latter has been accomplished by sustained cell surface receptor stimulation, such as constitutive high expression of a growth factor, or by sequestration with dominantly acting viral proteins. Taken together, chronic hyperphosphorlyation/inactivation of pRb, p107 and p130 is probably one of the key events in converting growth-factor dependent normal melanocytes, to autonomously growing melanoma cells. Since all pocket proteins are regulated by CDKs activity, it is likely that agents that inhibit this class of enzymes will be effective in treating melanoma patients.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Melanoma/patología , Proteína de Retinoblastoma/fisiología , Factores de Transcripción/fisiología , Animales , Ciclo Celular , División Celular , Quinasas Ciclina-Dependientes/fisiología , Factores de Transcripción E2F , Humanos , Melanocitos/fisiología , Fosforilación , Proteína 1 de Unión a Retinoblastoma , Transducción de Señal , Factor de Transcripción DP1
10.
Int J Mol Med ; 1(2): 419-25, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9852245

RESUMEN

Transformation of melanocytes to metastatic melanoma cells is characterized by unrestricted proliferation under growth-factor-deprived conditions, genetic loss of cyclin dependent kinase (CDK) inhibitors (CKI, e.g. p16INK4A), and aberrant production of autocrine growth factors (e.g. basic fibroblast growth factor). The latter induces increased expression of positive CDK regulators (e.g. cyclin D1) and reduced expression of additional CKIs (e.g. p27KIP1). Combined, these events lead to sustained CDK activity and hyperphosphorylation/inactivation of the retinoblastoma tumor suppressor protein (Rb). The persistent Rb phosphorylation causes the accumulation of E2F and the transcription of its target genes whose products promote cell cycle progression.


Asunto(s)
Melanoma , Ciclo Celular , Humanos
11.
Proc Natl Acad Sci U S A ; 95(24): 14361-6, 1998 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-9826705

RESUMEN

The microrchidia, or morc, autosomal recessive mutation results in the arrest of spermatogenesis early in prophase I of meiosis. The morc mutation arose spontaneously during the development of a mouse strain transgenic for a tyrosinase cDNA construct. Morc -/- males are infertile and have grossly reduced testicular mass, whereas -/- females are normal, indicating that the Morc gene acts specifically during male gametogenesis. Immunofluorescence to synaptonemal complex antigens demonstrated that -/- male germ cells enter meiosis but fail to progress beyond zygotene or leptotene stage. An apoptosis assay revealed massive numbers of cells undergoing apoptosis in testes of -/- mice. No other abnormal phenotype was observed in mutant animals, with the exception of eye pigmentation caused by transgene expression in the retina. Spermatogenesis is normal in +/- males, despite significant transgene expression in germ cells. Genomic analysis of -/- animals indicates the presence of a deletion adjacent to the transgene. Identification of the gene inactivated by the transgene insertion may define a novel biochemical pathway involved in mammalian germ cell development and meiosis.


Asunto(s)
Genes Recesivos , Infertilidad Masculina/genética , Espermatogénesis/genética , Espermatozoides/fisiología , Testículo/anomalías , Animales , Apoptosis , Secuencia de Bases , Cartilla de ADN , Color del Ojo/genética , Eliminación de Gen , Infertilidad Masculina/patología , Masculino , Meiosis , Ratones , Ratones Endogámicos , Ratones Transgénicos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , ARN Polimerasa II/genética , Secuencias Reguladoras de Ácidos Nucleicos , Retina/fisiología , Espermatozoides/citología , Testículo/patología
12.
Oncogene ; 16(19): 2489-501, 1998 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-9627115

RESUMEN

Compared to normal melanocytes, melanoma cell lines exhibit overexpression of hyperphosphorylated retinoblastoma tumor suppressor protein (Rb) or a marked decrease in, or lack of, expression of Rb. Hyperphosphorylation of Rb results in increased E2F-mediated transactivation of target genes and cell cycle progression. Using a combination of gene disruption and ectopic expression in growth factor-dependent mouse melanocytes, we studied the roles of E2F1 and the p16INK4A and p21WAF1/CIP1 CKIs (cyclin dependent kinase inhibitors) in the acquisition of TPA (12-O-tetradecanoyl phorbol-13-acetate)-independent growth in culture, a hallmark of melanomas. Surprisingly, melanocytes from p16INK4A- or p21WAF1/CIP1-null mice remained TPA-dependent, and disruption of p21WAF1/CIP1 accelerated cell death in the absence of this mitogen. Disruption of E2F1 had the most profound effect on melanocyte growth, resulting in a fourfold decrease in growth rate in the presence of TPA. Furthermore, enforced overexpression of the DNA-binding-defective E2F1E132 mutant conferred TPA-independence upon melanocytes and was associated with sequestration of Rb and constitutive expression of E2F1 target genes, including p21WAF1/CIP1. We conclude that neutralization of Rb by E2F1E132, but not the disruption of p16INK4A or p21WAF1/CIP1, resulted in the accumulation of free E2F and cell cycle progression. Thus, mechanisms other than the loss of p16INK4A or p21WAF1/CIP1 that activate E2F may play an important role in melanomas.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Ciclo Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Ciclinas/fisiología , Proteínas de Unión al ADN , Melanocitos/citología , Factores de Transcripción/fisiología , Animales , Supervivencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Regulación de la Expresión Génica , Humanos , Melanocitos/metabolismo , Ratones , Ratones Desnudos , Mutagénesis , Biosíntesis de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteína 1 de Unión a Retinoblastoma , Acetato de Tetradecanoilforbol/farmacología , Factor de Transcripción DP1 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Proc Natl Acad Sci U S A ; 94(17): 9017-21, 1997 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-9256427

RESUMEN

Stem cell factor (SCF) is produced by stromal cells as a membrane-bound molecule, which may be proteolytically cleaved at a site close to the membrane to produce a soluble bioactive form. The proteases producing this cleavage are unknown. In this study, we demonstrate that human mast cell chymase, a chymotrypsin-like protease, cleaves SCF at a novel site. Cleavage is at the peptide bond between Phe-158 and Met-159, which are encoded by exon 6 of the SCF gene. This cleavage results in a soluble bioactive product that is 7 amino acids shorter at the C terminus than previously identified soluble SCF. This research shows the identification of a physiologically relevant enzyme that specifically cleaves SCF. Because mast cells express the KIT protein, the receptor for SCF, and respond to SCF by proliferation and degranulation, this observation identifies a possible feedback loop in which chymase released from mast cell secretory granules may solubilize SCF bound to the membrane of surrounding stromal cells. The liberated soluble SCF may in turn stimulate mast cell proliferation and differentiated functions; this loop could contribute to abnormal accumulations of mast cells in the skin and hyperpigmentation at sites of chronic cutaneous inflammation.


Asunto(s)
Mastocitos/metabolismo , Serina Endopeptidasas/metabolismo , Factor de Células Madre/metabolismo , Sitios de Unión , Quimasas , Humanos , Hidrólisis , Especificidad por Sustrato
14.
Leukemia ; 11(8): 1338-46, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9264390

RESUMEN

Malignant cells of patients with cutaneous T cell lymphoma (CTCL) are of monoclonal origin and of the CD4+/CD45RO+ subset. Since unlike their normal counterparts, triggering of their TCR/CD3 in vitro elicits only a weak mitogenic response, we set out to determine which of the signal transduction molecules initiated by anti-CD3E antibodies are affected in neoplastic cells. The results obtained from analysis of tumor cells from four patients show a general reduction in basal and induced tyrosine phosphorylation of a wide range of signaling proteins. Furthermore, the function of members from distinct families of protein tyrosine kinases was altered in neoplastic cells. The enzymatic activity of the membrane-bound fraction of Csk was suppressed, and its association with other cellular proteins was altered. There was a decline in the amount and activity of Syk, and a slight decrease in the specific activity of Lck kinases. Zap70 tyrosyl phosphorylation was reduced or undetectable and the kinase associated weakly, or not at all, with the TCR zeta chain. We propose that dampened TCR-triggered responses in CTCL are caused by suppression of an array of effector molecules required for coupling cell surface receptors to early and late signaling events.


Asunto(s)
Linfoma Cutáneo de Células T/fisiopatología , Receptores de Antígenos de Linfocitos T/fisiología , Neoplasias Cutáneas/fisiopatología , Linfocitos T/fisiología , Adulto , Proteína Tirosina Quinasa CSK , Activación Enzimática , Precursores Enzimáticos/metabolismo , Humanos , Inmunofenotipificación , Péptidos y Proteínas de Señalización Intracelular , Fosfotirosina/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Quinasa Syk , Proteína Tirosina Quinasa ZAP-70 , Familia-src Quinasas/metabolismo
15.
Oncogene ; 14(25): 2999-3009, 1997 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-9223663

RESUMEN

Basic Fibroblast Growth Factor (bFGF/FGF2) is thought to play a decisive role in malignant progression. Aberrant expression of bFGF causes constitutive autocrine activation of its cognate receptor and autonomous growth of human melanoma cells or bFGF transformed fibroblasts in culture. It remains to be determined, however, whether the endogenous bFGF confers growth advantage to tumors and what are the downstream targets of the activated FGF receptor critical for its transforming capacity. We therefore transfected metastatic melanoma cells and bFGF transformed mouse fibroblasts with a dominant-negative mutant of the murine FGF receptor 1 (fgfr1/flg), comprising the extracellular and transmembrane domains but lacking the intracellular kinase domain (dnflg). Reverse transcriptase-PCR, 125I-bFGF binding and affinity labeling analyses show that the truncated receptor is targeted to the membrane and is expressed at much higher levels than the endogenous receptor in all of the selected clones. Expression of the dnflg dramatically reduces the basal as well as bFGF induced growth of these cells in vitro and also suppresses their tumorigenic potential in nude mice. The expression of the dnflg does not significantly alter the general level of tyrosyl-phosphorylated proteins in the trunsduced melanoma cells. Rather, a major downstream affected target is a Src-family kinase, whose activity, determined by an in vitro immune kinase assay, is stimulated in normal melanocytes by exogenous bFGF, and is markedly reduced in the dnflg-expressing melanoma cells. The present study demonstrates that direct interference with the activity of FGF receptors has a deleterious effect on cell proliferation and survival in vitro and in vivo leading to the suppression of melanoma tumor progression possibly through the inactivation of a Src-family kinase.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/genética , Melanoma/genética , Melanoma/patología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras , Receptores de Factores de Crecimiento de Fibroblastos/genética , Células 3T3/patología , Animales , División Celular/efectos de los fármacos , División Celular/genética , Línea Celular Transformada , Fibroblastos/patología , Proteínas Filagrina , Genes Dominantes , Humanos , Melanoma/enzimología , Ratones , Fenotipo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Tumorales Cultivadas , Dominios Homologos src
16.
Proc Natl Acad Sci U S A ; 94(12): 6210-5, 1997 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-9177196

RESUMEN

The loss of tyrosinase, the key enzyme in melanin synthesis, has been implicated in the dedifferentiation of malignant melanocytes. The presence of tyrosinase transcripts and antigenic peptides in melanoma tumors prompted us to investigate whether the basis for the loss of the enzyme was proteolytic degradation. Toward this aim, we followed the kinetics of synthesis, degradation, processing, chaperone binding, inhibitor sensitivity, and subcellular localization of tyrosinase in normal and malignant melanocytes. We found that, in amelanotic melanoma cell lines, tyrosinase failed to reach the melanosome, the organelle for melanin synthesis, because it was retained in the endoplasmic reticulum (ER) and then degraded. Tyrosinase appeared mostly as a 70-kDa core-glycosylated, endoglycosidase H-sensitive, immature form bound to the ER chaperone calnexin and had a life-span of only 25% of normal. Maturation and transit from the ER to the Golgi compartment was facilitated by lowering the temperature of incubation to 31 degrees C. Several proteasome inhibitors caused the accumulation of an approximately 60-kDa tyrosinase doublet that was more prominent in malignant than in normal melanocytes and promoted, to various degrees, the maturation of tyrosinase in melanoma cells and the translocation of the enzyme to melanosomes. The appearance of ubiquitinated tyrosinase after treatment of normal melanocytes with N-acetyl-L-leucinyl-L-leucinal-L-norleucinal reinforced our notion that some tyrosinase is normally degraded by proteasomes. Proteolysis of tyrosinase by proteasomes is consistent with the production of antigenic tyrosinase peptides that are presented to the immune system by major histocompatibility complex class I molecules.


Asunto(s)
Retículo Endoplásmico/enzimología , Melanocitos/enzimología , Melanoma/enzimología , Monofenol Monooxigenasa/metabolismo , Neoplasias Cutáneas/enzimología , Adulto , Proteínas de Unión al Calcio/metabolismo , Calnexina , Calreticulina , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Precursores Enzimáticos/metabolismo , Humanos , Recién Nacido , Cinética , Melanocitos/citología , Melanoma/genética , Melanoma/patología , Chaperonas Moleculares/metabolismo , Peso Molecular , Monofenol Monooxigenasa/biosíntesis , Fenotipo , Ribonucleoproteínas/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Factores de Tiempo , Células Tumorales Cultivadas
17.
Oncogene ; 14(21): 2553-61, 1997 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-9191055

RESUMEN

The macrophage colony stimulating factor receptor (CSF-1R), the product of the c-fms proto-oncogene, plays an important role in regulating the normal proliferation and differentiation of macrophages and trophoblasts. However, the abnormal expression of CSF-1R transcripts and protein by human breast carcinomas has been shown to correlate with advanced stage and poor prognosis. Ligand activated CSF-1R dimers transphosphorylate several tyrosines in their cytoplasmic domains which provide recognition sites for various effector proteins in multiple signal transduction pathways. In cells transformed by the c-fms oncogene, one of the major CSF-1R phosphotyrosines, pTyr723 is important for phenotypic expression of anchorage-independent growth and metastasis. In order to investigate the relationship between receptor activation/phosphorylation and cellular phenotypes in vitro and in vivo, we prepared a CSF-1R phosphorylation-state specific antibody raised against a specific phosphopeptide of CSF-1R, which included phosphorylated tyrosine 723. On immunoblots of lysates from cells expressing CSF-1R, this antibody recognizes phosphorylated CSF-IR in CSF-1 stimulated cells but not in unstimulated cells. As an immunohistochemical reagent, this antibody stained 52% of invasive human breast tumors (72% of CSF-1R positive cases) in a sample of 114 cases and 38% of carcinoma in situ. This data represents the first direct evidence of in vivo phosphorylation of CSF-1R in human breast carcinomas.


Asunto(s)
Anticuerpos/química , Neoplasias de la Mama/metabolismo , Fosfopéptidos/inmunología , Fosfotirosina/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos , Neoplasias de la Mama/diagnóstico , Carcinoma in Situ/metabolismo , Células Cultivadas , Reacciones Cruzadas , Epítopos , Fibroblastos/metabolismo , Genes fms/fisiología , Humanos , Inmunohistoquímica , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Fosforilación , Proto-Oncogenes Mas , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Células Tumorales Cultivadas
18.
Cell Growth Differ ; 8(11): 1199-210, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9372243

RESUMEN

Dysregulated expression of basic fibroblast growth factor [fibroblast growth factor 2 (FGF-2)] mediates autocrine growth of melanoma cells. The presence of a consensus Myb binding site in the human FGF-2 promoter prompted us to investigate whether this transcription factor could regulate FGF-2 expression in melanomas. We report that c-MYB mRNA is overexpressed in melanoma cell lines compared to normal melanocytes and that ectopic expression of murine c-Myb in SK-MEL-2 human melanoma cells resulted in increased expression of FGF-2 mRNA and FGF-2 protein. Furthermore, murine c-Myb transactivated a reporter plasmid containing the human FGF-2 promoter region in contransfected SK-MEL-2 human melanoma cells. Although a functional DNA-binding domain was required for transactivation, responsiveness to c-Myb was independent of the putative Myb binding site and mapped to two regions of the FGF-2 promoter that did not bind c-Myb in vitro. We suggest that c-MYB contributes to FGF-2-mediated autocrine growth of melanomas by indirectly regulating the FGF-2 promoter.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Melanoma/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Animales , Sitios de Unión , Proteínas de Unión al ADN/genética , Factor 2 de Crecimiento de Fibroblastos/genética , Humanos , Ratones , Regiones Promotoras Genéticas , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-myb , ARN Mensajero/metabolismo , Transactivadores/genética , Activación Transcripcional
19.
Semin Oncol ; 23(6): 673-81, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8970586

RESUMEN

Understanding the growth constraints imposed on normal human melanocytes may help to elucidate the processes conferring growth advantage to melanoma cells. Several synergistic growth factors have been identified for normal human melanocytes. They include fibroblast growth factors (FGF), hepatocyte growth factor/scatter factor, mast/stem cell growth factor, and the neuropeptides endothelin-1, 2 and 3 (ET-1, ET-2, ET-3). From this group of peptides, only basic FGF (bFGF/FGF2) appears, so far, to play a role in autonomous growth of melanoma cells. Aberrant expression of FGF2 is due to activation of an otherwise repressed gene by a mechanism that may involve the transcriptional activity of wild-type p53. The growth factors and activated receptors aberrantly expressed in melanoma cells act in concert with molecules that control cell cycle progression. These proteins bind to, and regulate cyclin-dependent kinase (CDK), such as CDK4, responsible for phosphorylation of retinoblastoma (RB) and dissociation of RB-E2F1 inhibitory complexes, thereby allowing progression through the cell cycle. Constitutive CDK4 activity in melanomas may be the results of inactivation of the negative regulators known as CDK inhibitor p16INK4, and/or p21; and/or overexpression of cyclin D, the positive CDK4 regulator. This complex set of changes in melanoma cells can lift growth constraints by inducing unregulated expression of genes promoting transition from GI to S phase of the cell cycle.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Sustancias de Crecimiento/fisiología , Melanoma/fisiopatología , Proteínas Portadoras/fisiología , Transformación Celular Neoplásica , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Endotelinas/fisiología , Factor 2 de Crecimiento de Fibroblastos/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos , Melanocitos/citología , Melanocitos/enzimología , Melanoma/enzimología , Melanoma/patología , Receptores de Factores de Crecimiento/fisiología , Transducción de Señal
20.
Proc Natl Acad Sci U S A ; 93(21): 11586-90, 1996 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-8876179

RESUMEN

Damage to actively transcribed DNA is preferentially repaired by the transcription-coupled repair (TCR) system. TCR requires RNA polymerase II (Pol II), but the mechanism by which repair enzymes preferentially recognize and repair DNA lesions on Pol II-transcribed genes is incompletely understood. Herein we demonstrate that a fraction of the large subunit of Pol II (Pol II LS) is ubiquitinated after exposing cells to UV-radiation or cisplatin but not several other DNA damaging agents. This novel covalent modification of Pol II LS occurs within 15 min of exposing cells to UV-radiation and persists for about 8-12 hr. Ubiquitinated Pol II LS is also phosphorylated on the C-terminal domain. UV-induced ubiquitination of Pol II LS is deficient in fibroblasts from individuals with two forms of Cockayne syndrome (CS-A and CS-B), a rare disorder in which TCR is disrupted. UV-induced ubiquitination of Pol II LS can be restored by introducing cDNA constructs encoding the CSA or CSB genes, respectively, into CS-A or CS-B fibroblasts. These results suggest that ubiquitination of Pol II LS plays a role in the recognition and/or repair of damage to actively transcribed genes. Alternatively, these findings may reflect a role played by the CSA and CSB gene products in transcription.


Asunto(s)
Síndrome de Cockayne/enzimología , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Ubiquitinas/metabolismo , Rayos Ultravioleta , Línea Celular , Cisplatino/farmacología , Síndrome de Cockayne/genética , Daño del ADN , Reparación del ADN , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , Fosforilación , ARN Polimerasa II/efectos de la radiación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/efectos de la radiación , Transcripción Genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA