Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 210
Filtrar
2.
Oncogene ; 33(7): 823-31, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-23376846

RESUMEN

Pancreatic cancer remains as one of the most deadly cancers with few treatment options at late stages and little information about how it develops through earlier stages. Activating mutation of the Kras gene has been implicated in, but is not sufficient for, tumorigenesis. In mouse models of pancreatic cancer, loss of tumor suppressor genes in conjunction with Kras mutation leads to gradual stochastic acquisition of neoplastic precursors and carcinomas, whereas many cells remain phenotypically unaltered in younger mice. Here, we demonstrate that two oncogenic events, mutation of Kras and production of the growth factor heparin-binding epidermal growth factor-like growth factor (HB-EGF), are sufficient for rapid and complete neoplastic transformation of the exocrine pancreas. We found that macrophages are the major source of HB-EGF production in pancreatic cancer tissue samples, and that macrophages are present in high density and in close association with human pancreatic cancer lesions. In a mouse model, high macrophage density was observed at the earliest stages of neoplastic transformation. The consequence of elevated HB-EGF signaling was investigated without the confounding effects of other macrophage-produced factors via transgenic overexpression of the active form of HB-EGF. In this model, HB-EGF was sufficient to promote Kras-initiated tumorigenesis, inducing rapid and complete neoplastic transformation of the entire exocrine pancreas shortly after birth. HB-EGF overexpression and Kras(G12D) together, but neither alone, increased proliferation with increased cyclinD1 and decreased Cdkn2a/2d (p16/p19(Ink4A/Arf)). These findings establish the importance of oncogenic synergy in cancer initiation and promotion, and establish a molecular link between inflammation and the earliest stages of tumor induction.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Proliferación Celular , Células Cultivadas , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense , Páncreas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Proteínas ras/metabolismo
3.
Br J Cancer ; 108(9): 1765-70, 2013 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-23558895

RESUMEN

In baseball parlance, a triple threat is a person who can run, hit and throw with aplomb. Leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) is a cell surface protein that antagonises ERBB receptor signalling by downregulating receptor levels. Over 10 years ago, Hedman et al postulated that LRIG1 might be a tumour suppressor. Recently, Powell et al provided in vivo evidence substantiating that claim by demonstrating that Lrig1 loss in mice leads to spontaneously arising, highly penetrant intestinal adenomas. Interestingly, Lrig1 also marks stem cells in the gut, suggesting a potential role for Lrig1 in maintaining intestinal epithelial homeostasis. In this review, we will discuss the ability of LRIG1 to act as a triple threat: pan-ERBB negative regulator, intestinal stem cell marker and tumour suppressor. We will summarise studies of LRIG1 expression in human cancers and discuss possible related roles for LRIG2 and LRIG3.


Asunto(s)
Receptores ErbB/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Neoplasias/genética , Proteínas del Tejido Nervioso/metabolismo , Células Madre/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adenoma/genética , Animales , Biomarcadores/metabolismo , Receptores ErbB/genética , Genes Supresores de Tumor , Humanos , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas Oncogénicas v-erbB/antagonistas & inhibidores , Proteínas Oncogénicas v-erbB/metabolismo , Células Madre/citología , Proteínas Supresoras de Tumor/genética
5.
Oncogene ; 26(21): 3051-9, 2007 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-17130841

RESUMEN

Activating mutations in members of the RAS family of genes are among the most common genetic events in human tumorigenesis. Once thought to be functionally interchangeable, it is increasingly recognized that the classical members of this protein family (H-RAS, N-RAS and K-RAS4B) exhibit unique and shared functions that are highly context-dependent. Herein, we demonstrate that the presence of an oncogenic KRAS allele results in elevated levels of GTP-bound N-RAS (N-RAS.GTP) in two human colorectal cancer cell lines, HCT 116 and DLD-1, compared to their isogenic counterparts in which the mutant KRAS allele has been disrupted by homologous recombination. N-RAS subserves an antiapoptotic role in cells expressing wild-type K-RAS; this function is compromised, however, by the presence of mutant K-RAS, and these cells display increased sensitivity to apoptotic stimuli. We additionally identify a physical interaction between N-RAS and gelsolin, a factor that has been shown to promote survival and show that the N-RAS:gelsolin complex is modulated differently in wild-type and mutant K-RAS environments following apoptotic challenge. These findings represent the first biochemical evidence of a functional relationship between endogenous RAS proteins and identify a dynamic physical interaction between endogenous N-RAS and gelsolin that correlates with survival.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Apoptosis/genética , Gelsolina/metabolismo , Genes ras/fisiología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Proteínas ras/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/fisiología , Células CACO-2 , Línea Celular , Supervivencia Celular/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Gelsolina/fisiología , Células HCT116 , Humanos , Ratones , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas ras/genética
6.
Mol Biol Cell ; 15(6): 2853-62, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15075369

RESUMEN

Epithelial cells in vivo form tight cell-cell associations that spatially separate distinct apical and basolateral domains. These domains provide discrete cellular processes essential for proper tissue and organ development. Using confocal imaging and selective plasma membrane domain activation, the type I and type II transforming growth factor-beta (TGFbeta) receptors were found to be localized specifically at the basolateral surfaces of polarized Madin-Darby canine kidney (MDCK) cells. Receptors concentrated predominantly at the lateral sites of cell-cell contact, adjacent to the gap junctional complex. Cytoplasmic domain truncations for each receptor resulted in the loss of specific lateral domain targeting and dispersion to both the apical and basal domains. Whereas receptors concentrate basolaterally in regions of direct cell-cell contact in nonpolarized MDCK cell monolayers, receptor staining was absent from areas of noncell contact. In contrast to the defined basolateral polarity observed for the TGFbeta receptor complex, TGFbeta ligand secretion was found to be from the apical surfaces. Confocal imaging of MDCK cells with an antibody to TGFbeta1 confirmed a predominant apical localization, with a stark absence at the basal membrane. These findings indicate that cell adhesion regulates the localization of TGFbeta receptors in polarized epithelial cultures and that the response to TGFbeta is dependent upon the spatial distribution and secretion of TGFbeta receptors and ligand, respectively.


Asunto(s)
Polaridad Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Uniones Adherentes/metabolismo , Animales , Adhesión Celular , Línea Celular , Membrana Celular/metabolismo , Polaridad Celular/efectos de los fármacos , Perros , Humanos , Ligandos , Transporte de Proteínas , Receptores de Factores de Crecimiento Transformadores beta/química , Receptores de Factores de Crecimiento Transformadores beta/genética , Eliminación de Secuencia/genética , Transfección , Factor de Crecimiento Transformador beta/farmacología
7.
J Neurosurg ; 94(5): 831-5, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11354419

RESUMEN

The authors report the case of a man who suffered from progressive, disseminated posttraumatic dural arteriovenous fistulas (DAVFs) resulting in death, despite aggressive endovascular, surgical, and radiosurgical treatment. This 31-year-old man was struck on the head while playing basketball. Two weeks later a soft, pulsatile mass developed at his vertex, and the man began to experience pulsatile tinnitus and progressive headaches. Magnetic resonance imaging and subsequent angiography revealed multiple AVFs in the scalp, calvaria, and dura, with drainage into the superior sagittal sinus. The patient was treated initially with transarterial embolization in five stages, followed by vertex craniotomy and surgical resection of the AVFs. However, multiple additional DAVFs developed over the bilateral convexities, the falx, and the tentorium. Subsequent treatment entailed 15 stages of transarterial embolization; seven stages of transvenous embolization, including complete occlusion of the sagittal sinus and partial occlusion of the straight sinus; three stages of stereotactic radiosurgery; and a second craniotomy with aggressive disconnection of the DAVFs. Unfortunately, the fistulas continued to progress, resulting in diffuse venous hypertension, multiple intracerebral hemorrhages in both hemispheres, and, ultimately, death nearly 5 years after the initial trauma. Endovascular, surgical, and radiosurgical treatments are successful in curing most patients with DAVFs. The failure of multimodal therapy and the fulminant progression and disseminated nature of this patient's disease are unique.


Asunto(s)
Malformaciones Vasculares del Sistema Nervioso Central/cirugía , Hemorragia Cerebral Traumática/cirugía , Adulto , Baloncesto/lesiones , Edema Encefálico/diagnóstico , Edema Encefálico/fisiopatología , Edema Encefálico/cirugía , Malformaciones Vasculares del Sistema Nervioso Central/diagnóstico , Malformaciones Vasculares del Sistema Nervioso Central/fisiopatología , Angiografía Cerebral , Hemorragia Cerebral Traumática/diagnóstico , Hemorragia Cerebral Traumática/fisiopatología , Terapia Combinada , Progresión de la Enfermedad , Embolización Terapéutica , Resultado Fatal , Humanos , Masculino , Radiocirugia , Insuficiencia del Tratamiento
8.
J Biol Chem ; 276(31): 29538-49, 2001 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-11382759

RESUMEN

In this study, the role of the amphiregulin precursor (pro-AR) cytoplasmic domain in the basolateral sorting and cell-surface processing of pro-AR in polarized epithelial cells has been investigated using Madin-Darby canine kidney cells stably expressing various human pro-AR forms. Our results demonstrate that newly synthesized wild-type pro-AR (50 kDa) is delivered directly to the basolateral membrane domain with >95% efficiency, where it is sequentially cleaved within the ectodomain to release several soluble amphiregulin (AR) forms. Analyses of a pro-AR cytoplasmic domain truncation mutant (ARTL27) and two pro-AR secretory mutants (ARsec184 and ARsec190) indicated that the pro-AR cytoplasmic domain is not required for efficient delivery to the plasma membrane, but does contain essential basolateral sorting information. We show that the pro-AR cytoplasmic domain truncation mutant (ARTL27) is not sorted in polarized Madin-Darby canine kidney cells, with approximately 65% of the newly synthesized protein delivered to the apical cell surface. Under base-line conditions, ARTL27 was preferentially cleaved from the basolateral surface with 4-fold greater efficiency compared with cleavage from the apical membrane domain. However, ARTL27 ectodomain cleavage could be stimulated equivalently from either membrane domain by a variety of different stimuli. The metalloprotease inhibitor BB-94 could inhibit both base-line and stimulus-induced ectodomain cleavage of wild-type pro-AR and ARTL27. These results indicate that the pro-AR cytoplasmic domain is required for basolateral sorting, but is not essential for ectodomain processing. Preferential constitutive cleavage of ARTL27 from the basolateral cell surface also suggests that the metalloprotease activity involved in base-line and stimulus-induced ARTL27 ectodomain cleavage may be regulated differently in the apical and basolateral membrane domains of polarized epithelial cells.


Asunto(s)
Membrana Celular/metabolismo , Polaridad Celular/fisiología , Glicoproteínas/metabolismo , Sustancias de Crecimiento/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Precursores de Proteínas/química , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Anfirregulina , Animales , Línea Celular , Citoplasma/metabolismo , ADN Complementario , Perros , Familia de Proteínas EGF , Glicoproteínas/química , Glicoproteínas/genética , Sustancias de Crecimiento/química , Sustancias de Crecimiento/genética , Humanos , Riñón , Cinética , Mutagénesis , Precursores de Proteínas/genética , Transporte de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Eliminación de Secuencia , Transfección
9.
Oncogene ; 20(11): 1287-99, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11313873

RESUMEN

The Nuclear Factor (NF)-kappaB family of transcription factors controls expression of genes which promote cell growth, survival, and neoplastic transformation. Recently we demonstrated aberrant constitutive activation of NF-kappaB in primary human and rat breast cancer specimens and in cell lines. Overexpression of the epidermal growth factor receptor (EGFR) family member Her-2/neu, seen in approximately 30% of breast cancers, is associated with poor prognosis. Previously, Her-2/neu has been shown to signal via a phosphatidylinositol 3 (PI3)-kinase to Akt/protein kinase B (PKB) pathway. Since this signaling pathway was recently shown to activate NF-kappaB, here we have tested the hypothesis that Her-2/neu can activate NF-kappaB in breast cancer. Overexpression of Her-2/neu and EGFR-4 in Ba/F3 cells led to constitutive PI3- and Akt kinase activities, and induction of classical NF-kappaB (p50/p65). Similarly, a tumor cell line and tumors derived from MMTV-Her-2/neu transgenic mice displayed elevated levels of classical NF-kappaB. Engagement of Her-2/neu receptor downregulated the level of NF-kappaB. NF-kappaB binding and activity in the cultured cells was reduced upon inhibition of the PI3- to Akt kinase signaling pathway via ectopic expression of kinase inactive mutants, incubation with wortmannin, or expression of the tumor suppressor phosphatase PTEN. Inhibitors of calpain, but not the proteasome, blocked IkappaB-alpha degradation. Inhibition of Akt did not affect IKK activity. These results indicate that Her-2/neu activates NF-kappaB via a PI3- to Akt kinase signaling pathway that can be inhibited via the tumor suppressor PTEN, and is mediated by calpain rather than the IkappaB kinase complex.


Asunto(s)
Neoplasias de la Mama/metabolismo , Genes Supresores de Tumor , Proteínas I-kappa B , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Serina-Treonina Quinasas , Receptor ErbB-2/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor , Animales , Calpaína/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Transgénicos , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Fosfohidrolasa PTEN , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Células Tumorales Cultivadas
10.
FASEB J ; 15(2): 403-15, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11156956

RESUMEN

The vitamin E analog alpha-tocopheryl succinate (alpha-TOS) can induce apoptosis. We show that the proapoptotic activity of alpha-TOS in hematopoietic and cancer cell lines involves inhibition of protein kinase C (PKC), since phorbol myristyl acetate prevented alpha-TOS-triggered apoptosis. More selective effectors indicated that alpha-TOS reduced PKCalpha isotype activity by increasing protein phosphatase 2A (PP2A) activity. The role of PKCalpha inhibition in alpha-TOS-induced apoptosis was confirmed using antisense oligonucleotides or PKCalpha overexpression. Gain- or loss-of-function bcl-2 mutants implied modulation of bcl-2 activity by PKC/PP2A as a mitochondrial target of alpha-TOS-induced proapoptotic signals. Structural analogs revealed that alpha-tocopheryl and succinyl moieties are both required for maximizing these effects. In mice with colon cancer xenografts, alpha-TOS suppressed tumor growth by 80%. This epitomizes cancer cell killing by a pharmacologically relevant compound without known side effects.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias del Colon/patología , Neoplasias Colorrectales/patología , Vitamina E/análogos & derivados , Vitamina E/toxicidad , Animales , Anticuerpos Monoclonales/farmacología , Caspasa 3 , Caspasas/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Genes bcl-2 , Humanos , Isoenzimas/antagonistas & inhibidores , Células Jurkat , Cinética , Ratones , Ratones Desnudos , Oligodesoxirribonucleótidos Antisentido/farmacología , Fosfoproteínas Fosfatasas/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C-alfa , Proteína Fosfatasa 2 , Acetato de Tetradecanoilforbol/farmacología , Tocoferoles , Trasplante Heterólogo , Células Tumorales Cultivadas , Vitamina E/uso terapéutico , Receptor fas/inmunología , Receptor fas/fisiología
12.
Pain Med ; 2(3): 183-92, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15102250

RESUMEN

OBJECTIVES: A U.S. Food and Drug Administration ruling required clinical trials to evaluate the safety and efficacy of deep brain stimulation devices, thereby limiting treatment to the investigational setting. INTRODUCTION: As an investigator in two clinical trials of deep brain stimulation, I sought to determine why pain remained an unapproved indication despite regulatory approval of the same device for tremor. METHODS: The results of two multicenter trials of deep brain stimulation for pain were analyzed, and the pertinent literature was reviewed using published guidelines for the evaluation of clinical trial reports. RESULTS: The first-generation Model 3380 lead trial enrolled 196 patients; the current Model 3387 trial enrolled 50 patients. Prospectively defined criteria for success included at least half of patients reporting >/=50% pain relief at 1 year. Manufacture of the Model 3380 lead was discontinued, and the 3387 trial closed early because of slow enrollment, high attrition, and low efficacy. When results were analyzed according to the study plan, neither trial was successful. Consequently, deep brain stimulation has not been approved for pain control by the U.S. Food and Drug Administration. CONCLUSIONS: Deep brain stimulation has not been shown to produce effective long-term pain relief. Future studies of motor cortex stimulation and similar therapies will require appropriate control groups and accepted methods of data collection and analysis to support claims that predictable and reliable analgesic effects are produced in humans.

14.
J Clin Invest ; 106(12): 1447-55, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11120752

RESUMEN

The KvLQT1 gene encodes a voltage-gated potassium channel. Mutations in KvLQT1 underlie the dominantly transmitted Ward-Romano long QT syndrome, which causes cardiac arrhythmia, and the recessively transmitted Jervell and Lange-Nielsen syndrome, which causes both cardiac arrhythmia and congenital deafness. KvLQT1 is also disrupted by balanced germline chromosomal rearrangements in patients with Beckwith-Wiedemann syndrome (BWS), which causes prenatal overgrowth and cancer. Because of the diverse human disorders and organ systems affected by this gene, we developed an animal model by inactivating the murine Kvlqt1. No electrocardiographic abnormalities were observed. However, homozygous mice exhibited complete deafness, as well as circular movement and repetitive falling, suggesting imbalance. Histochemical study revealed severe anatomic disruption of the cochlear and vestibular end organs, suggesting that Kvlqt1 is essential for normal development of the inner ear. Surprisingly, homozygous mice also displayed threefold enlargement by weight of the stomach resulting from mucous neck cell hyperplasia. Finally, there were no features of BWS, suggesting that Kvlqt1 is not responsible for BWS.


Asunto(s)
Sordera/genética , Hiperplasia/genética , Síndrome de QT Prolongado/genética , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/deficiencia , Canales de Potasio/metabolismo , Estómago/patología , Animales , Tronco Encefálico/fisiología , Cóclea/patología , Cóclea/fisiopatología , Sordera/fisiopatología , Modelos Animales de Enfermedad , Oído Interno/patología , Oído Interno/fisiopatología , Electrocardiografía , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Histocitoquímica , Humanos , Hiperplasia/patología , Canales de Potasio KCNQ , Canal de Potasio KCNQ1 , Locomoción/fisiología , Masculino , Ratones , Ratones Noqueados , Mutación/genética , Tamaño de los Órganos , Fenotipo , Canales de Potasio/genética
15.
Neoplasia ; 2(4): 357-64, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11005570

RESUMEN

Ras-transformed intestinal epithelial cells are resistant to the growth inhibitory actions of TGFbeta and have a marked decrease in expression of the TGFbeta type II receptor (TGFbetaRII). Rat intestinal epithelial cells (RIE) were stably transfected with activated Ras, Sos and Raf constructs and tested for expression of TGFbetaRII and sensitivity to growth inhibition by TGFbeta. The parental RIE line and the RIE-Raf cells were non-transformed in morphology and were sensitive to TGFbeta (70-90% inhibited). In contrast, the RIE-Ras and RIE-Sos lines were transformed, resistant to TGFbeta and expressed 5- to 10-fold decreased levels of the TGFbetaRII mRNA and protein. Cyclin D1 protein expression was repressed by TGFbeta treatment in parental RIE and RIE-Raf cells, whereas levels of cyclin D1 in RIE-Ras and RIE-Sos cells remained unchanged. Treatment of RIE-Ras cells with 25 microM farnesyl transferase inhibitor, FTI L739,749, for 48 hours restored expression of TGFbetaRII to levels equivalent to control cells. In addition, treatment of RIE-Ras cells for 48 hours with PD-98059, a specific MAPKK inhibitor, also increased expression of TGFbetaRII to control levels. Collectively these results suggest that downregulation of TGFbetaRII and loss of sensitivity to growth inhibition by TGFbeta in Ras-transformed intestinal epithelial cells is not mediated exclusively by the conventional Ras/Raf/MAPKK/MAPK pathway. However, activation of MAPK, perhaps by an alternate Ras effector pathway, appears to be necessary for Ras-mediated downregulation of TGFbetaRII.


Asunto(s)
Regulación de la Expresión Génica , Genes ras , Mucosa Intestinal/fisiología , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal/fisiología , Transferasas Alquil y Aril/antagonistas & inhibidores , Animales , División Celular , Línea Celular , Línea Celular Transformada , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa , Flavonoides/farmacología , Mucosa Intestinal/citología , Oligopéptidos/farmacología , Proteínas Serina-Treonina Quinasas , Ratas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Transfección , Factor de Crecimiento Transformador beta/farmacología
16.
Proc Natl Acad Sci U S A ; 97(17): 9609-14, 2000 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-10931950

RESUMEN

Overexpression of ErbB-2/Neu has been causally associated with mammary epithelial transformation. Here we report that blockade of the epidermal growth factor receptor (EGFR) kinase with AG-1478 markedly delays breast tumor formation in mouse mammary tumor virus (MMTV)/Neu + MMTV/transforming growth factor alpha bigenic mice. This delay was associated with inhibition of EGFR and Neu signaling, reduction of cyclin-dependent kinase 2 (Cdk2) and mitogen-activated protein kinase (MAPK) activities and cyclin D1, and an increase in the levels of the Cdk inhibitor p27(Kip1). In addition, BrdUrd incorporation into tumor cell nuclei was prevented with no signs of tumor cell apoptosis. These observations prompted us to investigate the stability of p27. Recombinant p27 was degraded rapidly in vitro by untreated but not by AG-1478-treated tumor lysates. Proteasome depletion of the tumor lysates, addition of the specific MEK1/2 inhibitor U-0126, or a T187A mutation in recombinant p27 all prevented p27 degradation. Cdk2 and MAPK precipitates from untreated tumor lysates phosphorylated recombinant wild-type p27 but not the T187A mutant in vitro. Cdk2 and MAPK precipitates from AG-1478-treated tumors were unable to phosphorylate p27 in vitro. These data suggest that increased signaling by ErbB receptors up-regulates MAPK activity, which, in turn, phosphorylates and destabilizes p27, thus contributing to dysregulated cell cycle progression.


Asunto(s)
Quinasas CDC2-CDC28 , Proteínas de Ciclo Celular , Transformación Celular Neoplásica , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Neoplasias Mamarias Experimentales/patología , Receptor ErbB-2/metabolismo , Factor de Crecimiento Transformador alfa/metabolismo , Proteínas Supresoras de Tumor , Animales , Butadienos/farmacología , División Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Cisteína Endopeptidasas/metabolismo , ADN/biosíntesis , Dimerización , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Virus del Tumor Mamario del Ratón/genética , Virus del Tumor Mamario del Ratón/fisiología , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Complejos Multienzimáticos/metabolismo , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal , Proteínas Serina-Treonina Quinasas/metabolismo , Quinazolinas , Receptor ErbB-2/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factor de Crecimiento Transformador alfa/genética , Células Tumorales Cultivadas , Tirfostinos/farmacología
17.
Neoplasia ; 2(3): 261-72, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10935512

RESUMEN

Pancreatic ductal adenocarcinoma is a highly lethal malignancy that is resistant to traditional cytotoxic therapy. High rates of activating codon 12 K-Ras mutations in this disease have generated considerable interest in the therapeutic application of novel farnesyl transferase inhibitors (FTIs). However, a comprehensive analysis of the effects of FTI treatment on pancreatic cancer cells has not been performed. Treatment of five different human pancreatic cancer cell lines with FTI L-744,832 resulted in inhibition of anchorage-dependent growth, with wide variation in sensitivity among different lines. Effective growth inhibition by L-744,832 correlated with accumulation of cells with a tetraploid (4N) DNA content and high levels of cyclin B1/cdc2 kinase activity, implying cell cycle arrest downstream from the DNA damage-inducible G2/M cell cycle checkpoint. In addition, sensitive cell lines underwent apoptosis as evidenced by changes in nuclear morphology and internucleosomal DNA fragmentation. L-744,832 at a concentration of 1 microM additively enhanced the cytotoxic effect of ionizing radiation, apparently by overriding G2/M checkpoint activation. The effects of FTI treatment on cell growth and cell cycle regulation were associated with changes in posttranslational processing of H-Ras and N-Ras, but not K-Ras. The results confirm the potential therapeutic efficacy of FTI treatment in pancreatic cancer, and suggest that farnesylated proteins other than K-Ras may act as important regulators of G2/M cell cycle kinetics.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Ductal de Mama/patología , Ciclina B/metabolismo , Inhibidores Enzimáticos/farmacología , Genes ras/fisiología , Metionina/análogos & derivados , Neoplasias Pancreáticas/patología , Ciclina B1 , Fase G2/efectos de los fármacos , Humanos , Metionina/farmacología , Mitosis/efectos de los fármacos
18.
Exp Dermatol ; 9(3): 192-9, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10839717

RESUMEN

The induction of epidermal differentiation by extracellular Ca2+ involves activation of both tyrosine kinase and protein kinase C (PKC) signaling cascades. To determine if the differentiation-dependent activation of tyrosine kinase signaling can influence the PKC pathway, we examined the tyrosine phosphorylation status of PKC isoforms in primary mouse keratinocytes stimulated to terminally differentiate with Ca2+. Elevation of extracellular Ca2+ induced tyrosine phosphorylation of PKC-delta, but not the other keratinocyte PKC isoforms (alpha, epsilon, eta, zeta). We have previously demonstrated that activation of the epidermal growth factor receptor (EGFR) pathway induces PKC-delta tyrosine phosphorylation in basal keratinocytes (Denning M F, Dlugosz A A, Threadgill D W, Magnuson T, Yuspa S H (1996) J Biol Chem 271: 5325-5331). When basal keratinocytes were stimulated to differentiate by Ca2+, the level of cell-associated transforming growth factor-alpha (TGF-alpha) increased 30-fold, while no increase in secreted TGF-alpha was detected. Furthermore, Ca2+-induced tyrosine phosphorylation of PKC-delta and phosphotyrosine-association of the receptor adapter protein Shc was diminished in EGFR -/- keratinocytes, suggesting that EGFR activation may occur during keratinocyte differentiation. Tyrosine phosphorylated PKC-delta was also detected in mouse epidermis, suggesting that this differentiation-associated signaling pathway is physiological. These results establish a requirement for the EGFR in Ca2+-induced tyrosine phosphorylation of PKC-delta, and document the production of cell-associated TGF-alpha in differentiated keratinocytes which may function independent of its usual mitogenic effects.


Asunto(s)
Receptores ErbB/metabolismo , Isoenzimas/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Proteína Quinasa C/metabolismo , Animales , Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Células Epidérmicas , Epidermis/metabolismo , Receptores ErbB/genética , Queratinocitos/efectos de los fármacos , Ratones , Ratones Noqueados , Fosforilación , Proteína Quinasa C-delta , Receptor Cross-Talk , Factor de Crecimiento Transformador alfa/biosíntesis , Tirosina/metabolismo
19.
Gastroenterology ; 118(6): 1080-93, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10833483

RESUMEN

BACKGROUND & AIMS: Oxyntic atrophy is the hallmark of chronic gastritis. Many studies have sought to develop animal models for oxyntic atrophy, but none of them are reversible. We now report that rats administered high doses of DMP 777 demonstrate reversible oxyntic atrophy. METHODS: DMP 777 was administered to CD-1 rats by oral gavage (200 mg. kg(-1). day(-1)). Serum gastrin level, in vivo acid secretion, and gastric histological changes were evaluated in DMP 777-dosed animals. Direct effects of DMP 777 on parietal cells were evaluated by assessment of aminopyrine accumulation into isolated rabbit parietal cells, as well as by assessment of DMP 777 effects on acridine orange fluorescence and H(+),K(+)-adenosine triphosphatase (ATPase) activity in isolated tubulovesicles. RESULTS: Oral dosing with DMP 777 caused a rapid increase in serum gastrin levels and severe hypochlorhydria. DMP 777 inhibited aminopyrine accumulation into rabbit parietal cells stimulated with either histamine or forskolin. DMP 777 reversed a stimulated proton gradient in isolated parietal cell tubulovesicles. Oral dosing with DMP 777 led to rapid loss of parietal cells from the gastric mucosa. In response to the acute loss of parietal cells, there was an increase in the activity of the progenitor zone along with rapid expansion of the foveolar cell compartment. DMP 777 treatment also led to the emergence of bromodeoxyuridine-labeled cells and cells positive for periodic acid-Schiff in the basal region of fundic glands. With extended dosing over 3-6 months, foveolar hyperplasia and oxyntic atrophy were sustained while chief cell, enterochromaffin-like cell, and somatostatin cell populations were decreased. No histological evidence of neoplastic transformation was observed with dosing up to 6 months. Withdrawal of the drug after 3 or 6 months of dosing led to complete restitution of the normal mucosal lineages within 3 months. CONCLUSIONS: DMP 777 acts as a protonophore with specificity for parietal cell acid-secretory membranes. DMP 777 in high doses leads to the specific loss of parietal cells. Foveolar hyperplasia, loss of normal gland lineages, and the emergence of basal mucous cells appear as sequelae of the absence of parietal cells. The results suggest that parietal cells are critical for the maintenance of the normal mucosal lineage repertoire.


Asunto(s)
Gastritis/patología , Células Parietales Gástricas/patología , Estómago/patología , Estómago/fisiología , Naranja de Acridina , Aminopirina/farmacocinética , Animales , Antiinflamatorios no Esteroideos/farmacocinética , Atrofia , Azetidinas , Radioisótopos de Carbono , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Colorantes Fluorescentes , Ácido Gástrico/metabolismo , Gastrinas/sangre , Gastritis/inducido químicamente , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Ionóforos/farmacología , Elastasa de Leucocito/antagonistas & inhibidores , Elastasa de Leucocito/metabolismo , Masculino , Necrosis , Nigericina/farmacología , Células Parietales Gástricas/enzimología , Células Parietales Gástricas/metabolismo , Piperazinas , Conejos , Ratas , Ratas Sprague-Dawley , Regeneración
20.
Exp Cell Res ; 256(1): 168-78, 2000 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-10739664

RESUMEN

Crk-associated substrate (p130(Cas), Cas) is a docking protein first recognized as having elevated phosphotyrosine content in mammalian cells transformed by v-Src and v-Crk oncoproteins. Subsequent studies have implicated Cas in the control of normal cell behavior through its roles in integrin-mediated signal transduction and organization of the actin cytoskeleton at sites of cell adhesion. In this study, we sought to gain new insight into normal Cas function by identifying previously unrecognized interacting proteins. A yeast two-hybrid screen using the C-terminal region of Cas as a bait identified the Src homology 3 (SH3) domain of the mouse "nephrocystin" protein-orthologous to a human protein whose loss of function leads to the cystic kidney disease familial juvenile nephronophthisis. The putative full-length mouse and partial canine nephrocystin sequences were deduced from cDNA clones. Additional studies using epitope-tagged mouse nephrocystin indicated that nephrocystin and Cas can interact in mammalian cells and revealed that both proteins prominently localize at or near sites of cell-cell contact in polarized Madin-Darby canine kidney epithelial cells. Our findings provide novel insight into the normal cellular activities regulated by both Cas and nephrocystin, and raise the possibility that these proteins have a related function in polarized epithelial cells.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/fisiología , Uniones Intercelulares/fisiología , Fosfoproteínas/metabolismo , Proteínas/metabolismo , Proteínas Oncogénicas de Retroviridae/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Animales , Línea Celular , Polaridad Celular , Transformación Celular Neoplásica , Proteína Sustrato Asociada a CrK , Proteínas del Citoesqueleto , Perros , Células Epiteliales/ultraestructura , Genes src , Humanos , Uniones Intercelulares/ultraestructura , Riñón , Proteínas de la Membrana , Ratones , Datos de Secuencia Molecular , Proteína Oncogénica v-crk , Fosfoproteínas/análisis , Fosfoproteínas/química , Proteínas/análisis , Proteínas/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteína p130 Similar a la del Retinoblastoma , Proteínas Oncogénicas de Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transcripción Genética , Transfección , Dominios Homologos src
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...