Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
MMWR Morb Mortal Wkly Rep ; 67(26): 738-741, 2018 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-29975678

RESUMEN

Chagas disease, a potentially life-threatening disease caused by the protozoan parasite Trypanosoma cruzi, has become a concern in the United States as a result of human emigration from Latin America where Chagas disease is endemic (1). It is estimated that as many as 8 million people living in Mexico, and Central and South America have Chagas disease.* Most cases of Chagas disease in the United States are chronic infections; however, rare cases of acute congenital infections and autochthonous vectorborne transmission have been reported (2). To understand how data are collected and used, a review of state-level public health surveillance for Chagas disease was conducted through semistructured interviews with health officials in six states (Arizona, Arkansas, Louisiana, Mississippi Tennessee, and Texas) where Chagas disease is reportable and one (Massachusetts) where it was previously reportable. States implemented surveillance in response to blood donor screening for Chagas disease and to identify the route of disease transmission. Many states reported primarily chronic cases and had limited ability to respond to local transmission because acute cases were infrequently reported. Surveillance remains important in states with large populations of immigrants or frequent travelers from countries with endemic disease and for states with a risk for local transmission. Surveillance efforts can also help increase awareness among providers and assist in linking patients with Chagas disease to treatment to help prevent cardiac and gastrointestinal complications.


Asunto(s)
Enfermedad de Chagas/epidemiología , Emigrantes e Inmigrantes , Vigilancia de la Población , Emigración e Inmigración/estadística & datos numéricos , Enfermedades Endémicas , Humanos , América Latina/epidemiología , América Latina/etnología , Trypanosoma cruzi/aislamiento & purificación , Estados Unidos/epidemiología
2.
Nat Genet ; 24(3): 291-5, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10700186

RESUMEN

Type 2 diabetes is a polygenic and genetically heterogeneous disease . The age of onset of the disease is usually late and environmental factors may be required to induce the complete diabetic phenotype. Susceptibility genes for diabetes have not yet been identified. Islet-brain-1 (IB1, encoded by MAPK8IP1), a novel DNA-binding transactivator of the glucose transporter GLUT2 (encoded by SLC2A2), is the homologue of the c-Jun amino-terminal kinase-interacting protein-1 (JIP-1; refs 2-5). We evaluated the role of IBi in beta-cells by expression of a MAPK8IP1 antisense RNA in a stable insulinoma beta-cell line. A 38% decrease in IB1 protein content resulted in a 49% and a 41% reduction in SLC2A2 and INS (encoding insulin) mRNA expression, respectively. In addition, we detected MAPK8IP1 transcripts and IBi protein in human pancreatic islets. These data establish MAPK8IP1 as a candidate gene for human diabetes. Sibpair analyses performed on i49 multiplex French families with type 2 diabetes excluded MAPK8IP1 as a major diabetogenic locus. We did, however, identify in one family a missense mutation located in the coding region of MAPK8IP1 (559N) that segregated with diabetes. In vitro, this mutation was associated with an inability of IB1 to prevent apoptosis induced by MAPK/ERK kinase kinase 1 (MEKK1) and a reduced ability to counteract the inhibitory action of the activated c-JUN amino-terminal kinase (JNK) pathway on INS transcriptional activity. Identification of this novel non-maturity onset diabetes of the young (MODY) form of diabetes demonstrates that IB1 is a key regulator of 3-cell function.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Diabetes Mellitus Tipo 2/genética , Islotes Pancreáticos/metabolismo , Proteínas Nucleares/genética , Transactivadores/genética , Edad de Inicio , Apoptosis/genética , Ensayo de Unidades Formadoras de Colonias , Diabetes Mellitus Tipo 2/epidemiología , Femenino , Efecto Fundador , Francia/epidemiología , Predisposición Genética a la Enfermedad , Genotipo , Transportador de Glucosa de Tipo 2 , Humanos , Insulina/metabolismo , Secreción de Insulina , Insulinoma/genética , Insulinoma/metabolismo , Insulinoma/patología , Proteínas Quinasas JNK Activadas por Mitógenos , Escala de Lod , Sistema de Señalización de MAP Quinasas , Masculino , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas Nucleares/fisiología , Obesidad/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Linaje , Transactivadores/fisiología , Transcripción Genética , Células Tumorales Cultivadas/metabolismo
3.
Neuropathol Appl Neurobiol ; 36(3): 211-24, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19849792

RESUMEN

AIM: Both hyperbaric oxygenation (HBO) and inhibition of the c-Jun N-terminal kinases (JNKs) by the peptide inhibitor XG-102 (D-JNKI-1) are efficient protective strategies against ischaemia-induced neurodegeneration. The present study investigated whether the combination of HBO and JNK inhibitor, XG-102, provides additive neuroprotection against cerebral ischaemia. METHODS: Rat middle cerebral artery was occluded (MCAO) for 90 min. XG-102 [2 mg/kg, intraperitoneally] or HBO (3 ATA, 60 min) was applied 3 h after the onset of MCAO. For the combination treatment, HBO was started 10 min after the injection of XG-102. Twenty-four hours after MCAO, the infarct area, the neurological score and the immunohistochemistry staining in brain slices for cleaved-PARP, transferase-mediated biotinylated UTP nick end labelling, c-Jun and phosphorylated (activated) c-Jun were observed. RESULTS: XG-102 or HBO alone reduced the total infarct area by 43% and 63%, respectively. The combination diminished total infarct area by 78%, improved the neurological function and reduced brain oedema. Co-application of HBO and XG-102 also significantly reduced the cleavage of PARP, by 96% and 91% in cortical penumbra and ischaemic core, respectively. Moreover, cotreatment significantly attenuated the number of cells labelled with transferase-mediated biotinylated UTP nick end labelling and phosphorylated c-Jun. CONCLUSION: Our study demonstrates that HBO reinforces the efficiency of neuroprotective drugs such as XG-102 and vice versa. Both treatments, physical HBO and pharmacological XG-102, are already in phase I/II studies and promising strategies for clinical use.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Oxigenoterapia Hiperbárica/métodos , Infarto de la Arteria Cerebral Media/terapia , Péptidos/uso terapéutico , Envejecimiento , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/patología , Edema Encefálico/terapia , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Inhibidores Enzimáticos/administración & dosificación , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Masculino , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Péptidos/administración & dosificación , Fosforilación , Proteínas Proto-Oncogénicas c-jun/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
4.
Diabetologia ; 52(9): 1871-80, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19609503

RESUMEN

AIMS/HYPOTHESIS: In insulin-secreting cells, activation of the c-Jun NH(2)-terminal kinase (JNK) pathway triggers apoptosis. Whereas JNK1 and JNK2 are ubiquitously produced, JNK3 has been described exclusively in neurons. This report aims to characterise the expression and role in apoptosis of the three JNK isoforms in insulin-secreting cells exposed to cytokines. METHODS: Sections of human and mouse pancreases were used for immunohistochemistry studies with isoform-specific anti-JNK antibodies. Human, pig, mouse and rat pancreatic islets were isolated by enzymatic digestion and RNA or protein extracts were prepared. RNA and protein levels were determined by quantitative RT-PCR and western blotting respectively, using JNK-isoform-specific primers and isoform-specific antibodies; activities of the three JNK isoforms were determined by kinase assays following quantitative immunoprecipitation/depletion of JNK3. JNK silencing was performed with small interfering RNAs and apoptotic rates were determined in INS-1E cells by scoring cells displaying pycnotic nuclei. RESULTS: JNK3 and JNK2 mRNAs are the predominant isoforms expressed in human pancreatic islets. JNK3 is nuclear while JNK2 is also cytoplasmic. In INS-1E cells, JNK3 knockdown increases c-Jun levels and caspase-3 cleavage and sensitises cells to cytokine-induced apoptosis; in contrast, JNK1 or JNK2 knockdown is protective. CONCLUSIONS/INTERPRETATION: In insulin-secreting cells, JNK3 plays an active role in preserving pancreatic beta cell mass from cytokine attacks. The specific localisation of JNK3 in the nucleus, its recruitment by cytokines, and its effects on key transcription factors such as c-Jun, indicate that JNK3 is certainly an important player in the transcriptional control of genes expressed in insulin-secreting cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Citocinas/farmacología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Animales , Cartilla de ADN , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Humanos , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Masculino , Ratones , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Páncreas/citología , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Reacción en Cadena de la Polimerasa/métodos , ARN Mensajero/genética , Ratas , Ratas Wistar , Porcinos , Donantes de Tejidos , Venas Umbilicales
5.
Neuroscience ; 159(1): 94-103, 2009 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-19135136

RESUMEN

The c-Jun N-terminal kinase (JNK) is a mitogen-activated protein kinase (MAPK) activated by stress-signals and involved in many different diseases. Previous results proved the powerful effect of the cell permeable peptide inhibitor d-JNKI1 (d-retro-inverso form of c-Jun N-terminal kinase-inhibitor) against neuronal death in CNS diseases, but the precise features of this neuroprotection remain unclear. We here performed cell-free and in vitro experiments for a deeper characterization of d-JNKI1 features in physiological conditions. This peptide works by preventing JNK interaction with its c-Jun N-terminal kinase-binding domain (JBD) dependent targets. We here focused on the two JNK upstream MAPKKs, mitogen-activated protein kinase kinase 4 (MKK4) and mitogen-activated protein kinase kinase 7 (MKK7), because they contain a JBD homology domain. We proved that d-JNKI1 prevents MKK4 and MKK7 activity in cell-free and in vitro experiments: these MAPKK could be considered not only activators but also substrates of JNK. This means that d-JNKI1 can interrupt downstream but also upstream events along the JNK cascade, highlighting a new remarkable feature of this peptide. We also showed the lack of any direct effect of the peptide on p38, MEK1, and extracellular signal-regulated kinase (ERK) in cell free, while in rat primary cortical neurons JNK inhibition activates the MEK1-ERK-Ets1/c-Fos cascade. JNK inhibition induces a compensatory effect and leads to ERK activation via MEK1, resulting in an activation of the survival pathway-(MEK1/ERK) as a consequence of the death pathway-(JNK) inhibition. This study should hold as an important step to clarify the strong neuroprotective effect of d-JNKI1.


Asunto(s)
Corteza Cerebral/citología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa 4/metabolismo , MAP Quinasa Quinasa 7/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Factor de Transcripción Activador 2/metabolismo , Secuencia de Aminoácidos , Análisis de Varianza , Animales , Animales Recién Nacidos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular , L-Lactato Deshidrogenasa/metabolismo , Péptidos/farmacología , Fosforilación , Unión Proteica/fisiología , Dominios y Motivos de Interacción de Proteínas , Ratas , Transducción de Señal/efectos de los fármacos , Proteína Elk-1 con Dominio ets/metabolismo
6.
Cell Death Differ ; 14(2): 240-53, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16794604

RESUMEN

Excitotoxic insults induce c-Jun N-terminal kinase (JNK) activation, which leads to neuronal death and contributes to many neurological conditions such as cerebral ischemia and neurodegenerative disorders. The action of JNK can be inhibited by the D-retro-inverso form of JNK inhibitor peptide (D-JNKI1), which totally prevents death induced by N-methyl-D-aspartate (NMDA) in vitro and strongly protects against different in vivo paradigms of excitotoxicity. To obtain optimal neuroprotection, it is imperative to elucidate the prosurvival action of D-JNKI1 and the death pathways that it inhibits. In cortical neuronal cultures, we first investigate the pathways by which NMDA induces JNK activation and show a rapid and selective phosphorylation of mitogen-activated protein kinase kinase 7 (MKK7), whereas the only other known JNK activator, mitogen-activated protein kinase kinase 4 (MKK4), was unaffected. We then analyze the action of D-JNKI1 on four JNK targets containing a JNK-binding domain: MAPK-activating death domain-containing protein/differentially expressed in normal and neoplastic cells (MADD/DENN), MKK7, MKK4 and JNK-interacting protein-1 (IB1/JIP-1).


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , N-Metilaspartato/toxicidad , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neurotoxinas/toxicidad , Proteínas Adaptadoras Transductoras de Señales/aislamiento & purificación , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Calcio/metabolismo , Corteza Cerebral/enzimología , Cicloheximida/farmacología , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Electroforesis en Gel Bidimensional , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , MAP Quinasa Quinasa 7/metabolismo , Neuronas/citología , Neuronas/patología , Fosforilación/efectos de los fármacos , Proteómica , Ratas , Transducción de Señal/efectos de los fármacos
7.
Neuroscience ; 152(2): 308-20, 2008 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-18262367

RESUMEN

The c-Jun-N-terminal kinase (JNK) pathway has been shown to play an important role in excitotoxic neuronal death and several studies have demonstrated a neuroprotective effect of D-JNKi, a peptide inhibitor of JNK, in various models of cerebral ischemia. We have now investigated the effect of D-JNKi in a model of transient focal cerebral ischemia (90 min) induced by middle cerebral artery occlusion (MCAo) in adult male rats. D-JNKi (0.1 mg/kg), significantly decreased the volume of infarct, 3 days after cerebral ischemia. Sensorimotor and cognitive deficits were then evaluated over a period of 6 or 10 days after ischemia and infarct volumes were measured after behavioral testing. In behavioral studies, D-JNKi improved the general state of the animals as demonstrated by the attenuation of body weight loss and improvement in neurological score, as compared with animals receiving the vehicle. Moreover, D-JNKi decreased sensorimotor deficits in the adhesive removal test and improved cognitive function in the object recognition test. In contrast, D-JNKi did not significantly affect the infarct volume at day 6 and at day 10. This study shows that D-JNKi can improve functional recovery after transient focal cerebral ischemia in the rat and therefore supports the use of this molecule as a potential therapy for stroke.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Ataque Isquémico Transitorio/tratamiento farmacológico , Péptidos/uso terapéutico , Recuperación de la Función/efectos de los fármacos , Análisis de Varianza , Animales , Conducta Animal , Infarto Cerebral/etiología , Infarto Cerebral/prevención & control , Circulación Cerebrovascular/efectos de los fármacos , Modelos Animales de Enfermedad , Maleato de Dizocilpina/uso terapéutico , Lateralidad Funcional , Ataque Isquémico Transitorio/complicaciones , Masculino , Examen Neurológico/métodos , Fármacos Neuroprotectores/uso terapéutico , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reconocimiento en Psicología/efectos de los fármacos
8.
J Wildl Dis ; 54(1): 85-94, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29077543

RESUMEN

: Estimates of the distribution and prevalence of the sinus roundworm ( Skrjabingylus chitwoodorum) have been based largely on the inspection of skunk (Mephitidae) skulls showing damage from infections. We examined 595 striped skunks ( Mephitis mephitis) and nine hog-nosed skunks ( Conepatus leuconotus) that had tested negative for rabies by the Texas Department of State Health Services (US) between November 2010 and April 2015 to determine species of Skrjabingylus, prevalence and intensity of infection, and distribution of infection in Texas by county. We expected ecoregions with more precipitation to have higher rates of infection than more-arid ecoregions. Prevalence of S. chitwoodorum in striped skunks was 48.7%, with a mean intensity of 19.4 (SD=24.44, range=1-181) nematodes. There was a bias for the left sinus. The prevalence of infection varied among ecoregions of Texas, but it was not correlated with precipitation. Infection intensity did not vary among ecoregions. The prevalence of sinus roundworms in rabies-negative skunks suggested that behavioral changes because of skrjabingylosis might have been responsible for the submission by the public of some skunks for rabies testing.


Asunto(s)
Mephitidae , Metastrongyloidea/aislamiento & purificación , Senos Paranasales/parasitología , Infecciones por Strongylida/veterinaria , Animales , Animales Salvajes , Infecciones por Strongylida/epidemiología , Infecciones por Strongylida/parasitología , Texas/epidemiología
9.
Neuroscience ; 150(1): 40-9, 2007 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-17900813

RESUMEN

The c-Jun N-terminal kinase (JNK) signaling pathway plays a critical role in ischemic brain injury. The d-retro-inverso form of c-Jun N-terminal kinase-inhibitor (D-JNKI1), a cell-permeable inhibitor of JNK, powerfully reduces neuronal death induced by permanent and transient ischemia, even when administered 6 h after the ischemic insult, offering a clinically relevant window. We investigated the JNK molecular cascade activation in rat cerebral ischemia and the effects of D-JNKI1 on this cascade. c-Jun activation starts after 3 h after ischemia and peaks at 6 h in the ischemic core and in the penumbra at 1 h and at 6 h respectively. The 6 h c-Jun activation peak correlates well with that of P-JNK. We also examined the activation of the two direct JNK activators, MAP kinase kinase 4 (MKK4) and MAP kinase kinase 7 (MKK7). MKK4 showed the same time course as JNK in both core and penumbra, reaching peak activation at 6 h. MKK7 did not show any significant increase of phosphorylation in either core or penumbra. D-JNKI1 markedly prevented the increase of P-c-Jun in both core and penumbra and powerfully inhibited caspase-3 activation in the core. These results confirm that targeting the JNK cascade using the TAT cell-penetrating peptide offers a promising therapeutic approach for ischemia, raising hopes for human neuroprotection, and elucidates the molecular pathways leading to and following JNK activation.


Asunto(s)
Caspasa 3/metabolismo , Infarto de la Arteria Cerebral Media/enzimología , Infarto de la Arteria Cerebral Media/prevención & control , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Péptidos/administración & dosificación , Proteínas Proto-Oncogénicas c-jun/metabolismo , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
10.
Aliment Pharmacol Ther ; 26(10): 1437-46, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17900267

RESUMEN

BACKGROUND: We previously reported high prevalence of hepatitis C virus genotype 5a (HCV 5) (14%) in Central France. AIM: To identify the risk factors associated with HCV5 infection and to characterize local HCV5 lineages. METHOD: A case-control study and phylogenetic analysis were conducted. RESULTS: In all, 131 HCV5 and 343 HCV non 5 infected patients were enrolled. No HCV5 patient was born in sub-Saharan Africa and only two were injection drug user. HCV5 contamination was associated with living in a rural area called Vic le Comte (VLC) in non-transfused patients (OR = 17.7), with transfusion in patients living outside VLC (OR = 3.8) and with receiving injections in patients from VLC (OR = 3.1). More than 80% of the patients from outside VLC were contaminated by transfusion and those from VLC mainly by an iatrogenic factor - injections performed before 1972 by the local physician. Phylogenetic analysis of HCV5 isolates evidenced no distinct genetic cluster, but close relationships between the isolates of spouse pairs and between blood donors and recipients. CONCLUSIONS: Our results suggest that HCV5 spread in our district by iatrogenic route before 1972 and then via transfusion to the whole district. Collaborative studies are underway to study viral sequences from different parts of Africa and Europe to estimate the origin of our HCV 5a strains.


Asunto(s)
Hepacivirus/metabolismo , Hepatitis C/virología , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Francia/epidemiología , Genotipo , Hepatitis C/epidemiología , Hepatitis C/transmisión , Anticuerpos contra la Hepatitis C/análisis , Anticuerpos contra la Hepatitis C/sangre , Humanos , Masculino , Persona de Mediana Edad , Filogenia , Prevalencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Riesgo
11.
Mol Cell Biol ; 21(21): 7256-67, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11585908

RESUMEN

Islet-brain 1 (IB1) is the human and rat homologue of JIP-1, a scaffold protein interacting with the c-Jun amino-terminal kinase (JNK). IB1 expression is mostly restricted to the endocrine pancreas and to the central nervous system. Herein, we explored the transcriptional mechanism responsible for this preferential islet and neuronal expression of IB1. A 731-bp fragment of the 5' regulatory region of the human MAPK8IP1 gene was isolated from a human BAC library and cloned upstream of a luciferase reporter gene. This construct drove high transcriptional activity in both insulin-secreting and neuron-like cells but not in unrelated cell lines. Sequence analysis of this promoter region revealed the presence of a neuron-restrictive silencer element (NRSE) known to bind repressor zinc finger protein REST. This factor is not expressed in insulin-secreting and neuron-like cells. By mobility shift assay, we confirmed that REST binds to the NRSE present in the IB1 promoter. Once transiently transfected in beta-cell lines, the expression vector encoding REST repressed IB1 transcriptional activity. The introduction of a mutated NRSE in the 5' regulating region of the IB1 gene abolished the repression activity driven by REST in insulin-secreting beta cells and relieved the low transcriptional activity of IB1 observed in unrelated cells. Moreover, transfection in non-beta and nonneuronal cell lines of an expression vector encoding REST lacking its transcriptional repression domain relieved IB1 promoter activity. Last, the REST-mediated repression of IB1 could be abolished by trichostatin A, indicating that deacetylase activity is required to allow REST repression. Taken together, these data establish a critical role for REST in the control of the tissue-specific expression of the human IB1 gene.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Regulación Enzimológica de la Expresión Génica , Proteínas Represoras/metabolismo , Proteínas Represoras/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Células 3T3 , Animales , Secuencia de Bases , Northern Blotting , Proteínas Portadoras/metabolismo , ADN Complementario/metabolismo , Inhibidores Enzimáticos/farmacología , Biblioteca de Genes , Células HeLa , Humanos , Ácidos Hidroxámicos/farmacología , Ratones , Datos de Secuencia Molecular , Mutación , Neuronas/metabolismo , Células PC12 , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Activación Transcripcional , Transfección , Células Tumorales Cultivadas , Dedos de Zinc
12.
Aliment Pharmacol Ther ; 24(4): 593-600, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16907892

RESUMEN

AIM: To assess the rate of sustained virological response in naïve hepatitis C virus-type 5 patients treated by standard interferon or pegylated-interferon [corrected] (peg-interferon) and ribavirin combination for 48 weeks. PATIENTS AND METHODS: A total of 87 hepatitis C virus patients were included from 12 centres in France; 28 patients received interferon plus ribavirin and 59 were treated with peg-interferon plus ribavirin. RESULTS: Baseline characteristics were: mean age 58 +/- 11 years, sex ratio 1, 66% had metavir fibrosis score >or=F2, 21% were cirrhotics and 53% had pretherapeutic viral load >or=800,000 IU/mL. Sustained virological response was achieved in 64% and 58% of hepatitis C virus-5 patients treated with interferon and peg-interferon, respectively (NS). In adherent patients, sustained virological response was obtained in 75% of patients. Sustained virological response in hepatitis C virus-5 patients (60%) was significantly higher than sustained virological response in hepatitis C virus-1 patients (37%) (P = 0.0499) and not significantly different from sustained virological response in hepatitis C virus-2-3 patients (63%) (P = 0.8098). CONCLUSIONS: Combination therapy is effective in 60% of hepatitis C virus-5-infected patients. Sustained virological response seems better in hepatitis C virus-5 patients than in hepatitis C virus-1 patients, and is similar to that of hepatitis C virus-2-3 patients. More studies are needed to determine optimal duration of treatment in hepatitis C virus-5 patients.


Asunto(s)
Antivirales/uso terapéutico , Hepatitis C/tratamiento farmacológico , Interferones/uso terapéutico , Ribavirina/uso terapéutico , Combinación de Medicamentos , Femenino , Francia , Humanos , Masculino , Persona de Mediana Edad , Cooperación del Paciente , Estudios Retrospectivos , Resultado del Tratamiento
13.
Acta Trop ; 164: 259-266, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27647574

RESUMEN

In contrast to other mammalian reservoirs, many bat species migrate long-distances and have the potential to introduce exotic pathogens to new areas. Bats have long been associated with blood-borne protozoal trypanosomes of the Schizotrypanum subgenus, which includes the zoonotic parasite Trypanosoma cruzi, agent of Chagas disease. Another member of the subgenus, Trypanosoma dionisii, infects bats of Europe and South America, and genetic similarities between strains from the two continents suggest transcontinental movement of this parasite via bats. Despite the known presence of diverse trypanosomes in bats of Central and South America, and the presence of T. cruzi-infected vectors and wildlife in the US, the role of bats in maintaining and dispersing trypanosomes in the US has not yet been reported. We collected hearts and blood from 8 species of insectivorous bats from 30 counties across Texas. Using PCR and DNA sequencing, we tested 593 bats for trypanosomes and found 1 bat positive for T. cruzi (0.17%), 9 for T. dionisii (1.5%), and 5 for Blastocrithidia spp. (0.8%), a group of insect trypanosomes. The T. cruzi-infected bat was carrying TcI, the strain type associated with human disease in the US. In the T. dionisii-infected bats, we detected three unique variants associated with the three infected bat species. These findings represent the first report of T. cruzi in a bat in the US, of T. dionisii in North America, and of Blastocrithidia spp. in mammals, and underscore the importance of bats in the maintenance of trypanosomes, including agents of human and animal disease, across broad geographic locales.


Asunto(s)
Enfermedad de Chagas/transmisión , Quirópteros/parasitología , Trypanosoma cruzi/aislamiento & purificación , Animales , Enfermedad de Chagas/parasitología , ADN Protozoario/genética , ADN Ribosómico/genética , Humanos , Filogenia , Análisis de Secuencia de ADN , Texas/epidemiología , Trypanosoma cruzi/genética
14.
J Neurosci ; 23(24): 8596-607, 2003 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-13679429

RESUMEN

Hearing loss can be caused by a variety of insults, including acoustic trauma and exposure to ototoxins, that principally effect the viability of sensory hair cells via the MAP kinase (MAPK) cell death signaling pathway that incorporates c-Jun N-terminal kinase (JNK). We evaluated the otoprotective efficacy of D-JNKI-1, a cell permeable peptide that blocks the MAPK-JNK signal pathway. The experimental studies included organ cultures of neonatal mouse cochlea exposed to an ototoxic drug and cochleae of adult guinea pigs that were exposed to either an ototoxic drug or acoustic trauma. Results obtained from the organ of Corti explants demonstrated that the MAPK-JNK signal pathway is associated with injury and that blocking of this signal pathway prevented apoptosis in areas of aminoglycoside damage. Treatment of the neomycin-exposed organ of Corti explants with D-JNKI-1 completely prevented hair cell death initiated by this ototoxin. Results from in vivo studies showed that direct application of D-JNKI-1 into the scala tympani of the guinea pig cochlea prevented nearly all hair cell death and permanent hearing loss induced by neomycin ototoxicity. Local delivery of D-JNKI-1 also prevented acoustic trauma-induced permanent hearing loss in a dose-dependent manner. These results indicate that the MAPK-JNK signal pathway is involved in both ototoxicity and acoustic trauma-induced hair cell loss and permanent hearing loss. Blocking this signal pathway with D-JNKI-1 is of potential therapeutic value for long-term protection of both the morphological integrity and physiological function of the organ of Corti during times of oxidative stress.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Células Ciliadas Auditivas/efectos de los fármacos , Pérdida Auditiva Provocada por Ruido/prevención & control , Pérdida Auditiva/prevención & control , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Órgano Espiral/efectos de los fármacos , Péptidos/farmacología , Estimulación Acústica , Aminoglicósidos/antagonistas & inhibidores , Aminoglicósidos/toxicidad , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Evaluación Preclínica de Medicamentos , Cobayas , Células Ciliadas Auditivas/citología , Pérdida Auditiva/inducido químicamente , Pruebas Auditivas , Técnicas In Vitro , Proteínas Quinasas JNK Activadas por Mitógenos , Ligandos , Ratones , Fármacos Neuroprotectores/farmacología , Órgano Espiral/citología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal/efectos de los fármacos
15.
Biochim Biophys Acta ; 1089(2): 213-9, 1991 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-1711372

RESUMEN

We previously reported about Escherichia coli transformation experiments yielding streptomycin-resistant cells carrying a C912 to T transition in a plasmid-born 16S rRNA gene. These experiments were based on results obtained with streptomycin-resistant Euglena chloroplasts bearing an equivalent mutation in the single chloroplast 16S rRNA gene. We extended this study and transformed E. coli with plasmid constructs having a mutated 16S rRNA gene at position 914 (A to C) or a double mutation at positions 912 and 888 (C to T:G to A) or a mutation in the S12 gene (Lys-42 to Thr). We tested the transformed cells before and after a screening procedure in the presence of streptomycin. We find that the plasmid-born mutations protect colonies against a short streptomycin exposure, but ribosomes carrying mutated 16S rRNA do not significantly reduce codon misreading in vitro. However, ribosomes isolated from transformed cells after the screening procedure resist misreading. These ribosomes have acquired a second mutation in the S12 protein as shown in one case by sequencing and by transformation experiments. Furthermore, we show that the A914 to C mutation prevents (strongly reduces) base methylation in the central domain of 16S rRNA.


Asunto(s)
Farmacorresistencia Microbiana/genética , Escherichia coli/genética , Mutación , Estreptomicina/farmacología , Secuencia de Bases , Cloroplastos , Escherichia coli/efectos de los fármacos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Plásmidos , ARN Bacteriano/genética , ARN Ribosómico 16S/genética , Proteínas Ribosómicas/genética
16.
Diabetes ; 50(1): 77-82, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11147798

RESUMEN

Stress conditions and proinflammatory cytokines activate the c-Jun NH2-terminal kinase (JNK), a member of the stress-activated group of mitogen-activated protein kinases (MAPKs). We recently demonstrated that inhibition of JNK signaling with the use of the islet-brain (IB) 1 and 2 proteins prevented interleukin (IL)-1beta-induced pancreatic beta-cell death. Bioactive cell-permeable peptide inhibitors of JNK were engineered by linking the minimal 20-amino acid inhibitory domains of the IB proteins to the 10-amino acid HIV-TAT sequence that rapidly translocates inside cells. Kinase assays indicate that the inhibitors block activation of the transcription factor c-Jun by JNK. Addition of the peptides to the insulin-secreting betaTC-3 cell line results in a marked inhibition of IL-1beta-induced c-jun and c-fos expression. The peptides protect betaTC-3 cells against apoptosis induced by IL-1beta. All-D retro-inverso peptides penetrate cells as efficiently as the L-enantiomers, decrease c-Jun activation by JNK, and remain highly stable inside cells. These latter peptides confer full protection against IL-1beta-induced apoptosis for up to 2 weeks of continual treatment with IL-1beta. These data establish these bioactive cell-permeable peptides as potent pharmacological compounds that decrease intracellular JNK signaling and confer long-term protection to pancreatic beta-cells from IL-1beta-induced apoptosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/farmacocinética , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/fisiología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Péptidos/farmacología , Péptidos/farmacocinética , Secuencia de Aminoácidos/genética , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Permeabilidad de la Membrana Celular , Secuencia Conservada/genética , Humanos , Interleucina-1/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Péptidos/síntesis química , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-jun/antagonistas & inhibidores , Transactivadores/genética
17.
Diabetes ; 49(9): 1468-76, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10969830

RESUMEN

To characterize the differentiation events that selectively target insulin-producing cells to interleukin (IL)-1beta-induced apoptosis, we studied IL-1beta signaling via mitogen-activated protein kinase (MAPK) and stress-activated protein kinase in 2 pancreatic endocrine cell lines. We studied the glucagon-secreting AN-glu cell line and the insulin and the islet amyloid polypeptide-producing beta-cell line (AN-ins cells), which is derived by stable transfection of AN-glu cells with the transcription factor pancreatic duodenal homeobox factor-1. AN-ins cells were more sensitive to the cytotoxic action of IL-1beta. This increased sensitivity was not associated with a more pronounced IL-l-induced nitric oxide production in AN-ins cells, but it correlated with a more marked activation of the 3 MAPKs extracellular signal-regulated kinases (ERKs)-1/2, c-Jun NH2-terminal kinase (JNK), and p38 MAPK (p38). This led to increased phosphorylation of the transcription factors c-Jun, Elk-1, and ATF2 and of heat shock protein 25. Inhibition of ERK-1/2 and p38 did not prevent but aggravated IL-1beta-induced cell death. In contrast, inhibition of JNK by transfection with the dominant negative inhibitor of the JNK-binding domain prevented apoptosis in both cell types. Cell death could be elicited by overexpressing the catalytic domain of MAPK kinase kinase 1, a specific activator of JNK and nuclear factor-kappaB, which does not recruit ERK-1/2 or p38. Coactivation of ERK-1/2 with JNK did not prevent apoptosis. In conclusion, increased MAPK signaling in response to IL-1beta may represent a novel molecular marker of beta-cell differentiation. JNK inhibition represents an effective means of preventing IL-1beta-activated beta-cell destruction.


Asunto(s)
Apoptosis/fisiología , Diferenciación Celular/fisiología , Interleucina-1/farmacología , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Proteínas de Homeodominio/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Islotes Pancreáticos/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Óxido Nítrico/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transactivadores/metabolismo , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos
18.
Mol Endocrinol ; 9(10): 1413-26, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8544849

RESUMEN

A defect in glucose sensing of the pancreatic beta-cells has been observed in several animal models of type II diabetes and has been correlated with a reduced gene expression of the glucose transporter type 2 (Glut2). In a transgenic mouse model, expression of Glut2 antisense RNA in pancreatic beta-cells has recently been shown to be associated with an impaired glucose-induced insulin secretion and the development of diabetes. To identify factors that may be involved in the specific decrease of Glut2 in the beta-cells of the diabetic animal, an attempt was made to localize the cis-elements and trans-acting factors involved in the control of Glut2 expression in the endocrine pancreas. It was demonstrated by transient transfection studies that only 338 base pairs (bp) of the murine Glut2 proximal promoter are needed for reporter gene expression in pancreatic islet-derived cell lines, whereas no activity was detected in nonpancreatic cells. Three cis-elements, GTI, GTII, and GTIII, have been identified by DNAse I footprinting and gel retardation experiments within these 338 bp. GTI and GTIII bind distinct but ubiquitously expressed trans-acting factors. On the other hand, nuclear proteins specifically expressed in pancreatic cell lines interact with GTII, and their relative abundance correlates with endogenous Glut2 expression. These GTII-binding factors correspond to nuclear proteins of 180 and 90 kilodaltons as defined by Southwestern analysis. The 180-kilodalton factor is present in pancreatic beta-cell lines but not in an alpha-cell line. Mutation of the GTI or GTIII cis-elements decreases transcriptional activity directed by the 338-bp promoter, whereas mutation of GTII increases gene transcription. Thus negative and positive regulatory sequences are identified within the proximal 338 bp of the GLUT2 promoter and may participate in the islet-specific expression of the gene by binding beta-cell specific trans-acting factors.


Asunto(s)
Islotes Pancreáticos/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Transactivadores/genética , Activación Transcripcional , Animales , Secuencia de Bases , Huella de ADN , Transportador de Glucosa de Tipo 2 , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/metabolismo , Ratas , Análisis de Secuencia , Células Tumorales Cultivadas
19.
Mol Endocrinol ; 10(11): 1327-34, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8923459

RESUMEN

The homeodomain protein PDX-1, referred as IPF-1/STF-1/IDX-1, is a transcriptional factor that plays a critical role in the control of several genes expressed in the pancreatic islet. PDX-1 gene expression has been previously shown to be reduced in cultured beta-cell lines chronically exposed to high glucose concentrations. As the glucose transporter type 2 (GLUT2) gene expression is selectively decreased in the beta-pancreatic cells of experimental models of diabetes, we postulated that the loss of GLUT2 gene expression in the pancreatic islets of diabetic animals may be due to the loss of PDX-1 transacting function on the GLUT2 gene. We, therefore, investigated the potential role of PDX-1 in the transcriptional control of GLUT2. We have identified a repeat of a TAAT motif (5'-TAATA-ATAACA-3') conserved in the sequence of the human and murine GLUT2 promoters. Recombinant PDX-1 binds to this GLUT2TAAT motif in electrophoretic mobility shift experiments. PDX-1 antiserum detects the formation of the complex of PDX-1 with the GLUT2TAAT motif in nuclear extracts from the pancreatic insulin-secreting cell line, beta TC3. The GLUT2TAAT motif was mutated in the murine GLUT2 promoter (-1308/+49 bp) linked to a luciferase reporter gene and transfected into beta TC3 cells. Compared with the transcriptional activity of the wild type promoter, that of the mutated promoter decreases by 41%. Multiple copies of the GLUT2TAAT motif were ligated 5' to a heterologous promoter and transfected into a PDX-1-expressing cell line (beta TC3) and into cell lines lacking the homeobox factor (InR1-G9 and JEG-3). The GLUT2TAAT motif mediates the activation of the heterologous promoter in the PDX-1-expressing cell line but not in InR1-G9 or JEG-3 cell lines. Furthermore, cotransfection in a PDX-1-deficient cell line with the expression vector encoding PDX-1 transactivates specifically the heterologous promoter containing the multimerized GLUT2TAAT motif. These data demonstrate that the murine GLUT2 promoter is controlled by the PDX-1 homeobox factor through the identified GLUT2TAAT motif.


Asunto(s)
Proteínas de Homeodominio , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Transactivadores/metabolismo , Activación Transcripcional , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Elementos de Facilitación Genéticos , Transportador de Glucosa de Tipo 2 , Humanos , Insulina/genética , Insulina/metabolismo , Ratones , Regiones Promotoras Genéticas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Somatostatina/genética , Somatostatina/metabolismo , Transactivadores/genética , Transfección
20.
Mol Cell Endocrinol ; 114(1-2): 205-15, 1995 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-8674846

RESUMEN

The high Km glucose transporter GLUT2 is a membrane protein expressed in tissues involved in maintaining glucose homeostasis, and in cells where glucose-sensing is necessary. In many experimental models of diabetes, GLUT2 gene expression is decreased in pancreatic beta-cells, which could lead to a loss of glucose-induced insulin secretion. In order to identify factors involved in pancreatic beta-cell specific expression of GLUT2, we have recently cloned the murine GLUT2 promoter and identified cis-elements within the 338-bp of the proximal promoter capable of binding islet-specific trans-acting factors. Furthermore, in transient transfection studies, this 338-bp fragment could efficiently drive the expression of the chloramphenicol acetyl transferase (CAT) gene in cell lines derived from the endocrine pancreas, but displayed no promoter activity in non-pancreatic cells. In this report, we tested the cell-specific expression of a CAT reporter gene driven by a short (338 bp) and a larger (1311 bp) fragment of the GLUT2 promoter in transgenic mice. We generated ten transgenic lines that integrated one of the constructs. CAT mRNA expression in transgenic tissues was assessed using the RNAse protection assay and the quantitative reverse transcribed polymerase chain reaction (RT-PCR). Overall CAT mRNA expression for both constructs was low compared to endogenous GLUT2 mRNA levels but the reporter transcript could be detected in all animals in the pancreatic islets and the liver, and in a few transgenic lines in the kidney and the small intestine. The CAT protein was also present in Langerhans islets and in the liver for both constructs by immunocytochemistry. These findings suggest that the proximal 338 bp of the murine GLUT2 promoter contain cis-elements required for the islet-specific expression of GLUT2.


Asunto(s)
Islotes Pancreáticos/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Regiones Promotoras Genéticas , Animales , Secuencia de Bases , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Clonación Molecular , Cartilla de ADN/genética , Expresión Génica , Genes Reporteros , Transportador de Glucosa de Tipo 2 , Inmunohistoquímica , Hígado/metabolismo , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA