Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 39(49): 9702-9715, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31685654

RESUMO

Nerve growth factor (NGF) is a key mediator of nociception, acting during the development and differentiation of dorsal root ganglion (DRG) neurons, and on adult DRG neuron sensitization to painful stimuli. NGF also has central actions in the brain, where it regulates the phenotypic maintenance of cholinergic neurons. The physiological function of NGF as a pain mediator is altered in patients with Hereditary Sensory and Autonomic Neuropathy type V (HSAN V), caused by the 661C>T transition in the Ngf gene, resulting in the R100W missense mutation in mature NGF. Homozygous HSAN V patients present with congenital pain insensitivity, but are cognitively normal. This led us to hypothesize that the R100W mutation may differentially affect the central and peripheral actions of NGF. To test this hypothesis and provide a mechanistic basis to the HSAN V phenotype, we generated transgenic mice harboring the human 661C>T mutation in the Ngf gene and studied both males and females. We demonstrate that heterozygous NGFR100W/wt mice display impaired nociception. DRG neurons of NGFR100W/wt mice are morphologically normal, with no alteration in the different DRG subpopulations, whereas skin innervation is reduced. The NGFR100W protein has reduced capability to activate pain-specific signaling, paralleling its reduced ability to induce mechanical allodynia. Surprisingly, however, NGFR100W/wt mice, unlike heterozygous mNGF+/- mice, show no learning or memory deficits, despite a reduction in secretion and brain levels of NGF. The results exclude haploinsufficiency of NGF as a mechanistic cause for heterozygous HSAN V mice and demonstrate a specific effect of the R100W mutation on nociception.SIGNIFICANCE STATEMENT The R100W mutation in nerve growth factor (NGF) causes Hereditary Sensory and Autonomic Neuropathy type V, a rare disease characterized by impaired nociception, even in apparently clinically silent heterozygotes. For the first time, we generated and characterized heterozygous knock-in mice carrying the human R100W-mutated allele (NGFR100W/wt). Mutant mice have normal nociceptor populations, which, however, display decreased activation of pain transduction pathways. NGFR100W interferes with peripheral and central NGF bioavailability, but this does not impact on CNS function, as demonstrated by normal learning and memory, in contrast with heterozygous NGF knock-out mice. Thus, a point mutation allows neurotrophic and pronociceptive functions of NGF to be split, with interesting implications for the treatment of chronic pain.


Assuntos
Cognição , Neuropatias Hereditárias Sensoriais e Autônomas/genética , Neuropatias Hereditárias Sensoriais e Autônomas/fisiopatologia , Mutação/genética , Fator de Crescimento Neural/genética , Nociceptividade , Animais , Comportamento Animal , Feminino , Gânglios Espinais/patologia , Técnicas de Introdução de Genes , Neuropatias Hereditárias Sensoriais e Autônomas/psicologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto/genética , Medição da Dor , Percepção da Dor , Desempenho Psicomotor , Ratos , Ratos Wistar , Pele/inervação
2.
Int J Mol Sci ; 20(17)2019 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-31450553

RESUMO

The correct morphofunctional shaping of the cerebral cortex requires a continuous interaction between intrinsic (genes/molecules expressed within the tissue) and extrinsic (e.g., neural activity) factors at all developmental stages. Forkhead Box G1 (FOXG1) is an evolutionarily conserved transcription factor, essential for the cerebral cortex patterning and layering. FOXG1-related disorders, including the congenital form of Rett syndrome, can be caused by deletions, intragenic mutations or duplications. These genetic alterations are associated with a complex phenotypic spectrum, spanning from intellectual disability, microcephaly, to autistic features, and epilepsy. We investigated the functional correlates of dysregulated gene expression by performing electrophysiological assays on FoxG1+/- mice. Local Field Potential (LFP) recordings on freely moving animals detected cortical hyperexcitability. On the other hand, patch-clamp recordings showed a downregulation of spontaneous glutamatergic transmission. These findings were accompanied by overactivation of Akt/S6 signaling. Furthermore, the expression of vesicular glutamate transporter 2 (vGluT2) was increased, whereas the level of the potassium/chloride cotransporter KCC2 was reduced, thus indicating a higher excitation/inhibition ratio. Our findings provide evidence that altered expression of a key gene for cortical development can result in specific alterations in neural circuit function at the macro- and micro-scale, along with dysregulated intracellular signaling and expression of proteins controlling circuit excitability.


Assuntos
Córtex Cerebral/metabolismo , Córtex Cerebral/fisiopatologia , Epilepsia/genética , Epilepsia/metabolismo , Fatores de Transcrição Forkhead/genética , Proteínas do Tecido Nervoso/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Transmissão Sináptica , Animais , Modelos Animais de Doenças , Suscetibilidade a Doenças , Epilepsia/fisiopatologia , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Camundongos , Camundongos Knockout , Fenótipo , Convulsões , Transdução de Sinais , Potenciais Sinápticos
3.
Neuropharmacology ; 148: 305-310, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30639390

RESUMO

The Forkhead Box G1 (FOXG1) gene encodes a transcription factor with an essential role in mammalian telencephalon development. FOXG1-related disorders, caused by deletions, intragenic mutations or duplications, are usually associated with severe intellectual disability, autistic features, and, in 87% of subjects, epileptiform manifestations. In a subset of patients with FoxG1 mutations, seizures remain intractable, prompting the need for novel therapeutic options. To address this issue, we took advantage of a haploinsufficient animal model, the FoxG1+/- mouse. In vivo electrophysiological analyses of FoxG1+/- mice detected hippocampal hyperexcitability, which turned into overt seizures upon delivery of the proconvulsant kainic acid, as confirmed by behavioral observations. These alterations were associated with decreased expression of the chloride transporter KCC2. Next, we tested whether a triheptanoin-based anaplerotic diet could have an impact on the pathological phenotype of FoxG1+/- mice. This manipulation abated altered neural activity and normalized enhanced susceptibility to proconvulsant-induced seizures, in addition to rescuing altered expression of KCC2 and increasing the levels of the GABA transporter vGAT. In conclusion, our data show that FoxG1 haploinsufficiency causes dysfunction of hippocampal circuits and increases the susceptibility to a proconvulsant insult, and that these alterations are rescued by triheptanoin dietary treatment.


Assuntos
Suscetibilidade a Doenças/dietoterapia , Fatores de Transcrição Forkhead/genética , Haploinsuficiência , Proteínas do Tecido Nervoso/genética , Convulsões/dietoterapia , Triglicerídeos/uso terapêutico , Animais , Suscetibilidade a Doenças/fisiopatologia , Fatores de Transcrição Forkhead/fisiologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Ácido Caínico , Camundongos , Proteínas do Tecido Nervoso/fisiologia , Convulsões/induzido quimicamente , Convulsões/fisiopatologia , Convulsões/prevenção & controle , Simportadores/biossíntese , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Cotransportadores de K e Cl-
4.
Nanomedicine ; 14(7): 2191-2203, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30016718

RESUMO

To improve the efficiency of pancreatic islet transplantation, we performed in-vitro and in-vivo experiments with isolated human pancreatic islets coated by multi-layer nano-encapsulation using differently charged polymers [chitosan and poly(sodium styrene sulfonate)] to obtain up to 9 layers. The islet coating (thickness: 104.2 ±â€¯4.2 nm) was uniform, with ≥ 90% cell viability and well preserved beta- and alpha-cell ultrastructure. Nano-encapsulated islets maintained physiological glucose-stimulated insulin secretion by both static incubation and perifusion studies. Notably, palmitate- or cytokine-induced toxicity was significantly reduced in nano-coated islets. Xenotransplantation of nano-encapsulated islets under the kidney capsule of streptozotocin-induced C57Bl/6J diabetic mice allowed long term normal or near normal glycemia, associated with minimal infiltration of immune cell into the grafts, well preserved islet morphology and signs of re-vascularization. In summary, the multi-layer nano-encapsulation approach described in the present study provides a promising tool to effectively protect human islets both in-vitro andin-vivo conditions.


Assuntos
Materiais Revestidos Biocompatíveis/química , Diabetes Mellitus Experimental/prevenção & controle , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/citologia , Nanoestruturas/administração & dosagem , Animais , Glicemia/análise , Células Cultivadas , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanoestruturas/química , Transplante Heterólogo
5.
Oncotarget ; 8(14): 23539-23550, 2017 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-28212563

RESUMO

Glioblastoma (GBM) is the most aggressive type of brain tumor. In this context, animal models represent excellent tools for the early detection and longitudinal mapping of neuronal dysfunction, that are critical in the preclinical validation of new therapeutic strategies. In a mouse glioma model, we developed sensitive behavioral readouts that allow early recognizing and following neurological symptoms. We injected GL261 cells into the primary motor cortex of syngenic mice and we used a battery of behavioral tests to longitudinally monitor the dysfunction induced by tumor growth. Grip strength test revealed an early onset of functional deficit associated to the glioma growth, with a significant forelimb weakness appearing 9 days after tumor inoculation. A later deficit was observed in the rotarod and in the grid walk tasks. Using this model, we found reduced tumor growth and maintenance of behavioral functions following treatment with Cytotoxic Necrotizing Factor 1 (CNF1) at a symptomatic stage. Our data provide a detailed and precise examination of how tumor growth reverberates on the behavioral functions of glioma-bearing mice, providing normative data for the study of therapeutic strategies for glioma treatment. The reduced tumor volume and robust functional sparing observed in CNF1-treated, glioma-bearing mice strengthen the notion that CNF1 delivery is a promising strategy for glioma therapy.


Assuntos
Toxinas Bacterianas/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Proteínas de Escherichia coli/farmacologia , Glioma/tratamento farmacológico , Transtornos Motores/tratamento farmacológico , Análise de Variância , Animais , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/fisiopatologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Proteína 4 Homóloga a Disks-Large , Glioma/patologia , Glioma/fisiopatologia , Guanilato Quinases/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Transtornos Motores/fisiopatologia , Fatores de Tempo , Carga Tumoral/efeitos dos fármacos
6.
Neuro Oncol ; 18(12): 1634-1643, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27298309

RESUMO

BACKGROUND: Glioblastomas are the most aggressive type of brain tumor. A successful treatment should aim at halting tumor growth and protecting neuronal cells to prevent functional deficits and cognitive deterioration. Here, we exploited a Rho GTPase-activating bacterial protein toxin, cytotoxic necrotizing factor 1 (CNF1), to interfere with glioma cell growth in vitro and vivo. We also investigated whether this toxin spares neuron structure and function in peritumoral areas. METHODS: We performed a microarray transcriptomic and in-depth proteomic analysis to characterize the molecular changes triggered by CNF1 in glioma cells. We also examined tumor cell senescence and growth in vehicle- and CNF1-treated glioma-bearing mice. Electrophysiological and morphological techniques were used to investigate neuronal alterations in peritumoral cortical areas. RESULTS: Administration of CNF1 triggered molecular and morphological hallmarks of senescence in mouse and human glioma cells in vitro. CNF1 treatment in vivo induced glioma cell senescence and potently reduced tumor volumes. In peritumoral areas of glioma-bearing mice, neurons showed a shrunken dendritic arbor and severe functional alterations such as increased spontaneous activity and reduced visual responsiveness. CNF1 treatment enhanced dendritic length and improved several physiological properties of pyramidal neurons, demonstrating functional preservation of the cortical network. CONCLUSIONS: Our findings demonstrate that CNF1 reduces glioma volume while at the same time maintaining the physiological and structural properties of peritumoral neurons. These data indicate a promising strategy for the development of more effective antiglioma therapies.


Assuntos
Toxinas Bacterianas/administração & dosagem , Neoplasias Encefálicas/fisiopatologia , Proteínas de Escherichia coli/administração & dosagem , Proteínas Ativadoras de GTPase/metabolismo , Glioblastoma/fisiopatologia , Neurônios/fisiologia , Animais , Toxinas Bacterianas/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Córtex Cerebral/fisiologia , Eletrofisiologia , Proteínas de Escherichia coli/farmacologia , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Humanos , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Proteômica , Transcriptoma
7.
J Neurosci ; 36(12): 3430-40, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-27013673

RESUMO

During an early phase of enhanced sensitivity called the critical period (CP), monocular deprivation causes a shift in the response of visual cortex binocular neurons in favor of the nondeprived eye, a process named ocular dominance (OD) plasticity. While the time course of the CP for OD plasticity can be modulated by genetic/pharmacological interventions targeting GABAergic inhibition, whether an increased sensory-motor experience can affect this major plastic phenomenon is not known. We report that exposure to environmental enrichment (EE) accelerated the closure of the CP for OD plasticity in the rat visual cortex. Histone H3 acetylation was developmentally regulated in primary visual cortex, with enhanced levels being detectable early in enriched pups, and chromatin immunoprecipitation revealed an increase at the level of the BDNF P3 promoter. Administration of the histone deacetylase inhibitor SAHA (suberoylanilide hydroxamic acid) to animals reared in a standard cage mimicked the increase in H3 acetylation observed in the visual cortex and resulted in an accelerated decay of OD plasticity. Finally, exposure to EE in adulthood upregulated H3 acetylation and was paralleled by a reopening of the CP. These findings demonstrate a critical involvement of the epigenetic machinery as a mediator of visual cortex developmental plasticity and of the impact of EE on OD plasticity. SIGNIFICANCE STATEMENT: While it is known that an epigenetic remodeling of chromatin structure controls developmental plasticity in the visual cortex, three main questions have remained open. Which is the physiological time course of histone modifications? Is it possible, by manipulating the chromatin epigenetic state, to modulate plasticity levels during the critical period? How can we regulate histone acetylation in the adult brain in a noninvasive manner? We show that the early exposure of rat pups to enriching environmental conditions accelerates the critical period for plasticity in the primary visual cortex, linking this effect to increased histone acetylation, specifically at the BDNF gene level. Moreover, we report that the exposure of adult animals to environmental enrichment enhances histone acetylation and reopens juvenile-like plasticity.


Assuntos
Período Crítico Psicológico , Epigênese Genética/genética , Histonas/fisiologia , Plasticidade Neuronal/genética , Processamento de Proteína Pós-Traducional/genética , Córtex Visual/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Feminino , Aprendizagem/fisiologia , Masculino , Ratos
8.
Eur J Hum Genet ; 24(6): 871-80, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26443267

RESUMO

Rett syndrome (RTT) is a severe neurodevelopmental disorder associated with mutations in either MECP2, CDKL5 or FOXG1. The precise molecular mechanisms that lead to the pathogenesis of RTT have yet to be elucidated. We recently reported that expression of GluD1 (orphan glutamate receptor δ-1 subunit) is increased in iPSC-derived neurons obtained from patients with mutations in either MECP2 or CDKL5. GluD1 controls synaptic differentiation and shifts the balance between excitatory and inhibitory synapses toward the latter. Thus, an increase in GluD1 might be a critical factor in the etiology of RTT by affecting the excitatory/inhibitory balance in the developing brain. To test this hypothesis, we generated iPSC-derived neurons from FOXG1(+/-) patients. We analyzed mRNA and protein levels of GluD1 together with key markers of excitatory and inhibitory synapses in these iPSC-derived neurons and in Foxg1(+/-) mouse fetal (E11.5) and adult (P70) brains. We found strong correlation between iPSC-derived neurons and fetal mouse brains, where GluD1 and inhibitory synaptic markers (GAD67 and GABA AR-α1) were increased, whereas the levels of a number of excitatory synaptic markers (VGLUT1, GluA1, GluN1 and PSD-95) were decreased. In adult mice, GluD1 was decreased along with all GABAergic and glutamatergic markers. Our findings further the understanding of the etiology of RTT by introducing a new pathological event occurring in the brain of FOXG1(+/-) patients during embryonic development and its time-dependent shift toward a general decrease in brain synapses.


Assuntos
Fatores de Transcrição Forkhead/genética , Glutamato Desidrogenase/metabolismo , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Receptores de Glutamato/metabolismo , Síndrome de Rett/metabolismo , Adolescente , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Células Cultivadas , Feminino , Glutamato Desidrogenase/genética , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Lactente , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese , Neurônios/citologia , Receptores de Glutamato/genética , Síndrome de Rett/genética , Sinapses/metabolismo
9.
Proc Natl Acad Sci U S A ; 112(45): 13910-5, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26508630

RESUMO

Forkhead box g1 (Foxg1) is a nuclear-cytosolic transcription factor essential for the forebrain development and involved in neurodevelopmental and cancer pathologies. Despite the importance of this protein, little is known about the modalities by which it exerts such a large number of cellular functions. Here we show that a fraction of Foxg1 is localized within the mitochondria in cell lines, primary neuronal or glial cell cultures, and in the mouse cortex. Import of Foxg1 in isolated mitochondria appears to be membrane potential-dependent. Amino acids (aa) 277-302 were identified as critical for mitochondrial localization. Overexpression of full-length Foxg1 enhanced mitochondrial membrane potential (ΔΨm) and promoted mitochondrial fission and mitosis. Conversely, overexpression of the C-term Foxg1 (aa 272-481), which is selectively localized in the mitochondrial matrix, enhanced organelle fusion and promoted the early phase of neuronal differentiation. These findings suggest that the different subcellular localizations of Foxg1 control the machinery that brings about cell differentiation, replication, and bioenergetics, possibly linking mitochondrial functions to embryonic development and pathological conditions.


Assuntos
Diferenciação Celular , Metabolismo Energético , Fatores de Transcrição Forkhead/metabolismo , Mitocôndrias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Linhagem Celular , Fatores de Transcrição Forkhead/genética , Proteínas de Fluorescência Verde/genética , Potencial da Membrana Mitocondrial , Camundongos , Proteínas do Tecido Nervoso/genética
10.
Diabetologia ; 57(2): 362-5, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24233056

RESUMO

AIMS/HYPOTHESIS: Previous work has demonstrated that beta cell amount (whether measured as beta cell mass, beta cell volume or insulin-positive area) is decreased in type 2 diabetes; however, recent findings suggest that mechanisms other than death may contribute to beta cell failure in this disease. To better characterise beta cell mass and function in type 2 diabetes, we performed morphological, ultra-structural and functional studies using histological samples and isolated islets. METHODS: Pancreases from ten non-diabetic (ND) and ten matched type 2 diabetic organ donors were studied by insulin, glucagon and chromogranin A immunocytochemistry and electron microscopy (EM). Glucose-stimulated insulin secretion was assessed using isolated islets and studies were performed using independent ND islet preparations after 24 h exposure to 22.2 mmol/l glucose. RESULTS: Immunocytochemistry showed that the fractional islet insulin-positive area was lower in type 2 diabetic islets (54.9 ± 6.3% vs 72.1 ± 8.7%, p < 0.01), whereas glucagon (23.3 ± 5.4% vs 20.2 ± 5.3%) and chromogranin A (86.4 ± 6.1% vs 89.0 ± 5.5%) staining was similar between the two groups. EM showed that the proportion of beta cells in type 2 diabetic islets was only marginally decreased; marked beta cell degranulation was found in diabetic beta cells; these findings were all reproduced after exposing isolated ND islets to high glucose. Glucose-stimulated insulin secretion was 40­50% lower from type 2 diabetic islets (p < 0.01), which again was mimicked by culturing non-diabetic islets in high glucose. CONCLUSIONS/INTERPRETATION: These results suggest that, at least in subgroups of type 2 diabetic patients, the loss of beta cells as assessed so far might be overestimated, possibly due to changes in beta cell phenotype other than death, also contributing to beta cell failure in type 2 diabetes.


Assuntos
Cromogranina A/metabolismo , Diabetes Mellitus Tipo 2/patologia , Glucagon/metabolismo , Células Secretoras de Insulina/patologia , Insulina/metabolismo , Pâncreas/patologia , Idoso , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Microscopia Eletrônica
11.
J Biol Chem ; 282(51): 37146-57, 2007 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-17942396

RESUMO

The long terminal repeat of human immunodeficiency virus, type 1 (HIV-1) contains an NF-kappaB enhancer and is potently inhibited by IkappaB-alphaS32/36A, a proteolysis-resistant inhibitor of NF-kappaB transacting factors. The evidence that NF-kappaB is dispensable for HIV-1 expression raises the question of whether IkappaB-alpha represses the HIV-1 transcription by mechanisms distinct from NF-kappaB inhibition. Here, we report that IkappaB-alpha negatively regulates the HIV-1 expression and replication in an NF-kappaB-independent manner by directly binding to Tat, which results in the nuclear export and cytoplasmic sequestration of the viral transactivator. The sequence of IkappaB-alpha required for Tat inhibition spans from amino acids 72 to 287 and includes the nuclear localization signal, the carboxyl-terminal nuclear export signal, and the binding site for the arginine-rich domain of Tat. This novel mechanism of cross-talk between Tat and IkappaB-alpha provides further insights into the mechanisms of HIV-1 regulation and could assist in the development of novel strategies for AIDS therapy.


Assuntos
Síndrome da Imunodeficiência Adquirida/metabolismo , Núcleo Celular/metabolismo , Elementos Facilitadores Genéticos , Regulação Viral da Expressão Gênica , HIV-1/metabolismo , Proteínas I-kappa B/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Síndrome da Imunodeficiência Adquirida/genética , Síndrome da Imunodeficiência Adquirida/terapia , Transporte Ativo do Núcleo Celular/genética , Sítios de Ligação/genética , Núcleo Celular/genética , Núcleo Celular/virologia , Elementos Facilitadores Genéticos/genética , Regulação Viral da Expressão Gênica/genética , Células HeLa , Humanos , Proteínas I-kappa B/genética , Inibidor de NF-kappaB alfa , NF-kappa B/genética , NF-kappa B/metabolismo , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Ligação Proteica/genética , Transcrição Gênica/genética , Replicação Viral/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
12.
J Biol Chem ; 277(35): 31448-58, 2002 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-12055184

RESUMO

Tat protein of the human immunodeficiency virus type-1 (HIV-1) plays a critical role in the regulation of viral transcription and replication. In addition, Tat regulates the expression of a variety of cellular genes and could account for AIDS-associated diseases including Kaposi's Sarcoma and non-Hodgkin's lymphoma by interfering with cellular processes such as proliferation, differentiation, and apoptosis. The molecular mechanisms underlying the pleiotropic activities of Tat may include the generation of functional heterodimers of Tat with cellular proteins. By screening a human B-lymphoblastoid cDNA library in the yeast two-hybrid system, we identified E2F-4, a member of E2F family of transcription factors, as a Tat-binding protein. The interaction between Tat and E2F-4 was confirmed by GST pull-down experiments performed with cellular extracts as well as with in vitro translated E2F-4. The physical association of Tat and E2F-4 was confirmed by in vivo binding experiments where Tat.E2F-4 heterodimers were recovered from Jurkat cells by immunoprecipitation and immunoblotting. By using plasmids expressing mutant forms of Tat and E2F-4, the domains involved in Tat.E2F-4 interaction were identified as the regions encompassing amino acids 1-49 of Tat and amino acids 1-184 of E2F-4. Tat x E2F-4 complexes were shown to bind to E2F cis-regions with increased efficiency compared with E2F-4 alone and to mediate the activity of E2F-dependent promoters including HIV-1 long terminal repeat and cyclin A. The data point to Tat as an adaptor protein that recruits cellular factors such as E2F-4 to exert its multiple biological activities.


Assuntos
HIV-1/metabolismo , Linfócitos B , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Clonagem Molecular , Dimerização , Biblioteca Gênica , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Repetição Terminal Longa de HIV , Humanos , Dados de Sequência Molecular , Plasmídeos , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...