Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 31(12): 1212-1227, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28724615

RESUMEN

In glioblastoma (GBM), heterogeneous expression of amplified and mutated epidermal growth factor receptor (EGFR) presents a substantial challenge for the effective use of EGFR-directed therapeutics. Here we demonstrate that heterogeneous expression of the wild-type receptor and its constitutively active mutant form, EGFRvIII, limits sensitivity to these therapies through an interclonal communication mechanism mediated by interleukin-6 (IL-6) cytokine secreted from EGFRvIII-positive tumor cells. IL-6 activates a NF-κB signaling axis in a paracrine and autocrine manner, leading to bromodomain protein 4 (BRD4)-dependent expression of the prosurvival protein survivin (BIRC5) and attenuation of sensitivity to EGFR tyrosine kinase inhibitors (TKIs). NF-κB and survivin are coordinately up-regulated in GBM patient tumors, and functional inhibition of either protein or BRD4 in in vitro and in vivo models restores sensitivity to EGFR TKIs. These results provide a rationale for improving anti-EGFR therapeutic efficacy through pharmacological uncoupling of a convergence point of NF-κB-mediated survival that is leveraged by an interclonal circuitry mechanism established by intratumoral mutational heterogeneity.


Asunto(s)
Resistencia a Antineoplásicos/genética , Glioblastoma/fisiopatología , FN-kappa B/genética , FN-kappa B/metabolismo , Transducción de Señal/genética , Animales , Comunicación Celular , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Interleucina-6/metabolismo , Ratones , Ratones Desnudos , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Brain ; 145(10): 3608-3621, 2022 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-35603900

RESUMEN

The lipid phosphatase PTEN (phosphatase and tensin homologue on chromosome 10) is a key tumour suppressor gene and an important regulator of neuronal signalling. PTEN mutations have been identified in patients with autism spectrum disorders, characterized by macrocephaly, impaired social interactions and communication, repetitive behaviour, intellectual disability, and epilepsy. PTEN enzymatic activity is regulated by a cluster of phosphorylation sites at the C-terminus of the protein. Here, we focused on the role of PTEN T366 phosphorylation and generated a knock-in mouse line in which Pten T366 was substituted with alanine (PtenT366A/T366A). We identify that phosphorylation of PTEN at T366 controls neuron size and connectivity of brain circuits involved in sensory processing. We show in behavioural tests that PtenT366/T366A mice exhibit cognitive deficits and selective sensory impairments, with significant differences in male individuals. We identify restricted cellular overgrowth of cortical neurons in PtenT366A/T366A brains, linked to increases in both dendritic arborization and soma size. In a combinatorial approach of anterograde and retrograde monosynaptic tracing using rabies virus, we characterize differences in connectivity to the primary somatosensory cortex of PtenT366A/T366A brains, with imbalances in long-range cortico-cortical input to neurons. We conclude that phosphorylation of PTEN at T366 controls neuron size and connectivity of brain circuits involved in sensory processing and propose that PTEN T366 signalling may account for a subset of autism-related functions of PTEN.


Asunto(s)
Fosfohidrolasa PTEN , Treonina , Animales , Ratones , Masculino , Treonina/metabolismo , Tensinas/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Neuronas/metabolismo , Alanina/metabolismo , Lípidos
3.
Mol Microbiol ; 107(3): 330-343, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29152799

RESUMEN

VieA is a cyclic diguanylate phosphodiesterase that modulates biofilm development and motility in Vibrio cholerae O1 of the classical biotype. vieA is part of an operon encoding the VieSAB signal transduction pathway that is nearly silent in V. cholerae of the El Tor biotype. A DNA pull-down assay for proteins interacting with the vieSAB promoter identified the LysR-type regulator LeuO. We show that in classical biotype V. cholerae, LeuO cooperates with the nucleoid-associated protein H-NS to repress vieSAB transcription. LeuO and H-NS interacted with the vieSAB promoter of both biotypes with similar affinities and protected overlapping DNA sequences. H-NS was expressed at similar levels in both cholera biotypes. In contrast, El Tor biotype strains expressed negligible LeuO under identical conditions. In El Tor biotype vibrios, transcription of vieSAB is repressed by the quorum sensing regulator HapR, which is absent in classical biotype strains. Restoring HapR expression in classical biotype V. cholerae repressed vieSAB transcription by binding to its promoter. We propose that double locking of the vieSAB promoter by H-NS and HapR in the El Tor biotype prior to the cessation of exponential growth results in a more pronounced decline in VieA specific activity compared to the classical biotype.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Proteínas de Unión al ADN/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Operón/genética , Regiones Promotoras Genéticas/genética , Percepción de Quorum/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Vibrio cholerae/genética , Vibrio cholerae/metabolismo , Vibrio cholerae O1/genética , Vibrio cholerae O1/metabolismo , Virulencia/genética
4.
Microbiology (Reading) ; 164(7): 998-1003, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29813015

RESUMEN

Hypervirulent atypical El Tor biotype Vibrio cholerae O1 isolates harbour mutations in the DNA-binding domain of the nucleoid-associated protein H-NS and the receiver domain of the response regulator VieA. Here, we provide two examples in which inactivation of H-NS in El Tor biotype vibrios unmasks hidden regulatory connections. First, deletion of the helix-turn-helix domain of VieA in an hns mutant background diminished biofilm formation and exopolysaccharide gene expression, a function that phenotypically opposes its phosphodiesterase activity. Second, deletion of vieA in an hns mutant diminished the expression of σE, a virulence determinant that mediates the envelope stress response. hns mutants were highly sensitive to envelope stressors compared to wild-type. However, deletion of vieA in the hns mutant restored or exceeded wild-type resistance. These findings suggest an evolutionary path for the emergence of hypervirulent strains starting from nucleotide sequence diversification affecting the interaction of H-NS with DNA.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas de Unión al ADN/genética , Regulación Bacteriana de la Expresión Génica , Vibrio cholerae O1/genética , Vibrio cholerae O1/patogenicidad , Biopelículas/crecimiento & desarrollo , Eliminación de Gen , Mutación , Polisacáridos Bacterianos/genética , Factor sigma/genética , Estrés Fisiológico/genética , Vibrio cholerae O1/fisiología , Virulencia/genética
5.
Microb Pathog ; 113: 17-24, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29038053

RESUMEN

Vibrio cholerae of serogroups O1 and O139, the causative agent of Asiatic cholera, continues to be a major global health threat. This pathogen utilizes substratum-specific pili to attach to distinct surfaces in the aquatic environment and the human small intestine and detaches when conditions become unfavorable. Both attachment and detachment are critical to bacterial environmental survival, pathogenesis and disease transmission. However, the factors that promote detachment are less understood. In this study, we examine the role of flagellar motility and hemagglutinin/protease (HapA) in vibrio detachment from a non-degradable abiotic surface and from the suckling mouse intestine. Flagellar motility facilitated V. cholerae detachment from abiotic surfaces. HapA had no effect on the stability of biofilms formed on abiotic surfaces despite representing >50% of the proteolytic activity present in the extracellular matrix. We developed a balanced lethal plasmid system to increase the bacterial cyclic diguanylate (c-di-GMP) pool late in infection, a condition that represses motility and HapA expression. Increasing the c-di-GMP pool enhanced V. cholerae colonization of the suckling mouse intestine. The c-di-GMP effect was fully abolished in hapA isogenic mutants. These results suggest that motility facilitates detachment in a substratum-independent manner. Instead, HapA appears to function as a substratum-specific detachment factor.


Asunto(s)
Adhesión Bacteriana/fisiología , Biopelículas/crecimiento & desarrollo , Flagelos/fisiología , Mucosa Intestinal/microbiología , Metaloendopeptidasas/metabolismo , Movimiento/fisiología , Vibrio cholerae/metabolismo , Animales , Cólera/microbiología , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Fimbrias Bacterianas/fisiología , Regulación Bacteriana de la Expresión Génica , Intestino Delgado/microbiología , Metaloendopeptidasas/genética , Ratones , Poliestirenos , Vibrio cholerae/genética
6.
Mol Microbiol ; 97(4): 630-45, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25982817

RESUMEN

Expression of Vibrio cholerae genes required for the biosynthesis of exopolysacchide (vps) and protein (rbm) components of the biofilm matrix is enhanced by cyclic diguanylate (c-di-GMP). In a previous study, we reported that the histone-like nucleoid structuring (H-NS) protein represses the transcription of vpsA, vpsL and vpsT. Here we demonstrate that the regulator VpsT can disrupt repressive H-NS nucleoprotein complexes at the vpsA and vpsL promoters in the presence of c-di-GMP, while H-NS could disrupt the VpsT-promoter complexes in the absence of c-di-GMP. Chromatin immunoprecipitation-Seq showed a remarkable trend for H-NS to cluster at loci involved in biofilm development such as the rbmABCDEF genes. We show that the antagonistic relationship between VpsT and H-NS regulates the expression of the rbmABCDEF cluster. Epistasis analysis demonstrated that VpsT functions as an antirepressor at the rbmA/F, vpsU and vpsA/L promoters. Deletion of vpsT increased H-NS occupancy at these promoters while increasing the c-di-GMP pool had the opposite effect and included the vpsT promoter. The negative effect of c-di-GMP on H-NS occupancy at the vpsT promoter required the regulator VpsR. These results demonstrate that c-di-GMP activates the transcription of genes required for the biosynthesis of the biofilm matrix by triggering a coordinated VpsR- and VpsT-dependent H-NS antirepression cascade.


Asunto(s)
Proteínas Bacterianas/genética , Biopelículas/crecimiento & desarrollo , GMP Cíclico/análogos & derivados , Proteínas de Unión al ADN/genética , Vibrio cholerae/fisiología , Proteínas Bacterianas/metabolismo , GMP Cíclico/metabolismo , Proteínas de Unión al ADN/metabolismo , Matriz Extracelular/metabolismo , Regulación Bacteriana de la Expresión Génica , Unión Proteica , Biosíntesis de Proteínas , Sistemas de Mensajero Secundario , Vibrio cholerae/genética , Vibrio cholerae/metabolismo
7.
Biochem Biophys Res Commun ; 461(1): 65-9, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25849889

RESUMEN

In Vibrio cholerae, the genes required for biofilm development are repressed by quorum sensing at high cell density due to the accumulation in the medium of two signaling molecules, cholera autoinducer 1 (CAI-1) and autoinducer 2 (AI-2). A significant fraction of toxigenic V. cholerae isolates, however, exhibit dysfunctional quorum sensing pathways. It was reported that transition state analogs of the enzyme methylthioadenosine/S-adenosylhomocysteine nucleosidase (MtnN) required to make AI-2 inhibited biofilm formation in the prototype quorum sensing-deficient strain N16961. This finding prompted us to examine the role of both autoinducers and MtnN in biofilm development and virulence gene expression in a quorum sensing-deficient genetic background. Here we show that deletion of mtnN encoding methylthioadenosine/S-adenosylhomocysteine nucleosidase, cqsA (CAI-1), and/or luxS (AI-2) do not prevent biofilm development. However, two independent mtnN mutants exhibited diminished growth rate and motility in swarm agar plates suggesting that, under certain conditions, MtnN could influence biofilm formation indirectly. Nevertheless, MtnN is not required for the development of a mature biofilm.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Liasas de Carbono-Azufre/metabolismo , Cetonas/metabolismo , N-Glicosil Hidrolasas/metabolismo , Purina-Nucleósido Fosforilasa/metabolismo , Percepción de Quorum/fisiología , Vibrio cholerae/fisiología , Movimiento Celular/fisiología
8.
J Bacteriol ; 196(5): 1020-30, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24363348

RESUMEN

Cholera is a waterborne diarrheal disease caused by Vibrio cholerae strains of serogroups O1 and O139. Expression of the general stress response regulator RpoS and formation of biofilm communities enhance the capacity of V. cholerae to persist in aquatic environments. The transition of V. cholerae between free-swimming (planktonic) and biofilm life-styles is regulated by the second messenger cyclic di-GMP (c-di-GMP). We previously reported that increasing the c-di-GMP pool by overexpression of a diguanylate cyclase diminished RpoS expression. Here we show that c-di-GMP repression of RpoS expression is eliminated by deletion of the genes vpsR and vpsT, encoding positive regulators of biofilm development. To determine the mechanism of this regulation, we constructed a strain expressing a vpsT-FLAG allele from native transcription and translation signals. Increasing the c-di-GMP pool induced vpsT-FLAG expression. The interaction between VpsT-FLAG and the rpoS promoter was demonstrated by chromatin immunoprecipitation. Furthermore, purified VpsT interacted with the rpoS promoter in a c-di-GMP-dependent manner. Primer extension analysis identified two rpoS transcription initiation sites located 43 bp (P1) and 63 bp (P2) upstream of the rpoS start codon. DNase I footprinting showed that the VpsT binding site at the rpoS promoter overlaps the primary P1 transcriptional start site. Deletion of vpsT significantly enhanced rpoS expression in V. cholerae biofilms that do not make HapR. This result suggests that VpsT and c-di-GMP contribute to the transcriptional silencing of rpoS in biofilms prior to cells entering the quorum-sensing mode.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Regulación Bacteriana de la Expresión Génica/fisiología , Factor sigma/metabolismo , Vibrio cholerae/metabolismo , Vibrio cholerae/fisiología , Proteínas Bacterianas/genética , GMP Cíclico/análogos & derivados , GMP Cíclico/genética , GMP Cíclico/metabolismo , Factor sigma/genética , Estrés Fisiológico/fisiología , Transcripción Genética/fisiología , Vibrio cholerae/genética
9.
Antimicrob Agents Chemother ; 57(8): 3950-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23733460

RESUMEN

Vibrio cholerae strains of serogroups O1 and O139, the causative agents of the diarrheal illness cholera, express a single polar flagellum powered by sodium motive force and require motility to colonize and spread along the small intestine. In a previous study, we described a high-throughput assay for screening for small molecules that selectively inhibit bacterial motility and identified a family of quinazoline-2,4-diamino analogs (Q24DAs) that (i) paralyzed the sodium-driven polar flagellum of Vibrios and (ii) diminished cholera toxin secreted by El Tor biotype V. cholerae. In this study, we provide evidence that a Q24DA paralyzes the polar flagellum by interacting with the motor protein PomB. Inhibition of motility with the Q24DA enhanced the transcription of the cholera toxin genes in both biotypes. We also show that the Q24DA interacts with outer membrane protein OmpU and other porins to induce envelope stress and expression of the extracellular RNA polymerase sigma factor σ(E). We suggest that Q24DA-induced envelope stress could affect the correct folding, assembly, and secretion of pentameric cholera toxin in El Tor biotype V. cholerae independently of its effect on motility.


Asunto(s)
Proteínas Bacterianas/metabolismo , Flagelos/efectos de los fármacos , Quinazolinas/farmacología , Canales de Sodio/metabolismo , Estrés Fisiológico , Vibrio cholerae/efectos de los fármacos , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Sustitución de Aminoácidos , Proteínas Bacterianas/genética , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Toxina del Cólera/genética , Toxina del Cólera/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Flagelos/fisiología , Genes Bacterianos , Pliegue de Proteína , Mapeo de Interacción de Proteínas , Canales de Sodio/efectos de los fármacos , Canales de Sodio/genética , Transcripción Genética , Vibrio cholerae/genética , Vibrio cholerae/metabolismo
10.
J Bacteriol ; 194(5): 1205-15, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22194453

RESUMEN

The bacterium Vibrio cholerae colonizes the human small intestine and secretes cholera toxin (CT) to cause the rice-watery diarrhea characteristic of this illness. The ability of this pathogen to colonize the small bowel, express CT, and return to the aquatic environment is controlled by a complex network of regulatory proteins. Two global regulators that participate in this process are the histone-like nucleoid structuring protein (H-NS) and the general stress response regulator RpoS. In this study, we address the role of RpoS and H-NS in the coordinate regulation of motility and hemagglutinin (HA)/protease expression. In addition to initiating transcription of hapA encoding HA/protease, RpoS enhanced flrA and rpoN transcription to increase motility. In contrast, H-NS was found to bind to the flrA, rpoN, and hapA promoters and represses their expression. The strength of H-NS repression at the above-mentioned promoters was weaker for hapA, which exhibited the strongest RpoS dependency, suggesting that transcription initiation by RNA polymerase containing σ(S) could be more resistant to H-NS repression. Occupancy of the flrA and hapA promoters by H-NS was demonstrated by chromatin immunoprecipitation (ChIP). We show that the expression of RpoS in the stationary phase significantly diminished H-NS promoter occupancy. Furthermore, RpoS enhanced the transcription of integration host factor (IHF), which positively affected the expression of flrA and rpoN by diminishing the occupancy of H-NS at these promoters. Altogether, we propose a model for RpoS regulation of motility gene expression that involves (i) attenuation of H-NS repression by IHF and (ii) RpoS-dependent transcription initiation resistant to H-NS.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Bacteriana de la Expresión Génica , Locomoción , Metaloendopeptidasas/biosíntesis , Mapeo de Interacción de Proteínas , Factor sigma/metabolismo , Vibrio cholerae/fisiología , Inmunoprecipitación de Cromatina , ADN Bacteriano/metabolismo , Perfilación de la Expresión Génica , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Transcripción Genética , Vibrio cholerae/genética
12.
J Bacteriol ; 193(23): 6529-38, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21965573

RESUMEN

Vibrio cholerae secretes the Zn-dependent metalloprotease hemagglutinin (HA)/protease (mucinase), which is encoded by hapA and displays a broad range of potential pathogenic activities. Expression of HA/protease has a stringent requirement for the quorum-sensing regulator HapR and the general stress response regulator RpoS. Here we report that the second messenger cyclic diguanylic acid (c-di-GMP) regulates the production of HA/protease in a negative manner. Overexpression of a diguanylate cyclase to increase the cellular c-di-GMP pool resulted in diminished expression of HA/protease and its positive regulator, HapR. The effect of c-di-GMP on HapR was independent of LuxO but was abolished by deletion of the c-di-GMP binding protein VpsT, the LuxR-type regulator VqmA, or a single-base mutation in the hapR promoter that prevents autorepression. Though expression of HapR had a positive effect on RpoS biosynthesis, direct manipulation of the c-di-GMP pool at a high cell density did not significantly impact RpoS expression in the wild-type genetic background. In contrast, increasing the c-di-GMP pool severely inhibited RpoS expression in a ΔhapR mutant that is locked in a regulatory state mimicking low cell density. Based on the above findings, we propose a model for the interplay between HapR, RpoS, and c-di-GMP in the regulation of HA/protease expression.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Metaloendopeptidasas/genética , Factor sigma/metabolismo , Vibrio cholerae/enzimología , Proteínas Bacterianas/genética , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Metaloendopeptidasas/metabolismo , Factor sigma/genética , Vibrio cholerae/genética , Vibrio cholerae/metabolismo
13.
Antimicrob Agents Chemother ; 55(9): 4134-43, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21709090

RESUMEN

Numerous bacterial pathogens, particularly those that colonize fast-flow areas in the bladder and gastrointestinal tract, require motility to establish infection and spread beyond the initially colonized tissue. Vibrio cholerae strains of serogroups O1 and O139, the causative agents of the diarrheal illness cholera, express a single polar flagellum powered by sodium motive force and require motility to colonize and spread along the small intestine. Therefore, motility may be an attractive target for small molecules that can prevent and/or block the infective process. In this study, we describe a high-throughput screening (HTS) assay to identify small molecules that selectively inhibit bacterial motility. The HTS assay was used to screen an ∼8,000-compound structurally diverse chemical library for inhibitors of V. cholerae motility. The screen identified a group of quinazoline-2,4-diamino analogs that completely suppressed motility without affecting the growth rate in broth. A further study on the effects of one analog, designated Q24DA, showed that it induces a flagellated but nonmotile (Mot(-)) phenotype and is specific for the Na(+)-driven flagellar motor of pathogenic Vibrio species. A mutation conferring phenamil-resistant motility did not eliminate inhibition of motility by Q24DA. Q24DA diminished the expression of cholera toxin and toxin-coregulated pilus as well as biofilm formation and fluid secretion in the rabbit ileal loop model. Furthermore, treatment of V. cholerae with Q24DA impacted additional phenotypes linked to Na(+) bioenergetics, such as the function of the primary Na(+) pump, Nqr, and susceptibility to fluoroquinolones. The above results clearly show that the described HTS assay is capable of identifying small molecules that specifically block bacterial motility. New inhibitors such as Q24DA may be instrumental in probing the molecular architecture of the Na(+)-driven polar flagellar motor and in studying the role of motility in the expression of other virulence factors.


Asunto(s)
Antibacterianos/uso terapéutico , Flagelos/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Vibrio cholerae/efectos de los fármacos , Vibrio cholerae/patogenicidad , Virulencia/efectos de los fármacos , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cólera/tratamiento farmacológico , Cólera/microbiología , Fluoroquinolonas/uso terapéutico , Masculino , Microscopía Electrónica de Transmisión , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
14.
Cell Mol Neurobiol ; 31(8): 1229-43, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21681558

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder characterized by motor, cognitive, and psychiatric symptoms. The most characteristic structural feature of this disease is neurodegeneration accompanied by gliosis in the striatum. BDNF has been proposed to protect striatal neurons from degeneration, because it is an important survival factor for these neurons from development to adulthood. Considering the extensive gliosis and the survival effects of BDNF, we constructed an adenovirus to express a BDNF cDNA in astrocyte cells using a promoter of the glial fibrillary acidic protein gene. Cells stably transfected in vitro with a BDNF cDNA driven by this promoter expressed BDNF and responded to external stimuli increasing BDNF production. When the vector was applied into the striata of mice transgenic for HD, long-term expression of the transgene was observed, associated with a delay of onset of the motor phenotype of the R6/2 HD transgenic mice. The present data indicate that the striatal expression of BDNF is a potential adjuvant for the treatment of HD.


Asunto(s)
Astrocitos/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cuerpo Estriado/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Astrocitos/citología , Conducta Animal/fisiología , Factor Neurotrófico Derivado del Encéfalo/genética , Células Cultivadas , Cuerpo Estriado/citología , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Ratones , Ratones Transgénicos , Pruebas Neuropsicológicas , Transgenes
15.
Neuro Oncol ; 23(7): 1072-1086, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33428749

RESUMEN

BACKGROUND: Glioblastoma (GBM) is the most common primary brain tumor in adults with a median survival of approximately 15 months; therefore, more effective treatment options for GBM are required. To identify new drugs targeting GBMs, we performed a high-throughput drug screen using patient-derived neurospheres cultured to preferentially retain their glioblastoma stem cell (GSC) phenotype. METHODS: High-throughput drug screening was performed on GSCs followed by a dose-response assay of the 5 identified original "hits." A PI3K/mTOR dependency to a proteasome inhibitor (carfilzomib), was confirmed by genetic and pharmacologic experiments. Proteasome Inhibition Response Signatures were derived from proteomic and bioinformatic analysis. Molecular mechanism of action was determined using three-dimensional (3D) GBM-organoids and preclinical orthotopic models. RESULTS: We found that GSCs were highly sensitive to proteasome inhibition due to an underlying dependency on an increased protein synthesis rate, and loss of autophagy, associated with PTEN loss and activation of the PI3K/mTOR pathway. In contrast, combinatory inhibition of autophagy and the proteasome resulted in enhanced cytotoxicity specifically in GSCs that did express PTEN. Finally, proteasome inhibition specifically increased cell death markers in 3D GBM-organoids, suppressed tumor growth, and increased survival of mice orthotopically engrafted with GSCs. As perturbations of the PI3K/mTOR pathway occur in nearly 50% of GBMs, these findings suggest that a significant fraction of these tumors could be vulnerable to proteasome inhibition. CONCLUSIONS: Proteasome inhibition is a potential synthetic lethal therapeutic strategy for GBM with proteasome addiction due to a high protein synthesis rate and autophagy deficiency.


Asunto(s)
Antineoplásicos , Neoplasias Encefálicas , Glioblastoma , Animales , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Glioblastoma/tratamiento farmacológico , Humanos , Ratones , Células Madre Neoplásicas , Fosfohidrolasa PTEN/genética , Complejo de la Endopetidasa Proteasomal , Proteómica
17.
Nat Commun ; 11(1): 550, 2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-31992716

RESUMEN

Many cellular models aimed at elucidating cancer biology do not recapitulate pathobiology including tumor heterogeneity, an inherent feature of cancer that underlies treatment resistance. Here we introduce a cancer modeling paradigm using genetically engineered human pluripotent stem cells (hiPSCs) that captures authentic cancer pathobiology. Orthotopic engraftment of the neural progenitor cells derived from hiPSCs that have been genome-edited to contain tumor-associated genetic driver mutations revealed by The Cancer Genome Atlas project for glioblastoma (GBM) results in formation of high-grade gliomas. Similar to patient-derived GBM, these models harbor inter-tumor heterogeneity resembling different GBM molecular subtypes, intra-tumor heterogeneity, and extrachromosomal DNA amplification. Re-engraftment of these primary tumor neurospheres generates secondary tumors with features characteristic of patient samples and present mutation-dependent patterns of tumor evolution. These cancer avatar models provide a platform for comprehensive longitudinal assessment of human tumor development as governed by molecular subtype mutations and lineage-restricted differentiation.


Asunto(s)
Ingeniería Genética , Glioblastoma/genética , Glioblastoma/patología , Células Madre Pluripotentes/patología , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Diferenciación Celular , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Genoma , Glioblastoma/metabolismo , Glioma/genética , Glioma/patología , Humanos , Ratones , Ratones SCID , Mutación , Trasplante de Neoplasias , Células Madre Neoplásicas/patología , Neurofibromina 1/genética , Fosfohidrolasa PTEN/genética , Trasplante Heterólogo , Proteína p53 Supresora de Tumor/genética
18.
J Microbiol Methods ; 165: 105702, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31454505

RESUMEN

We describe a proteomic approach to identify transcription factors binding to a target promoter. The method's usefulness was tested by identifying proteins binding to the Vibrio cholerae rpoS promoter in response to cell density. Proteins identified in this screen included the nucleoid-associated protein Fis and the quorum sensing regulator HapR.


Asunto(s)
Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Factor sigma/genética , Factores de Transcripción/genética , Vibrio cholerae/genética , Regiones Promotoras Genéticas , Proteoma/metabolismo , Percepción de Quorum , Transcripción Genética
19.
Cancer Cell ; 35(3): 504-518.e7, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30827889

RESUMEN

Ionizing radiation (IR) and chemotherapy are standard-of-care treatments for glioblastoma (GBM) patients and both result in DNA damage, however, the clinical efficacy is limited due to therapeutic resistance. We identified a mechanism of such resistance mediated by phosphorylation of PTEN on tyrosine 240 (pY240-PTEN) by FGFR2. pY240-PTEN is rapidly elevated and bound to chromatin through interaction with Ki-67 in response to IR treatment and facilitates the recruitment of RAD51 to promote DNA repair. Blocking Y240 phosphorylation confers radiation sensitivity to tumors and extends survival in GBM preclinical models. Y240F-Pten knockin mice showed radiation sensitivity. These results suggest that FGFR-mediated pY240-PTEN is a key mechanism of radiation resistance and is an actionable target for improving radiotherapy efficacy.


Asunto(s)
Neoplasias Encefálicas/terapia , Núcleo Celular/metabolismo , Glioma/terapia , Fosfohidrolasa PTEN/metabolismo , Pirimidinas/administración & dosificación , Tolerancia a Radiación/efectos de los fármacos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Reparación del ADN/efectos de los fármacos , Femenino , Glioma/metabolismo , Humanos , Masculino , Ratones , Fosforilación/efectos de los fármacos , Pirimidinas/farmacología , Recombinasa Rad51/metabolismo , Tirosina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Bacteriol ; 190(22): 7335-45, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18790865

RESUMEN

Production of the Zn-metalloprotease hemagglutinin (HA)/protease by Vibrio cholerae has been reported to enhance enterotoxicity in rabbit ileal loops and the reactogenicity of live cholera vaccine candidates. Expression of HA/protease requires the alternate sigma factor sigma(S) (RpoS), encoded by rpoS. The histone-like nucleoid structuring protein (H-NS) has been shown to repress rpoS expression in Escherichia coli. In V. cholerae strains of the classical biotype, H-NS has been reported to silence virulence gene expression. In this study we examined the role of H-NS in the expression of HA/protease and motility in an El Tor biotype strain by constructing a Deltahns mutant. The Deltahns mutant exhibited multiple phenotypes, such as production of cholera toxin in nonpermissive LB medium, reduced resistance to high osmolarity, enhanced resistance to low pH and hydrogen peroxide, and reduced motility. Depletion of H-NS by overexpression of a dominant-negative allele or by deletion of hns resulted in diminished expression of HA/protease. Epistasis analysis of HA/protease expression in Deltahns, DeltarpoS, and Deltahns DeltarpoS mutants, analysis of RpoS reporter fusions, quantitative reverse transcription-PCR measurements, and ectopic expression of RpoS in DeltarpoS and DeltarpoS Deltahns mutants showed that H-NS posttranscriptionally enhances RpoS expression. The Deltahns mutant exhibited a lower degree of motility and lower levels of expression of flaA, flaC, cheR-2, and motX mRNAs than the wild type. Comparison of the mRNA abundances of these genes in wild-type, Deltahns, DeltarpoS, and Deltahns DeltarpoS strains revealed that deletion of rpoS had a more severe negative effect on their expression. Interestingly, deletion of hns in the rpoS background resulted in higher expression levels of flaA, flaC, and motX, suggesting that H-NS represses the expression of these genes in the absence of sigma(S). Finally, we show that the cyclic AMP receptor protein and H-NS act along the same pathway to positively affect RpoS expression.


Asunto(s)
Proteínas Bacterianas/fisiología , Regulación Bacteriana de la Expresión Génica , Factor sigma/fisiología , Vibrio cholerae/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Quimiotaxis , Perfilación de la Expresión Génica , Mutagénesis Insercional , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Eliminación de Secuencia , Factor sigma/genética , Factor sigma/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Vibrio cholerae/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA