Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Neuroendocrinology ; 111(4): 370-387, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32335558

RESUMO

INTRODUCTION: Metabolic dysfunction is now recognized as a pivotal component of Alzheimer's disease (AD), the most common dementia worldwide. However, the precise molecular mechanisms linking metabolic dysfunction to AD remain elusive. OBJECTIVE: Here, we investigated the direct impact of soluble oligomeric amyloid beta (Aß) peptides, the main molecular hallmark of AD, on the leptin system, a major component of central energy metabolism regulation. METHODS: We developed a new time-resolved fluorescence resonance energy transfer-based Aß binding assay for the leptin receptor (LepR) and studied the effect of Aß on LepR function in several in vitro assays. The in vivo effect of Aß on LepR function was studied in an Aß-specific AD mouse model and in pro-opiomelanocortin (POMC) neurons of the hypothalamic arcuate nucleus. RESULTS: We revealed specific and high-affinity (Ki = 0.1 nM) binding of Aß to LepR. Pharmacological characterization of this interaction showed that Aß binds allosterically to the extracellular domain of LepR and negatively affects receptor function. Negative allosteric modulation of LepR by Aß was detected at the level of signaling pathways (STAT-3, AKT, and ERK) in vitroand in vivo. Importantly, the leptin-induced response of POMC neurons, key players in the regulation of metabolic function, was completely abolished in the presence of Aß. CONCLUSION: Our data indicate that Aß is a negative allosteric modulator of LepR, resulting in impaired leptin action, and qualify LepR as a new and direct target of Aß oligomers. Preventing the interaction of Aß with LepR might improve both the metabolic and cognitive dysfunctions in AD condition.


Assuntos
Regulação Alostérica/fisiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Leptina/metabolismo , Pró-Opiomelanocortina/metabolismo , Receptores para Leptina/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Células HEK293 , Humanos , Masculino , Camundongos , Transdução de Sinais/fisiologia
2.
Cell Mol Life Sci ; 76(6): 1201-1214, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30659329

RESUMO

Leptin links body energy stores to high energy demanding processes like reproduction and immunity. Based on leptin's role in autoimmune diseases and cancer, several leptin and leptin receptor (LR) antagonists have been developed, but these intrinsically lead to unwanted weight gain. Here, we report on the uncoupling of leptin's metabolic and immune functions based on the cross talk with the epidermal growth factor receptor (EGFR). We show that both receptors spontaneously interact and, remarkably, that this complex can partially overrule the lack of LR activation by a leptin antagonistic mutein. Moreover, this leptin mutant induces EGFR phosphorylation comparable to wild-type leptin. Exploiting this non-canonical leptin signalling pathway, we identified a camelid single-domain antibody that selectively inhibits this LR-EGFR cross talk without interfering with homotypic LR signalling. Administration in vivo showed that this single-domain antibody did not interfere with leptin's metabolic functions, but could reverse the leptin-driven protection against starvation-induced thymic and splenic atrophy. These findings offer new opportunities for the design and clinical application of selective leptin and LR antagonists that avoid unwanted metabolic side effects.


Assuntos
Leptina/imunologia , Leptina/metabolismo , Receptores para Leptina/antagonistas & inibidores , Receptores para Leptina/metabolismo , Anticorpos de Domínio Único/farmacologia , Animais , Camelídeos Americanos/imunologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Células HEK293 , Humanos , Leptina/genética , Ligantes , Camundongos Endogâmicos C57BL , Mutação , Ligação Proteica/efeitos dos fármacos , Receptor Cross-Talk/efeitos dos fármacos , Receptores para Leptina/genética , Transdução de Sinais
3.
Mol Microbiol ; 107(3): 330-343, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29152799

RESUMO

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.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Proteínas de Ligação a DNA/metabolismo , Regulação Bacteriana da Expressão Gênica/genética , Óperon/genética , Regiões Promotoras Genéticas/genética , Percepção de Quorum/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Vibrio cholerae/genética , Vibrio cholerae/metabolismo , Vibrio cholerae O1/genética , Vibrio cholerae O1/metabolismo , Virulência/genética
4.
Microbiology (Reading) ; 164(7): 998-1003, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29813015

RESUMO

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.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Ligação a DNA/genética , Regulação Bacteriana da Expressão Gênica , Vibrio cholerae O1/genética , Vibrio cholerae O1/patogenicidade , Biofilmes/crescimento & desenvolvimento , Deleção de Genes , Mutação , Polissacarídeos Bacterianos/genética , Fator sigma/genética , Estresse Fisiológico/genética , Vibrio cholerae O1/fisiologia , Virulência/genética
5.
Microb Pathog ; 113: 17-24, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29038053

RESUMO

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.


Assuntos
Aderência Bacteriana/fisiologia , Biofilmes/crescimento & desenvolvimento , Flagelos/fisiologia , Mucosa Intestinal/microbiologia , Metaloendopeptidases/metabolismo , Movimento/fisiologia , Vibrio cholerae/metabolismo , Animais , Cólera/microbiologia , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Fímbrias Bacterianas/fisiologia , Regulação Bacteriana da Expressão Gênica , Intestino Delgado/microbiologia , Metaloendopeptidases/genética , Camundongos , Poliestirenos , Vibrio cholerae/genética
6.
Mol Microbiol ; 97(4): 630-45, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25982817

RESUMO

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.


Assuntos
Proteínas de Bactérias/genética , Biofilmes/crescimento & desenvolvimento , GMP Cíclico/análogos & derivados , Proteínas de Ligação a DNA/genética , Vibrio cholerae/fisiologia , Proteínas de Bactérias/metabolismo , GMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Matriz Extracelular/metabolismo , Regulação Bacteriana da Expressão Gênica , Ligação Proteica , Biossíntese de Proteínas , Sistemas do Segundo Mensageiro , Vibrio cholerae/genética , Vibrio cholerae/metabolismo
7.
Biochem Biophys Res Commun ; 461(1): 65-9, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25849889

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Liases de Carbono-Enxofre/metabolismo , Cetonas/metabolismo , N-Glicosil Hidrolases/metabolismo , Purina-Núcleosídeo Fosforilase/metabolismo , Percepção de Quorum/fisiologia , Vibrio cholerae/fisiologia , Movimento Celular/fisiologia
8.
J Bacteriol ; 196(5): 1020-30, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24363348

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Regulação Bacteriana da Expressão Gênica/fisiologia , Fator sigma/metabolismo , Vibrio cholerae/metabolismo , Vibrio cholerae/fisiologia , Proteínas de Bactérias/genética , GMP Cíclico/análogos & derivados , GMP Cíclico/genética , GMP Cíclico/metabolismo , Fator sigma/genética , Estresse Fisiológico/fisiologia , Transcrição Gênica/fisiologia , Vibrio cholerae/genética
9.
Antimicrob Agents Chemother ; 57(8): 3950-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23733460

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Flagelos/efeitos dos fármacos , Quinazolinas/farmacologia , Canais de Sódio/metabolismo , Estresse Fisiológico , Vibrio cholerae/efeitos dos fármacos , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Substituição de Aminoácidos , Proteínas de Bactérias/genética , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Toxina da Cólera/genética , Toxina da Cólera/metabolismo , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Flagelos/fisiologia , Genes Bacterianos , Dobramento de Proteína , Mapeamento de Interação de Proteínas , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/genética , Transcrição Gênica , Vibrio cholerae/genética , Vibrio cholerae/metabolismo
10.
J Bacteriol ; 194(5): 1205-15, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22194453

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Bacteriana da Expressão Gênica , Locomoção , Metaloendopeptidases/biossíntese , Mapeamento de Interação de Proteínas , Fator sigma/metabolismo , Vibrio cholerae/fisiologia , Imunoprecipitação da Cromatina , DNA Bacteriano/metabolismo , Perfilação da Expressão Gênica , Humanos , Regiões Promotoras Genéticas , Ligação Proteica , Transcrição Gênica , Vibrio cholerae/genética
12.
J Bacteriol ; 193(23): 6529-38, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21965573

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Metaloendopeptidases/genética , Fator sigma/metabolismo , Vibrio cholerae/enzimologia , Proteínas de Bactérias/genética , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Metaloendopeptidases/metabolismo , Fator sigma/genética , Vibrio cholerae/genética , Vibrio cholerae/metabolismo
13.
Antimicrob Agents Chemother ; 55(9): 4134-43, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21709090

RESUMO

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.


Assuntos
Antibacterianos/uso terapêutico , Flagelos/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Vibrio cholerae/efeitos dos fármacos , Vibrio cholerae/patogenicidade , Virulência/efeitos dos fármacos , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cólera/tratamento farmacológico , Cólera/microbiologia , Fluoroquinolonas/uso terapêutico , Masculino , Microscopia Eletrônica de Transmissão , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
14.
Endocrinology ; 162(11)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34388249

RESUMO

The brain influences liver metabolism through many neuroendocrine and autonomic mechanisms that have evolved to protect the organism against starvation and hypoglycemia. Unfortunately, this effective way of preventing death has become dysregulated in modern obesogenic environments, although the pathophysiological mechanisms behind metabolic dyshomeostasis are still unclear. In this Mini-Review, we provide our thoughts regarding obesity and type 2 diabetes as diseases of the autonomic nervous system. We discuss the pathophysiological mechanisms that alter the autonomic brain-liver communication in these diseases, and how they could represent important targets to prevent or treat metabolic dysfunctions. We discuss how sympathetic hyperactivity to the liver may represent an early event in the progression of metabolic diseases and could progressively lead to hepatic neuropathy. We hope that this discussion will inspire and help to frame a model based on better understanding of the chronology of autonomic dysfunctions in the liver, enabling the application of the right strategy at the right time.


Assuntos
Sistema Nervoso Autônomo/fisiopatologia , Encéfalo/fisiopatologia , Fígado/fisiopatologia , Doenças Metabólicas , Animais , Sistema Nervoso Autônomo/patologia , Encéfalo/metabolismo , Comunicação Celular , Humanos , Fígado/inervação , Fígado/metabolismo , Doenças Metabólicas/fisiopatologia , Doenças Metabólicas/psicologia , Sistemas Neurossecretores/fisiopatologia
15.
Nat Metab ; 3(8): 1071-1090, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34341568

RESUMO

Metabolic health depends on the brain's ability to control food intake and nutrient use versus storage, processes that require peripheral signals such as the adipocyte-derived hormone, leptin, to cross brain barriers and mobilize regulatory circuits. We have previously shown that hypothalamic tanycytes shuttle leptin into the brain to reach target neurons. Here, using multiple complementary models, we show that tanycytes express functional leptin receptor (LepR), respond to leptin by triggering Ca2+ waves and target protein phosphorylation, and that their transcytotic transport of leptin requires the activation of a LepR-EGFR complex by leptin and EGF sequentially. Selective deletion of LepR in tanycytes blocks leptin entry into the brain, inducing not only increased food intake and lipogenesis but also glucose intolerance through attenuated insulin secretion by pancreatic ß-cells, possibly via altered sympathetic nervous tone. Tanycytic LepRb-EGFR-mediated transport of leptin could thus be crucial to the pathophysiology of diabetes in addition to obesity, with therapeutic implications.


Assuntos
Encéfalo/metabolismo , Células Ependimogliais/metabolismo , Receptores ErbB/metabolismo , Leptina/metabolismo , Metabolismo dos Lipídeos , Pâncreas/metabolismo , Receptores para Leptina/metabolismo , Diabetes Mellitus/etiologia , Diabetes Mellitus/metabolismo , Metabolismo Energético , Células Secretoras de Insulina/metabolismo , Fosforilação
16.
J Microbiol Methods ; 165: 105702, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31454505

RESUMO

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.


Assuntos
Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Fator sigma/genética , Fatores de Transcrição/genética , Vibrio cholerae/genética , Regiões Promotoras Genéticas , Proteoma/metabolismo , Percepção de Quorum , Transcrição Gênica
17.
J Bacteriol ; 190(22): 7335-45, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18790865

RESUMO

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.


Assuntos
Proteínas de Bactérias/fisiologia , Regulação Bacteriana da Expressão Gênica , Fator sigma/fisiologia , Vibrio cholerae/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Quimiotaxia , Perfilação da Expressão Gênica , Mutagênese Insercional , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deleção de Sequência , Fator sigma/genética , Fator sigma/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Vibrio cholerae/metabolismo
18.
FEBS Lett ; 582(27): 3744-50, 2008 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-18930049

RESUMO

In Vibrio cholerae, expression of the quorum sensing regulator HapR is induced by the accumulation of a major autoinducer synthesized by the activity of CqsA. Here we show that the cAMP-cAMP receptor protein complex regulates cqsA expression at the post-transcriptional level. This conclusion is supported by the analysis of cqsA-lacZ fusions, the ectopic expression of cqsA in Deltacrp mutants and by Northern blot analysis showing that cqsA mRNA is unstable in Deltacrp and Deltacya (adenylate cyclase) mutants. Addition of cAMP to the culture of a Deltacya mutant restored cqsA mRNA stability and cholera autoinducer 1 production. Lowering intracellular cAMP levels by addition of d-glucose increased the cell density required to activate HapR. These results indicate that cAMP acts as a quorum modulator.


Assuntos
Proteínas de Bactérias/biossíntese , AMP Cíclico/metabolismo , Cetonas/metabolismo , Percepção de Quorum , Vibrio cholerae/metabolismo , Proteínas de Bactérias/genética , AMP Cíclico/farmacologia , Proteína Receptora de AMP Cíclico/genética , Proteína Receptora de AMP Cíclico/metabolismo , Biossíntese de Proteínas , Estabilidade de RNA , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transcrição Gênica , Vibrio cholerae/genética , Vibrio cholerae/fisiologia
19.
Methods Mol Biol ; 1839: 65-75, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30047055

RESUMO

Chromatin immunoprecipitation (ChIP) measures the physical association between a protein and DNA in the cell. In combination with next-generation sequencing, the technique enables the identification of DNA targets for the corresponding protein across an entire genome. Here we describe the immunoprecipitation of Vibrio cholerae DNA bound to the histone-like nucleoid structuring protein (H-NS) tagged with the Flag epitope. The quality of the DNA obtained in this protocol is suitable for next-generation sequencing. The procedure described herein can be readily adapted to other bacteria and DNA-binding proteins.


Assuntos
Imunoprecipitação da Cromatina , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA , Sequenciamento de Nucleotídeos em Larga Escala , Ligação Proteica , Reação em Cadeia da Polimerase em Tempo Real , Vibrio cholerae/genética , Vibrio cholerae/metabolismo
20.
Res Microbiol ; 168(1): 16-25, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27492955

RESUMO

Vibrio cholerae has become a model organism for studies connecting virulence, pathogen evolution and infectious disease ecology. The coordinate expression of motility, virulence and biofilm enhances its pathogenicity, environmental fitness and fecal-oral transmission. The histone-like nucleoid structuring protein negatively regulates gene expression at multiple phases of the V. cholerae life cycle. Here we discuss: (i) the regulatory and structural implications of H-NS chromatin-binding in the two-chromosome cholera bacterium; (ii) the factors that counteract H-NS repression; and (iii) a model for the regulation of the V. cholerae life cycle that integrates H-NS repression, cyclic diguanylic acid signaling and the general stress response.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Bacteriana da Expressão Gênica , Vibrio cholerae/genética , Vibrio cholerae/fisiologia , Biofilmes/crescimento & desenvolvimento , Cromatina/metabolismo , Locomoção , Ligação Proteica , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA