Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cell ; 187(4): 897-913.e18, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38280374

RESUMO

Canonically, the complement system is known for its rapid response to remove microbes in the bloodstream. However, relatively little is known about a functioning complement system on intestinal mucosal surfaces. Herein, we report the local synthesis of complement component 3 (C3) in the gut, primarily by stromal cells. C3 is expressed upon commensal colonization and is regulated by the composition of the microbiota in healthy humans and mice, leading to an individual host's specific luminal C3 levels. The absence of membrane attack complex (MAC) components in the gut ensures that C3 deposition does not result in the lysis of commensals. Pathogen infection triggers the immune system to recruit neutrophils to the infection site for pathogen clearance. Basal C3 levels directly correlate with protection against enteric infection. Our study reveals the gut complement system as an innate immune mechanism acting as a vigilant sentinel that combats pathogens and spares commensals.


Assuntos
Complemento C3 , Mucosa Intestinal , Microbiota , Animais , Humanos , Camundongos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Neutrófilos , Complemento C3/metabolismo , Células Estromais/metabolismo
2.
Cell ; 179(4): 803-805, 2019 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-31675492

RESUMO

How does stress promote anxiety? In this issue of Cell, Fan et al. report that immune cells have a direct role in this process. They show that chronic stress promotes mitochondrial fission in CD4+ T cells, causing increased synthesis of xanthine, which acts on the brain and induces anxiety-like behaviors.


Assuntos
Linfócitos T CD4-Positivos , Doenças Metabólicas , Ansiedade , Humanos , Dinâmica Mitocondrial
4.
Nature ; 576(7785): 143-148, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31776512

RESUMO

Bile acids are abundant in the mammalian gut, where they undergo bacteria-mediated transformation to generate a large pool of bioactive molecules. Although bile acids are known to affect host metabolism, cancer progression and innate immunity, it is unknown whether they affect adaptive immune cells such as T helper cells that express IL-17a (TH17 cells) or regulatory T cells (Treg cells). Here we screen a library of bile acid metabolites and identify two distinct derivatives of lithocholic acid (LCA), 3-oxoLCA and isoalloLCA, as T cell regulators in mice. 3-OxoLCA inhibited the differentiation of TH17 cells by directly binding to the key transcription factor retinoid-related orphan receptor-γt (RORγt) and isoalloLCA increased the differentiation of Treg cells through the production of mitochondrial reactive oxygen species (mitoROS), which led to increased expression of FOXP3. The isoalloLCA-mediated enhancement of Treg cell differentiation required an intronic Foxp3 enhancer, the conserved noncoding sequence (CNS) 3; this represents a mode of action distinct from that of previously identified metabolites that increase Treg cell differentiation, which require CNS1. The administration of 3-oxoLCA and isoalloLCA to mice reduced TH17 cell differentiation and increased Treg cell differentiation, respectively, in the intestinal lamina propria. Our data suggest mechanisms through which bile acid metabolites control host immune responses, by directly modulating the balance of TH17 and Treg cells.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Ácido Litocólico/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Animais , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Ácido Litocólico/química , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th17/citologia , Células Th17/imunologia , Células Th17/metabolismo
5.
PLoS Pathog ; 13(6): e1006428, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28586382

RESUMO

Vibrio cholerae is a diarrheal pathogen that induces accumulation of lipid droplets in enterocytes, leading to lethal infection of the model host Drosophila melanogaster. Through untargeted lipidomics, we provide evidence that this process is the product of a host phospholipid degradation cascade that induces lipid droplet coalescence in enterocytes. This infection-induced cascade is inhibited by mutation of the V. cholerae glycine cleavage system due to intestinal accumulation of methionine sulfoxide (MetO), and both dietary supplementation with MetO and enterocyte knock-down of host methionine sulfoxide reductase A (MsrA) yield increased resistance to infection. MsrA converts both free and protein-associated MetO to methionine. These findings support a model in which dietary MetO competitively inhibits repair of host proteins by MsrA. Bacterial virulence strategies depend on functional host proteins. We propose a novel virulence paradigm in which an intestinal pathogen ensures the repair of host proteins essential for pathogenesis through consumption of dietary MetO.


Assuntos
Cólera , Interações Hospedeiro-Patógeno/fisiologia , Metionina/análogos & derivados , Vibrio cholerae/patogenicidade , Virulência/fisiologia , Animais , Western Blotting , Modelos Animais de Doenças , Drosophila melanogaster , Imunofluorescência , Metionina/metabolismo , Coelhos , Vibrio cholerae/metabolismo
6.
bioRxiv ; 2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36778396

RESUMO

Canonically, complement is a serum-based host defense system that protects against systemic microbial invasion. Little is known about the production and function of complement components on mucosal surfaces. Here we show gut complement component 3 (C3), central to complement function, is regulated by the composition of the microbiota in healthy humans and mice, leading to host-specific gut C3 levels. Stromal cells in intestinal lymphoid follicles (LFs) are the predominant source of intestinal C3. During enteric infection with Citrobacter rodentium or enterohemorrhagic Escherichia coli, luminal C3 levels increase significantly and are required for protection. C. rodentium is remarkably more invasive to the gut epithelium of C3-deficient mice than of wild-type mice. In the gut, C3-mediated phagocytosis of C. rodentium functions to clear pathogens. Our study reveals that variations in gut microbiota determine individuals’ intestinal mucosal C3 levels, dominantly produced by LF stromal cells, which directly correlate with protection against enteric infection. Highlights: Gut complement component 3 (C3) is induced by the microbiome in healthy humans and mice at a microbiota-specific level.Gut stromal cells located in intestinal lymphoid follicles are a major source of luminal C3 During enteric infections with Citrobacter rodentium or enterohemorrhagic Escherichia coli, gut luminal C3 levels increase and are required for protection. C. rodentium is significantly more invasive of the gut epithelium in C3-deficient mice when compared to WT mice. In the gut, C3-mediated opsonophagocytosis of C. rodentium functions to clear pathogens.

7.
Nat Microbiol ; 7(9): 1404-1418, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35982310

RESUMO

Members of the human gut microbiome enzymatically process many bioactive molecules in the gastrointestinal tract. Most gut bacterial modifications characterized so far are hydrolytic or reductive in nature. Here we report that abundant human gut bacteria from the phylum Bacteroidetes perform conjugative modifications by selectively sulfonating steroidal metabolites. While sulfonation is a ubiquitous biochemical modification, this activity has not yet been characterized in gut microbes. Using genetic and biochemical approaches, we identify a widespread biosynthetic gene cluster that encodes both a sulfotransferase (BtSULT, BT0416) and enzymes that synthesize the sulfonate donor adenosine 3'-phosphate-5'-phosphosulfate (PAPS), including an APS kinase (CysC, BT0413) and an ATP sulfurylase (CysD and CysN, BT0414-BT0415). BtSULT selectively sulfonates steroidal metabolites with a flat A/B ring fusion, including cholesterol. Germ-free mice monocolonized with Bacteroides thetaiotaomicron ΔBT0416 exhibited reduced gastrointestinal levels of cholesterol sulfate (Ch-S) compared with wild-type B. thetaiotaomicron-colonized mice. The presence of BtSULT and BtSULT homologues in bacteria inhibited leucocyte migration in vitro and in vivo, and abundances of cluster genes were significantly reduced in patients with inflammatory bowel disease. Together, these data provide a mechanism by which gut bacteria sulfonate steroidal metabolites and suggest that these compounds can modulate immune cell trafficking in the host.


Assuntos
Bacteroides thetaiotaomicron , Vias Biossintéticas , Animais , Bactérias , Trato Gastrointestinal , Humanos , Camundongos , Sulfato Adenililtransferase
8.
Cell Host Microbe ; 29(9): 1366-1377.e9, 2021 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-34416161

RESUMO

Bile acids act as signaling molecules that regulate immune homeostasis, including the differentiation of CD4+ T cells into distinct T cell subsets. The bile acid metabolite isoallolithocholic acid (isoalloLCA) enhances the differentiation of anti-inflammatory regulatory T cells (Treg cells) by facilitating the formation of a permissive chromatin structure in the promoter region of the transcription factor forkhead box P3 (Foxp3). Here, we identify gut bacteria that synthesize isoalloLCA from 3-oxolithocholic acid and uncover a gene cluster responsible for the conversion in members of the abundant human gut bacterial phylum Bacteroidetes. We also show that the nuclear hormone receptor NR4A1 is required for the effect of isoalloLCA on Treg cells. Moreover, the levels of isoalloLCA and its biosynthetic genes are significantly reduced in patients with inflammatory bowel diseases, suggesting that isoalloLCA and its bacterial producers may play a critical role in maintaining immune homeostasis in humans.


Assuntos
Bacteroidetes/metabolismo , Ácidos e Sais Biliares/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Fenantrenos/metabolismo , Linfócitos T Reguladores/imunologia , Diferenciação Celular/fisiologia , Cromatina/metabolismo , Fatores de Transcrição Forkhead/genética , Humanos , Doenças Inflamatórias Intestinais/patologia , Família Multigênica/genética , Regiões Promotoras Genéticas/genética , Transdução de Sinais/fisiologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/citologia
9.
Nat Microbiol ; 3(2): 243-252, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29180725

RESUMO

Vibrio cholerae colonizes the human terminal ileum to cause cholera, and the arthropod intestine and exoskeleton to persist in the aquatic environment. Attachment to these surfaces is regulated by the bacterial quorum-sensing signal transduction cascade, which allows bacteria to assess the density of microbial neighbours. Intestinal colonization with V. cholerae results in expenditure of host lipid stores in the model arthropod Drosophila melanogaster. Here we report that activation of quorum sensing in the Drosophila intestine retards this process by repressing V. cholerae succinate uptake. Increased host access to intestinal succinate mitigates infection-induced lipid wasting to extend survival of V. cholerae-infected flies. Therefore, quorum sensing promotes a more favourable interaction between V. cholerae and an arthropod host by reducing the nutritional burden of intestinal colonization.


Assuntos
Artrópodes/microbiologia , Intestinos/microbiologia , Percepção de Quorum/fisiologia , Vibrio cholerae/metabolismo , Vibrio cholerae/patogenicidade , Tecido Adiposo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Drosophila melanogaster/microbiologia , Feminino , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Técnicas de Silenciamento de Genes , Interações Hospedeiro-Patógeno/fisiologia , Lipólise , Tamanho do Órgão , Transdução de Sinais , Somatomedinas/genética , Ácido Succínico/metabolismo , Triglicerídeos/metabolismo , Vibrio cholerae/genética , Vibrio cholerae/crescimento & desenvolvimento , Virulência/genética
10.
J Exp Med ; 215(12): 3136-3150, 2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30442645

RESUMO

An inducible program of inflammatory gene expression is a hallmark of antimicrobial defenses. Recently, cellular nucleic acid-binding protein (CNBP) was identified as a regulator of nuclear factor-kappaB (NF-κB)-dependent proinflammatory cytokine gene expression. Here, we generated mice lacking CNBP and found that CNBP regulates a very restricted gene signature that includes IL-12ß. CNBP resides in the cytosol of macrophages and translocates to the nucleus in response to diverse microbial pathogens and pathogen-derived products. Cnbp-deficient macrophages induced canonical NF-κB/Rel signaling normally but were impaired in their ability to control the activation of c-Rel, a key driver of IL-12ß gene transcription. The nuclear translocation and DNA-binding activity of c-Rel required CNBP. Lastly, Cnbp-deficient mice were more susceptible to acute toxoplasmosis associated with reduced production of IL-12ß, as well as a reduced T helper type 1 (Th1) cell IFN-γ response essential to controlling parasite replication. Collectively, these findings identify CNBP as important regulator of c-Rel-dependent IL-12ß gene transcription and Th1 immunity.


Assuntos
Imunidade Celular , Subunidade p40 da Interleucina-12/imunologia , Proteínas de Ligação a RNA/imunologia , Células Th1/imunologia , Transcrição Gênica/imunologia , Animais , Interferon gama/genética , Interferon gama/imunologia , Subunidade p40 da Interleucina-12/genética , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/imunologia , Proteínas Proto-Oncogênicas c-rel/genética , Proteínas Proto-Oncogênicas c-rel/imunologia , Proteínas de Ligação a RNA/genética , Células Th1/citologia
11.
Mol Biol Cell ; 28(5): 681-691, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28077616

RESUMO

The initial metameric expression of the Drosophila sloppy paired 1 (slp1) gene is controlled by two distinct cis-regulatory DNA elements that interact in a nonadditive manner to integrate inputs from transcription factors encoded by the pair-rule segmentation genes. We performed chromatin immunoprecipitation on reporter genes containing these elements in different embryonic genotypes to investigate the mechanism of their regulation. The distal early stripe element (DESE) mediates both activation and repression by Runt. We find that the differential response of DESE to Runt is due to an inhibitory effect of Fushi tarazu (Ftz) on P-TEFb recruitment and the regulation of RNA polymerase II (Pol II) pausing. The proximal early stripe element (PESE) is also repressed by Runt, but in this case, Runt prevents PESE-dependent Pol II recruitment and preinitiation complex (PIC) assembly. PESE is also repressed by Even-skipped (Eve), but, of interest, this repression involves regulation of P-TEFb recruitment and promoter-proximal Pol II pausing. These results demonstrate that the mode of slp1 repression by Runt is enhancer specific, whereas the mode of repression of the slp1 PESE enhancer is transcription factor specific. We propose a model based on these differential regulatory interactions that accounts for the nonadditive interactions between the PESE and DESE enhancers during Drosophila segmentation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Padronização Corporal/genética , Proteínas de Ligação a DNA/genética , Drosophila/embriologia , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Embrião não Mamífero/metabolismo , Feminino , Fatores de Transcrição Fushi Tarazu/genética , Fatores de Transcrição Fushi Tarazu/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Proteínas de Homeodomínio/genética , Masculino , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Sequências Reguladoras de Ácido Nucleico , Fatores de Transcrição/genética
12.
Cell Host Microbe ; 16(5): 592-604, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25525791

RESUMO

Vibrio cholerae is lethal to the model host Drosophila melanogaster through mechanisms not solely attributable to cholera toxin. To examine additional virulence determinants, we performed a genetic screen in V. cholerae-infected Drosophila and identified the two-component system CrbRS. CrbRS controls transcriptional activation of acetyl-CoA synthase-1 (ACS-1) and thus regulates the acetate switch, in which bacteria transition from excretion to assimilation of environmental acetate. The resultant loss of intestinal acetate leads to deactivation of host insulin signaling and lipid accumulation in enterocytes, resulting in host lethality. These metabolic effects are not observed upon infection with ΔcrbS or Δacs1 V. cholerae mutants. Additionally, uninfected flies lacking intestinal commensals, which supply short chain fatty acids (SCFAs) such as acetate, also exhibit altered insulin signaling and intestinal steatosis, which is reversed upon acetate supplementation. Thus, acetate consumption by V. cholerae alters host metabolism, and dietary acetate supplementation may ameliorate some sequelae of cholera.


Assuntos
Acetatos/metabolismo , Interações Hospedeiro-Patógeno , Insulinas/metabolismo , Intestinos/microbiologia , Metabolismo dos Lipídeos , Vibrio cholerae/patogenicidade , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Toxina da Cólera/toxicidade , Coenzima A Ligases/genética , Coenzima A Ligases/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/microbiologia , Enterócitos/metabolismo , Imunidade Inata , Microbiota , Transdução de Sinais , Fatores de Virulência
13.
mBio ; 4(3): e00337-13, 2013 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-23781070

RESUMO

Vibrio cholerae is an estuarine bacterium and an intestinal pathogen of humans that causes severe epidemic diarrhea. In the absence of adequate mammalian models in which to study the interaction of V. cholerae with the host intestinal innate immune system, we have implemented Drosophila melanogaster as a surrogate host. We previously showed that immune deficiency pathway loss-of-function and mustard gain-of-function mutants are less susceptible to V. cholerae infection. We find that although the overall burden of intestinal bacteria is not significantly different from that of control flies, intestinal stem cell (ISC) division is increased in these mutants. This led us to examine the effect of V. cholerae on ISC division. We report that V. cholerae infection and cholera toxin decrease ISC division. Because IMD pathway and Mustard mutants, which are resistant to V. cholerae, maintain higher levels of ISC division during V. cholerae infection, we hypothesize that suppression of ISC division is a virulence strategy of V. cholerae and that accelerated epithelial regeneration protects the host against V. cholerae. Extension of these findings to mammals awaits the development of an adequate experimental model.


Assuntos
Divisão Celular/efeitos dos fármacos , Toxina da Cólera/toxicidade , Drosophila melanogaster/microbiologia , Células-Tronco/fisiologia , Vibrio cholerae/patogenicidade , Animais , Cólera/microbiologia , Cólera/patologia , Toxina da Cólera/metabolismo , Modelos Animais de Doenças , Drosophila melanogaster/fisiologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/fisiologia , Imunossupressores/metabolismo , Imunossupressores/toxicidade , Células-Tronco/microbiologia , Vibrio cholerae/metabolismo
14.
Cell Host Microbe ; 14(3): 294-305, 2013 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-24034615

RESUMO

Cholera toxin (CT), a virulence factor elaborated by Vibrio cholerae, is sufficient to induce the severe diarrhea characteristic of cholera. The enzymatic moiety of CT (CtxA) increases cAMP synthesis in intestinal epithelial cells, leading to chloride ion (Cl(-)) efflux through the CFTR Cl(-) channel. To preserve electroneutrality and osmotic balance, sodium ions and water also flow into the intestinal lumen via a paracellular route. We find that CtxA-driven cAMP increase also inhibits Rab11/exocyst-mediated trafficking of host proteins including E-cadherin and Notch signaling components to cell-cell junctions in Drosophila, human intestinal epithelial cells, and ligated mouse ileal loops, thereby disrupting barrier function. Additionally, CtxA induces junctional damage, weight loss, and dye leakage in the Drosophila gut, contributing to lethality from live V. cholerae infection, all of which can be rescued by Rab11 overexpression. These barrier-disrupting effects of CtxA may act in parallel with Cl(-) secretion to drive the pathophysiology of cholera.


Assuntos
Toxina da Cólera/metabolismo , Células Epiteliais/fisiologia , Exossomos/efeitos dos fármacos , Interações Hospedeiro-Patógeno , Proteínas de Junções Íntimas/antagonistas & inibidores , Junções Íntimas/fisiologia , Vibrio cholerae/fisiologia , Animais , Linhagem Celular , Cloro/metabolismo , AMP Cíclico/metabolismo , Modelos Animais de Doenças , Drosophila , Células Epiteliais/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Humanos , Camundongos , Modelos Biológicos , Sódio/metabolismo , Análise de Sobrevida , Junções Íntimas/efeitos dos fármacos , Água/metabolismo
15.
Mol Biol Cell ; 22(8): 1364-74, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21325629

RESUMO

Runt is a vital transcriptional regulator in the developmental pathway responsible for segmentation in the Drosophila embryo. Runt activates or represses transcription in a manner that is dependent on both cellular context and the specific downstream target. Here we identify Hairless (H) as a Runt-interacting molecule that functions during segmentation. We find that H is important for maintenance of engrailed (en) repression as was previously demonstrated for Groucho (Gro), Rpd3, and CtBP. H also contributes to the Runt-dependent repression of sloppy-paired-1 (slp1), a role that is not shared with these other corepressors. We further find distinct roles for these different corepressors in the regulation of other Runt targets in the early Drosophila embryo. These findings, coupled with observations on the distinct functional requirements for Runt in regulating these several different targets, indicate that Runt-dependent regulation in the Drosophila blastoderm embryo relies on unique, target-gene-specific molecular interactions.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Blastoderma/fisiologia , Padronização Corporal/fisiologia , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Embrião não Mamífero/metabolismo , Feminino , Proteínas de Homeodomínio/genética , Proteínas Nucleares/genética , Mapeamento de Interação de Proteínas , Análise de Sequência , Deleção de Sequência , Fatores de Transcrição/genética , Técnicas do Sistema de Duplo-Híbrido
16.
PLoS One ; 5(7): e11498, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20634899

RESUMO

A hallmark of genes that are subject to developmental regulation of transcriptional elongation is association of the negative elongation factor NELF with the paused RNA polymerase complex. Here we use a combination of biochemical and genetic experiments to investigate the in vivo function of NELF in the Drosophila embryo. NELF associates with different gene promoter regions in correlation with the association of RNA polymerase II (Pol II) and the initial activation of gene expression during the early stages of embryogenesis. Genetic experiments reveal that maternally provided NELF is required for the activation, rather than the repression of reporter genes that emulate the expression of key developmental control genes. Furthermore, the relative requirement for NELF is dictated by attributes of the flanking cis-regulatory information. We propose that NELF-associated paused Pol II complexes provide a platform for high fidelity integration of the combinatorial spatial and temporal information that is central to the regulation of gene expression during animal development.


Assuntos
Drosophila/metabolismo , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição/metabolismo , Animais , Western Blotting , Imunoprecipitação da Cromatina , Drosophila/embriologia , Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hibridização In Situ , Regiões Promotoras Genéticas/genética , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética
17.
Mol Biol Cell ; 21(13): 2315-26, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20462957

RESUMO

Runx proteins play vital roles in regulating transcription in numerous developmental pathways throughout the animal kingdom. Two Runx protein hallmarks are the DNA-binding Runt domain and a C-terminal VWRPY motif that mediates interaction with TLE/Gro corepressor proteins. A phylogenetic analysis of Runt, the founding Runx family member, identifies four distinct regions C-terminal to the Runt domain that are conserved in Drosophila and other insects. We used a series of previously described ectopic expression assays to investigate the functions of these different conserved regions in regulating gene expression during embryogenesis and in controlling axonal projections in the developing eye. The results indicate each conserved region is required for a different subset of activities and identify distinct regions that participate in the transcriptional activation and repression of the segmentation gene sloppy-paired-1 (slp1). Interestingly, the C-terminal VWRPY-containing region is not required for repression but instead plays a role in slp1 activation. Genetic experiments indicating that Groucho (Gro) does not participate in slp1 regulation further suggest that Runt's conserved C-terminus interacts with other factors to promote transcriptional activation. These results provide a foundation for further studies on the molecular interactions that contribute to the context-dependent properties of Runx proteins as developmental regulators.


Assuntos
Subunidades alfa de Fatores de Ligação ao Core/genética , Transcrição Gênica , Sequência de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Subunidades alfa de Fatores de Ligação ao Core/classificação , Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Insetos/classificação , Insetos/genética , Insetos/metabolismo , Dados de Sequência Molecular , Filogenia , Estrutura Terciária de Proteína , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Alinhamento de Sequência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
19.
Acta Biochim Biophys Sin (Shanghai) ; 37(10): 694-701, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16215637

RESUMO

4-1BBL (TNFSF9) is a member of the tumor necrosis factor (TNF) ligand superfamily, which is expressed on some activated antigen presenting cells and B cells. We isolated a rat cDNA clone encoding the rat homologue of the human 4-1BBL (GenBank accession No. AY259541). The deduced rat 4-1BBL protein, consisting of 308 amino acids with a molecular weight of 33,469 Da, was a typical type II transmembrane glycoprotein, the same as human and murine 4-1BBL. "SDAA" in the cytoplasmic domain of rat 4-1BBL was deduced to act as the phosphorylation site for casein kinase I ("SXXS" motif), which is present in the cytoplasmic domains of human and murine 4-1BBL, and all other TNF ligand family members known to utilize reverse signaling. The two introns of 4-1BBL were also cloned (GenBank accession No. AY332409). Rat 4-1BBL is much more homologous with murine 4-1BBL than with human 4-1BBL, in both nucleotide and amino acid sequences. Rat 4-1BBL was expressed in all tested tissues: brain, lung, colon, liver, thymus, testicle, kidney, adrenal, stomach, spleen and heart. The chromosomal location of rat 4-1BBL was first identified by bioinformatics, then by fluorescence in situ hybridization at 9q11 (GenBank accession UniGene No. Rn.46783). Rat, murine and human 4-1BBL genes are evolved from a common gene. The identification and characterization of the rat counterpart of human 4-1BBL will facilitate studies of the biological function of this molecule.


Assuntos
Fator de Necrose Tumoral alfa/genética , Ligante 4-1BB , Sequência de Aminoácidos , Animais , Sequência de Bases , Mapeamento Cromossômico , Clonagem Molecular , Hibridização in Situ Fluorescente , Masculino , Dados de Sequência Molecular , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/biossíntese
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa