Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Gastroenterology ; 148(7): 1405-1416.e3, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25701737

RESUMO

BACKGROUND & AIMS: Defects in colonic epithelial barrier defenses are associated with ulcerative colitis (UC). The proteins that regulate bacterial clearance in the colonic epithelium have not been completely identified. The Drosophila chromosome-associated protein D3 (dCAP-D3) regulates responses to bacterial infection. We examined whether CAP-D3 promotes bacterial clearance in human colonic epithelium. METHODS: Clearance of Salmonella or adherent-invasive Escherichia coli LF82 was assessed by gentamycin protection assays in HT-29 and Caco-2 cells expressing small hairpin RNAs against CAP-D3. We used immunoblot assays to measure levels of CAP-D3 in colonic epithelial cells from patients with UC and healthy individuals (controls). RNA sequencing identified genes activated by CAP-D3. We analyzed the roles of CAP-D3 target genes in bacterial clearance using gentamycin protection and immunofluorescence assays and studies with pharmacologic inhibitors. RESULTS: CAP-D3 expression was reduced in colonic epithelial cells from patients with active UC. Reduced CAP-D3 expression decreased autophagy and impaired intracellular bacterial clearance by HT-29 and Caco-2 colonic epithelial cells. Lower levels of CAP-D3 increased transcription of genes encoding SLC7A5 and SLC3A2, the products of which heterodimerize to form an amino acid transporter in HT-29 cells after bacterial infection; levels of SLC7A5-SLC3A2 were increased in tissues from patients with UC compared with controls. Reduced CAP-D3 in HT-29 cells resulted in earlier recruitment of SLC7A5 to Salmonella-containing vacuoles, increased activity of mTORC1, and increased survival of bacteria. Inhibition of SLC7A5-SLC3A2 or mTORC1 activity rescued the bacterial clearance defects of CAP-D3-deficient cells. CONCLUSIONS: CAP-D3 down-regulates transcription of genes that encode amino acid transporters (SLC7A5 and SLC3A2) to promote bacterial autophagy by colon epithelial cells. Levels of CAP-D3 protein are reduced in patients with active UC; strategies to increase its levels might restore mucosal homeostasis to patients with active UC.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Escherichia coli/fisiologia , Cadeia Pesada da Proteína-1 Reguladora de Fusão/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Salmonella/fisiologia , Adenosina Trifosfatases , Autofagia , Células CACO-2 , Proteínas de Ciclo Celular/genética , Colite Ulcerativa/imunologia , Colite Ulcerativa/metabolismo , Colite Ulcerativa/microbiologia , Doença de Crohn/imunologia , Doença de Crohn/metabolismo , Doença de Crohn/microbiologia , Proteínas de Drosophila , Células Epiteliais/imunologia , Escherichia coli/imunologia , Cadeia Pesada da Proteína-1 Reguladora de Fusão/genética , Regulação da Expressão Gênica , Células HT29 , Humanos , Imunidade Inata , Mucosa Intestinal/imunologia , Transportador 1 de Aminoácidos Neutros Grandes/genética , Alvo Mecanístico do Complexo 1 de Rapamicina , Viabilidade Microbiana , Complexos Multiproteicos/metabolismo , Interferência de RNA , Salmonella/imunologia , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo , Transcrição Gênica , Transfecção
2.
Am J Pathol ; 185(6): 1624-37, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25864926

RESUMO

Intestinal epithelial cell (IEC) death is typical of inflammatory bowel disease (IBD). We investigated: i) whether IEC-released necrotic cell products (proinflammatory mediators) amplify mucosal inflammation, ii) the capacity of necrotic cell lysates from HT29 cells or human IECs to induce human intestinal fibroblasts' (HIF) production of IL-6 and IL-8, and iii) whether IL-1α, released by injured colonocytes, exacerbated experimental IBD. Necrotic cell lysates potently induced HIF IL-6 and IL-8 production independent of Toll-like receptors 2 and 4, receptor for advanced glycation end-products, high-mobility group box 1, uric acid, IL-33, or inflammasome activation. IL-1α was the key IEC-derived necrotic cell product involved in HIF cytokine production. IL-1α-positive cells were identified in the epithelium in human IBD and dextran sulfate sodium (DSS)-induced colitis. IL-1α was detected in the stool of colitic mice before IL-1ß. IL-1α enemas reactivated inflammation after DSS colitis recovery, induced IL-1 receptor expression in subepithelial fibroblasts, and activated de novo inflammation even in mice without overt colitis, after the administration of low-dose DSS. IL-1α amplifies gut inflammation by inducing cytokine production by mesenchymal cells. IL-1α-mediated IEC-fibroblast interaction may be involved in amplifying and perpetuating inflammation, even without obvious intestinal damage. IL-1α may be a target for treating early IBD or preventing the reactivation of IBD.


Assuntos
Colite/metabolismo , Fibroblastos/metabolismo , Inflamação/metabolismo , Interleucina-1alfa/metabolismo , Mucosa Intestinal/metabolismo , Animais , Colite/induzido quimicamente , Colite/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Fibroblastos/patologia , Células HT29 , Humanos , Inflamação/patologia , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Intestinos/patologia , Camundongos
3.
J Biol Chem ; 288(40): 29090-104, 2013 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-23950179

RESUMO

Breast-feeding is associated with enhanced protection from gastrointestinal disease in infants, mediated in part by an array of bioactive glycan components in milk that act through molecular mechanisms to inhibit enteric pathogen infection. Human milk contains hyaluronan (HA), a glycosaminoglycan polymer found in virtually all mammalian tissues. We have shown that synthetic HA of a specific size range promotes expression of antimicrobial peptides in intestinal epithelium. We hypothesize that hyaluronan from human milk also enhances innate antimicrobial defense. Here we define the concentration of HA in human milk during the first 6 months postpartum. Importantly, HA isolated from milk has a biological function. Treatment of HT-29 colonic epithelial cells with human milk HA at physiologic concentrations results in time- and dose-dependent induction of the antimicrobial peptide human ß-defensin 2 and is abrogated by digestion of milk HA with a specific hyaluronidase. Milk HA induction of human ß-defensin 2 expression is also reduced in the presence of a CD44-blocking antibody and is associated with a specific increase in ERK1/2 phosphorylation, suggesting a role for the HA receptor CD44. Furthermore, oral administration of human milk-derived HA to adult, wild-type mice results in induction of the murine Hß D2 ortholog in intestinal mucosa and is dependent upon both TLR4 and CD44 in vivo. Finally, treatment of cultured colonic epithelial cells with human milk HA enhances resistance to infection by the enteric pathogen Salmonella typhimurium. Together, our observations suggest that maternally provided HA stimulates protective antimicrobial defense in the newborn.


Assuntos
Ácido Hialurônico/farmacologia , Imunidade Inata/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Leite Humano/química , Administração Oral , Animais , Anticorpos/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Contagem de Colônia Microbiana , Resistência à Doença/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Células HT29 , Humanos , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/administração & dosagem , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Lactação/efeitos dos fármacos , Camundongos , Microscopia de Fluorescência , Fosforilação/efeitos dos fármacos , Período Pós-Parto , Transporte Proteico/efeitos dos fármacos , Salmonelose Animal/imunologia , Salmonelose Animal/patologia , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/fisiologia , Homologia de Sequência de Aminoácidos , Receptor 4 Toll-Like/metabolismo , beta-Defensinas/metabolismo
4.
J Biol Chem ; 287(30): 25565-76, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22665475

RESUMO

Autophagy is triggered by the intracellular bacterial sensor NOD2 (nucleotide-binding, oligomerization domain 2) as an anti-bacterial response. Defects in autophagy have been implicated in Crohn's disease susceptibility. The molecular mechanisms of activation and regulation of this process by NOD2 are not well understood, with recent studies reporting conflicting requirements for RIP2 (receptor-interacting protein kinase 2) in autophagy induction. We examined the requirement of NOD2 signaling mediated by RIP2 for anti-bacterial autophagy induction and clearance of Salmonella typhimurium in the intestinal epithelial cell line HCT116. Our data demonstrate that NOD2 stimulates autophagy in a process dependent on RIP2 tyrosine kinase activity. Autophagy induction requires the activity of the mitogen-activated protein kinases MEKK4 and p38 but is independent of NFκB signaling. Activation of autophagy was inhibited by a PP2A phosphatase complex, which interacts with both NOD2 and RIP2. PP2A phosphatase activity inhibited NOD2-dependent autophagy but not activation of NFκB or p38. Upon stimulation of NOD2, the phosphatase activity of the PP2A complex is inhibited through tyrosine phosphorylation of the catalytic subunit in a process dependent on RIP2 activity. These findings demonstrate that RIP2 tyrosine kinase activity is not only required for NOD2-dependent autophagy but plays a dual role in this process. RIP2 both sends a positive autophagy signal through activation of p38 MAPK and relieves repression of autophagy mediated by the phosphatase PP2A.


Assuntos
Autofagia , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína Adaptadora de Sinalização NOD2/metabolismo , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Ativação Enzimática/genética , Células Epiteliais/microbiologia , Células HEK293 , Humanos , Mucosa Intestinal/microbiologia , MAP Quinase Quinase Quinase 4/genética , MAP Quinase Quinase Quinase 4/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD2/genética , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/genética , Infecções por Salmonella/genética , Infecções por Salmonella/metabolismo , Salmonella typhimurium , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Gastroenterology ; 142(7): 1483-92.e6, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22387394

RESUMO

BACKGROUND & AIMS: Polymorphisms that reduce the function of nucleotide-binding oligomerization domain (NOD)2, a bacterial sensor, have been associated with Crohn's disease (CD). No proteins that regulate NOD2 activity have been identified as selective pharmacologic targets. We sought to discover regulators of NOD2 that might be pharmacologic targets for CD therapies. METHODS: Carbamoyl phosphate synthetase/aspartate transcarbamylase/dihydroorotase (CAD) is an enzyme required for de novo pyrimidine nucleotide synthesis; it was identified as a NOD2-interacting protein by immunoprecipitation-coupled mass spectrometry. CAD expression was assessed in colon tissues from individuals with and without inflammatory bowel disease by immunohistochemistry. The interaction between CAD and NOD2 was assessed in human HCT116 intestinal epithelial cells by immunoprecipitation, immunoblot, reporter gene, and gentamicin protection assays. We also analyzed human cell lines that express variants of NOD2 and the effects of RNA interference, overexpression and CAD inhibitors. RESULTS: CAD was identified as a NOD2-interacting protein expressed at increased levels in the intestinal epithelium of patients with CD compared with controls. Overexpression of CAD inhibited NOD2-dependent activation of nuclear factor κB and p38 mitogen-activated protein kinase, as well as intracellular killing of Salmonella. Reduction of CAD expression or administration of CAD inhibitors increased NOD2-dependent signaling and antibacterial functions of NOD2 variants that are and are not associated with CD. CONCLUSIONS: The nucleotide synthesis enzyme CAD is a negative regulator of NOD2. The antibacterial function of NOD2 variants that have been associated with CD increased in response to pharmacologic inhibition of CAD. CAD is a potential therapeutic target for CD.


Assuntos
Aspartato Carbamoiltransferase/fisiologia , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/fisiologia , Doença de Crohn/imunologia , Desoxirribonucleases/fisiologia , Di-Hidro-Orotase/fisiologia , Mucosa Intestinal/microbiologia , Proteína Adaptadora de Sinalização NOD2/imunologia , Aspartato Carbamoiltransferase/antagonistas & inibidores , Aspartato Carbamoiltransferase/uso terapêutico , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/antagonistas & inibidores , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/uso terapêutico , Linhagem Celular , Células Cultivadas , Doença de Crohn/tratamento farmacológico , Doença de Crohn/microbiologia , Di-Hidro-Orotase/antagonistas & inibidores , Di-Hidro-Orotase/uso terapêutico , Inibidores Enzimáticos/farmacologia , Humanos , Imuno-Histoquímica , Imunoprecipitação , Mucosa Intestinal/imunologia , Espectrometria de Massas , NF-kappa B/fisiologia , Proteína Adaptadora de Sinalização NOD2/fisiologia , Salmonella/crescimento & desenvolvimento , Salmonella/imunologia , Transdução de Sinais
6.
J Biol Chem ; 286(3): 1938-50, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21097508

RESUMO

The Crohn's disease and early onset sarcoidosis susceptibility protein, NOD2, coordinates innate immune signaling pathways. Because dysregulation of this coordination can lead to inflammatory disease, maintaining appropriate activation of the NOD2 signaling pathway is paramount in immunologic homeostasis. In this work, we identify the atypical tumor necrosis factor-associated factor (TRAF) family member, TRAF4, as a key negative regulator of NOD2 signaling. TRAF4 inhibits NOD2-induced NF-κB activation and directly binds to NOD2 to inhibit NOD2-induced bacterial killing. We find that two consecutive glutamate residues in NOD2 are required for interaction with TRAF4 and inhibition of NOD2 signaling because mutation of these residues abrogated both TRAF4 binding and inhibition of NOD2. This work identifies a novel negative regulator of NOD2 signaling. Additionally, it defines a TRAF4 binding motif within NOD2 involved in termination of innate immune signaling responses.


Assuntos
Doença de Crohn/metabolismo , Regulação para Baixo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Fator 4 Associado a Receptor de TNF/metabolismo , Motivos de Aminoácidos , Animais , Doença de Crohn/genética , Doença de Crohn/imunologia , Predisposição Genética para Doença/genética , Células HEK293 , Humanos , Imunidade Inata , Camundongos , NF-kappa B/genética , NF-kappa B/imunologia , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD2/genética , Proteína Adaptadora de Sinalização NOD2/imunologia , Ligação Proteica , Infecções por Salmonella/imunologia , Infecções por Salmonella/metabolismo , Salmonella typhimurium/imunologia , Salmonella typhimurium/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fator 4 Associado a Receptor de TNF/genética , Fator 4 Associado a Receptor de TNF/imunologia
7.
Gastroenterology ; 139(5): 1630-41, 1641.e1-2, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20637199

RESUMO

BACKGROUND & AIMS: The identification of numerous genes that confer susceptibility to Crohn's disease (CD) indicates that this complex disease might arise from alterations in several genes with related functions. We examined the functional interaction between the CD risk genes ATG16L1 and NOD2 to identify an autophagy-dependent pathway that is altered by disease-associated variants. METHODS: We assessed Nod2 signaling and autophagy activation in response to muramyl dipeptide (MDP) by immunoblot, confocal microscopy, flow cytometry, reporter gene, and gentamicin protection assays in human epithelial cell lines and primary human macrophages and dendritic cells from healthy individuals. The requirement of Nod2 and ATG16L1 expression and the effects of CD-associated variants in MDP-stimulated autophagy and Nod2-dependent signaling were assessed in cell lines manipulated by RNA interference, inhibitors, or ATG16L1 or NOD2 variants and in primary macrophages and dendritic cells from healthy genotyped donors. RESULTS: MDP stimulation of epithelial cells, macrophages, and dendritic cells activated autophagy and nuclear factor κB and mitogen-activated protein kinase signaling; it also increased killing of Salmonella. These responses depended on ATG16L1 and Nod2 expression and were impaired by CD-associated NOD2 variants. Nod2-dependent signaling was not impaired in cells with the ATG16L1 T300A genotype, which is associated with CD. However, the ATG16L1 T300A variant blocked the increase in MDP-mediated killing of Salmonella only in epithelial cell lines and not primary macrophages or dendritic cells. CONCLUSIONS: ATG16L1 and NOD2 are components of an autophagy-mediated antibacterial pathway that is altered in a cell- and function-specific manner by CD-associated mutations.


Assuntos
Autofagia , Proteínas de Transporte/genética , Doença de Crohn/genética , Expressão Gênica , Predisposição Genética para Doença , Proteína Adaptadora de Sinalização NOD2/genética , RNA/genética , Proteínas Relacionadas à Autofagia , Proteínas de Transporte/metabolismo , Linhagem Celular , Doença de Crohn/metabolismo , Doença de Crohn/patologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Genótipo , Humanos , Immunoblotting , Microscopia Confocal , Proteína Adaptadora de Sinalização NOD2/metabolismo , Fagocitose
8.
Sci Rep ; 8(1): 8708, 2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29880914

RESUMO

Multidrug-resistant bacterial strains are a rapidly emerging healthcare threat; therefore it is critical to develop new therapies to combat these organisms. Prior antibacterial strategies directly target pathogen growth or viability. Host-directed strategies to increase antimicrobial defenses may be an effective alternative to antibiotics and reduce development of resistant strains. In this study, we demonstrated the efficacy of a pyrimidine synthesis inhibitor, N-phosphonacetyl-L-aspartate (PALA), to enhance clearance of methicillin-resistant Staphylococcus aureus (MRSA), Pseudomonas aeruginosa, and Acinetobacter baumannii strains by primary human dermal fibroblasts in vitro. PALA did not have a direct bactericidal effect, but enhanced cellular secretion of the antimicrobial peptides human ß-defensin 2 (HBD2) and HBD3 from fibroblasts. When tested in porcine and human skin explant models, a topical PALA formulation was efficacious to enhance MRSA, P. aeruginosa, and A. baumannii clearance. Topical PALA treatment of human skin explants also resulted in increased HBD2 and cathelicidin (LL-37) production. The antimicrobial actions of PALA required expression of nucleotide-binding, oligomerization domain 2 (NOD2), receptor-interacting serine/threonine-protein kinase 2 (RIP2), and carbamoyl phosphatase synthase II/aspartate transcarbamylase/dihydroorotase (CAD). Our results indicate that PALA may be a new option to combat multidrug-resistant bacterial infections of the skin through enhancement of an integral pathway of the cutaneous innate immune defense system.


Assuntos
Ácido Aspártico/análogos & derivados , Bactérias/imunologia , Derme/imunologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Proteína Adaptadora de Sinalização NOD2/imunologia , Ácido Fosfonoacéticos/análogos & derivados , Pirimidinas/imunologia , Transdução de Sinais/efeitos dos fármacos , Dermatopatias Bacterianas/tratamento farmacológico , Animais , Ácido Aspártico/farmacologia , Bactérias/patogenicidade , Derme/microbiologia , Derme/patologia , Farmacorresistência Bacteriana Múltipla/imunologia , Células HEK293 , Humanos , Proteína Adaptadora de Sinalização NOD2/metabolismo , Ácido Fosfonoacéticos/farmacologia , Pirimidinas/biossíntese , Transdução de Sinais/imunologia , Dermatopatias Bacterianas/enzimologia , Dermatopatias Bacterianas/imunologia , Dermatopatias Bacterianas/microbiologia , Suínos
9.
Matrix Biol ; 62: 28-39, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-27845198

RESUMO

Maintaining a healthy intestinal barrier, the primary physical barrier between intestinal microbiota and the underlying lamina propria, is critical for optimal health. Epithelial integrity is essential for the prevention of the entrance of luminal contents, such as bacteria and their products, through the large intestinal barrier. In this study, we investigated the protective functions of biosynthetic, specific sized, hyaluronan around 35kDa (HA35) on intestinal epithelium in healthy mice, as well as mice infected Citrobacter rodentium, an established model that mimics infection with a serious human pathogen, enteropathogenic E. coli (EPEC). Our results reveal that treatment with HA35 protects mice from Citrobacter infection and enhances the epithelial barrier function. In particular, we have found that HA35 induces the expression of tight junction protein zonula occludens (ZO)-1 in both healthy and Citrobacter infected mice, as demonstrated by immunoflurorescence and Western blot analyses. Furthermore, we determined that HA35 treatment enhances ZO-1 expression and reduces intestinal permeability at the early stages of dextran sulfate sodium (DSS)-induced colitis in mice. Together, our data demonstrate that the expression and functionality of tight junctions, are increased by HA35 treatment, suggesting a novel mechanism for the protection from Citrobacter infection.


Assuntos
Colite/metabolismo , Infecções por Enterobacteriaceae/prevenção & controle , Ácido Hialurônico/administração & dosagem , Mucosa Intestinal/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo , Administração Oral , Animais , Citrobacter rodentium/efeitos dos fármacos , Colite/induzido quimicamente , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Infecções por Enterobacteriaceae/metabolismo , Regulação da Expressão Gênica , Ácido Hialurônico/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
10.
Oncogene ; 24(56): 8314-25, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16158057

RESUMO

Nuclear localization and high levels of the Y-box-binding protein YB1 appear to be important indicators of drug resistance and tumor prognosis. YB1 also interacts with the p53 tumor suppressor protein. In this paper, we have continued to explore YB1/p53 interactions. We report that transcriptionally active p53 is required for nuclear localization of YB1. We go on to show that nuclear YB1 regulates p53 function. Our data demonstrate that YB1 inhibits the ability of p53 to cause cell death and to transactivate cell death genes, but does not interfere with the ability of p53 to transactivate the CDKN1A gene, encoding the kinase p21(WAF1/CIP1) required for cell cycle arrest, nor the MDM2 gene. We also show that nuclear YB1 is associated with a failure to increase the level of the Bax protein in normal mammary epithelial cells after stress activation of p53. Together these data suggest that (nuclear) YB1 selectively alters p53 activity, which may in part provide an explanation for the correlation of nuclear YB1 with drug resistance and poor tumor prognosis.


Assuntos
Apoptose/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Humanos , Glândulas Mamárias Animais/metabolismo , Regiões Promotoras Genéticas , Transporte Proteico/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/genética , Ratos , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2/antagonistas & inibidores , Proteína X Associada a bcl-2/genética
11.
Oncogene ; 22(18): 2782-94, 2003 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-12743601

RESUMO

Nuclear localization and high levels of the Y-box binding protein YB1 appear to be important indicators of drug resistance and tumor prognosis. YB1 also interacts with the p53 tumor suppressor protein. In this paper, we explore a role for p53 in the nuclear localization of YB1. We report that various genotoxic stresses induce nuclear localization of YB1 in a small proportion of treated cells, but only in cells with wild-type p53. We go on to show directly that functional p53 is required for YB1 to translocate to the nucleus. Tumor-associated p53 mutants however are attenuated for YB1 nuclear localization as are mutants mutated in the proline-rich domain of p53. These data link the DNA-damage response of p53 to YB1 nuclear translocation. In addition, we find that YB1 inhibits p53-induced cell death and its ability to trans-activate promoters of genes involved in cell death signaling. Together these data suggest that some forms of p53 cause YB1 to accumulate in the nucleus, which in turn inhibits p53 activity. These results provide a possible explanation for the correlation of nuclear YB1 with drug resistance and poor prognosis in some tumor types, and for the first time implicate p53 in the process of nuclear translocation.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas de Ligação a DNA , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Transporte Ativo do Núcleo Celular , Adenovírus Humanos , Antineoplásicos/farmacologia , Proteínas Estimuladoras de Ligação a CCAAT/genética , Transformação Celular Viral , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Humanos , Cinética , Fatores de Transcrição NFI , Proteínas Nucleares , Plasmídeos , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/genética , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação a Y-Box
12.
PLoS One ; 9(7): e101789, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25000398

RESUMO

In the latter half of the 20th century, societal and technological changes led to a shift in the composition of the American diet to include a greater proportion of processed, pre-packaged foods high in fat and carbohydrates, and low in dietary fiber (a "Western diet"). Over the same time period, there have been parallel increases in Salmonella gastroenteritis cases and a broad range of chronic inflammatory diseases associated with intestinal dysbiosis. Several polysaccharide food additives are linked to bacterially-driven intestinal inflammation and may contribute to the pathogenic effects of a Western diet. Therefore, we examined the effect of a ubiquitous polysaccharide food additive, maltodextrin (MDX), on clearance of the enteric pathogen Salmonella using both in vitro and in vivo infection models. When examined in vitro, murine bone marrow-derived macrophages exposed to MDX had altered vesicular trafficking, suppressed NAPDH oxidase expression, and reduced recruitment of NADPH oxidase to Salmonella-containing vesicles, which resulted in persistence of Salmonella in enlarged Rab7+ late endosomal vesicles. In vivo, mice consuming MDX-supplemented water had a breakdown of the anti-microbial mucous layer separating gut bacteria from the intestinal epithelium surface. Additionally, oral infection of these mice with Salmonella resulted in increased cecal bacterial loads and enrichment of lamina propria cells harboring large Rab7+ vesicles. These findings indicate that consumption of processed foods containing the polysaccharide MDX contributes to suppression of intestinal anti-microbial defense mechanisms and may be an environmental priming factor for the development of chronic inflammatory disease.


Assuntos
Carboidratos da Dieta/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Viabilidade Microbiana/efeitos dos fármacos , Polissacarídeos/farmacologia , Salmonella typhi/efeitos dos fármacos , Salmonella typhi/fisiologia , Animais , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Camundongos , NADPH Oxidases/metabolismo , Transporte Proteico/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
13.
Inflamm Bowel Dis ; 18(4): 782-92, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21936032

RESUMO

The success of genetic analyses identifying multiple loci associated with inflammatory bowel disease (IBD) susceptibility has resulted in the identification of several risk genes linked to a common cellular process called autophagy. Autophagy is a process involving the encapsulation of cytosolic cellular components in double-membrane vesicles, their subsequent lysosomal degradation, and recycling of the degraded components for use by the cell. It plays an important part in the innate immune response to a variety of intracellular pathogens, and it is this component of autophagy that appears to be defective in IBD. This has lead to the hypothesis that Crohn's disease may result from an impaired antibacterial response, which leads to ineffective control of bacterial infection, dysbiosis of the intestinal microbiota, and chronic inflammation. Several recurrent themes have surfaced from studies examining the function of autophagy-related genes in the context of IBD, with cellular context, disease status, risk variant effect, and risk gene interplay all affecting the interpretation of these studies. The identification of autophagy as a major risk pathway in IBD is a significant step forward and may lead to pathway-focused therapy in the future; however, there is more to understand in order to unravel the complexity of this disease.


Assuntos
Autofagia/genética , Doenças Inflamatórias Intestinais/genética , Fagossomos/genética , Proteínas Relacionadas à Autofagia , Proteínas de Transporte/genética , Feminino , Proteínas de Ligação ao GTP/genética , Predisposição Genética para Doença , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Masculino , Proteína Adaptadora de Sinalização NOD2/genética , Polimorfismo Genético , Proteínas Serina-Treonina Quinases/genética , Receptores de Calcitriol/genética , Risco
14.
Mol Cell Biol ; 32(13): 2479-89, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22547678

RESUMO

Despite their homology, IκB kinase α (IKKα) and IKKß have divergent roles in NF-κB signaling. IKKß strongly activates NF-κB while IKKα can downregulate NF-κB under certain circumstances. Given this, identifying independent substrates for these kinases could help delineate their divergent roles. Peptide substrate array technology followed by bioinformatic screening identified TRAF4 as a substrate for IKKα. Like IKKα, TRAF4 is atypical within its family because it is the only TRAF family member to negatively regulate innate immune signaling. IKKα's phosphorylation of serine-426 on TRAF4 was required for this negative regulation. Binding to the Crohn's disease susceptibility protein, NOD2, is required for TRAF4 phosphorylation and subsequent inhibition of NOD2 signaling. Structurally, serine-426 resides within an exaggerated ß-bulge in TRAF4 that is not present in the other TRAF proteins, and phosphorylation of this site provides a structural basis for the atypical function of TRAF4 and its atypical role in NOD2 signaling.


Assuntos
Quinase I-kappa B/metabolismo , Imunidade Inata , Fator 4 Associado a Receptor de TNF/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Doença de Crohn/genética , Doença de Crohn/imunologia , Doença de Crohn/metabolismo , Primers do DNA/genética , Regulação para Baixo , Células HCT116 , Células HEK293 , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/imunologia , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD2/genética , Proteína Adaptadora de Sinalização NOD2/metabolismo , Biblioteca de Peptídeos , Fosforilação , Estabilidade Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Serina/química , Transdução de Sinais/imunologia , Especificidade por Substrato , Fator 4 Associado a Receptor de TNF/química , Fator 4 Associado a Receptor de TNF/imunologia
15.
J Biol Chem ; 278(37): 35516-23, 2003 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-12835324

RESUMO

The p53 tumor suppressor plays a major role in preventing tumor development by transactivating genes to remove or repair potentially tumorigenic cells. Here we show that the Y-box-binding protein, YB1, acts as a negative regulator of p53. Using reporter assays we show that YB1 represses transcription of the p53 promoter in a sequence-specific manner. We also show that YB1 reduces endogenous levels of p53, which in turn reduces p53 activity. Conversely, inhibiting YB1 in a variety of tumor cell lines induces p53 activity, resulting in significant apoptosis via a p53-dependent pathway. These data suggest that YB1 may, in some situations, protect cells from p53-mediated apoptosis, indicating that YB1 may be a good target for the development of new therapeutics.


Assuntos
Apoptose/fisiologia , Proteínas Estimuladoras de Ligação a CCAAT/fisiologia , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/fisiologia , Genes p53/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Animais , Apoptose/genética , Clonagem Molecular , Proteínas de Ligação a DNA/fisiologia , Humanos , Fatores de Transcrição NFI , Proteínas Nucleares , Regiões Promotoras Genéticas , Ratos , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Proteína 1 de Ligação a Y-Box
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA