Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mediators Inflamm ; 2023: 2899271, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36926280

RESUMO

Toll-like receptors (TLRs) are the most studied receptors among the pattern recognition receptors (PRRs). They act as microbial sensors, playing major roles in the regulation of the innate immune system. TLRs mediate their cellular functions through the activation of MyD88-dependent or MyD88-independent signaling pathways. Myd88, or myeloid differentiation primary response 88, is a cytosolic adaptor protein essential for the induction of proinflammatory cytokines by all TLRs except TLR3. While the crucial role of Myd88 is well described, the contribution of other adaptors in mediating TLR signaling and function has been underestimated. In this review, we highlight important results demonstrating that TIRAP and TRAM adaptors are also required for full signaling activity and responses induced by most TLRs.


Assuntos
Fator 88 de Diferenciação Mieloide , Receptor 4 Toll-Like , Receptor 3 Toll-Like , Receptores Toll-Like , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal
2.
Cell Commun Signal ; 19(1): 10, 2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33494775

RESUMO

Over the past 2 decades, pattern recognition receptors (PRRs) have been shown to be on the front line of many illnesses such as autoimmune, inflammatory, and neurodegenerative diseases as well as allergies and cancer. Among PRRs, toll-like receptors (TLRs) are the most studied family. Dissecting TLRs signaling turned out to be advantageous to elaborate efficient treatments to cure autoimmune and chronic inflammatory disorders. However, a broad understanding of TLR effectors is required to propose a better range of cures. In addition to kinases and E3 ubiquitin ligases, phosphatases emerge as important regulators of TLRs signaling mediated by NF-κB, type I interferons (IFN I) and Mitogen-Activated Protein Kinases signaling pathways. Here, we review recent knowledge on TLRs signaling modulation by different classes and subclasses of phosphatases. Thus, it becomes more and more evident that phosphatases could represent novel therapeutic targets to control pathogenic TLRs signaling. Video Abstract.


Assuntos
Monoéster Fosfórico Hidrolases/metabolismo , Receptores Toll-Like/metabolismo , Animais , COVID-19/metabolismo , Humanos , SARS-CoV-2 , Transdução de Sinais
3.
J Nutr ; 150(10): 2673-2686, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-32886125

RESUMO

BACKGROUND: Recent meta-analyses suggest that the consumption of fermented dairy products reduces type 2 diabetes and cardiovascular disease (CVD) risk, although the underlying mechanisms remain unclear. OBJECTIVE: We evaluated whether dairy protein products modulated gut microbiota and cardiometabolic features in mouse models of diet-induced obesity and CVD. METHODS: Eight-week-old C57BL/6J wild-type (WT) and LDLr-/-ApoB100/100 (LRKO) male mice were fed for 12 and 24 wk, respectively, with a high-fat/high-sucrose diet [66% kcal lipids, 22% kcal carbohydrates (100% sucrose), 12% kcal proteins]. The protein sources of the 4 diets were 100% nondairy protein (NDP), or 50% of the NDP energy replaced by milk (MP), milk fermented by Lactobacillus helveticus (FMP), or Greek-style yogurt (YP) protein. Fecal 16S rRNA gene-based amplicon sequencing, intestinal gene expression, and glucose tolerance test were conducted. Hepatic inflammation and circulating adhesion molecules were measured by multiplex assays. RESULTS: Feeding WT mice for 12 wk led to a 74% increase in body weight, whereas after 24 wk the LRKO mice had a 101.5% increase compared with initial body weight. Compared with NDP and MP, the consumption of FMP and YP modulated the gut microbiota composition in a similar clustering pattern, upregulating the Streptococcus genus in both genotypes. In WT mice, feeding YP compared with NDP increased the expression of genes involved in jejunal (Reg3b, 7.3-fold, P = 0.049) and ileal (Ocln, 1.7-fold, P = 0.047; Il1-ß,1.7-fold, P = 0.038; Nos2, 3.8-fold, P = 0.018) immunity and integrity. In LRKO mice, feeding YP compared with MP improved insulin sensitivity by 65% (P = 0.039). In LRKO mice, feeding with FMP versus NDP attenuated hepatic inflammation (monocyte chemoattractant protein 1, 2.1-fold, P ˂ 0.0001; IL1-ß, 5.7-fold, P = 0.0003; INF-γ, 1.7-fold, P = 0.002) whereas both FMP [vascular adhesion molecule 1 (VCAM1), 1.3-fold, P = 0.0003] and YP (VCAM1, 1.04-fold, P = 0.013; intracellular adhesion molecule 1, 1.4-fold, P = 0.028) decreased circulating adhesion molecules. CONCLUSION: Both fermented dairy protein products reduce cardiometabolic risk factors in diet-induced obese mice, possibly by modulating the gut microbiota.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Produtos Fermentados do Leite/análise , Microbioma Gastrointestinal/efeitos dos fármacos , Doenças Metabólicas/prevenção & controle , Proteínas do Leite/farmacologia , Obesidade/induzido quimicamente , Animais , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Bactérias/classificação , Bactérias/efeitos dos fármacos , Biomarcadores/sangue , Dieta , Dieta Hiperlipídica , Sacarose Alimentar/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Leite/química , Proteínas do Leite/química , Receptores de LDL/genética , Receptores de LDL/metabolismo
4.
Proteomics ; 17(20)2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28643936

RESUMO

The development of 3D cell cultures into self-organizing organ-like structures named organoids provides a model that better reflects in vivo organ physiology and their functional properties. Organoids have been established from several organs, such as the intestine, prostate, brain, liver, kidney and pancreas. With recent advances in high-throughput and -omics profiling technologies, it is now possible to study the mechanisms of cellular organisation at the systems level. It is therefore not surprising that these methods are now used to characterize organoids at the transcriptomic, proteomic, chromatin state and transcription factor DNA-binding levels. These approaches can therefore provide a wealth of information regarding both the mechanisms involved in different diseases, and those involved in cell responses to different conditions, in a more in vivo setting. The authors provide an overview of the potential applications of quantitative mass spectrometry with organoid culture, and how the use of large-scale proteome measurements is emerging in different organoid systems.


Assuntos
Técnicas de Cultura de Órgãos/métodos , Organoides/crescimento & desenvolvimento , Proteômica/métodos , Animais , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Sistema Digestório/citologia , Sistema Digestório/crescimento & desenvolvimento , Humanos , Rim/citologia , Rim/crescimento & desenvolvimento , Espectrometria de Massas , Organogênese , Organoides/citologia , Peptídeos/análise , Fenótipo , Proteoma/análise
5.
Biochim Biophys Acta ; 1849(12): 1411-22, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26477491

RESUMO

Intestinal epithelial cells are exposed to luminal bacterial threat and require adequate defense mechanisms to ensure host protection and epithelium regeneration against possible deleterious damage. Differentiated intestinal epithelial cells produce antimicrobial and regenerative components that protect against such challenges. Few intestinal specific transcription factors have been identified to control the switching from repression to activation of this class of gene. Herein, we show that gene transcription of some regenerating islet-derived (REG) family members is dependent on the transcription factor GATA-4. Silencing of GATA-4 expression in cultured intestinal epithelial cells identified Reg3ß as a target gene using an unbiased approach of gene expression profiling. Co-transfection and RNA interference assays identified complex GATA-4-interactive transcriptional components required for the activation or repression of Reg3ß gene activity. Conditional deletion of Gata4 in the mouse intestinal epithelium supported its regulatory role for Reg1, Reg3α, Reg3ß and Reg3γ genes. Reg1 dramatic down-modulation of expression in Gata4 conditional null mice was associated with a significant decrease in intestinal epithelial cell migration. Altogether, these results identify a novel and complex role for GATA-4 in the regulation of REG family members gene expression.


Assuntos
Células Epiteliais/metabolismo , Fator de Transcrição GATA4/fisiologia , Regulação da Expressão Gênica/genética , Mucosa Intestinal/citologia , Família Multigênica , Transcrição Gênica , Animais , Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Fator de Transcrição CDX2 , Diferenciação Celular/genética , Linhagem Celular , Técnicas de Cocultura , Fator de Transcrição GATA4/classificação , Fator de Transcrição GATA4/deficiência , Fator de Transcrição GATA4/genética , Genes Reporter , Proteínas de Homeodomínio/metabolismo , Lectinas Tipo C/metabolismo , Litostatina/metabolismo , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Proteínas Associadas a Pancreatite , Estrutura Terciária de Proteína , Proteínas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia
6.
J Cell Physiol ; 231(2): 436-48, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26174178

RESUMO

The intestinal epithelium responds to and transmits signals from the microbiota and the mucosal immune system to insure intestinal homeostasis. These interactions are in part conveyed by epigenetic modifications, which respond to environmental changes. Protein acetylation is an epigenetic signal regulated by histone deacetylases, including Hdac1 and Hdac2. We have previously shown that villin-Cre-inducible intestinal epithelial cell (IEC)-specific Hdac1 and Hdac2 deletions disturb intestinal homeostasis. To determine the role of Hdac1 and Hdac2 in the regulation of IEC function and the establishment of the dual knockout phenotype, we have generated villin-Cre murine models expressing one Hdac1 allele without Hdac2, or one Hdac2 allele without Hdac1. We have also investigated the effect of short-term deletion of both genes in naphtoflavone-inducible Ah-Cre and tamoxifen-inducible villin-Cre(ER) mice. Mice with one Hdac1 allele displayed normal tissue architecture, but increased sensitivity to DSS-induced colitis. In contrast, mice with one Hdac2 allele displayed intestinal architecture defects, increased proliferation, decreased goblet cell numbers as opposed to Paneth cells, increased immune cell infiltration associated with fibrosis, and increased sensitivity to DSS-induced colitis. In comparison to dual knockout mice, intermediary activation of Notch, mTOR, and Stat3 signaling pathways was observed. While villin-Cre(ER) Hdac1 and Hdac2 deletions led to an impaired epithelium and differentiation defects, Ah-Cre-mediated deletion resulted in blunted proliferation associated with the induction of a DNA damage response. Our results suggest that IEC determination and intestinal homeostasis are highly dependent on Hdac1 and Hdac2 activity levels, and that changes in the IEC acetylome may alter the mucosal environment.


Assuntos
Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Mucosa Intestinal/enzimologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células/genética , Proliferação de Células/fisiologia , Colite/enzimologia , Colite/genética , Colite/patologia , Dano ao DNA , Modelos Animais de Doenças , Células Epiteliais/enzimologia , Células Caliciformes/citologia , Células Caliciformes/enzimologia , Histona Desacetilase 1/deficiência , Histona Desacetilase 1/genética , Histona Desacetilase 2/deficiência , Histona Desacetilase 2/genética , Homeostase , Imunidade nas Mucosas , Mucosa Intestinal/anormalidades , Mucosa Intestinal/citologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Notch/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
7.
Am J Physiol Gastrointest Liver Physiol ; 306(7): G594-605, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24525021

RESUMO

Histone deacetylases (Hdac) remove acetyl groups from proteins, influencing global and specific gene expression. Hdacs control inflammation, as shown by Hdac inhibitor-dependent protection from dextran sulfate sodium (DSS)-induced murine colitis. Although tissue-specific Hdac knockouts show redundant and specific functions, little is known of their intestinal epithelial cell (IEC) role. We have shown previously that dual Hdac1/Hdac2 IEC-specific loss disrupts cell proliferation and determination, with decreased secretory cell numbers and altered barrier function. We thus investigated how compound Hdac1/Hdac2 or Hdac2 IEC-specific deficiency alters the inflammatory response. Floxed Hdac1 and Hdac2 and villin-Cre mice were interbred. Compound Hdac1/Hdac2 IEC-deficient mice showed chronic basal inflammation, with increased basal disease activity index (DAI) and deregulated Reg gene colonic expression. DSS-treated dual Hdac1/Hdac2 IEC-deficient mice displayed increased DAI, histological score, intestinal permeability, and inflammatory gene expression. In contrast to double knockouts, Hdac2 IEC-specific loss did not affect IEC determination and growth, nor result in chronic inflammation. However, Hdac2 disruption protected against DSS colitis, as shown by decreased DAI, intestinal permeability and caspase-3 cleavage. Hdac2 IEC-specific deficient mice displayed increased expression of IEC gene subsets, such as colonic antimicrobial Reg3b and Reg3g mRNAs, and decreased expression of immune cell function-related genes. Our data show that Hdac1 and Hdac2 are essential IEC homeostasis regulators. IEC-specific Hdac1 and Hdac2 may act as epigenetic sensors and transmitters of environmental cues and regulate IEC-mediated mucosal homeostatic and inflammatory responses. Different levels of IEC Hdac activity may lead to positive or negative outcomes on intestinal homeostasis during inflammation.


Assuntos
Colite/enzimologia , Colo/enzimologia , Células Epiteliais/enzimologia , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Mucosa Intestinal/enzimologia , Animais , Colite/genética , Colite/imunologia , Colite/patologia , Colo/imunologia , Colo/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Epigênese Genética , Células Epiteliais/imunologia , Células Epiteliais/patologia , Regulação da Expressão Gênica , Genótipo , Histona Desacetilase 1/deficiência , Histona Desacetilase 1/genética , Histona Desacetilase 2/deficiência , Histona Desacetilase 2/genética , Homeostase , Imunidade nas Mucosas , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , Permeabilidade , Fenótipo , Fatores de Tempo
8.
J Cell Biochem ; 114(5): 1203-15, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23192652

RESUMO

Polycomb-group proteins form multimeric protein complexes involved in transcriptional silencing. The Polycomb Repressive complex 2 (PRC2) contains the Suppressor of Zeste-12 protein (Suz12) and the histone methyltransferase Enhancer of Zeste protein-2 (Ezh2). This complex, catalyzing the di- and tri-methylation of histone H3 lysine 27, is essential for embryonic development and stem cell renewal. However, the role of Polycomb-group protein complexes in the control of the intestinal epithelial cell (IEC) phenotype is not known. We show that Suz12 and Ezh2 were differentially expressed along the intestinal crypt-villus axis. ShRNA-mediated Suz12 depletion in the IEC-6 rat crypt-derived cell line decreased Ezh2 expression and H3K27 di-trimethylation. Suz12-depleted cells achieved higher cell densities after confluence, with increased cyclin D2 and cyclin D3 protein levels, and increased STAT3 activation in post-confluent cells. Suz12 depletion specifically increased mostly developmental, cell adhesion and immune response gene expression, including neuronal and inflammatory genes. Suz12 depletion directly and indirectly de-regulated the IL-1ß-dependent inflammatory response, as demonstrated by decreased MAPK p38 activation as opposed to JNK activation, and altered basal and stimulated expression of inflammatory genes, including transcription factors such as C/EBPß. Of note, this positive effect on cell proliferation and inflammatory gene expression was revealed in the absence of the cyclin-dependent kinase inhibitor p16, a main target negatively regulated by PRC2. These results demonstrate that the PRC2 complex, in addition to keeping in check non-IEC differentiation pathways, insures the proper IEC response to cell density as well as to external growth and inflammatory signals, by controlling specific signaling pathways.


Assuntos
Células Epiteliais/metabolismo , Células Epiteliais/patologia , Histonas/metabolismo , Inflamação/patologia , Intestinos/patologia , Lisina/metabolismo , Animais , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Proteína Potenciadora do Homólogo 2 de Zeste , Células Epiteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/genética , Interleucina-1beta/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Metilação/efeitos dos fármacos , Camundongos , Microvilosidades/efeitos dos fármacos , Microvilosidades/metabolismo , Microvilosidades/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Complexo Repressor Polycomb 2/metabolismo , Ratos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , beta Catenina/metabolismo
9.
Methods Mol Biol ; 2603: 151-161, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36370277

RESUMO

Stable isotope labeling by amino acids in cell culture (SILAC) is a strategic quantitative mass spectrometry method to analyze multiple protein samples in different conditions simultaneously. In recent years, 3D cell growth culture conditions have been developed to establish intestinal organoids from isolated crypts, which mimic the intestine's cell composition and organization. Organoids, isolated from normal or diseased tissues, can be used to compare cell distribution and differentiation, signaling pathways, and cell responses to pharmacological agents, therapeutic drugs, endogenous or exogenous metabolites, and environmental stresses, among others. Here, we describe the process of generating SILAC organoids from the mouse small intestine.


Assuntos
Organoides , Proteômica , Camundongos , Animais , Proteômica/métodos , Marcação por Isótopo/métodos , Organoides/metabolismo , Fluxo de Trabalho , Aminoácidos/química , Intestinos
10.
Anat Rec (Hoboken) ; 306(5): 1111-1130, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-35899872

RESUMO

Inflammatory bowel diseases (IBDs) are characterized by abnormal, non-antigen specific chronic inflammation of unknown etiology. Genome-wide association studies show that many IBD genetic susceptibility loci map to immune function genes and compelling evidence indicate that environmental factors play a critical role in IBD pathogenesis. Clinical and experimental evidence implicate the pro-inflammatory cytokine IL-15 in the pathogenesis of IBD. IL-15 and IL-15α expression is increased in the inflamed mucosa of IBD patients. IL-15 contributes to the maintenance of different cell subsets in the intestinal mucosa. However, very few studies have addressed the role of IL-15 in pre-clinical models of colitis. In this study, we use three well-characterized models of experimental colitis to determine the contribution of IL-15 to pathological intestinal inflammation.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Humanos , Animais , Camundongos , Interleucina-15/genética , Interleucina-15/metabolismo , Modelos Animais de Doenças , Estudo de Associação Genômica Ampla , Colite/genética , Colite/metabolismo , Colite/patologia , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/metabolismo , Mucosa Intestinal , Inflamação/metabolismo
11.
Am J Physiol Gastrointest Liver Physiol ; 301(4): G719-30, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21737780

RESUMO

The Ras/Raf/MEK/ERK cascade regulates intestinal epithelial cell proliferation. Indeed, while barely detectable in differentiated cells of the villi, ERK1/2-activated forms are detected in the nucleus of undifferentiated human intestinal crypt cells. In addition, we and others have reported that ERKs are selectively inactivated during enterocyte differentiation. However, whether inactivation of the ERK pathway is necessary for inhibition of both proliferation and induction of differentiation of intestinal epithelial cells is unknown. Human Caco-2/15 cells, undifferentiated crypt IEC-6 cells, and differentiating Cdx3-expressing IEC-6 cells were infected with retroviruses encoding either a hemagglutinin (HA)-tagged MEK1 wild type (wtMEK) or a constitutively active S218D/S222D MEK1 mutant (caMEK). Protein and gene expression was assessed by Western blotting, semiquantitative RT-PCR, and real-time PCR. Morphology was analyzed by transmission electron microscopy. We found that 1) IEC-6/Cdx3 cells formed multicellular layers after confluence and differentiated after 30 days in culture, as assessed by increased polarization, microvilli formation, expression of differentiation markers, and ERK1/2 inhibition; 2) while activated MEK prevented neither the inhibition of ERK1/2 activities nor the differentiation process in postconfluent Caco-2/15 cells, caMEK expression prevented ERK inhibition in postconfluent IEC-6/Cdx3 cells, thus leading to maintenance of elevated ERK1/2 activities; 3) caMEK-expressing IEC-6/Cdx3 cells exhibited altered multicellular structure organization, poorly defined tight junctions, reduced number of microvilli on the apical surface, and decreased expression of the hepatocyte nuclear factor 1α transcription factor and differentiation markers, namely apolipoprotein A-4, fatty acid-binding protein, calbindin-3, mucin 2, alkaline phosphatase, and sucrase-isomaltase; and 4) increased Cdx3 phosphorylation on serine-60 (S60) in IEC-6/Cdx3 cells expressing caMEK led to decreased Cdx2 transactivation potential. These results indicate that inactivation of the ERK pathway is required to ensure the full Cdx2/3 transcriptional activity necessary for intestinal epithelial cell terminal differentiation.


Assuntos
Diferenciação Celular/fisiologia , Mucosa Intestinal/citologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Animais , Fator de Transcrição CDX2 , Células CACO-2 , Células Cultivadas , Células Epiteliais/metabolismo , Proteínas de Homeodomínio/fisiologia , Humanos , MAP Quinase Quinase 1/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Ratos , Transativadores/fisiologia
12.
Cells ; 10(2)2021 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-33498747

RESUMO

We have previously reported that histone deacetylase epigenetic regulator Hdac1 and Hdac2 deletion in intestinal epithelial cells (IEC) disrupts mucosal tissue architecture and barrier, causing chronic inflammation. In this study, proteome and transcriptome analysis revealed the importance of signaling pathways induced upon genetic IEC-Hdac1 and Hdac2 deletion. Indeed, Gene Ontology biological process analysis of enriched deficient IEC RNA and proteins identified common pathways, including lipid metabolic and oxidation-reduction process, cell adhesion, and antigen processing and presentation, related to immune responses, correlating with dysregulation of major histocompatibility complex (MHC) class II genes. Top upstream regulators included regulators associated with environmental sensing pathways to xenobiotics, microbial and diet-derived ligands, and endogenous metabolites. Proteome analysis revealed mTOR signaling IEC-specific defects. In addition to mTOR, the STAT and Notch pathways were dysregulated specifically in jejunal IEC. To determine the impact of pathway dysregulation on mutant jejunum alterations, we treated mutant mice with Tofacitinib, a JAK inhibitor. Treatment with the inhibitor partially corrected proliferation and tight junction defects, as well as niche stabilization by increasing Paneth cell numbers. Thus, IEC-specific histone deacetylases 1 (HDAC1) and 2 (HDAC2) support intestinal homeostasis by regulating survival and translation processes, as well as differentiation and metabolic pathways. HDAC1 and HDAC2 may play an important role in the regulation of IEC-specific inflammatory responses by controlling, directly or indirectly, the JAK/STAT pathway. IEC-specific JAK/STAT pathway deregulation may be, at least in part, responsible for intestinal homeostasis disruption in mutant mice.


Assuntos
Células Epiteliais/metabolismo , Histona Desacetilase 1/deficiência , Histona Desacetilase 2/deficiência , Homeostase , Intestinos/citologia , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Deleção de Genes , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/genética , Histona Desacetilase 2/metabolismo , Homeostase/efeitos dos fármacos , Contagem de Linfócitos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Organoides/efeitos dos fármacos , Organoides/crescimento & desenvolvimento , Celulas de Paneth/efeitos dos fármacos , Celulas de Paneth/metabolismo , Piperidinas/farmacologia , Pirimidinas/farmacologia , Linfócitos T/efeitos dos fármacos
13.
J Biol Chem ; 284(37): 25220-9, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19608741

RESUMO

Stem cells of the gut epithelium constantly produce precursors that progressively undergo a succession of molecular changes resulting in growth arrest and commitment to a specific differentiation program. Few transcriptional repressors have been identified that maintain the normal intestinal epithelial cell (IEC) proliferation state. Herein, we show that the nuclear receptor co-repressor (NCoR1) is differentially expressed during the proliferation-to-differentiation IEC transition. Silencing of NCoR1 expression in proliferating cells of crypt origin resulted in a rapid growth arrest without associated cell death. A genechip profiling analysis identified several candidate genes to be up-regulated in NCoR1-deficient IEC. Pigment epithelium-derived factor (PEDF, also known as serpinf1), a suspected tumor suppressor gene that plays a key role in the inhibition of epithelial tissue growth, was significantly up-regulated in these cells. Chromatin immunoprecipitation experiments showed that the PEDF gene promoter was occupied by NCoR1 in proliferating epithelial cells. Multiple retinoid X receptor (RXR) heterodimers interacting sites of the PEDF promoter were confirmed to interact with RXR and retinoid acid receptor (RAR). Cotransfection assays showed that RXR and RAR were able to transactivate the PEDF promoter and that NCoR1 was repressing this effect. Finally, forced expression of PEDF in IEC resulted in a slower rate of proliferation. These observations suggest that NCoR1 expression is required to maintain IEC in a proliferative state and identify PEDF as a novel transcriptional target for NCoR1 repressive action.


Assuntos
Células Epiteliais/metabolismo , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica , Intestinos/citologia , Fatores de Crescimento Neural/metabolismo , Proteínas Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Serpinas/metabolismo , Sequência de Aminoácidos , Animais , Células CACO-2 , Proliferação de Células , Humanos , Modelos Biológicos , Dados de Sequência Molecular , Correpressor 1 de Receptor Nuclear , Ratos , Frações Subcelulares/metabolismo
14.
Pediatr Obes ; 15(8): e12642, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32351036

RESUMO

BACKGROUND: Introducing complementary foods other than breastmilk or formula acutely changes the infant gut microbiota composition. However, it is unknown whether the timing of introduction to complementary foods (early vs. late) in infancy is associated with early childhood gut microbiota and BMI, and if these associations depend on breastfeeding duration. OBJECTIVE: Our primary objective was to investigate whether timing of infant complentary feeding with solid foods is associated with early childhood gut microbiota composition and BMI-z, and whether these associations differ by duration of breastfeeding. METHODS: We used data from a Canadian pre-birth cohort followed till age 5 years. We examined timing of introduction to solid foods with the gut microbiota, determined by 16S rRNA gene sequencing of stool collected at 5 years of age, and age-and-sex specific BMI-z. We conducted analyses before and after stratifying by breastfeeding duration, and adjusted for delivery mode, gestational age and birth weight. RESULTS: Of the 392 children in the analysis, 109 (27.8%) had early (≤4 months) solids. The association between early (vs later) solids and BMI-z at 5 years was modified by breastfeeding status at 4 months (P = .06). Among children breastfed >4 months, early (vs later) solids were associated with differential relative abundance of 6 bacterial taxa, including lower Roseburia, and 0.30 higher BMI-z (95% CI: 0.05, 0.55) at 5 years. In children breastfed <4 months, early solids were associated with differential relative abundance of 9 taxa, but not with child BMI-z. CONCLUSIONS: Early (vs. later) introduction to solid foods in infancy is associated with altered gut microbiota composition and BMI in early childhood, however these associations differ by duration of breastfeeding.


Assuntos
Índice de Massa Corporal , Aleitamento Materno , Microbioma Gastrointestinal , Fenômenos Fisiológicos da Nutrição do Lactente , Adulto , Pré-Escolar , Estudos de Coortes , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Fatores de Tempo
15.
Infect Immun ; 77(12): 5400-10, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19797069

RESUMO

It has been well established that Clostridium difficile toxin A (TcdA) induces cell death in human epithelial cells. However, the mechanism of TcdA-induced cell death remains to be fully characterized. Here, we show that TcdA induces dose-dependent cell death in ovarian carcinoma and colonic carcinoma cell lines. TcdA-mediated cell death, as well as caspase 8 and caspase 3 activation, were specifically abrogated by anti-toxin antibodies. Although caspase 8 and caspase 3 were activated by TcdA in OVCAR3 ovarian carcinoma and T84 colonic cancer cells, pancaspase and caspase 8, 3, and 9 inhibitors did not block TcdA-induced cell death. In contrast, tumor necrosis factor-related apoptosis-inducing ligand-induced cell death was nearly completely blocked by caspase inhibitors in OVCAR3 cells. In these cells, TcdA induces the mitochondrial pathway of apoptosis, as demonstrated by changes in mitochondrial outer membrane permeabilization (MOMP). Furthermore, overexpression of the antiapoptotic proteins Bcl-2 and Bcl-X(L) significantly inhibited TcdA-induced cell death, as well as TcdA-induced MOMP. Conversely, small interfering RNA-mediated inhibition of Bcl-X(L) in TcdA-resistant SKOV3ip1 cells enhanced TcdA-induced cell death. Overexpression of the antiapoptotic proteins Bcl-2 and Bcl-X(L) in T84 cells also inhibited TcdA-induced cell death. Altogether, our data demonstrate that TcdA induces cell death in both ovarian and colonic cancer cells preferentially via the mitochondrial pathway of apoptosis by a death receptor-independent and a caspase-independent mechanism. This process is regulated by antiapoptotic members of the Bcl-2 family.


Assuntos
Toxinas Bacterianas/toxicidade , Morte Celular , Clostridioides difficile/patogenicidade , Enterotoxinas/toxicidade , Células Epiteliais/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Proteína bcl-X/imunologia , Caspase 3/biossíntese , Caspase 8/biossíntese , Linhagem Celular Tumoral , Humanos , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/fisiologia , Permeabilidade/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Ligante Indutor de Apoptose Relacionado a TNF/imunologia
16.
Int J Cancer ; 125(7): 1575-86, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19462441

RESUMO

Constitutive activation of the MAP kinase kinase MEK1 induces oncogenic transformation in intestinal epithelial cells. Loss of cell-cell adhesion followed by the dissociation of epithelial structures is a prerequisite for increased cell motility and tumor invasion. This phenotypic switch is designated epithelial-to-mesenchymal transition (EMT). EMT also plays an important role in determining the dissemination of tumors. However, the role of MEK1 in intestinal EMT, tumor invasion and metastasis has not been elucidated. To determine the functions of activated MEK1 in intestinal tumorigenesis, we established intestinal epithelial cell lines that overexpress wild-type MEK1 (wtMEK) or activated MEK1 (caMEK). Our results indicate that expression of caMEK is sufficient to induce EMT as confirmed with the induction of N-cadherin, vimentin, Snail1 and Snail2, whereas a reduction in E-cadherin, occludin, ZO-1 and cortical F-actin was noted. The Snail1 and Snail2 promoter analyses revealed that Egr-1 and Fra-1, an AP-1 protein, are responsible for MEK1-induced Snail1 and Snail2 expression, respectively. Cells expressing activated MEK1 clearly acquired an invasive capacity when compared to wtMEK-expressing cells. Zymography studies confirmed elevated levels of MMP2 and MMP9 activities in media of caMEK-expressing cells. Importantly, cells expressing activated MEK1 induced tumors with short latency in correlation with their ability to induce experimental metastasis in vivo and to express factors known to promote colorectal cancer cell metastasis. In conclusion, our results demonstrate, for the first time, that constitutive activation of MEK1 in intestinal epithelial cells is sufficient to induce an EMT associated with tumor invasion and metastasis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Células Epiteliais/patologia , MAP Quinase Quinase 1/metabolismo , Mesoderma/patologia , Invasividade Neoplásica , Metástase Neoplásica , Animais , Imunoprecipitação da Cromatina , Neoplasias Colorretais/metabolismo , Células Epiteliais/enzimologia , Imunofluorescência , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Intestinos , MAP Quinase Quinase 1/genética , Sistema de Sinalização das MAP Quinases , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Mesoderma/enzimologia , Camundongos , Camundongos Nus , Mutação , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo
17.
Sci Rep ; 9(1): 5363, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30926862

RESUMO

Both HDAC1 and HDAC2 are class I deacetylases acting as erasers of lysine-acetyl marks on histones and non-histone proteins. Several histone deacetylase inhibitors, either endogenous to the cell, such as the ketogenic ß-hydroxybutyrate metabolite, or exogenous, such as butyrate, a microbial-derived metabolite, regulate HDAC activity. Different combinations of intestinal epithelial cell (IEC)-specific Hdac1 and/or Hdac2 deletion differentially alter mucosal homeostasis in mice. Thus, HDAC1 and HDAC2 could act as sensors and transmitters of environmental signals to the mucosa. In this study, enteroid culture models deleted for Hdac1 or Hdac2 were established to determine IEC-specific function as assessed by global transcriptomic and proteomic approaches. Results show that Hdac1 or Hdac2 deficiency altered differentiation of Paneth and goblet secretory cells, which sustain physical and chemical protection barriers, and increased intermediate secretory cell precursor numbers. Furthermore, IEC Hdac1- and Hdac2-dependent common and specific biological processes were identified, including oxidation-reduction, inflammatory responses, and lipid-related metabolic processes, as well as canonical pathways and upstream regulators related to environment-dependent signaling through steroid receptor pathways, among others. These findings uncover unrecognized regulatory similarities and differences between Hdac1 and Hdac2 in IEC, and demonstrate how HDAC1 and HDAC2 may complement each other to regulate the intrinsic IEC phenotype.


Assuntos
Enterócitos/metabolismo , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Animais , Camundongos
18.
J Cell Biochem ; 103(5): 1573-83, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17910034

RESUMO

The C/EBPdelta transcription factor is involved in the positive regulation of the intestinal epithelial cell acute phase response. C/EBPdelta regulation by histone deacetylases (HDACs) during the course of inflammation remains to be determined. Our aim was to examine the effect of HDACs on C/EBPdelta-dependent regulation of haptoglobin, an acute phase protein induced in intestinal epithelial cells in response to pro-inflammatory cytokines. HDAC1, HDAC3, and HDAC4 were expressed in intestinal epithelial cells, as determined by Western blot. GST pull-down assays showed specific HDAC1 interactions with the transcriptional activation and the b-ZIP C/EBPdelta domains, while the co-repressor mSin3A interacts with the C-terminal domain. Immunoprecipitation assays confirmed the interaction between HDAC1 and the N-terminal C/EBPdelta amino acid 36-164 domain. HDAC1 overexpression decreased C/EBPdelta transcriptional activity of the haptoglobin promoter, as assessed by transient transfection and luciferase assays. Chromatin immunoprecipitation analysis showed a displacement of HDAC1 from the haptoglobin promoter in response to inflammatory stimuli and an increased acetylation of histone H3 and H4. HDAC1 silencing by shRNA expression increased both basal and IL-1beta-induced haptoglobin mRNA levels in epithelial intestinal cells. Our results suggest that interactions between C/EBPs and HDAC1 negatively regulate C/EBPdelta-dependent haptoglobin expression in intestinal epithelial cells.


Assuntos
Reação de Fase Aguda/metabolismo , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Haptoglobinas/biossíntese , Histona Desacetilases/metabolismo , Mucosa Intestinal/metabolismo , Ativação Transcricional , Acetilação/efeitos dos fármacos , Animais , Proteína delta de Ligação ao Facilitador CCAAT/antagonistas & inibidores , Células CACO-2 , Inibidores de Histona Desacetilases , Histonas/metabolismo , Humanos , Interleucina-1beta/farmacologia , Regiões Promotoras Genéticas , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , RNA Interferente Pequeno , Ratos , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos
19.
Biochem Biophys Res Commun ; 370(2): 371-5, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18387357

RESUMO

C/EBP transcription factors are involved in the regulation of the intestinal epithelial cell response to inflammatory stimuli. GATA transcription factors modulate C/EBP-dependent transcriptional activation in various cell types. We thus determined whether GATA-4 whose expression is restricted to epithelial cells, modulate C/EBP transcriptional activity and C/EBP-dependent acute phase protein expression in intestinal epithelial cells. Interaction between C/EBPdelta and GATA-4 required both C/EBPdelta leucine zipper and basic DNA-binding domains, and the GATA-4 C-terminal region. C/EBP isoforms and GATA-4 synergistically activated the alpha-acid glycoprotein gene while GATA-4 repressed thiostatin and haptoglobin C/EBP-dependent transactivation. In GATA-4 expressing intestinal epithelial cells, GATA-4 led to decreased C/EBPbeta and C/EBPdelta protein levels, and decreased thiostatin expression in response to IL-1beta and dexamethasone. This correlated with decreased C/EBPdelta recruitment to the thiostatin promoter, as assessed by chromatin immunoprecipitation assays. In contrast, increased AGP expression in response to dexamethasone correlated with increased basal histone H4 acetylation. Thus, GATA-4 may be involved in the establishment of specific intestinal epithelial cell C/EBP-dependent and C/EBP-independent transcriptional responses.


Assuntos
Proteínas de Fase Aguda/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Fator de Transcrição GATA4/metabolismo , Ativação Transcricional , Animais , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Expressão Gênica , Genes Reporter , Humanos , Cininogênios/genética , Camundongos , Ratos
20.
Cells ; 7(12)2018 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-30544623

RESUMO

Dendritic cells (DCs) play a major role in innate and adaptive immunity and self-immune tolerance. Immunogenic versus tolerogenic DC functions are dictated by their levels of costimulatory molecules and their cytokine expression profile. The transcription factor C/EBPß regulates the expression of several inflammatory genes in many cell types including macrophages. However, little is known regarding the role of C/EBPß in tolerogenic versus immunogenic DCs functions. We have previously reported that bone marrow-derived DCs generated with GM-CSF (GM/DCs) acquire the signature of semi-mature tolerogenic IL-10-producing DCs as opposed to immunogenic DCs generated with GM-CSF and IL-4 (IL-4/DCs). Here, we show that tolerogenic GM/DCs exhibit higher levels of phosphorylation and enhanced DNA binding activity of C/EBPß and CREB than immunogenic IL-4/DCs. We also show that the p38 MAPK/CREB axis and GSK3 play an important role in regulating C/EBPß phosphorylation and DNA binding activity. Inhibition of p38 MAPK in GM/DCs resulted in a drastic decrease of C/EBPß and CREB DNA binding activities, a reduction of their IL-10 production and an increase of their IL-12p70 production, a characteristic of immunogenic IL-4/DCs. We also present evidence that GSK3 inhibition in GM/DCs reduced C/EBPß DNA binding activity and increased expression of costimulatory molecules in GM/DCs and their production of IL-10. Analysis of GM/DCs of C/EBPß-/- mice showed that C/EBPß was essential to maintain the semimature phenotype and the production of IL-10 as well as low CD4⁺ T cell proliferation. Our results highlight the importance of the p38MAPK-C/EBPß pathway in regulating phenotype and function of tolerogenic GM/DCs.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA