Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Adv Nutr ; 13(5): 1450-1461, 2022 10 02.
Article in English | MEDLINE | ID: mdl-35776947

ABSTRACT

Humans often show variable responses to dietary, prebiotic, and probiotic interventions. Emerging evidence indicates that the gut microbiota is a key determinant for this population heterogeneity. Here, we provide an overview of some of the major computational and experimental tools being applied to critical questions of microbiota-mediated personalized nutrition and health. First, we discuss the latest advances in in silico modeling of the microbiota-nutrition-health axis, including the application of statistical, mechanistic, and hybrid artificial intelligence models. Second, we address high-throughput in vitro techniques for assessing interindividual heterogeneity, from ex vivo batch culturing of stool and continuous culturing in anaerobic bioreactors, to more sophisticated organ-on-a-chip models that integrate both host and microbial compartments. Third, we explore in vivo approaches for better understanding of personalized, microbiota-mediated responses to diet, prebiotics, and probiotics, from nonhuman animal models and human observational studies, to human feeding trials and crossover interventions. We highlight examples of existing, consumer-facing precision nutrition platforms that are currently leveraging the gut microbiota. Furthermore, we discuss how the integration of a broader set of the tools and techniques described in this piece can generate the data necessary to support a greater diversity of precision nutrition strategies. Finally, we present a vision of a precision nutrition and healthcare future, which leverages the gut microbiota to design effective, individual-specific interventions.


Subject(s)
Gastrointestinal Microbiome , Probiotics , Animals , Artificial Intelligence , Diet , Humans , Prebiotics
2.
Cell Mol Life Sci ; 79(2): 80, 2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35044528

ABSTRACT

The gut and brain link via various metabolic and signalling pathways, each with the potential to influence mental, brain and cognitive health. Over the past decade, the involvement of the gut microbiota in gut-brain communication has become the focus of increased scientific interest, establishing the microbiota-gut-brain axis as a field of research. There is a growing number of association studies exploring the gut microbiota's possible role in memory, learning, anxiety, stress, neurodevelopmental and neurodegenerative disorders. Consequently, attention is now turning to how the microbiota can become the target of nutritional and therapeutic strategies for improved brain health and well-being. However, while such strategies that target the gut microbiota to influence brain health and function are currently under development with varying levels of success, still very little is yet known about the triggers and mechanisms underlying the gut microbiota's apparent influence on cognitive or brain function and most evidence comes from pre-clinical studies rather than well controlled clinical trials/investigations. Filling the knowledge gaps requires establishing a standardised methodology for human studies, including strong guidance for specific focus areas of the microbiota-gut-brain axis, the need for more extensive biological sample analyses, and identification of relevant biomarkers. Other urgent requirements are new advanced models for in vitro and in vivo studies of relevant mechanisms, and a greater focus on omics technologies with supporting bioinformatics resources (training, tools) to efficiently translate study findings, as well as the identification of relevant targets in study populations. The key to building a validated evidence base rely on increasing knowledge sharing and multi-disciplinary collaborations, along with continued public-private funding support. This will allow microbiota-gut-brain axis research to move to its next phase so we can identify realistic opportunities to modulate the microbiota for better brain health.


Subject(s)
Brain-Gut Axis , Brain/physiology , Gastrointestinal Microbiome , Animals , Brain/physiopathology , Cognition , Humans , Metabolic Networks and Pathways , Signal Transduction
3.
Food Chem Toxicol ; 145: 111709, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32866515

ABSTRACT

Food allergy affects up to 6% of Europeans. Allergen identification is important for the risk assessment and management of the inadvertent presence of allergens in foods. The VITAL® initiative for voluntary incidental trace allergen labeling suggests protein reference doses, based on clinical reactivity in food challenge studies, at or below which voluntary labelling is unnecessary. Here, we investigated if current analytical methodology could verify the published VITAL® 2.0 doses, that were available during this analysis, in serving sizes between 5 and 500 g. Available data on published and commercial ELISA, PCR and mass spectrometry methods, especially for the detection of peanuts, soy, hazelnut, wheat, cow's milk and hen's egg were reviewed in detail. Limit of detection, quantitative capability, matrix compatibility, and specificity were assessed. Implications by the recently published VITAL® 3.0 doses were also considered. We conclude that available analytical methods are capable of reasonably robust detection of peanut, soy, hazelnut and wheat allergens for levels at or below the VITAL® 2.0 and also 3.0 doses, with some methods even capable of achieving this in a large 500 g serving size. Cow's milk and hen's egg are more problematic, largely due to matrix/processing incompatibility. An unmet need remains for harmonized reporting units, available reference materials, and method ring-trials to enable validation and the provision of comparable measurement results.


Subject(s)
Allergens/analysis , Food Analysis/standards , Food Hypersensitivity/immunology , Food/adverse effects , Animals , Enzyme-Linked Immunosorbent Assay , Europe , Food Analysis/methods , Humans , Mass Spectrometry
4.
Cell Metab ; 28(4): 557-572.e6, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30017352

ABSTRACT

The gut microbiota is essential for the normal function of the gut immune system, and microbiota alterations are associated with autoimmune disorders. However, how the gut microbiota prevents autoimmunity in distant organs remains poorly defined. Here we reveal that gut microbiota conditioned innate lymphoid cells (ILCs) induce the expression of mouse ß-defensin 14 (mBD14) by pancreatic endocrine cells, preventing autoimmune diabetes in the non-obese diabetic (NOD) mice. MBD14 stimulates, via Toll-like receptor 2, interleukin-4 (IL-4)-secreting B cells that induce regulatory macrophages, which in turn induce protective regulatory T cells. The gut microbiota-derived molecules, aryl hydrocarbon receptor (AHR) ligands and butyrate, promote IL-22 secretion by pancreatic ILCs, which induce expression of mBD14 by endocrine cells. Dysbiotic microbiota and low-affinity AHR allele explain the defective pancreatic expression of mBD14 observed in NOD mice. Our study reveals a yet unidentified crosstalk between ILCs and endocrine cells in the pancreas that is essential for the prevention of autoimmune diabetes development.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Gastrointestinal Microbiome/immunology , Insulin-Secreting Cells/metabolism , Lymphocytes/metabolism , Pancreatic Polypeptide-Secreting Cells/metabolism , beta-Defensins/metabolism , Animals , B-Lymphocytes, Regulatory/metabolism , Female , Humans , Immunity, Innate , Interleukins/metabolism , Islets of Langerhans/metabolism , Kaplan-Meier Estimate , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Statistics, Nonparametric , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptor 2/metabolism , Interleukin-22
5.
J Mol Biol ; 430(18 Pt B): 3041-3050, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30036494

ABSTRACT

Diabetes is a metabolic disorder affecting more than 400 million individuals and their families worldwide. The major forms of diabetes (types 1 and 2) are characterized by pancreatic ß-cell dysfunction and, in some cases, loss of ß-cell mass causing hyperglycemia due to absolute or relative insulin deficiency. The BCL-2 homology 3 (BH3)-only protein BIM has a wide role in apoptosis induction in cells. In this review, we describe the apoptotic mechanisms mediated by BIM activation in ß cells in obesity and both forms of diabetes. We focus on molecular pathways triggered by inflammation, saturated fats, and high levels of glucose. Besides its role in cell death, BIM has been implicated in the regulation of mitochondrial oxidative phosphorylation and cellular metabolism in hepatocytes. BIM is both a key mediator of pancreatic ß-cell death and hepatic insulin resistance and is thus a potential therapeutic target for novel anti-diabetogenic drugs. We consider the implications and challenges of targeting BIM in the treatment of the disease.


Subject(s)
Bcl-2-Like Protein 11/metabolism , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Obesity/etiology , Obesity/metabolism , Peptide Fragments , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins , Animals , Apoptosis , Autoimmunity , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/chemistry , Diabetes Mellitus/diagnosis , Diabetes Mellitus/drug therapy , Hepatocytes/metabolism , Humans , Insulin/metabolism , Insulin Resistance , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mitochondria/metabolism , Molecular Targeted Therapy , Obesity/drug therapy , Peptide Fragments/chemistry , Proto-Oncogene Proteins/chemistry , Signal Transduction
6.
Pesqui. vet. bras ; 37(11): 1187-1192, Nov. 2017. tab
Article in English | LILACS, VETINDEX | ID: biblio-895366

ABSTRACT

Haemophilus parasuis is the etiological agent of Glässer's disease (GD), an ubiquitous infection of swine characterized by systemic fibrinous polyserositis, polyarthritis and meningitis. Intensive use of antimicrobial agents in swine husbandries during the last years triggered the development of antibiotic resistances in bacterial pathogens. Thus, regular susceptibility testing is crucial to ensure efficacy of different antimicrobial agents to this porcine pathogen. In this study, 50 clinical isolates from South Brazilian pig herds were characterized and analyzed for their susceptibility to commonly used antibiotic. The identification and typing of clinical isolates was carried out by a modified indirect hemagglutination assay combined with a multiplex PCR. The susceptibility of each isolate was analyzed by broth microdilution method against a panel of 21 antimicrobial compounds. We found that field isolates are highly resistance to gentamycin, bacitracin, lincomycin and tiamulin, but sensitive to ampicillin, clindamycin, neomycin, penicillin, danofloxacin and enrofloxacin. Furthermore, an individual susceptibility analysis indicated that enrofloxacin is effective to treat clinical isolates with the exception of those classified as serovar 1. The results presented here firstly demonstrate the susceptibility of Brazilian clinical isolates of H. parasuis to antimicrobials widely used by swine veterinary practitioners and strengthen the need to perform susceptibility test prior to antibiotic therapy during GD outbreaks. In addition, because only six antimicrobial drugs (28.6%) were found effective against field isolates, a continuous surveillance of the susceptibility profile should be of major concern to the swine industry.(AU)


Haemophilus parasuis é o agente etiológico da doença de Glässer (GD), um processo infeccioso que acomete suínos e que se caracteriza por poliserosites fibrinosas sistêmicas, poliartrites e meningites. O uso intensivo de agentes antimicrobianos na produção de suínos, durante os últimos anos, tem disparado a seleção de cepas bacterianas resistentes a antibióticos. Desta maneira, a avaliação rotineira de susceptibilidade torna-se crucial para assegurar a correta seleção de um antimicrobiano eficaz contra este patógeno. Neste estudo, analisou-se a susceptibilidade antimicrobiana de 50 isolados clínicos de H. parasuis procedentes de granjas localizadas na região sul do Brasil. A identificação e tipificação dos isolados clínicos foi realizada através de uma PCR multiplex combinada com o teste de hemaglutinação indireta modificada. A susceptibilidade de cada isolado foi analisada pelo método de microdiluição em caldo utilizando-se um painel composto por 21 agentes antimicrobianos. Os resultados deste estudo indicam que as cepas clínicas de H. parasuis apresentam alta resistência à gentamicina, bacitracina, lincomicina e tiamulina, no entanto, são susceptíveis a ampicilina, clindamicina, neomicina, penicilina, enrofloxacina e danofloxacina. A análise de susceptibilidade realizada dentro de cada grupo de cepas de um mesmo sorovar indicou que a enrofloxacina é o antibiótico mais efetivo para tratar todos isolados clínicos com exceção daqueles pertencentes ao sorovar 1. Em termos gerais, neste trabalho, demonstra-se o perfil de susceptibilidade de isolados clínicos de H. parasuis aos antimicrobianos comumente utilizados pelos médicos veterinários especialistas em suínos, e reforça-se a necessidade da realização de testes de susceptibilidade antes do início da terapia com antibióticos durante surtos de DG. Além disso, como somente seis antimicrobianos (28.6%) foram efetivos contra os isolados clínicos, uma vigilância contínua do perfil de susceptibilidade aos antimicrobianos deve ser de grande preocupação para a indústria de suínos.(AU)


Subject(s)
Animals , Swine Diseases/drug therapy , Drug Resistance, Microbial , Drug Resistance, Bacterial/drug effects , Haemophilus parasuis/drug effects , Haemophilus Infections/veterinary , Sus scrofa
7.
Mol Cell Endocrinol ; 448: 108-121, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28390953

ABSTRACT

Pannexins (Panx's) are membrane proteins involved in a variety of biological processes, including cell death signaling and immune functions. The role and functions of Panx's in pancreatic ß-cells remain to be clarified. Here, we show Panx1 and Panx2 expression in isolated islets, primary ß-cells, and ß-cell lines. The expression of Panx2, but not Panx1, was downregulated by interleukin-1ß (IL-1ß) plus interferon-γ (IFNγ), two pro-inflammatory cytokines suggested to contribute to ß-cell demise in type 1 diabetes (T1D). siRNA-mediated knockdown (KD) of Panx2 aggravated cytokine-induced apoptosis in rat INS-1E cells and primary rat ß-cells, suggesting anti-apoptotic properties of Panx2. An anti-apoptotic function of Panx2 was confirmed in isolated islets from Panx2-/- mice and in human EndoC-ßH1 cells. Panx2 KD was associated with increased cytokine-induced activation of STAT3 and higher expression of inducible nitric oxide synthase (iNOS). Glucose-stimulated insulin release was impaired in Panx2-/- islets, and Panx2-/- mice subjected to multiple low-dose Streptozotocin (MLDS) treatment, a model of T1D, developed more severe diabetes compared to wild type mice. These data suggest that Panx2 is an important regulator of the insulin secretory capacity and apoptosis in pancreatic ß-cells.


Subject(s)
Apoptosis/drug effects , Connexins/deficiency , Cytokines/pharmacology , Glucose Intolerance/metabolism , Insulin-Secreting Cells/metabolism , Animals , Connexins/metabolism , Gene Knockdown Techniques , Glucose Intolerance/pathology , Humans , Hyperglycemia/pathology , Inflammation/pathology , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Rats , STAT3 Transcription Factor/metabolism , Streptozocin
8.
FEMS Microbiol Lett ; 363(5): fnw021, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26832642

ABSTRACT

Hepatitis E, caused by hepatitis E virus (HEV), is a viral infectious pathology of great importance in the public health. Hepatitis E outbreaks were registered in developing countries with poor or no sanitation, where drinking water was contaminated with fecal material, but also in many industrialized countries probably due to consumption of HEV-positive swine meat. In this study, we present the development and characterization of a recombinant antigen from ORF2 HEV genotype 3. Viral RNA was extracted from swine feces infected with the native virus. A total of 267 residues from the C-terminal ORF2((394-661)) coding sequence were cloned into the pET20a vector and expressed in Escherichia coli ER2566. Recombinant protein was purified by liquid chromatography and the fragment obtained a 98% homology against other human or swine HEV genotype 3 ORF2 sequences. Wistar rats were inoculated with ORF2p, developing antibodies able to recognize both the homologous antigen and the native HEV genotype 3 ORF2 present in infected stool. In parallel, HEV-negative swine were experimentally challenged with HEV genotype 3. ORF2 was detected by PCR 14 days post-inoculation in three-fourth piglets' feces and one week later by dot blot. In conclusion, this study proved the immunogenic and antigenic properties of the recombinant protein ORF2p.


Subject(s)
Antigens, Viral/immunology , Feces/virology , Hepatitis E virus/immunology , Hepatitis E/diagnosis , Open Reading Frames/genetics , Amino Acid Sequence , Animals , Base Sequence , Brazil , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Hepatitis E virus/classification , Hepatitis E virus/genetics , Humans , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Viral/genetics , RNA, Viral/isolation & purification , Rats , Rats, Wistar , Sequence Alignment , Sequence Analysis, RNA , Swine , Swine Diseases
9.
Proc Natl Acad Sci U S A ; 111(28): 10305-10, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24982147

ABSTRACT

Over 40 susceptibility loci have been identified for type 1 diabetes (T1D). Little is known about how these variants modify disease risk and progression. Here, we combined in vitro and in vivo experiments with clinical studies to determine how genetic variation of the candidate gene cathepsin H (CTSH) affects disease mechanisms and progression in T1D. The T allele of rs3825932 was associated with lower CTSH expression in human lymphoblastoid cell lines and pancreatic tissue. Proinflammatory cytokines decreased the expression of CTSH in human islets and primary rat ß-cells, and overexpression of CTSH protected insulin-secreting cells against cytokine-induced apoptosis. Mechanistic studies indicated that CTSH exerts its antiapoptotic effects through decreased JNK and p38 signaling and reduced expression of the proapoptotic factors Bim, DP5, and c-Myc. CTSH overexpression also up-regulated Ins2 expression and increased insulin secretion. Additionally, islets from Ctsh(-/-) mice contained less insulin than islets from WT mice. Importantly, the TT genotype was associated with higher daily insulin dose and faster disease progression in newly diagnosed T1D patients, indicating agreement between the experimental and clinical data. In line with these observations, healthy human subjects carrying the T allele have lower ß-cell function, which was evaluated by glucose tolerance testing. The data provide strong evidence that CTSH is an important regulator of ß-cell function during progression of T1D and reinforce the concept that candidate genes for T1D may affect disease progression by modulating survival and function of pancreatic ß-cells, the target cells of the autoimmune assault.


Subject(s)
Cathepsin H/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Insulin-Secreting Cells/metabolism , Adolescent , Alleles , Animals , Apoptosis/genetics , Cathepsin H/genetics , Cell Line , Child , Child, Preschool , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Gene Expression Regulation/genetics , Genotype , Humans , Insulin-Secreting Cells/pathology , Mice , Mice, Knockout , Rats
10.
Genomics ; 103(4): 264-75, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24462878

ABSTRACT

Type 1 Diabetes (T1D) is an autoimmune disease where local release of cytokines such as IL-1ß and IFN-γ contributes to ß-cell apoptosis. To identify relevant genes regulating this process we performed a meta-analysis of 8 datasets of ß-cell gene expression after exposure to IL-1ß and IFN-γ. Two of these datasets are novel and contain time-series expressions in human islet cells and rat INS-1E cells. Genes were ranked according to their differential expression within and after 24 h from exposure, and characterized by function and prior knowledge in the literature. A regulatory network was then inferred from the human time expression datasets, using a time-series extension of a network inference method. The two most differentially expressed genes previously unknown in T1D literature (RIPK2 and ELF3) were found to modulate cytokine-induced apoptosis. The inferred regulatory network is thus supported by the experimental validation, providing a proof-of-concept for the proposed statistical inference approach.


Subject(s)
Cytokines/metabolism , Gene Expression Profiling/methods , Gene Regulatory Networks , Insulin-Secreting Cells/physiology , Animals , Cytokines/pharmacology , DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 1 , Humans , Insulin-Secreting Cells/drug effects , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Islets of Langerhans/physiology , Proto-Oncogene Proteins c-ets/genetics , Rats , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Reproducibility of Results , Transcription Factors/genetics
11.
Immunobiology ; 217(12): 1292-300, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22944250

ABSTRACT

The vitamin D receptor (VDR) is a hormone nuclear receptor regulating bone and calcium homeostasis. Studies revealing the expression of VDR on immune cells point toward a role for VDR-dependent signaling pathways in immunity. Here we verified the ability of the natural VDR ligand, 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) to interfere in inflammatory and T cell stimulatory capacity of macrophages, in particular within a chronic inflammatory disease features of experimental type 1 diabetes (T1D). We demonstrated that VDR is constitutively expressed in macrophages and both the levels of VDR and its downstream targets, are clearly induced by 1,25(OH)(2)D(3). In control mice, macrophage programming with 1,25(OH)(2)D(3) partially abrogated the activation-provoked expression of IL-12p40, TNFα and iNOS as well as the effector T cell-recruiting chemokines, CXCL9, CXCL10 and CXCL11. Targeting VDR signaling in macrophages counteracted their T-cell stimulatory ability despite essentially unaltered expression of antigen-presenting and costimulatory molecules. Furthermore, even in non-obese diabetic (NOD) mice, where macrophages/monocytes featured a heightened responsiveness toward danger signals and a superior T cell stimulatory capacity, 1,25(OH)(2)D(3) successfully curtailed these basic macrophage-mediated functions. Interestingly, the inhibitory action of the active compound was associated with an IL-10-dependent mechanism since 1,25(OH)(2)D(3)-treatment of IL-10-deficient macrophages failed to reproduce the characteristic repression on inflammatory mediators or T cell proliferation. Combined, these results highlight the possible therapeutic applicability of this natural immunomodulator, due to its ability to counteract macrophage inflammatory and T cell-activating pathways.


Subject(s)
Calcitriol/immunology , Inflammation/immunology , Interleukin-10/immunology , Macrophages/immunology , T-Lymphocytes/immunology , Animals , Calcitriol/metabolism , Cells, Cultured , Chemokines, CXC/immunology , Chemokines, CXC/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Female , Inflammation/metabolism , Interleukin-10/metabolism , Interleukin-12 Subunit p40/immunology , Interleukin-12 Subunit p40/metabolism , Lymphocyte Activation/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type II/metabolism , Receptors, Calcitriol/immunology , Receptors, Calcitriol/metabolism , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
12.
Endocrinology ; 153(7): 3017-28, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22529213

ABSTRACT

The prevalence of obesity and type 1 diabetes in children is increasing worldwide. Insulin resistance and augmented circulating free fatty acids associated with obesity may cause pancreatic ß-cell endoplasmic reticulum (ER) stress. We tested the hypothesis that mild ER stress predisposes ß-cells to an exacerbated inflammatory response when exposed to IL-1ß or TNF-α, cytokines that contribute to the pathogenesis of type 1 diabetes. INS-1E cells or primary rat ß-cells were exposed to a low dose of the ER stressor cyclopiazonic acid (CPA) or free fatty acids, followed by low-dose IL-1ß or TNF-α. ER stress signaling was inhibited by small interfering RNA. Cells were evaluated for proinflammatory gene expression by RT-PCR and ELISA, gene reporter activity, p65 activation by immunofluorescence, and apoptosis. CPA pretreatment enhanced IL-1ß- induced, but not TNF-α-induced, expression of chemokine (C-C motif) ligand 2, chemokine (C-X-C motif) ligand 1, inducible nitric oxide synthase, and Fas via augmented nuclear factor κB (NF-κB) activation. X-box binding protein 1 (XBP1) and inositol-requiring enzyme 1, but not CCAAT/enhancer binding protein homologous protein, knockdown prevented the CPA-induced exacerbation of NF-κB-dependent genes and decreased IL-1ß-induced NF-κB promoter activity. XBP1 modulated NF-κB activity via forkhead box O1 inhibition. In conclusion, rat ß-cells facing mild ER stress are sensitized to IL-1ß, generating a more intense and protracted inflammatory response through inositol-requiring enzyme 1/XBP1 activation. These observations link ß-cell ER stress to the triggering of exacerbated local inflammation.


Subject(s)
DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoribonucleases/metabolism , Insulin-Secreting Cells/cytology , Interleukin-1beta/metabolism , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Animals , Apoptosis , Genes, Reporter , Inflammation , Insulinoma/metabolism , Male , Microscopy, Fluorescence/methods , Models, Biological , NF-kappa B/metabolism , RNA Interference , Rats , Rats, Wistar , Regulatory Factor X Transcription Factors , X-Box Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...