Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 14(12): 838, 2023 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-38110339

RESUMO

Acute infection and chronic infection are the two most common fates of pathogenic virus infections. While several factors that contribute to these fates are described, the critical control points and the mechanisms that underlie infection fate regulation are incompletely understood. Using the acute and chronic lymphocytic choriomeningitis virus (LCMV) infection model of mice, we find that the early dynamic pattern of the IFN-I response is a differentiating trait between both infection fates. Acute-infected mice generate a 2-wave IFN-I response while chronic-infected mice generate only a 1-wave response. The underlying cause is a temporal difference in CD8 T cell-mediated killing of splenic marginal zone CD169+ macrophages. It occurs later in acute infection and thus enables CD169+ marginal zone macrophages to produce the 2nd IFN-I wave. This is required for subsequent immune events including induction of inflammatory macrophages, generation of effector CD8+ T cells and virus clearance. Importantly, these benefits come at a cost for the host in the form of spleen fibrosis. Due to an earlier marginal zone destruction, these ordered immune events are deregulated in chronic infection. Our findings demonstrate the critical importance of kinetically well-coordinated sequential immune events for acute infection control and highlights that it may come at a cost for the host organism.


Assuntos
Coriomeningite Linfocítica , Camundongos , Animais , Coriomeningite Linfocítica/patologia , Vírus da Coriomeningite Linfocítica/fisiologia , Infecção Persistente , Camundongos Endogâmicos C57BL , Linfócitos T CD8-Positivos , Macrófagos/patologia
2.
Cell Rep ; 42(2): 112123, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36795562

RESUMO

The contribution of cross-presenting XCR1+ dendritic cells (DCs) and SIRPα+ DCs in maintaining T cell function during exhaustion and immunotherapeutic interventions of chronic infections remains poorly characterized. Using the mouse model of chronic LCMV infection, we found that XCR1+ DCs are more resistant to infection and highly activated compared with SIRPα+ DCs. Exploiting XCR1+ DCs via Flt3L-mediated expansion or XCR1-targeted vaccination notably reinvigorates CD8+ T cells and improves virus control. Upon PD-L1 blockade, XCR1+ DCs are not required for the proliferative burst of progenitor exhausted CD8+ T (TPEX) cells but are indispensable to sustain the functionality of exhausted CD8+ T (TEX) cells. Combining anti-PD-L1 therapy with increased frequency of XCR1+ DCs improves functionality of TPEX and TEX subsets, while increase of SIRPα+ DCs dampened their proliferation. Together, this demonstrates that XCR1+ DCs are crucial for the success of checkpoint inhibitor-based therapies through differential activation of exhausted CD8+ T cell subsets.


Assuntos
Apresentação Cruzada , Viroses , Camundongos , Animais , Células Dendríticas , Linfócitos T CD8-Positivos , Imunoterapia
3.
Genome Res ; 29(6): 907-919, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31138618

RESUMO

The processes and mechanisms of virus infection fate decisions that are the result of a dynamic virus-immune system interaction with either an efficient effector response and virus elimination or an alleviated immune response and chronic infection are poorly understood. Here, we characterized the host response to acute and chronic lymphocytic choriomeningitis virus (LCMV) infections by gene coexpression network analysis of time-resolved splenic transcriptomes. First, we found an early attenuation of inflammatory monocyte/macrophage prior to the onset of T cell exhaustion, and second, a critical role of the XCL1-XCR1 communication axis during the functional adaptation of the T cell response to the chronic infection state. These findings not only reveal an important feedback mechanism that couples T cell exhaustion with the maintenance of a lower level of effector T cell response but also suggest therapy options to better control virus levels during the chronic infection phase.


Assuntos
Interações Hospedeiro-Patógeno , Modelos Biológicos , Biologia de Sistemas , Viroses/virologia , Fenômenos Fisiológicos Virais , Doença Aguda , Animais , Biomarcadores , Doença Crônica , Biologia Computacional/métodos , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Celular , Imunidade Humoral , Mediadores da Inflamação/metabolismo , Camundongos , Baço/imunologia , Baço/metabolismo , Biologia de Sistemas/métodos
4.
Front Immunol ; 10: 1002, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31130969

RESUMO

The host immune response against infection requires the coordinated action of many diverse cell subsets that dynamically adapt to a pathogen threat. Due to the complexity of such a response, most immunological studies have focused on a few genes, proteins, or cell types. With the development of "omic"-technologies and computational analysis methods, attempts to analyze and understand complex system dynamics are now feasible. However, the decomposition of transcriptomic data sets generated from complete organs remains a major challenge. Here, we combined Weighted Gene Coexpression Network Analysis (WGCNA) and Digital Cell Quantifier (DCQ) to analyze time-resolved mouse splenic transcriptomes in acute and chronic Lymphocytic Choriomeningitis Virus (LCMV) infections. This enabled us to generate hypotheses about complex immune functioning after a virus-induced perturbation. This strategy was validated by successfully predicting several known immune phenomena, such as effector cytotoxic T lymphocyte (CTL) expansion and exhaustion. Furthermore, we predicted and subsequently verified experimentally macrophage-CD8 T cell cooperativity and the participation of virus-specific CD8+ T cells with an early effector transcriptome profile in the host adaptation to chronic infection. Thus, the linking of gene expression changes with immune cell kinetics provides novel insights into the complex immune processes within infected tissues.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/imunologia , Macrófagos/imunologia , Transcriptoma , Doença Aguda , Animais , Doença Crônica , Citocinas/imunologia , Redes Reguladoras de Genes , Masculino , Camundongos Endogâmicos C57BL
5.
PLoS One ; 8(9): e73365, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24039925

RESUMO

T helper (Th) cells play a major role in the immune response and pathology at the gastric mucosa during Helicobacter pylori infection. There is a limited mechanistic understanding regarding the contributions of CD4+ T cell subsets to gastritis development during H. pylori colonization. We used two computational approaches: ordinary differential equation (ODE)-based and agent-based modeling (ABM) to study the mechanisms underlying cellular immune responses to H. pylori and how CD4+ T cell subsets influenced initiation, progression and outcome of disease. To calibrate the model, in vivo experimentation was performed by infecting C57BL/6 mice intragastrically with H. pylori and assaying immune cell subsets in the stomach and gastric lymph nodes (GLN) on days 0, 7, 14, 30 and 60 post-infection. Our computational model reproduced the dynamics of effector and regulatory pathways in the gastric lamina propria (LP) in silico. Simulation results show the induction of a Th17 response and a dominant Th1 response, together with a regulatory response characterized by high levels of mucosal Treg) cells. We also investigated the potential role of peroxisome proliferator-activated receptor γ (PPARγ) activation on the modulation of host responses to H. pylori by using loss-of-function approaches. Specifically, in silico results showed a predominance of Th1 and Th17 cells in the stomach of the cell-specific PPARγ knockout system when compared to the wild-type simulation. Spatio-temporal, object-oriented ABM approaches suggested similar dynamics in induction of host responses showing analogous T cell distributions to ODE modeling and facilitated tracking lesion formation. In addition, sensitivity analysis predicted a crucial contribution of Th1 and Th17 effector responses as mediators of histopathological changes in the gastric mucosa during chronic stages of infection, which were experimentally validated in mice. These integrated immunoinformatics approaches characterized the induction of mucosal effector and regulatory pathways controlled by PPARγ during H. pylori infection affecting disease outcomes.


Assuntos
Simulação por Computador , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Imunidade nas Mucosas , Modelos Imunológicos , Estômago/microbiologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Mucosa Gástrica/imunologia , Mucosa Gástrica/microbiologia , Helicobacter pylori/fisiologia , Interações Hospedeiro-Patógeno , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , PPAR gama/imunologia , Estômago/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/microbiologia , Células Th17/imunologia , Células Th17/microbiologia
6.
Infect Immun ; 81(10): 3803-13, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23897614

RESUMO

Helicobacter pylori infection is the leading cause for peptic ulcer disease and gastric adenocarcinoma. Mucosal T cell responses play an important role in mediating H. pylori-related gastric immunopathology. While induced regulatory T (iTreg) cells are required for chronic colonization without disease, T helper 1 (Th1) effector responses are associated with lower bacterial loads at the expense of gastric pathology. Pigs were inoculated with either H. pylori strain SS1 or J99. Phenotypic and functional changes in peripheral blood mononuclear cell (PBMC) populations were monitored weekly, and mucosal immune responses and bacterial loads were assessed up to 2 months postinfection. Both H. pylori strains elicited a Th1 response characterized by increased percentages of CD4(+)Tbet(+) cells and elevated gamma interferon (IFN-γ) mRNA in PBMCs. A subset of CD8(+) T cells expressing Tbet and CD16 increased following infection. Moreover, a significant increase in perforin and granzyme mRNA expression was observed in PBMCs of infected pigs, indicating a predominant cytotoxic immune response. Infiltration of B cells, myeloid cells, T cells expressing Treg- and Th17-associated transcription factors, and cytotoxic T cells was found in the gastric lamina propria of both infected groups. Interestingly, based on bacterial reisolation data, strain SS1 showed greater capacity to colonize and/or persist in the gastric mucosa than did strain J99. This novel pig model of infection closely mimics human gastric pathology and presents a suitable avenue for studying effector and regulatory responses toward H. pylori described in humans.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Infecções por Helicobacter/veterinária , Helicobacter pylori/fisiologia , Doenças dos Suínos/microbiologia , Células Th1/fisiologia , Animais , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Leucócitos Mononucleares , Linfonodos , Baço/metabolismo , Gastropatias/microbiologia , Gastropatias/veterinária , Suínos , Regulação para Cima
7.
PLoS Comput Biol ; 9(4): e1003027, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23592971

RESUMO

Differentiation of CD4+ T cells into effector or regulatory phenotypes is tightly controlled by the cytokine milieu, complex intracellular signaling networks and numerous transcriptional regulators. We combined experimental approaches and computational modeling to investigate the mechanisms controlling differentiation and plasticity of CD4+ T cells in the gut of mice. Our computational model encompasses the major intracellular pathways involved in CD4+ T cell differentiation into T helper 1 (Th1), Th2, Th17 and induced regulatory T cells (iTreg). Our modeling efforts predicted a critical role for peroxisome proliferator-activated receptor gamma (PPARγ) in modulating plasticity between Th17 and iTreg cells. PPARγ regulates differentiation, activation and cytokine production, thereby controlling the induction of effector and regulatory responses, and is a promising therapeutic target for dysregulated immune responses and inflammation. Our modeling efforts predict that following PPARγ activation, Th17 cells undergo phenotype switch and become iTreg cells. This prediction was validated by results of adoptive transfer studies showing an increase of colonic iTreg and a decrease of Th17 cells in the gut mucosa of mice with colitis following pharmacological activation of PPARγ. Deletion of PPARγ in CD4+ T cells impaired mucosal iTreg and enhanced colitogenic Th17 responses in mice with CD4+ T cell-induced colitis. Thus, for the first time we provide novel molecular evidence in vivo demonstrating that PPARγ in addition to regulating CD4+ T cell differentiation also plays a major role controlling Th17 and iTreg plasticity in the gut mucosa.


Assuntos
Linfócitos T CD4-Positivos/citologia , Biologia Computacional/métodos , Citocinas/metabolismo , Animais , Diferenciação Celular , Simulação por Computador , Relação Dose-Resposta a Droga , Citometria de Fluxo , Imunofenotipagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Modelos Moleculares , Modelos Teóricos , PPAR gama/metabolismo , Fenótipo , Transdução de Sinais , Células Th17/metabolismo
8.
PLoS One ; 8(2): e57812, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23469071

RESUMO

BACKGROUND: Enteroaggregative Escherichia coli (EAEC) is recognized as an emerging cause of persistent diarrhea and enteric disease worldwide. Mucosal immunity towards EAEC infections is incompletely understood due in part to the lack of appropriate animal models. This study presents a new mouse model and investigates the role of peroxisome proliferator-activated receptor gamma (PPARγ) in the modulation of host responses to EAEC in nourished and malnourished mice. METHODS/PRINCIPAL FINDINGS: Wild-type and T cell-specific PPARγ null C57BL/6 mice were fed protein-deficient diets at weaning and challenged with 5×10(9)cfu EAEC strain JM221 to measure colonic gene expression and immune responses to EAEC. Antigen-specific responses to E. coli antigens were measured in nourished and malnourished mice following infection and demonstrated the immunosuppressive effects of malnutrition at the cellular level. At the molecular level, both pharmacological blockade and deletion of PPARγ in T cells resulted in upregulation of TGF-ß, IL-6, IL-17 and anti-microbial peptides, enhanced Th17 responses, fewer colonic lesions, faster clearance of EAEC, and improved recovery. The beneficial effects of PPARγ blockade on weight loss and EAEC clearance were abrogated by neutralizing IL-17 in vivo. CONCLUSIONS: Our studies provide in vivo evidence supporting the beneficial role of mucosal innate and effector T cell responses on EAEC burden and suggest pharmacological blockade of PPARγ as a novel therapeutic intervention for EAEC infection.


Assuntos
Infecções por Escherichia coli/imunologia , Escherichia coli/fisiologia , PPAR gama/metabolismo , Anilidas/farmacologia , Animais , Antígenos de Bactérias/imunologia , Carga Bacteriana/genética , Carga Bacteriana/imunologia , Peso Corporal/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Escherichia coli/imunologia , Infecções por Escherichia coli/complicações , Infecções por Escherichia coli/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Técnicas de Inativação de Genes , Interleucina-17/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Desnutrição/complicações , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/antagonistas & inibidores , PPAR gama/deficiência , PPAR gama/genética , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/microbiologia , Fatores de Tempo
9.
PLoS One ; 7(11): e50069, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23166823

RESUMO

BACKGROUND: There is an inverse secular trend between the incidence of obesity and gastric colonization with Helicobacter pylori, a bacterium that can affect the secretion of gastric hormones that relate to energy homeostasis. H. pylori strains that carry the cag pathogenicity island (PAI) interact more intimately with gastric epithelial cells and trigger more extensive host responses than cag(-) strains. We hypothesized that gastric colonization with H. pylori strains differing in cag PAI status exert distinct effects on metabolic and inflammatory phenotypes. METHODOLOGY/PRINCIPAL FINDINGS: To test this hypothesis, we examined metabolic and inflammatory markers in db/db mice and mice with diet-induced obesity experimentally infected with isogenic forms of H. pylori strain 26695: the cag PAI wild-type and its cag PAI mutant strain 99-305. H. pylori colonization decreased fasting blood glucose levels, increased levels of leptin, improved glucose tolerance, and suppressed weight gain. A response found in both wild-type and mutant H. pylori strain-infected mice included decreased white adipose tissue macrophages (ATM) and increased adipose tissue regulatory T cells (Treg) cells. Gene expression analyses demonstrated upregulation of gastric PPAR γ-responsive genes (i.e., CD36 and FABP4) in H. pylori-infected mice. The loss of PPAR γ in immune and epithelial cells in mice impaired the ability of H. pylori to favorably modulate glucose homeostasis and ATM infiltration during high fat feeding. CONCLUSIONS/SIGNIFICANCE: Gastric infection with some commensal strains of H. pylori ameliorates glucose homeostasis in mice through a PPAR γ-dependent mechanism and modulates macrophage and Treg cell infiltration into the abdominal white adipose tissue.


Assuntos
Mucosa Gástrica/microbiologia , Ilhas Genômicas/genética , Infecções por Helicobacter/metabolismo , Helicobacter pylori/crescimento & desenvolvimento , Homeostase/fisiologia , Obesidade/microbiologia , PPAR gama/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/imunologia , Animais , Glicemia , Peso Corporal , Antígenos CD36/metabolismo , Ensaio de Imunoadsorção Enzimática , Proteínas de Ligação a Ácido Graxo/metabolismo , Citometria de Fluxo , Mucosa Gástrica/imunologia , Mucosa Gástrica/metabolismo , Perfilação da Expressão Gênica , Grelina/sangue , Infecções por Helicobacter/imunologia , Helicobacter pylori/genética , Insulina/sangue , Leptina/sangue , Macrófagos/imunologia , Camundongos , Linfócitos T Reguladores/imunologia
10.
PLoS One ; 7(10): e47525, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23071818

RESUMO

Clostridium difficile is an anaerobic bacterium that has re-emerged as a facultative pathogen and can cause nosocomial diarrhea, colitis or even death. Peroxisome proliferator-activated receptor (PPAR) γ has been implicated in the prevention of inflammation in autoimmune and infectious diseases; however, its role in the immunoregulatory mechanisms modulating host responses to C. difficile and its toxins remains largely unknown. To characterize the role of PPARγ in C. difficile-associated disease (CDAD), immunity and gut pathology, we used a mouse model of C. difficile infection in wild-type and T cell-specific PPARγ null mice. The loss of PPARγ in T cells increased disease activity and colonic inflammatory lesions following C. difficile infection. Colonic expression of IL-17 was upregulated and IL-10 downregulated in colons of T cell-specific PPARγ null mice. Also, both the loss of PPARγ in T cells and C. difficile infection favored Th17 responses in spleen and colonic lamina propria of mice with CDAD. MicroRNA (miRNA)-sequencing analysis and RT-PCR validation indicated that miR-146b was significantly overexpressed and nuclear receptor co-activator 4 (NCOA4) suppressed in colons of C. difficile-infected mice. We next developed a computational model that predicts the upregulation of miR-146b, downregulation of the PPARγ co-activator NCOA4, and PPARγ, leading to upregulation of IL-17. Oral treatment of C. difficile-infected mice with the PPARγ agonist pioglitazone ameliorated colitis and suppressed pro-inflammatory gene expression. In conclusion, our data indicates that miRNA-146b and PPARγ activation may be implicated in the regulation of Th17 responses and colitis in C. difficile-infected mice.


Assuntos
Clostridioides difficile/imunologia , Colo/patologia , Enterocolite Pseudomembranosa/imunologia , Regulação da Expressão Gênica/imunologia , MicroRNAs/imunologia , Modelos Imunológicos , PPAR gama/imunologia , Análise de Variância , Animais , Sequência de Bases , Colo/imunologia , Primers do DNA/genética , Enterocolite Pseudomembranosa/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Técnicas Histológicas , Imunofenotipagem , Interleucina-10/imunologia , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Dados de Sequência Molecular , PPAR gama/agonistas , Pioglitazona , Reação em Cadeia da Polimerase em Tempo Real , Células Th17/imunologia , Tiazolidinedionas/farmacologia
11.
PLoS One ; 7(4): e34676, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22511958

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) increases the risk of colorectal cancer. Probiotic bacteria produce immunoregulatory metabolites in vitro such as conjugated linoleic acid (CLA), a polyunsaturated fatty acid with potent anti-carcinogenic effects. This study aimed to investigate the cellular and molecular mechanisms underlying the efficacy of probiotic bacteria in mouse models of cancer. METHODOLOGY/PRINCIPAL FINDINGS: The immune modulatory mechanisms of VSL#3 probiotic bacteria and CLA were investigated in mouse models of inflammation-driven colorectal cancer. Colonic specimens were collected for histopathology, gene expression and flow cytometry analyses. Immune cell subsets in the mesenteric lymph nodes (MLN), spleen and colonic lamina propria lymphocytes (LPL) were phenotypically and functionally characterized. Mice treated with CLA or VSL#3 recovered faster from the acute inflammatory phase of disease and had lower disease severity in the chronic, tumor-bearing phase of disease. Adenoma and adenocarcinoma formation was also diminished by both treatments. VSL#3 increased the mRNA expression of TNF-α, angiostatin and PPAR γ whereas CLA decreased COX-2 levels. Moreover, VSL#3-treated mice had increased IL-17 expression in MLN CD4+ T cells and accumulation of Treg LPL and memory CD4+ T cells. CONCLUSIONS/SIGNIFICANCE: Both CLA and VSL#3 suppressed colon carcinogenesis, although VSL#3 showed greater anti-carcinogenic and anti-inflammatory activities than CLA. Mechanistically, CLA modulated expression of COX-2 levels in the colonic mucosa, whereas VSL#3 targeted regulatory mucosal CD4+ T cell responses.


Assuntos
Anticarcinógenos/uso terapêutico , Colite/complicações , Neoplasias Colorretais/prevenção & controle , Ácidos Linoleicos Conjugados/metabolismo , Probióticos/uso terapêutico , Animais , Anticarcinógenos/farmacologia , Linfócitos T CD4-Positivos/metabolismo , Colite/microbiologia , Colite/patologia , Neoplasias Colorretais/complicações , Citocinas/metabolismo , Imunidade nas Mucosas/efeitos dos fármacos , Ácidos Linoleicos Conjugados/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Probióticos/farmacologia
12.
PLoS One ; 7(4): e34643, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22509338

RESUMO

BACKGROUND: Lanthionine synthetase component C-like protein 2 (LANCL2) is a member of the eukaryotic lanthionine synthetase component C-Like protein family involved in signal transduction and insulin sensitization. Recently, LANCL2 is a target for the binding and signaling of abscisic acid (ABA), a plant hormone with anti-diabetic and anti-inflammatory effects. METHODOLOGY/PRINCIPAL FINDINGS: The goal of this study was to determine the role of LANCL2 as a potential therapeutic target for developing novel drugs and nutraceuticals against inflammatory diseases. Previously, we performed homology modeling to construct a three-dimensional structure of LANCL2 using the crystal structure of lanthionine synthetase component C-like protein 1 (LANCL1) as a template. Using this model, structure-based virtual screening was performed using compounds from NCI (National Cancer Institute) Diversity Set II, ChemBridge, ZINC natural products, and FDA-approved drugs databases. Several potential ligands were identified using molecular docking. In order to validate the anti-inflammatory efficacy of the top ranked compound (NSC61610) in the NCI Diversity Set II, a series of in vitro and pre-clinical efficacy studies were performed using a mouse model of dextran sodium sulfate (DSS)-induced colitis. Our findings showed that the lead compound, NSC61610, activated peroxisome proliferator-activated receptor gamma in a LANCL2- and adenylate cyclase/cAMP dependent manner in vitro and ameliorated experimental colitis by down-modulating colonic inflammatory gene expression and favoring regulatory T cell responses. CONCLUSIONS/SIGNIFICANCE: LANCL2 is a novel therapeutic target for inflammatory diseases. High-throughput, structure-based virtual screening is an effective computational-based drug design method for discovering anti-inflammatory LANCL2-based drug candidates.


Assuntos
Anti-Inflamatórios/farmacologia , Simulação por Computador , Receptores de Superfície Celular/metabolismo , Adenilil Ciclases/metabolismo , Animais , Anti-Inflamatórios/uso terapêutico , Linhagem Celular , Colo/efeitos dos fármacos , Colo/metabolismo , AMP Cíclico/metabolismo , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , PPAR gama/metabolismo , Fenótipo , Proteínas de Ligação a Fosfato , Conformação Proteica , Receptores de Superfície Celular/química , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Homologia de Sequência de Aminoácidos , Transdução de Sinais/efeitos dos fármacos , Interface Usuário-Computador
13.
PLoS One ; 7(2): e31238, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22363592

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) therapies are modestly successful and associated with significant side effects. Thus, the investigation of novel approaches to prevent colitis is important. Probiotic bacteria can produce immunoregulatory metabolites in vitro such as conjugated linoleic acid (CLA), a polyunsaturated fatty acid with potent anti-inflammatory effects. This study aimed to investigate the cellular and molecular mechanisms underlying the anti-inflammatory efficacy of probiotic bacteria using a mouse model of colitis. METHODOLOGY/PRINCIPAL FINDINGS: The immune modulatory mechanisms of VSL#3 probiotic bacteria and CLA were investigated in a mouse model of DSS colitis. Colonic specimens were collected for histopathology, gene expression and flow cytometry analyses. Immune cell subsets in the mesenteric lymph nodes (MLN), spleen, blood and colonic lamina propria cells were phenotypically and functionally characterized. Fecal samples and colonic contents were collected to determine the effect of VSL#3 and CLA on gut microbial diversity and CLA production. CLA and VSL#3 treatment ameliorated colitis and decreased colonic bacterial diversity, a finding that correlated with decreased gut pathology. Colonic CLA concentrations were increased in response to probiotic bacterial treatment, but without systemic distribution in blood. VSL#3 and CLA decreased macrophage accumulation in the MLN of mice with DSS colitis. The loss of PPAR γ in myeloid cells abrogated the protective effect of probiotic bacteria and CLA in mice with DSS colitis. CONCLUSIONS/SIGNIFICANCE: Probiotic bacteria modulate gut microbial diversity and favor local production of CLA in the colon that targets myeloid cell PPAR γ to suppress colitis.


Assuntos
Bactérias/metabolismo , Colite/tratamento farmacológico , Trato Gastrointestinal/metabolismo , Ácidos Linoleicos Conjugados/metabolismo , Macrófagos/metabolismo , PPAR gama/metabolismo , Probióticos/uso terapêutico , Animais , Bactérias/efeitos dos fármacos , Biodiversidade , Colite/sangue , Colite/genética , Colite/patologia , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Linfonodos/efeitos dos fármacos , Linfonodos/patologia , Macrófagos/efeitos dos fármacos , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Probióticos/farmacologia , Substâncias Protetoras/farmacologia , Substâncias Protetoras/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...