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










Base de dados
Intervalo de ano de publicação
1.
Immunohorizons ; 8(1): 89-96, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38226923

RESUMO

Klebsiella pneumoniae (KP) presents a global health threat, leading to significant morbidity and mortality due to its multidrug-resistant profile and the limited availability of therapeutic options. To eliminate KP lung infection, the host initiates a robust inflammatory response. One of the host's mechanisms for mitigating excessive inflammation involves the RNA-binding protein regnase-1 (Reg1, MCPIP1, or ZC3H12A). Reg1 has an RNA binding domain that recognizes stem-loop structures in the 3' untranslated region of various proinflammatory transcripts, leading to mRNA decay. However, excessive suppression of inflammation by Reg1 results in suboptimal KP control. Reg1 deficiency within the nonhematopoietic compartment confers resistance to KP in the lung. Given that lung epithelium is crucial for KP resistance, we hypothesized that selective deletion of Reg1 in lung epithelial cells might enhance proinflammatory signals, leading to a better control of KP. Our transcriptomic analysis of epithelial cells in KP-infected wild-type mice revealed the presence of three distinct alveolar type 2 cell (AT2) subpopulations (conventional, inflammatory, and cycling) and enrichment of Reg1 in inflammatory AT2 cells. We conditionally deleted Reg1 in lung AT2 cells (ΔReg1), which amplified the local inflammatory response in the lung and increased macrophage cell numbers compared with controls. However, when ΔReg1 mice were subjected to KP infection, there were no significant differences in bacterial burden or survival compared with controls. These findings suggest that the local inflammatory response enhanced by Reg1 deletion in AT2 cells is insufficient to control KP infection.


Assuntos
Células Epiteliais , Klebsiella pneumoniae , Ribonucleases , Animais , Camundongos , Regiões 3' não Traduzidas , Inflamação , Pulmão , Ribonucleases/genética
2.
iScience ; 25(9): 104900, 2022 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-36039299

RESUMO

Understanding lung immunity requires an unbiased profiling of tissue-resident T cells at their precise anatomical locations within the lung, but such information has not been characterized in the immunized mouse model. In this pilot study, using 10x Genomics Chromium and Visium platform, we performed an integrative analysis of spatial transcriptome with single-cell RNA-seq and single-cell ATAC-seq on lung cells from mice after immunization using a well-established Klebsiella pneumoniae infection model. We built an optimized deconvolution pipeline to accurately decipher specific cell-type compositions by anatomic location. We discovered that combining scATAC-seq and scRNA-seq data may provide more robust cell-type identification, especially for lineage-specific T helper cells. Combining all three modalities, we observed a dynamic change in the location of T helper cells as well as their corresponding chemokines. In summary, our proof-of-principle study demonstrated the power and potential of single-cell multi-omics analysis to uncover spatial- and cell-type-dependent mechanisms of lung immunity.

3.
Cell Host Microbe ; 30(4): 530-544.e6, 2022 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-35316647

RESUMO

Combating fungal pathogens poses metabolic challenges for neutrophils, key innate cells in anti-Candida albicans immunity, yet how host-pathogen interactions cause remodeling of the neutrophil metabolism is unclear. We show that neutrophils mediate renal immunity to disseminated candidiasis by upregulating glucose uptake via selective expression of glucose transporter 1 (Glut1). Mechanistically, dectin-1-mediated recognition of ß-glucan leads to activation of PKCδ, which triggers phosphorylation, localization, and early glucose transport by a pool of pre-formed Glut1 in neutrophils. These events are followed by increased Glut1 gene transcription, leading to more sustained Glut1 accumulation, which is also dependent on the ß-glucan/dectin-1/CARD9 axis. Card9-deficient neutrophils show diminished glucose incorporation in candidiasis. Neutrophil-specific Glut1-ablated mice exhibit increased mortality in candidiasis caused by compromised neutrophil phagocytosis, reactive oxygen species (ROS), and neutrophil extracellular trap (NET) formation. In human neutrophils, ß-glucan triggers metabolic remodeling and enhances candidacidal function. Our data show that the host-pathogen interface increases glycolytic activity in neutrophils by regulating Glut1 expression, localization, and function.


Assuntos
Candidíase , Transportador de Glucose Tipo 1 , Neutrófilos , beta-Glucanas , Animais , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Candida albicans , Candidíase/imunologia , Glucose/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Camundongos , Neutrófilos/imunologia , beta-Glucanas/metabolismo
4.
mBio ; 13(1): e0379221, 2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35100872

RESUMO

Excessive inflammation can cause tissue damage and autoimmunity, sometimes accompanied by severe morbidity or mortality. Numerous negative feedback mechanisms exist to prevent unchecked inflammation, but this restraint may come at the cost of suboptimal infection control. Regnase-1 (MCPIP1), a feedback regulator of IL-17 and LPS signaling, binds and degrades target mRNAs. Consequently, Reg1 deficiency exacerbates autoimmunity in multiple models. However, the role of Reg1 in bacterial immunity remains poorly defined. Here, we show that mice deficient in Reg1 are resistant to Klebsiella pneumoniae (KP). Reg1 deficiency did not accelerate bacterial eradication. Rather, Reg1-deficient alveolar macrophages had elevated Ifnb1 and enrichment of type I IFN genes. Blockade of IFNR during KP infection reversed disease improvement. Reg1 did not impact Ifnb1 stability directly, but Irf7 expression was affected. Thus, Reg1 suppresses type I IFN signaling restricting resistance to KP, suggesting that Reg1 could potentially be a target in severe bacterial infections. IMPORTANCE Klebsiella pneumoniae (KP) can cause life-threatening bacterial pneumonia and is the third most common cause of ventilator-associated pneumonia in the United States. Host inflammatory responses to infection are necessary to control disease, yet at the same time can cause collateral damage or immunopathology. During immune responses, many events are established within the infected tissue to limit unchecked inflammation. However, this restraint of immunity can impair infection control, and it is not fully understood how this balance is maintained during different infection settings. In this study we explored the possibility that a host-derived negative regulator of RNA, Regnase-1, limits immunity to KP by dampening inflammation. Indeed, mice with reduced Regnase-1 levels showed improved survival to KP infection, linked to regulation of type I interferons. Therefore, although restraint of Reg1 is beneficial to prevent immunopathology, temporary blockade of Reg1 could potentially be exploited to improve host defense during infectious settings such as KP.


Assuntos
Interferon Tipo I , Infecções por Klebsiella , Pneumonia Bacteriana , Animais , Camundongos , Klebsiella pneumoniae , Pneumonia Bacteriana/microbiologia , Endorribonucleases , Inflamação , Infecções por Klebsiella/microbiologia , Litostatina
5.
Sci Immunol ; 5(48)2020 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-32503875

RESUMO

Oropharyngeal candidiasis (OPC; thrush) is an opportunistic infection caused by the commensal fungus Candida albicans Interleukin-17 (IL-17) and IL-22 are cytokines produced by type 17 lymphocytes. Both cytokines mediate antifungal immunity yet activate quite distinct downstream signaling pathways. While much is now understood about how IL-17 promotes immunity in OPC, the activities of IL-22 are far less well delineated. We show that, despite having similar requirements for induction from type 17 cells, IL-22 and IL-17 function nonredundantly during OPC. We find that the IL-22 and IL-17 receptors are required in anatomically distinct locations within the oral mucosa; loss of IL-22RA1 or signal transducer and activator of transcription 3 (STAT3) in the oral basal epithelial layer (BEL) causes susceptibility to OPC, whereas IL-17RA is needed in the suprabasal epithelial layer (SEL). Transcriptional profiling of the tongue linked IL-22/STAT3 not only to oral epithelial cell proliferation and survival but also, unexpectedly, to driving an IL-17-specific gene signature. We show that IL-22 mediates regenerative signals on the BEL that replenish the IL-17RA-expressing SEL, thereby restoring the ability of the oral epithelium to respond to IL-17 and thus to mediate antifungal events. Consequently, IL-22 signaling in BEL "licenses" IL-17 signaling in the oral mucosa, revealing spatially distinct yet cooperative activities of IL-22 and IL-17 in oral candidiasis.


Assuntos
Candidíase Bucal/imunologia , Células Epiteliais/imunologia , Interleucina-17/imunologia , Interleucinas/imunologia , Mucosa Bucal/imunologia , Fator de Transcrição STAT3/imunologia , Animais , Candida albicans/imunologia , Feminino , Interleucina-17/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/imunologia , Interleucina 22
6.
Infect Immun ; 87(11)2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31451621

RESUMO

Streptococcus pneumoniae is the most common cause of community-acquired pneumonia worldwide, and interleukin-22 (IL-22) helps contain pneumococcal burden in lungs and extrapulmonary tissues. Administration of IL-22 increases hepatic complement 3 and complement deposition on bacteria and improves phagocytosis by neutrophils. The effects of IL-22 can be tempered by a secreted natural antagonist, known as IL-22 binding protein (IL-22BP), encoded by Il22ra2 To date, the degree to which IL-22BP controls IL-22 in pulmonary infection is not well defined. Here, we show that Il22ra2 inhibits IL-22 during S. pneumoniae lung infection and that Il22ra2 deficiency favors downregulation of oxidative phosphorylation (OXPHOS) genes in an IL-22-dependent manner. Il22ra2-/- mice are more resistant to S. pneumoniae infection, have increased IL-22 in lung tissues, and sustain longer survival upon infection than control mice. Transcriptome sequencing (RNA-seq) analysis of infected Il22ra2-/- mouse lungs revealed downregulation of genes involved in OXPHOS. Downregulation of this metabolic process is necessary for increased glycolysis, a crucial step for transitioning to a proinflammatory phenotype, in particular macrophages and dendritic cells (DCs). Accordingly, we saw that macrophages from Il22ra2-/- mice displayed reduced OXPHOS gene expression upon infection with S. pneumoniae, changes that were IL-22 dependent. Furthermore, we showed that macrophages express IL-22 receptor subunit alpha-1 (IL-22Ra1) during pneumococcal infection and that Il22ra2-/- macrophages rely more on the glycolytic pathway than wild-type (WT) controls. Together, these data indicate that IL-22BP deficiency enhances IL-22 signaling in the lung, thus contributing to resistance to pneumococcal pneumonia by downregulating OXPHOS genes and increasing glycolysis in macrophages.


Assuntos
Interleucinas/metabolismo , Pneumonia Pneumocócica/metabolismo , Receptores de Interleucina/metabolismo , Animais , Linhagem Celular , Suscetibilidade a Doenças , Células Epiteliais/fisiologia , Regulação da Expressão Gênica , Interleucinas/genética , Antígenos Comuns de Leucócito , Pulmão/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Oxirredução , Fosforilação , Pneumonia Pneumocócica/imunologia , Receptores de Interleucina/genética , Streptococcus pneumoniae , Interleucina 22
7.
JCI Insight ; 3(12)2018 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-29925696

RESUMO

Despite the discovery of key pattern recognition receptors and CD4+ T cell subsets in laboratory mice, there is ongoing discussion of the value of murine models to reflect human disease. Pneumocystis is an AIDS-defining illness, in which risk of infection is inversely correlated with peripheral CD4+ T cell counts. Due to medical advances in the control of HIV, the current epidemiology of Pneumocystis infection is predominantly due to primary human immunodeficiencies and immunosuppressive therapies. To this end, we found that every human genetic immunodeficiency associated with Pneumocystis infection that has been tested in mice recapitulated susceptibility. For example, humans with a loss-of-function IL21R mutation are severely immunocompromised. We found that IL-21R, in addition to CD4+ T cell intrinsic STAT3 signaling, were required for generating protective antifungal class-switched antibody responses, as well as effector T cell-mediated protection. Furthermore, CD4+ T cell intrinsic IL-21R/STAT3 signaling was required for CD4+ T cell effector responses, including IL-22 production. Recombinant IL-22 administration to Il21r-/- mice induced the expression of a fungicidal peptide, cathelicidin antimicrobial peptide, which showed in vitro fungicidal activity. In conclusion, SPF laboratory mice faithfully replicate many aspects of human primary immunodeficiency and provide useful tools to understand the generation and nature of effector CD4+ T cell immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Doenças do Sistema Imunitário/imunologia , Infecções por Pneumocystis/imunologia , Animais , Anti-Infecciosos/metabolismo , Antifúngicos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Subunidade alfa de Receptor de Interleucina-21/genética , Subunidade alfa de Receptor de Interleucina-21/metabolismo , Interleucinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumocystis/imunologia , Infecções por Pneumocystis/genética , Infecções por Pneumocystis/patologia , Fator de Transcrição STAT3 , Transdução de Sinais , Interleucina 22
8.
Cell Rep ; 18(13): 3078-3090, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28355561

RESUMO

Inducible bronchus-associated lymphoid tissue (iBALT) is an ectopic lymphoid structure composed of highly organized T cell and B cell zones that forms in the lung in response to infectious or inflammatory stimuli. Here, we develop a model for fungal-mediated iBALT formation, using infection with Pneumocystis that induces development of pulmonary lymphoid follicles. Pneumocystis-dependent iBALT structure formation and organization required CXCL13 signaling. Cxcl13 expression was regulated by interleukin (IL)-17 family members, as Il17ra-/-, Il17rb-/-, and Il17rc-/- mice failed to develop iBALT. Interestingly, Il17rb-/- mice have intact Th17 responses, but failed to generate an anti-Pneumocystis Th2 response. Given a role for Th2 and Th17 immunity in iBALT formation, we demonstrated that primary pulmonary fibroblasts synergistically upregulated Cxcl13 transcription following dual stimulation with IL-13 and IL-17A in a STAT3/GATA3-dependent manner. Together, these findings uncover a role for Th2/Th17 cells in regulating Cxcl13 expression and provide an experimental model for fungal-driven iBALT formation.


Assuntos
Brônquios/patologia , Imunidade , Tecido Linfoide/patologia , Infecções por Pneumocystis/imunologia , Pneumocystis/fisiologia , Células Th17/imunologia , Células Th2/imunologia , Animais , Quimiocina CXCL13/metabolismo , Fator de Transcrição GATA3/metabolismo , Interleucina-13/metabolismo , Interleucina-17/metabolismo , Linfotoxina-alfa/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções por Pneumocystis/microbiologia , Infecções por Pneumocystis/prevenção & controle , Receptores CXCR5/metabolismo , Receptores de Interleucina-17/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
9.
Cell Host Microbe ; 20(5): 596-605, 2016 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-27923703

RESUMO

The cytokine IL-17, and signaling via its heterodimeric IL-17RA/IL-17RC receptor, is critical for host defense against extracellular bacterial and fungal pathogens. Polarized lung epithelial cells express IL-17RA and IL-17RC basolaterally. However, their contribution to IL-17-dependent pulmonary defenses in vivo remains to be determined. To address this, we generated mice with conditional deletion of Il17ra or Il17rc in Scgb1a1-expressing club cells, a major component of the murine bronchiolar epithelium. These mice displayed an impaired ability to recruit neutrophils into the airway lumen in response to IL-17, a defect in bacterial clearance upon mucosal challenge with the pulmonary pathogen Klebsiella pneumoniae, and substantially reduced epithelial expression of the chemokine Cxcl5. Neutrophil recruitment and bacterial clearance were restored by intranasal administration of recombinant CXCL5. Our data show that IL-17R signaling in the lung epithelium plays a critical role in establishing chemokine gradients that are essential for mucosal immunity against pulmonary bacterial pathogens.


Assuntos
Quimiocinas/metabolismo , Infecções por Klebsiella/imunologia , Klebsiella pneumoniae/imunologia , Pulmão/imunologia , Receptores de Interleucina-17/metabolismo , Mucosa Respiratória/imunologia , Transdução de Sinais , Animais , Modelos Animais de Doenças , Pulmão/microbiologia , Camundongos , Camundongos Knockout , Neutrófilos/imunologia , Pneumonia Bacteriana/imunologia , Receptores de Interleucina-17/deficiência
10.
J Immunol ; 197(5): 1877-83, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27456484

RESUMO

IL-22-IL-22R signaling plays a crucial role in regulating host defenses against extracellular pathogens, particularly in the intestine, through the induction of antimicrobial peptides and chemotactic genes. However, the role of IL-22-IL-22R is understudied in Streptococcus pneumoniae lung infection, a prevalent pathogen of pneumonia. This paper presents the findings of IL-22 signaling during a murine model of pneumococcal pneumonia and improvement of bacterial burden upon IL-22 administration. IL-22 was rapidly induced in the lung during pneumococcal infection in wild-type mice, and Il22(-/-) mice had higher pneumococcal burdens compared with controls. Additionally, mice with hepatic-specific deletion of Il22ra1 also had higher bacterial burdens in lungs compared with littermate controls after intrapulmonary pneumococcal infection, suggesting that IL-22 signaling in the liver is important to control pneumococcal pneumonia. Thus, we hypothesized that enhancement of IL-22 signaling would control pneumococcal burden in lung tissues in an experimental pneumonia model. Administration of rIL-22 systemically to infected wild-type mice decreased bacterial burden in lung and liver at 24 h postinfection. Our in vitro studies also showed that mice treated with IL-22 had increased C3 expression in the liver compared with the isotype control group. Furthermore, serum from mice treated with IL-22 had improved opsonic capacity by increasing C3 binding on S. pneumoniae Taken together, endogenous IL-22 and hepatic IL-22R signaling play critical roles in controlling pneumococcal lung burden, and systemic IL-22 decreases bacterial burden in the lungs and peripheral organs by potentiating C3 opsonization on bacterial surfaces, through the increase of hepatic C3 expression.


Assuntos
Interleucinas/imunologia , Interleucinas/metabolismo , Pulmão/microbiologia , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/metabolismo , Receptores de Interleucina/metabolismo , Transdução de Sinais , Animais , Carga Bacteriana , Complemento C3/genética , Complemento C3/imunologia , Citocinas/biossíntese , Citocinas/imunologia , Interleucinas/administração & dosagem , Interleucinas/sangue , Fígado/imunologia , Fígado/microbiologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose , Pneumonia Pneumocócica/microbiologia , Receptores de Interleucina/imunologia , Streptococcus pneumoniae/imunologia , Streptococcus pneumoniae/patogenicidade , Interleucina 22
11.
Sci Rep ; 6: 30384, 2016 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-27461849

RESUMO

There remains a significant need for development of effective small molecules that can inhibit cytokine-mediated inflammation. Phosphoinositide 3 kinase (PI3K) is a direct upstream activator of AKT, and plays a critical role in multiple cell signaling pathways, cell cycle progression, and cell growth, and PI3K inhibitors have been approved or are in clinical development. We examined novel PI3Kdelta inhibitors, which are highly selective for the p110delta isoform of in CD3/CD28 stimulated T-cell cytokine production. In vitro generated CD4+ T effector cells stimulated in the presence of a PI3Kdelta inhibitor demonstrated a dose-dependent suppression of cytokines produced by Th1, Th2, and Th17 cells. This effect was T-cell intrinsic, and we observed similar effects on human PBMCs. Th17 cells expressing a constitutively activated form of AKT were resistant to PI3Kdelta inhibition, suggesting that the inhibitor is acting through AKT signaling pathways. Additionally, PI3Kdelta inhibition decreased IL-17 production in vivo and decreased neutrophil recruitment to the lung in a murine model of acute pulmonary inflammation. These experiments show that targeting PI3Kdelta activity can modulate T-cell cytokine production and reduce inflammation in vivo, suggesting that PI3Kdelta inhibition could have therapeutic potential in treating inflammatory diseases.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Interleucina-17/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Feminino , Interleucina-17/genética , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
12.
Alcohol Res ; 37(2): 177-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26695743

RESUMO

Physicians have recognized for more than a century that alcohol use is associated with infections and that alcoholics are especially at risk for pneumonia. Clear evidence now indicates that alcohol has a systemic effect on every organ. This review first presents a clinical case to describe a patient with immunity issues complicated by alcohol use-a setting familiar to many clinicians. This is followed by a description of the molecular mechanisms that explain the secondary immune deficiency produced by alcohol in the host, focusing mostly on the gut and lower respiratory mucosal immunity. The goal of this review is to increase awareness of the new mechanisms being investigated to understand how alcohol affects the human immune system and the development of new strategies to attenuate adverse outcomes in the affected population.


Assuntos
Consumo de Bebidas Alcoólicas/imunologia , Alcoolismo/imunologia , Imunidade nas Mucosas/imunologia , Mucosa Intestinal/imunologia , Pneumonia Bacteriana/imunologia , Mucosa Respiratória/imunologia , Dermatopatias Infecciosas/imunologia , Cicatrização/imunologia , Consumo de Bebidas Alcoólicas/epidemiologia , Alcoolismo/epidemiologia , Humanos , Pneumonia Bacteriana/epidemiologia , Fatores de Risco , Dermatopatias Infecciosas/epidemiologia
14.
Infect Immun ; 83(5): 2082-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25754201

RESUMO

Acute ethanol intoxication suppresses the host immune responses against Streptococcus pneumoniae. As interleukin 17 (IL-17) is a critical cytokine in host defense against extracellular pathogens, including S. pneumoniae, we hypothesized that ethanol impairs mucosal immunity against this pathogen by disrupting IL-17 production or IL-17 receptor (IL-17R) signaling. A chronic ethanol feeding model in simian immunodeficiency virus (SIV)-infected rhesus macaques and acute ethanol intoxication in a murine model were used. Transcriptome analysis of bronchial brushes in the nonhuman primate model showed downregulation of the expression of IL-17-regulated chemokines in ethanol-fed animals, a finding also replicated in the murine model. Surprisingly, recombinant CXCL1 and CXCL5 but not IL-17 or IL-23 plus IL-1ß rescued bacterial burden in the ethanol group to control levels. Taken together, the results of this study suggest that ethanol impairs IL-17-mediated chemokine production in the lung. Thus, exogenous luminal restoration of IL-17-related chemokines, CXCL1 and CXCL5, improves host defenses against S. pneumoniae.


Assuntos
Etanol/toxicidade , Expressão Gênica/efeitos dos fármacos , Imunidade nas Mucosas/efeitos dos fármacos , Interleucina-17/biossíntese , Mucosa/efeitos dos fármacos , Infecções Pneumocócicas/imunologia , Streptococcus pneumoniae/imunologia , Animais , Brônquios/imunologia , Estudos de Coortes , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Macaca mulatta , Masculino , Camundongos
15.
Infect Immun ; 82(4): 1402-7, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24421048

RESUMO

The incidence of community-associated methicillin-resistant Staphylococcus aureus (MRSA) pneumonia in previously healthy individuals has increased in the past 5 years. Such infections are associated with bronchiectasis and high mortality rates, making them a significant public health concern. The mechanisms of host defense against this pathogen are not well characterized. However, patients diagnosed with MRSA, as opposed to methicillin-susceptible S. aureus (MSSA), are more likely to have abused alcohol in the past, and these patients are more likely to die from sepsis. In the United States, USA300 is the predominant strain that causes necrotizing pneumonia. To investigate whether acute ethanol exacerbates MRSA pneumonia, mice were intraperitoneally (i.p.) administered 2 or 4 g/kg of ethanol 30 min prior to oropharyngeal inoculation of 2 × 10(7) CFU of USA300. An increased pulmonary bacterial burden was observed in alcohol-intoxicated mice at 16 and 24 h and was associated with decreased levels of interleukin 6 (IL-6). IL-6 activates signal transducer and activator of transcription 3 (STAT3) as part of an acute-phase response of infection. Reg3γ is an antimicrobial C-type lectin that is induced by STAT3 signaling in response to Gram-positive bacteria. Previously, in situ hybridization studies showed that Reg3g is highly expressed in lung epithelium. In the present study, we found that acute ethanol exacerbated USA300 in a murine model of USA300 pneumonia. This was associated with reduced IL-6 expression in vivo as well as inhibition of IL-6 induction of STAT3 signaling and Reg3g expression in mouse lung epithelial (MLE12) cells in vitro. Furthermore, recombinant Reg3γ administration 4 h after MRSA infection in alcohol-intoxicated mice rescued USA300 clearance in vivo. Therefore, acute alcohol intoxication leads to decreased MRSA clearance in part by inhibiting IL-6/STAT3 induction of the antimicrobial protein Reg3γ in the pulmonary epithelium.


Assuntos
Intoxicação Alcoólica , Staphylococcus aureus Resistente à Meticilina , Pneumonia Estafilocócica , Proteínas/metabolismo , Doença Aguda , Intoxicação Alcoólica/imunologia , Intoxicação Alcoólica/microbiologia , Análise de Variância , Animais , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Etanol/farmacologia , Interleucina-6/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas a Pancreatite , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/metabolismo , Pneumonia Estafilocócica/microbiologia , Mucosa Respiratória/citologia , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais
16.
Nat Immunol ; 13(12): 1178-86, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23104095

RESUMO

We report the clinical description and molecular dissection of a new fatal human inherited disorder characterized by chronic autoinflammation, invasive bacterial infections and muscular amylopectinosis. Patients from two kindreds carried biallelic loss-of-expression and loss-of-function mutations in HOIL1 (RBCK1), a component of the linear ubiquitination chain assembly complex (LUBAC). These mutations resulted in impairment of LUBAC stability. NF-κB activation in response to interleukin 1ß (IL-1ß) was compromised in the patients' fibroblasts. By contrast, the patients' mononuclear leukocytes, particularly monocytes, were hyper-responsive to IL-1ß. The consequences of human HOIL-1 and LUBAC deficiencies for IL-1ß responses thus differed between cell types, consistent with the unique association of autoinflammation and immunodeficiency in these patients. These data suggest that LUBAC regulates NF-κB-dependent IL-1ß responses differently in different cell types.


Assuntos
Doença de Depósito de Glicogênio Tipo IV/genética , Doenças Hereditárias Autoinflamatórias/genética , Síndromes de Imunodeficiência/genética , NF-kappa B/metabolismo , Ubiquitina-Proteína Ligases/genética , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Proteínas de Ciclo Celular/genética , Linhagem Celular , Fibroblastos/imunologia , Fibroblastos/metabolismo , Humanos , Síndromes de Imunodeficiência/metabolismo , Interleucina-1beta/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Repressoras/genética , Fatores de Transcrição , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
17.
J Microbiol Immunol Infect ; 43(2): 155-8, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20457433

RESUMO

Pasteurella multocida is a Gram-negative bacteria found in the oropharynx of many domestic animals. P. multocida can cause a variety of human infections, but it remains a rare cause of peritoneal dialysis-related peritonitis. We describe a severe case of peritoneal dialysis-related peritonitis due to P. multocida infection caused by close contact with a cat.


Assuntos
Animais Domésticos/microbiologia , Portador Sadio/veterinária , Infecções por Pasteurella/diagnóstico , Infecções por Pasteurella/microbiologia , Pasteurella multocida/isolamento & purificação , Diálise Peritoneal/efeitos adversos , Peritonite/microbiologia , Adulto , Animais , Antibacterianos/uso terapêutico , Portador Sadio/microbiologia , Gatos , Humanos , Masculino , Infecções por Pasteurella/transmissão
19.
J Immunol ; 179(4): 2509-19, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17675513

RESUMO

Mycobacterium tuberculosis (Mtb) is thought to reside in macrophages, although infected dendritic cells (DCs) have been observed. Thus, although cellular associations have been made, global characterization of the cells harboring Mtb is lacking. We have performed temporal and quantitative characterization of the cells harboring Mtb following aerosol infection of mice by using GFP-expressing bacteria and flow cytometry. We discovered that Mtb infects phagocytic cells of diverse phenotypes, that the predominant infected cell populations change with time, and that myeloid DCs are the major cell population infected with Mtb in the lungs and lymph nodes. We also found that the bacteria in the lung-draining lymph node are transported there from the lungs by a CCL19/21-dependent mechanism and that the transport of bacteria to the lymph node is a transient phenomenon despite chronic infection. In addition, we found that the lymph node cell subsets that are most efficacious in stimulating Mtb-specific, TCR-transgenic CD4(+) T lymphocytes are not infected with the bacteria and are scarce or absent from the lungs of infected mice. Finally, we found that the lung cell populations that are infected with Mtb at high frequency are relatively ineffective at stimulating Ag-specific CD4(+) T lymphocytes, and we have obtained evidence that live Mtb can inhibit MHC class II Ag presentation without a decrease in the surface expression of MHC class II. These results indicate that Mtb targets DC migration and Ag presentation in vivo to promote persistent infection.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Linfonodos/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose Pulmonar/imunologia , Animais , Apresentação de Antígeno/genética , Transporte Biológico Ativo/genética , Transporte Biológico Ativo/imunologia , Linfócitos T CD4-Positivos/microbiologia , Linfócitos T CD4-Positivos/patologia , Movimento Celular/genética , Movimento Celular/imunologia , Quimiocina CCL19 , Quimiocina CCL21 , Quimiocinas CC/imunologia , Células Dendríticas/microbiologia , Células Dendríticas/patologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Linfonodos/microbiologia , Linfonodos/patologia , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Mycobacterium tuberculosis/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Tuberculose Pulmonar/patologia , Tuberculose Pulmonar/veterinária
20.
J Clin Invest ; 117(7): 1738-45, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17607348

RESUMO

Tuberculosis kills nearly 2 million people annually, and current approaches to tuberculosis control are expensive, have limited efficacy, and are vulnerable to being overcome by extensively drug-resistant strains of Mycobacterium tuberculosis. Determination of the genome sequence of M. tuberculosis has revolutionized tuberculosis research, contributed to major advances in the understanding of the evolution and pathogenesis of M. tuberculosis, and facilitated development of new diagnostic tests with increased specificity for tuberculosis. In this review, we describe some of the major progress in tuberculosis research that has resulted from knowledge of the genome sequence and note some of the problems that remain unsolved.


Assuntos
Evolução Biológica , Genômica , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidade , Tuberculose/diagnóstico , Tuberculose/genética , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Mycobacterium tuberculosis/classificação , Tuberculose/microbiologia , Tuberculose/transmissão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...