Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Adv Immunol ; 151: 49-97, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34656288

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections trigger viral RNA sensors such as TLR7 and RIG-I, thereby leading to production of type I interferon (IFN) and other inflammatory mediators. Expression of viral proteins in the context of this inflammation leads to stereotypical antigen-specific antibody and T cell responses that clear the virus. Immunity is then maintained through long-lived antibody-secreting plasma cells and by memory B and T cells that can initiate anamnestic responses. Each of these steps is consistent with prior knowledge of acute RNA virus infections. Yet there are certain concepts, while not entirely new, that have been resurrected by the biology of severe SARS-CoV-2 infections and deserve further attention. These include production of anti-IFN autoantibodies, early inflammatory processes that slow adaptive humoral immunity, immunodominance of antibody responses, and original antigenic sin. Moreover, multiple different vaccine platforms allow for comparisons of pathways that promote robust and durable adaptive immunity.


Assuntos
COVID-19 , Vacinas , Imunidade Adaptativa , Humanos , Imunidade Humoral , SARS-CoV-2
3.
J Photochem Photobiol B ; 224: 112319, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34598020

RESUMO

The germicidal properties of short wavelength ultraviolet C (UVC) light are well established and used to inactivate many viruses and other microbes. However, much less is known about germicidal effects of terrestrial solar UV light, confined exclusively to wavelengths in the UVA and UVB regions. Here, we have explored the sensitivity of the human coronaviruses HCoV-NL63 and SARS-CoV-2 to solar-simulated full spectrum ultraviolet light (sUV) delivered at environmentally relevant doses. First, HCoV-NL63 coronavirus inactivation by sUV-exposure was confirmed employing (i) viral plaque assays, (ii) RT-qPCR detection of viral genome replication, and (iii) infection-induced stress response gene expression array analysis. Next, a detailed dose-response relationship of SARS-CoV-2 coronavirus inactivation by sUV was elucidated, suggesting a half maximal suppression of viral infectivity at low sUV doses. Likewise, extended sUV exposure of SARS-CoV-2 blocked cellular infection as revealed by plaque assay and stress response gene expression array analysis. Moreover, comparative (HCoV-NL63 versus SARS-CoV-2) single gene expression analysis by RT-qPCR confirmed that sUV exposure blocks coronavirus-induced redox, inflammatory, and proteotoxic stress responses. Based on our findings, we estimate that solar ground level full spectrum UV light impairs coronavirus infectivity at environmentally relevant doses. Given the urgency and global scale of the unfolding SARS-CoV-2 pandemic, these prototype data suggest feasibility of solar UV-induced viral inactivation, an observation deserving further molecular exploration in more relevant exposure models.


Assuntos
Infecções por Coronavirus/prevenção & controle , Coronavirus Humano NL63/efeitos da radiação , Infecções Respiratórias/prevenção & controle , SARS-CoV-2/efeitos da radiação , Luz Solar , Raios Ultravioleta , Animais , Linhagem Celular , Chlorocebus aethiops , Coronavirus Humano NL63/fisiologia , Células Epiteliais/virologia , Genoma Viral/efeitos da radiação , Humanos , SARS-CoV-2/fisiologia , Transcriptoma/efeitos da radiação , Ensaio de Placa Viral , Inativação de Vírus/efeitos da radiação , Replicação Viral/efeitos da radiação
4.
bioRxiv ; 2021 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-34282415

RESUMO

The germicidal properties of short wavelength ultraviolet C (UVC) light are well established and used to inactivate many viruses and other microbes. However, much less is known about germicidal effects of terrestrial solar UV light, confined exclusively to wavelengths in the UVA and UVB regions. Here, we have explored the sensitivity of the human coronaviruses HCoV-NL63 and SARS-CoV-2 to solar-simulated full spectrum ultraviolet light (sUV) delivered at environmentally relevant doses. First, HCoV-NL63 coronavirus inactivation by sUV-exposure was confirmed employing (i) viral plaque assays, (ii) RT-qPCR detection of viral genome replication, and (iii) infection-induced stress response gene expression array analysis. Next, a detailed dose-response relationship of SARS-CoV-2 coronavirus inactivation by sUV was elucidated, suggesting a half maximal suppression of viral infectivity at low sUV doses. Likewise, extended sUV exposure of SARS-CoV-2 blocked cellular infection as revealed by plaque assay and stress response gene expression array analysis. Moreover, comparative (HCoV-NL63 versus SARS-CoV-2) single gene expression analysis by RT-qPCR confirmed that sUV exposure blocks coronavirus-induced redox, inflammatory, and proteotoxic stress responses. Based on our findings, we estimate that solar ground level full spectrum UV light impairs coronavirus infectivity at environmentally relevant doses. Given the urgency and global scale of the unfolding SARS-CoV-2 pandemic, these prototype data suggest feasibility of solar UV-induced viral inactivation, an observation deserving further molecular exploration in more relevant exposure models.

5.
PLoS Pathog ; 16(10): e1009009, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33104760

RESUMO

A key difference that distinguishes viral infections from protein immunizations is the recognition of viral nucleic acids by cytosolic pattern recognition receptors (PRRs). Insights into the functions of cytosolic PRRs such as the RNA-sensing Rig-I-like receptors (RLRs) in the instruction of adaptive immunity are therefore critical to understand protective immunity to infections. West Nile virus (WNV) infection of mice deficent of RLR-signaling adaptor MAVS results in a defective adaptive immune response. While this finding suggests a role for RLRs in the instruction of adaptive immunity to WNV, it is difficult to interpret due to the high WNV viremia, associated exessive antigen loads, and pathology in the absence of a MAVS-dependent innate immune response. To overcome these limitations, we have infected MAVS-deficient (MAVSKO) mice with a single-round-of-infection mutant of West Nile virus. We show that MAVSKO mice failed to produce an effective neutralizing antibody response to WNV despite normal antibody titers against the viral WNV-E protein. This defect occurred independently of antigen loads or overt pathology. The specificity of the antibody response in infected MAVSKO mice remained unchanged and was still dominated by antibodies that bound the neutralizing lateral ridge (LR) epitope in the DIII domain of WNV-E. Instead, MAVSKO mice produced IgM antibodies, the dominant isotype controlling primary WNV infection, with lower affinity for the DIII domain. Our findings suggest that RLR-dependent signals are important for the quality of the humoral immune response to WNV.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Receptores de Reconhecimento de Padrão/imunologia , Imunidade Adaptativa/imunologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Formação de Anticorpos , Proteína DEAD-box 58/imunologia , Proteína DEAD-box 58/metabolismo , Feminino , Imunidade Humoral , Imunidade Inata/imunologia , Imunoglobulina M , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais/imunologia , Febre do Nilo Ocidental/imunologia , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/patogenicidade
6.
J Immunol ; 201(12): 3641-3650, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30455400

RESUMO

IL-1, generally considered an amplifier of adaptive immune responses, has been proposed for use as adjuvant during immunization with weak immunogens. However, its effects on memory T cell function remain largely undefined. Using the murine model of acute viral infection, in this paper, we show that in addition to augmenting the size of the Ag-specific pool, IL-1 signals act directly on CD8 T cells to promote the quality of effector and memory responses. Ablation of IL-1R1 or MyD88 signaling in T cells led to functional impairment; both the ability to produce multiple cytokines on a per cell basis (polyfunctionality) and the potential for recall proliferation in response to antigenic restimulation were compromised. IL-1 supplementation during priming augmented the expansion of Ag-specific CD8 T cells through the MyD88-IRAK1/4 axis, resulting in a larger memory pool capable of robust secondary expansion in response to rechallange. Together, these findings demonstrate a critical role of the IL-1-MyD88 axis in programming the quantity and quality of memory CD8 T cell responses and support the notion that IL-1 supplementation may be exploited to enhance adoptive T cell therapies against cancers and chronic infections.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Interleucina-1/metabolismo , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Reprogramação Celular , Técnicas de Reprogramação Celular , Humanos , Memória Imunológica , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/metabolismo , Receptores Tipo I de Interleucina-1/genética , Transdução de Sinais
7.
Nature ; 514(7524): 638-41, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25274297

RESUMO

Systemic infection induces conserved physiological responses that include both resistance and 'tolerance of infection' mechanisms. Temporary anorexia associated with an infection is often beneficial, reallocating energy from food foraging towards resistance to infection or depriving pathogens of nutrients. However, it imposes a stress on intestinal commensals, as they also experience reduced substrate availability; this affects host fitness owing to the loss of caloric intake and colonization resistance (protection from additional infections). We hypothesized that the host might utilize internal resources to support the gut microbiota during the acute phase of the disease. Here we show that systemic exposure to Toll-like receptor (TLR) ligands causes rapid α(1,2)-fucosylation of small intestine epithelial cells (IECs) in mice, which requires the sensing of TLR agonists, as well as the production of interleukin (IL)-23 by dendritic cells, activation of innate lymphoid cells and expression of fucosyltransferase 2 (Fut2) by IL-22-stimulated IECs. Fucosylated proteins are shed into the lumen and fucose is liberated and metabolized by the gut microbiota, as shown by reporter bacteria and community-wide analysis of microbial gene expression. Fucose affects the expression of microbial metabolic pathways and reduces the expression of bacterial virulence genes. It also improves host tolerance of the mild pathogen Citrobacter rodentium. Thus, rapid IEC fucosylation appears to be a protective mechanism that utilizes the host's resources to maintain host-microbial interactions during pathogen-induced stress.


Assuntos
Doença , Epitélio/metabolismo , Epitélio/microbiologia , Fucose/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Simbiose , Animais , Anorexia/complicações , Anorexia/microbiologia , Bactérias/genética , Bactérias/metabolismo , Bactérias/patogenicidade , Citrobacter rodentium/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Ingestão de Alimentos , Ácidos Graxos/química , Ácidos Graxos/metabolismo , Feminino , Fucosiltransferases/metabolismo , Regulação Bacteriana da Expressão Gênica , Glicosilação , Tolerância Imunológica , Imunidade Inata , Interleucinas/biossíntese , Interleucinas/imunologia , Ligantes , Masculino , Redes e Vias Metabólicas/genética , Camundongos , Microbiota/fisiologia , Fatores de Proteção , Receptores Toll-Like/agonistas , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Fatores de Virulência/genética , Interleucina 22 , Galactosídeo 2-alfa-L-Fucosiltransferase
8.
Elife ; 3: e01949, 2014 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-24842874

RESUMO

Innate immune recognition is critical for the induction of adaptive immune responses; however the underlying mechanisms remain incompletely understood. In this study, we demonstrate that T cell-specific deletion of the IL-6 receptor α chain (IL-6Rα) results in impaired Th1 and Th17 T cell responses in vivo, and a defect in Tfh function. Depletion of Tregs in these mice rescued the Th1 but not the Th17 response. Our data suggest that IL-6 signaling in effector T cells is required to overcome Treg-mediated suppression in vivo. We show that IL-6 cooperates with IL-1ß to block the suppressive effect of Tregs on CD4(+) T cells, at least in part by controlling their responsiveness to IL-2. In addition, although IL-6Rα-deficient T cells mount normal primary Th1 responses in the absence of Tregs, they fail to mature into functional memory cells, demonstrating a key role for IL-6 in CD4(+) T cell memory formation.DOI: http://dx.doi.org/10.7554/eLife.01949.001.


Assuntos
Imunidade Adaptativa , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Imunidade Inata , Memória Imunológica , Interleucina-6/metabolismo , Transdução de Sinais , Imunidade Adaptativa/efeitos dos fármacos , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Imunidade Inata/efeitos dos fármacos , Imunização , Memória Imunológica/efeitos dos fármacos , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacologia , Interleucina-6/imunologia , Interleucina-6/farmacologia , Subunidade alfa de Receptor de Interleucina-6/deficiência , Subunidade alfa de Receptor de Interleucina-6/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th17/imunologia , Células Th17/metabolismo
9.
PLoS Biol ; 12(1): e1001762, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24453940

RESUMO

Mast cells are implicated in the pathogenesis of inflammatory and autoimmune diseases. However, this notion based on studies in mast cell-deficient mice is controversial. We therefore established an in vivo model for hyperactive mast cells by specifically ablating the NF-κB negative feedback regulator A20. While A20 deficiency did not affect mast cell degranulation, it resulted in amplified pro-inflammatory responses downstream of IgE/FcεRI, TLRs, IL-1R, and IL-33R. As a consequence house dust mite- and IL-33-driven lung inflammation, late phase cutaneous anaphylaxis, and collagen-induced arthritis were aggravated, in contrast to experimental autoimmune encephalomyelitis and immediate anaphylaxis. Our results provide in vivo evidence that hyperactive mast cells can exacerbate inflammatory disorders and define diseases that might benefit from therapeutic intervention with mast cell function.


Assuntos
Anafilaxia/imunologia , Artrite Experimental/imunologia , Proteínas de Ligação a DNA/deficiência , Encefalomielite Autoimune Experimental/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Mastócitos/imunologia , Ubiquitina-Proteína Ligases/deficiência , Anafilaxia/induzido quimicamente , Anafilaxia/metabolismo , Anafilaxia/patologia , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Colágeno Tipo II/administração & dosagem , Cisteína Endopeptidases , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Dinitrofenóis/administração & dosagem , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Expressão Gênica , Imunoglobulina E/genética , Imunoglobulina E/imunologia , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33 , Interleucinas/genética , Interleucinas/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Masculino , Mastócitos/metabolismo , Mastócitos/patologia , Camundongos , Camundongos Transgênicos , Glicoproteína Mielina-Oligodendrócito/administração & dosagem , NF-kappa B/genética , NF-kappa B/imunologia , Fragmentos de Peptídeos/administração & dosagem , Pneumonia/induzido quimicamente , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/patologia , Pyroglyphidae/imunologia , Receptores de IgE/genética , Receptores de IgE/imunologia , Receptores de Interleucina/genética , Receptores de Interleucina/imunologia , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/imunologia , Soroalbumina Bovina/administração & dosagem , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
10.
Immunity ; 40(1): 78-90, 2014 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-24439266

RESUMO

Innate immune recognition controls adaptive immune responses through multiple mechanisms. The MyD88 signaling adaptor operates in many cell types downstream of Toll-like receptors (TLRs) and interleukin-1 (IL-1) receptor family members. Cell-type-specific functions of MyD88 signaling remain poorly characterized. Here, we have shown that the T cell-specific ablation of MyD88 in mice impairs not only T helper 17 (Th17) cell responses, but also Th1 cell responses. MyD88 relayed signals of TLR-induced IL-1, which became dispensable for Th1 cell responses in the absence of T regulatory (Treg) cells. Treg cell-specific ablation of MyD88 had no effect, suggesting that IL-1 acts on naive CD4(+) T cells instead of Treg cells themselves. Together, these findings demonstrate that IL-1 renders naive CD4(+) T cells refractory to Treg cell-mediated suppression in order to allow their differentiation into Th1 cells. In addition, IL-1 was also important for the generation of functional CD4(+) memory T cells.


Assuntos
Interleucina-1/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th17/imunologia , Imunidade Adaptativa , Animais , Células Cultivadas , Imunidade Inata , Memória Imunológica , Terapia de Imunossupressão , Interleucina-18/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/genética , Especificidade de Órgãos , Receptores de Interleucina-1/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia
11.
Immunity ; 39(5): 976-85, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24210353

RESUMO

Venoms consist of toxic components that are delivered to their victims via bites or stings. Venoms also represent a major class of allergens in humans. Phospholipase A2 (PLA2) is a conserved component of venoms from multiple species and is the major allergen in bee venom. Here we examined how bee venom PLA2 is sensed by the innate immune system and induces a type 2 immune response in mice. We found that bee venom PLA2 induced a T helper type 2 (Th2) cell-type response and group 2 innate lymphoid cell activation via the enzymatic cleavage of membrane phospholipids and release of interleukin-33. Furthermore, we showed that the IgE response to PLA2 could protect mice from future challenge with a near-lethal dose of PLA2. These data suggest that the innate immune system can detect the activity of a conserved component of venoms and induce a protective immune response against a venom toxin.


Assuntos
Venenos de Abelha/enzimologia , Imunidade Inata/imunologia , Imunoglobulina E/biossíntese , Proteínas de Insetos/imunologia , Lisofosfolipídeos/imunologia , Fosfolipases A2/imunologia , Receptores de Interleucina/imunologia , Células Th2/imunologia , Anafilaxia/etiologia , Anafilaxia/imunologia , Anafilaxia/prevenção & controle , Animais , Venenos de Abelha/toxicidade , Venenos de Crotalídeos/imunologia , Genes Reporter , Imunoglobulina E/imunologia , Imunoglobulina G/biossíntese , Imunoglobulina G/imunologia , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33 , Interleucina-4/biossíntese , Interleucina-4/genética , Interleucinas/imunologia , Ativação Linfocitária , Meliteno/imunologia , Lipídeos de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/fisiologia , Ovalbumina/imunologia , Fosfolipídeos/metabolismo , Receptores de IgE/imunologia
12.
Immunity ; 38(3): 528-40, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23499488

RESUMO

Detection of self nucleic acids by Toll-like receptors (TLR) preciptates autoimmune diseases, including systemic lupus erythematosus (SLE). It remains unknown how TLR signals in specific cell types contribute to distinct manifestations of SLE. Here, we demonstrate that formation of anti-nuclear antibodies in MRL.Fas(lpr) mice entirely depends on the TLR signaling adaptor MyD88 in B cells. Further, MyD88 deficiency in B cells ameliorated nephritis, including antibody-independent interstitial T cell infiltrates, suggesting that nucleic acid-specific B cells activate nephrotoxic T cells. Surprisingly, MyD88 deletion in dendritic cells (DCs) did not affect nephritis, despite the importance of DCs in renal inflammation. In contrast, MyD88 in DCs was critical for dermatitis, revealing a separate pathogenetic mechanism. DC-expressed MyD88 promoted interferon-α production by plasmacytoid DCs, which was associated with Death domain-associated protein 6 upregulation and B lymphopenia. Our findings thus reveal unique immunopathological consequences of MyD88 signaling in B cells and DCs in lupus.


Assuntos
Linfócitos B/imunologia , Células Dendríticas/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais/imunologia , Animais , Anticorpos Antinucleares/sangue , Anticorpos Antinucleares/imunologia , Autoimunidade/genética , Autoimunidade/imunologia , Linfócitos B/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/metabolismo , Nefrite Lúpica/genética , Nefrite Lúpica/imunologia , Nefrite Lúpica/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Células Th1/imunologia , Células Th1/metabolismo
13.
Nat Commun ; 3: 1120, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23047678

RESUMO

Commensal bacterial sensing by Toll-like receptors is critical for maintaining intestinal homeostasis, but can lead to colitis in the absence of interleukin-10. Although Toll-like receptors are expressed in multiple cell types in the colon, the cell type(s) responsible for the development of colitis are currently unknown. Here we generated mice that are selectively deficient in MyD88 in various cellular compartments in an interleukin-10(-/-) setting. Although epithelial expression of MyD88 was dispensable, MyD88 expression in the mononuclear phagocyte compartment was required for colitis development. Specifically, phenotypically distinct populations of colonic mononuclear phagocytes expressed high levels of interleukin-1ß, interleukin-23 and interleukin-6, and promoted T-helper 17 responses in the absence of interleukin-10. Thus, gut bacterial sensing through MyD88 in mononuclear phagocytes drives inflammatory bowel disease when unopposed by interleukin-10.


Assuntos
Colite/metabolismo , Colo/metabolismo , Colo/patologia , Interleucina-10/deficiência , Interleucina-10/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Fagócitos/metabolismo , Animais , Colite/genética , Colite/patologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Interleucina-10/genética , Interleucina-1beta/metabolismo , Interleucina-23/metabolismo , Interleucina-6/metabolismo , Camundongos , Fator 88 de Diferenciação Mieloide/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
14.
J Exp Med ; 209(9): 1689-702, 2012 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-22908325

RESUMO

Constitutively active RAS plays a central role in the development of human cancer and is sufficient to induce tumors in two-stage skin carcinogenesis. RAS-mediated tumor formation is commonly associated with up-regulation of cytokines and chemokines that mediate an inflammatory response considered relevant to oncogenesis. In this study, we report that mice lacking IL-1R or MyD88 are less sensitive to topical skin carcinogenesis than their respective wild-type (WT) controls. MyD88(-/-) or IL-1R(-/-) keratinocytes expressing oncogenic RAS are hyperproliferative and fail to up-regulate proinflammatory genes or down-regulate differentiation markers characteristic of RAS-expressing WT keratinocytes. Although RAS-expressing MyD88(-/-) keratinocytes form only a few small tumors in orthotopic grafts, IL-1R-deficient RAS-expressing keratinocytes retain the ability to form tumors in orthotopic grafts. Using both genetic and pharmacological approaches, we find that the differentiation and proinflammatory effects of oncogenic RAS in keratinocytes require the establishment of an autocrine loop through IL-1α, IL-1R, and MyD88 leading to phosphorylation of IκBα and NF-κB activation. Blocking IL-1α-mediated NF-κB activation in RAS-expressing WT keratinocytes reverses the differentiation defect and inhibits proinflammatory gene expression. Collectively, these results demonstrate that MyD88 exerts a cell-intrinsic function in RAS-mediated transformation of keratinocytes.


Assuntos
Queratinócitos/metabolismo , Queratinócitos/patologia , Fator 88 de Diferenciação Mieloide/metabolismo , Receptores de Interleucina-1/metabolismo , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Diferenciação Celular/genética , Transformação Celular Neoplásica/genética , Receptores ErbB/metabolismo , Genes ras , Proteínas I-kappa B/metabolismo , Inflamação/genética , Inflamação/metabolismo , Interleucina-1alfa/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fator 88 de Diferenciação Mieloide/genética , NF-kappa B/metabolismo , Fosforilação , Receptores de Interleucina-1/genética , Transdução de Sinais , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/metabolismo
15.
J Exp Med ; 209(6): 1075-81, 2012 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-22615128

RESUMO

Low-fidelity DNA polymerases introduce nucleotide substitutions in immunoglobulin variable regions during somatic hypermutation. Although DNA polymerase (pol) η is the major low-fidelity polymerase, other DNA polymerases may also contribute. Existing data are contradictory as to whether pol ζ is involved. We reasoned that the presence of pol η may mask the contribution of pol ζ, and therefore we generated mice deficient for pol η and heterozygous for pol ζ. The frequency and spectra of hypermutation was unaltered between Polζ(+/-) Polη(-/-) and Polζ(+/+) Polη(-/-) clones. However, there was a decrease in tandem double-base substitutions in Polζ(+/-) Polη(-/-) cells compared with Polζ(+/+) Polη(-/-) cells, suggesting that pol ζ generates tandem mutations. Contiguous mutations are consistent with the biochemical property of pol ζ to extend a mismatch with a second mutation. The presence of this unique signature implies that pol ζ contributes to mutational synthesis in vivo. Additionally, data on tandem mutations from wild type, Polζ(+/-), Polζ(-/-), Ung(-/-), Msh2(-/-), Msh6(-/-), and Ung(-/-) Msh2(-/-) clones suggest that pol ζ may function in the MSH2-MSH6 pathway.


Assuntos
DNA Polimerase Dirigida por DNA/genética , Região Variável de Imunoglobulina/genética , Mutação , Animais , Divisão Celular/genética , Reparo de Erro de Pareamento de DNA/genética , Proteínas de Ligação a DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Heterozigoto , Camundongos , Camundongos Mutantes , Proteína 2 Homóloga a MutS/genética , Hipermutação Somática de Imunoglobulina , Uracila-DNA Glicosidase/genética
16.
J Immunol ; 188(9): 4334-9, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22442445

RESUMO

Langerhans cells (LC) are a subset of skin-resident dendritic cells (DC) that reside in the epidermis as immature DC, where they acquire Ag. A key step in the life cycle of LC is their activation into mature DC in response to various stimuli, including epicutaneous sensitization with hapten and skin infection with Candida albicans. Mature LC migrate to the skin-draining LN, where they present Ag to CD4 T cells and modulate the adaptive immune response. LC migration is thought to require the direct action of IL-1ß and IL-18 on LC. In addition, TLR ligands are present in C. albicans, and hapten sensitization produces endogenous TLR ligands. Both could contribute to LC activation. We generated Langerin-Cre MyD88(fl) mice in which LC are insensitive to IL-1 family members and most TLR ligands. LC migration in the steady state, after hapten sensitization and postinfection with C. albicans, was unaffected. Contact hypersensitivity in Langerin-Cre MyD88(fl) mice was similarly unaffected. Interestingly, in response to C. albicans infection, these mice displayed reduced proliferation of Ag-specific CD4 T cells and defective Th17 subset differentiation. Surface expression of costimulatory molecules was intact on LC, but expression of IL-1ß, IL-6, and IL-23 was reduced. Thus, sensitivity to MyD88-dependent signals is not required for LC migration, but is required for the full activation and function of LC in the setting of fungal infection.


Assuntos
Antígenos de Fungos/imunologia , Candida albicans/imunologia , Candidíase/imunologia , Movimento Celular/imunologia , Dermatite de Contato/imunologia , Dermatomicoses/imunologia , Células de Langerhans/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais/imunologia , Animais , Antígenos de Fungos/genética , Antígenos de Fungos/metabolismo , Candida albicans/genética , Candida albicans/metabolismo , Candidíase/genética , Candidíase/metabolismo , Movimento Celular/genética , Dermatite de Contato/genética , Dermatite de Contato/metabolismo , Dermatomicoses/genética , Dermatomicoses/metabolismo , Haptenos/genética , Haptenos/imunologia , Interleucinas/genética , Interleucinas/imunologia , Interleucinas/metabolismo , Células de Langerhans/metabolismo , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais/genética , Células Th17/imunologia , Células Th17/metabolismo
17.
Arterioscler Thromb Vasc Biol ; 32(1): 103-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22034510

RESUMO

OBJECTIVE: Increased circulating cytokine levels are a prominent feature of aging that may contribute to atherosclerosis. However, the role vascular cells play in chronic inflammation induced by aging is not clear. Here, we examined the role of aging on inflammatory responses of vascular cells. METHODS AND RESULTS: In an ex vivo culture system, we examined the inflammatory response of aortas from young (2-4 months) and aged (16-18 months) mice under nonstimulatory conditions. We found that basal levels of interleukin-6 were increased in aged aortas. Aged aortic vascular smooth muscle cells (VSMC) exhibited a higher basal secretion of interleukin-6 than young VSMC. Gene and protein expression analysis revealed that aged VSMC exhibited upregulation of chemokines (eg, CCL2), adhesion molecules (eg, intracellular adhesion molecule 1), and innate immune receptors (eg, Toll-like receptor [TLR] 4), which all contribute to atherosclerosis. Using VSMC from aged TL4(-/-) and Myd88(-/-) mice, we demonstrate that signaling via TLR4 and its signal adaptor, MyD88, are in part responsible for the age-elevated basal interleukin-6 response. CONCLUSIONS: Aging induces a proinflammatory phenotype in VSMC due in part to increased signaling of TLR4 and MyD88. Our results provide a potential explanation as to why aging leads to chronic inflammation and enhanced atherosclerosis.


Assuntos
Envelhecimento/imunologia , Quimiocina CCL2/biossíntese , Interleucina-6/biossíntese , Miócitos de Músculo Liso/imunologia , Envelhecimento/genética , Animais , Aorta Torácica/imunologia , Aterosclerose/etiologia , Aterosclerose/genética , Aterosclerose/imunologia , Células Cultivadas , Quimiocina CCL2/genética , Feminino , Interleucina-6/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Transdução de Sinais , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética , Regulação para Cima
18.
Adv Immunol ; 109: 87-124, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21569913

RESUMO

The mammalian immune system comprises an adaptive and an innate component. The innate immune system employs a limited number of germ-line-encoded pattern-recognition receptors (PRRs) that recognize invariant pathogen-associated molecular patterns (PAMPs). In contrast, the adaptive immune system depends on the generation of a diverse repertoire of antigen receptors on T and B lymphocytes and subsequent activation and clonal expansion of cells carrying the appropriate antigen-specific receptors. Induction of adaptive immunity not only depends on direct antigen recognition by the antigen receptors but also relies on essential signals that are delivered by the innate immune system. In recent years, we have witnessed the discovery of a still expanding array of different PRR systems that govern the generation of adaptive immunity. Here, we review our current understanding of innate control of adaptive immunity. In particular, we discuss how PRRs initiate adaptive immune responses in general, discuss specific mechanisms that shape the ensuing T and B cell responses, and highlight open questions that are still awaiting answers.


Assuntos
Imunidade Adaptativa , Comunicação Celular , Imunidade Inata , Infecções/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Animais , Comunicação Celular/imunologia , Humanos , Infecções/genética , Mutação , Receptores de Antígenos/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
19.
Blood ; 117(7): 2227-36, 2011 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-21088135

RESUMO

The ubiquitin-editing enzyme A20/TNFAIP3 is essential for controlling signals inducing the activation of nuclear factor-κB transcription factors. Polymorphisms and mutations in the TNFAIP3 gene are linked to various human autoimmune conditions, and inactivation of A20 is a frequent event in human B-cell lymphomas characterized by constitutive nuclear factor-κB activity. Through B cell-specific ablation in the mouse, we show here that A20 is required for the normal differentiation of the marginal zone B and B1 cell subsets. However, loss of A20 in B cells lowers their activation threshold and enhances proliferation and survival in a gene-dose-dependent fashion. Through the expression of proinflammatory cytokines, most notably interleukin-6, A20-deficient B cells trigger a progressive inflammatory reaction in naive mice characterized by the expansion of myeloid cells, effector-type T cells, and regulatory T cells. This culminates in old mice in an autoimmune syndrome characterized by splenomegaly, plasma cell hyperplasia, and the presence of class-switched, tissue-specific autoantibodies.


Assuntos
Linfócitos B/imunologia , Linfócitos B/patologia , Cisteína Endopeptidases/deficiência , Cisteína Endopeptidases/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Envelhecimento/imunologia , Envelhecimento/patologia , Animais , Autoimunidade , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/patologia , Diferenciação Celular , Cisteína Endopeptidases/genética , Dosagem de Genes , Humanos , Técnicas In Vitro , Inflamação/etiologia , Inflamação/imunologia , Inflamação/patologia , Interleucina-6/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/patologia , NF-kappa B/metabolismo , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/imunologia
20.
Cell Metab ; 10(4): 249-59, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19808018

RESUMO

Obesity-associated activation of inflammatory pathways represents a key step in the development of insulin resistance in peripheral organs, partially via activation of TLR4 signaling by fatty acids. Here, we demonstrate that palmitate acting in the central nervous system (CNS) inhibits leptin-induced anorexia and Stat3 activation. To determine the functional significance of TLR signaling in the CNS in the development of leptin resistance and diet-induced obesity in vivo, we have characterized mice deficient for the TLR adaptor molecule MyD88 in the CNS (MyD88(DeltaCNS)). Compared to control mice, MyD88(DeltaCNS) mice are protected from high-fat diet (HFD)-induced weight gain, from the development of HFD-induced leptin resistance, and from the induction of leptin resistance by acute central application of palmitate. Moreover, CNS-restricted MyD88 deletion protects from HFD- and icv palmitate-induced impairment of peripheral glucose metabolism. Thus, we define neuronal MyD88-dependent signaling as a key regulator of diet-induced leptin and insulin resistance in vivo.


Assuntos
Sistema Nervoso Central/metabolismo , Dieta , Leptina/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Obesidade/metabolismo , Ácido Palmítico/metabolismo , Transdução de Sinais/fisiologia , Animais , Gorduras na Dieta/metabolismo , Ingestão de Alimentos , Metabolismo Energético , Ativação Enzimática , Feminino , Glucose/metabolismo , Homeostase , Humanos , Quinase I-kappa B/metabolismo , Insulina/metabolismo , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/fisiologia , Fator 88 de Diferenciação Mieloide/genética , Aumento de Peso
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...