Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Sci (Weinh) ; 10(31): e2302248, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37750461

RESUMO

New vaccine platforms that activate humoral immunity and generate neutralizing antibodies are required to combat emerging pathogens, including influenza virus. A slurry of antigen-loaded hydrogel microparticles that anneal to form a porous scaffold with high surface area for antigen uptake by infiltrating immune cells as the biomaterial degrades is demonstrated to enhance humoral immunity. Antigen-loaded-microgels elicited a robust cellular humoral immune response, with increased CD4+ T follicular helper (Tfh) cells and prolonged germinal center (GC) B cells comparable to the commonly used adjuvant, aluminum hydroxide (Alum). Increasing the weight fraction of polymer material led to increased material stiffness and antigen-specific antibody titers superior to Alum. Vaccinating mice with inactivated influenza virus loaded into this more highly cross-linked formulation elicited a strong antibody response and provided protection against a high dose viral challenge. By tuning physical and chemical properties, adjuvanticity can be enhanced leading to humoral immunity and protection against a pathogen, leveraging two different types of antigenic material: individual protein antigen and inactivated virus. The flexibility of the platform may enable design of new vaccines to enhance innate and adaptive immune cell programming to generate and tune high affinity antibodies, a promising approach to generate long-lasting immunity.


Assuntos
Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Orthomyxoviridae , Animais , Camundongos , Humanos , Imunidade Humoral , Porosidade , Anticorpos Antivirais , Antígenos
2.
iScience ; 23(5): 101050, 2020 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-32339990

RESUMO

Myeloid derived suppressor cells (MDSCs) are a population of immature myeloid cells that suppress adaptive immune function, yet the factors that regulate their suppressive function in patients with infection remain unclear. We studied MDSCs in patients with leprosy, a disease caused by Mycobacterium leprae, where clinical manifestations present on a spectrum that correlate with immunity to the pathogen. We found that HLA-DR-CD33+CD15+ MDSCs were increased in blood from patients with disseminated/progressive lepromatous leprosy and possessed T cell-suppressive activity as compared with self-limiting tuberculoid leprosy. Mechanistically, we found ER stress played a critical role in regulating the T cell suppressive activity in these MDSCs. Furthermore, ER stress augmented IL-10 production, contributing to MDSC activity, whereas IFN-γ allowed T cells to overcome MDSC suppressive activity. These studies highlight a regulatory mechanism that links ER stress to IL-10 in mediating MDSC suppressive function in human infectious disease.

3.
Cell ; 170(5): 973-985.e10, 2017 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-28841420

RESUMO

Mycobacterium leprae causes leprosy and is unique among mycobacterial diseases in producing peripheral neuropathy. This debilitating morbidity is attributed to axon demyelination resulting from direct interaction of the M. leprae-specific phenolic glycolipid 1 (PGL-1) with myelinating glia and their subsequent infection. Here, we use transparent zebrafish larvae to visualize the earliest events of M. leprae-induced nerve damage. We find that demyelination and axonal damage are not directly initiated by M. leprae but by infected macrophages that patrol axons; demyelination occurs in areas of intimate contact. PGL-1 confers this neurotoxic response on macrophages: macrophages infected with M. marinum-expressing PGL-1 also damage axons. PGL-1 induces nitric oxide synthase in infected macrophages, and the resultant increase in reactive nitrogen species damages axons by injuring their mitochondria and inducing demyelination. Our findings implicate the response of innate macrophages to M. leprae PGL-1 in initiating nerve damage in leprosy.


Assuntos
Antígenos de Bactérias/metabolismo , Modelos Animais de Doenças , Glicolipídeos/metabolismo , Hanseníase/microbiologia , Hanseníase/patologia , Macrófagos/imunologia , Mycobacterium leprae/fisiologia , Animais , Axônios/metabolismo , Axônios/patologia , Doenças Desmielinizantes , Larva/crescimento & desenvolvimento , Hanseníase/imunologia , Mycobacterium marinum/metabolismo , Bainha de Mielina/química , Bainha de Mielina/metabolismo , Bainha de Mielina/ultraestrutura , Neuroglia/metabolismo , Neuroglia/patologia , Óxido Nítrico/metabolismo , Peixe-Zebra
4.
PLoS Pathog ; 13(7): e1006496, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28704551

RESUMO

Successful host defense against pathogens requires innate immune recognition of the correct pathogen associated molecular patterns (PAMPs) by pathogen recognition receptors (PRRs) to trigger the appropriate gene program tailored to the pathogen. While many PRR pathways contribute to the innate immune response to specific pathogens, the relative importance of each pathway for the complete transcriptional program elicited has not been examined in detail. Herein, we used RNA-sequencing with wildtype and mutant macrophages to delineate the innate immune pathways contributing to the early transcriptional response to Staphylococcus aureus, a ubiquitous microorganism that can activate a wide variety of PRRs. Unexpectedly, two PRR pathways-the Toll-like receptor (TLR) and Stimulator of Interferon Gene (STING) pathways-were identified as dominant regulators of approximately 95% of the genes that were potently induced within the first four hours of macrophage infection with live S. aureus. TLR signaling predominantly activated a pro-inflammatory program while STING signaling activated an antiviral/type I interferon response with live but not killed S. aureus. This STING response was largely dependent on the cytosolic DNA sensor cyclic guanosine-adenosine synthase (cGAS). Using a cutaneous infection model, we found that the TLR and STING pathways played opposite roles in host defense to S. aureus. TLR signaling was required for host defense, with its absence reducing interleukin (IL)-1ß production and neutrophil recruitment, resulting in increased bacterial growth. In contrast, absence of STING signaling had the opposite effect, enhancing the ability to restrict the infection. These results provide novel insights into the complex interplay of innate immune signaling pathways triggered by S. aureus and uncover opposing roles of TLR and STING in cutaneous host defense to S. aureus.


Assuntos
Citosol/imunologia , Proteínas de Membrana/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/fisiologia , Receptores Toll-Like/imunologia , Animais , Citosol/microbiologia , DNA , DNA Bacteriano/genética , DNA Bacteriano/imunologia , Feminino , Humanos , Imunidade Inata , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Nucleotidiltransferases/genética , Nucleotidiltransferases/imunologia , Transdução de Sinais , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia , Receptores Toll-Like/genética
5.
PLoS Pathog ; 12(6): e1005705, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27355424

RESUMO

Triggering antimicrobial mechanisms in macrophages infected with intracellular pathogens, such as mycobacteria, is critical to host defense against the infection. To uncover the unique and shared antimicrobial networks induced by the innate and adaptive immune systems, gene expression profiles generated by RNA sequencing (RNAseq) from human monocyte-derived macrophages (MDMs) activated with TLR2/1 ligand (TLR2/1L) or IFN-γ were analyzed. Weighed gene correlation network analysis identified modules of genes strongly correlated with TLR2/1L or IFN-γ that were linked by the "defense response" gene ontology term. The common TLR2/1L and IFN-γ inducible human macrophage host defense network contained 16 antimicrobial response genes, including S100A12, which was one of the most highly induced genes by TLR2/1L. There is limited information on the role of S100A12 in infectious disease, leading us to test the hypothesis that S100A12 contributes to host defense against mycobacterial infection in humans. We show that S100A12 is sufficient to directly kill Mycobacterium tuberculosis and Mycobacterium leprae. We also demonstrate that S100A12 is required for TLR2/1L and IFN-γ induced antimicrobial activity against M. leprae in infected macrophages. At the site of disease in leprosy, we found that S100A12 was more strongly expressed in skin lesions from tuberculoid leprosy (T-lep), the self-limiting form of the disease, compared to lepromatous leprosy (L-lep), the progressive form of the disease. These data suggest that S100A12 is part of an innate and adaptive inducible antimicrobial network that contributes to host defense against mycobacteria in infected macrophages.


Assuntos
Hanseníase/imunologia , Macrófagos/imunologia , Proteína S100A12/imunologia , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Humanos , Macrófagos/microbiologia , Infecções por Mycobacterium/imunologia , Mycobacterium leprae/imunologia , Mycobacterium tuberculosis/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Transcriptoma
6.
J Invest Dermatol ; 135(10): 2410-2417, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26030183

RESUMO

The mechanisms by which intracellular pathogens trigger immunosuppressive pathways are critical for understanding the pathogenesis of microbial infection. One pathway that inhibits host defense responses involves the induction of type I interferons and subsequently IL-10, yet the mechanism by which type I IFN induces IL-10 remains unclear. Our studies of gene expression profiles derived from leprosy skin lesions suggested a link between IL-27 and the IFN-ß induced IL-10 pathway. Here, we demonstrate that the IL-27p28 subunit is upregulated following treatment of monocytes with IFN-ß and Mycobacterium leprae, the intracellular bacterium that causes leprosy. The ability of IFN-ß and M. leprae to induce IL-10 was diminished by IL-27 knockdown. Additionally, treatment of monocytes with recombinant IL-27 was sufficient to induce the production of IL-10. Functionally, IL-27 inhibited the ability of IFN-γ to trigger antimicrobial activity against M. leprae in infected monocytes. At the site of disease, IL-27 was more strongly expressed in skin lesions of patients with progressive lepromatous leprosy, correlating and colocalizing with IFN-ß and IL-10 in macrophages. Together, these data provide evidence that in the human cutaneous immune responses to microbial infection, IL-27 contributes to the suppression of host antimicrobial responses.


Assuntos
Interferon beta/farmacologia , Interleucina-10/metabolismo , Interleucina-27/metabolismo , Hanseníase Virchowiana/tratamento farmacológico , Hanseníase Virchowiana/metabolismo , Mycobacterium leprae/metabolismo , Animais , Biomarcadores/metabolismo , Células Cultivadas , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Imunossupressores/farmacologia , Interleucina-27/farmacologia , Hanseníase Virchowiana/patologia , Camundongos , Microscopia Confocal , Modelos Animais , Monócitos/citologia , Monócitos/efeitos dos fármacos , Mycobacterium leprae/patogenicidade , Prognóstico , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real/métodos , Estudos de Amostragem , Sensibilidade e Especificidade , Transfecção
7.
J Immunol ; 194(3): 1169-77, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25548220

RESUMO

Current evidence suggests that neonatal immunity is functionally distinct from adults. Although TLR signaling through the adaptor protein, MyD88, has been shown to be critical for survival to sepsis in adults, little is known about the role of MyD88 or TRIF in neonatal sepsis. We demonstrate that TRIF(-/-) but not MyD88(-/-) neonates are highly susceptible to Escherichia coli peritonitis and bacteremia. This was associated with decreased innate immune recruitment and function. Importantly, we found that the reverse was true in adults that MyD88(-/-) but not TRIF(-/-) or wild-type adults are susceptible to E. coli peritonitis and bacteremia. In addition, we demonstrate that TRIF but not MyD88 signaling is critical for the TLR4 protective adjuvant effect we have previously demonstrated. These data suggest a differential requirement for the survival of neonates versus adults to Gram-negative infection, and that modulation of TRIF in neonates can be used to augment survival to neonatal sepsis.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Infecções por Bactérias Gram-Negativas/genética , Infecções por Bactérias Gram-Negativas/imunologia , Imunidade Inata , Sepse/genética , Sepse/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Animais Recém-Nascidos , Quimiocina CXCL10/metabolismo , Quimiocinas/biossíntese , Citocinas/biossíntese , Modelos Animais de Doenças , Suscetibilidade a Doenças/imunologia , Escherichia coli/imunologia , Feminino , Predisposição Genética para Doença , Infecções por Bactérias Gram-Negativas/metabolismo , Infecções por Bactérias Gram-Negativas/microbiologia , Infecções por Bactérias Gram-Negativas/mortalidade , Granulócitos/imunologia , Granulócitos/metabolismo , Interferon Tipo I/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/metabolismo , Fagocitose/genética , Fagocitose/imunologia , Espécies Reativas de Oxigênio/metabolismo , Sepse/metabolismo , Sepse/microbiologia , Sepse/mortalidade , Receptores Toll-Like/metabolismo
8.
J Immunol ; 192(12): 6111-9, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24829407

RESUMO

Cancer progression is associated with inflammation, increased metabolic demand, infection, cachexia, and eventually death. Myeloid-derived suppressor cells (MDSCs) commonly expand during cancer and are associated with adaptive immune suppression and inflammatory metabolite production. We propose that cancer-induced cachexia is driven at least in part by the expansion of MDSCs. MDSC expansion in 4T1 mammary carcinoma-bearing hosts is associated with induction of a hepatic acute-phase protein response and altered host energy and fat metabolism, and eventually reduced survival to polymicrobial sepsis and endotoxemia. Similar results are also seen in mice bearing a Lewis lung carcinoma and a C26 colon adenocarcinoma. However, a similar cachexia response is not seen with equivalent growth of the 66C4 subclone of 4T1, in which MDSC expansion does not occur. Importantly, reducing MDSC numbers in 4T1-bearing animals can ameliorate some of these late responses and reduce susceptibility to inflammation-induced organ injury and death. In addition, administering MDSCs from both tumor- and nontumor-bearing mice can produce an acute-phase response. Thus, we propose a previously undescribed mechanism for the development of cancer cachexia, whereby progressive MDSC expansion contributes to changes in host protein and energy metabolism and reduced resistance to infection.


Assuntos
Caquexia/imunologia , Tolerância Imunológica , Células Mieloides/imunologia , Neoplasias Experimentais/imunologia , Animais , Caquexia/etiologia , Linhagem Celular Tumoral , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/patologia , Neoplasias Experimentais/patologia
9.
J Immunol ; 190(8): 3916-27, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23509349

RESUMO

Although ectopic lymphoid tissue formation is associated with many autoimmune diseases, it is unclear whether it serves a functional role in autoimmune responses. 2,6,10,14-Tetramethylpentadecane causes chronic peritoneal inflammation and lupus-like disease with autoantibody production and ectopic lymphoid tissue (lipogranuloma) formation. A novel transplantation model was used to show that transplanted lipogranulomas retain their lymphoid structure over a prolonged period in the absence of chronic peritoneal inflammation. Recipients of transplanted lipogranulomas produced anti-U1A autoantibodies derived exclusively from the donor, despite nearly complete repopulation of the transplanted lipogranulomas by host lymphocytes. The presence of ectopic lymphoid tissue alone was insufficient, as an anti-U1A response was not generated by the host in the absence of ongoing peritoneal inflammation. Donor-derived anti-U1A autoantibodies were produced for up to 2 mo by plasma cells/plasmablasts recruited to the ectopic lymphoid tissue by CXCR4. Although CD4(+) T cells were not required for autoantibody production from the transplanted lipogranulomas, de novo generation of anti-U1A plasma cells/plasmablasts was reduced following T cell depletion. Significantly, a population of memory B cells was identified in the bone marrow and spleen that did not produce anti-U1A autoantibodies unless stimulated by LPS to undergo terminal differentiation. We conclude that 2,6,10,14-tetramethylpentadecane promotes the T cell-dependent development of class-switched, autoreactive memory B cells and plasma cells/plasmablasts. The latter home to ectopic lymphoid tissue and continue to produce autoantibodies after transplantation and in the absence of peritoneal inflammation. However, peritoneal inflammation appears necessary to generate autoreactive B cells de novo.


Assuntos
Autoanticorpos/biossíntese , Subpopulações de Linfócitos B/imunologia , Células da Medula Óssea/imunologia , Memória Imunológica , Tecido Linfoide/imunologia , Plasmócitos/imunologia , Ribonucleoproteínas Nucleares Pequenas/imunologia , Animais , Autoanticorpos/metabolismo , Subpopulações de Linfócitos B/citologia , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Coristoma/imunologia , Feminino , Granuloma/sangue , Granuloma/imunologia , Granuloma/patologia , Tecido Linfoide/citologia , Tecido Linfoide/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasmócitos/metabolismo
10.
Lab Invest ; 91(12): 1787-95, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21931299

RESUMO

Gene expression analysis can be a powerful tool in predicting patient outcomes and identifying patients who may benefit from targeted therapies. However, isolating human blood polymorphonuclear cells (PMNs) for genomic analysis has been challenging. We used a novel microfluidic technique that isolates PMNs by capturing CD66b(+) cells and compared it with dextran-Ficoll gradient isolation. We also used microfluidic isolation techniques for blood and bronchoalveolar lavage (BAL) samples of patients with acute respiratory distress syndrome (ARDS) to evaluate PMN genomic alterations secondary to pulmonary sequestration. PMNs obtained from ex vivo lipopolysaccharide (LPS)-stimulated or -unstimulated whole blood from five healthy volunteers were isolated by either dextran-Ficoll gradient, microfluidics capture, or a combination of the two techniques. Blood and BAL fluid PMNs were also isolated using microfluidics from seven hospitalized patients with ARDS. Gene expression was inferred from extracted RNA using Affymetrix U133 Plus 2.0 GeneChips. All methods of PMN isolation produced similar quantities of high-quality RNA, when adjusted for recovered cell number. Unsupervised analysis and hierarchical clustering indicated that LPS stimulation was the primary factor affecting gene expression patterns among all ex vivo samples. Patterns of gene expression from blood and BAL PMNs differed significantly from each other in the patients with ARDS. Isolation of PMNs by microfluidics can be applied to both blood and BAL specimens from critically ill, hospitalized patients. Unique genomic expression patterns are obtained from the blood and BAL fluid of critically ill patients with ARDS, and these differ significantly from genomic patterns seen after ex vivo LPS stimulation.


Assuntos
Lesão Pulmonar Aguda/patologia , Antígenos CD/análise , Líquido da Lavagem Broncoalveolar/citologia , Moléculas de Adesão Celular/análise , Neutrófilos/patologia , Síndrome do Desconforto Respiratório/patologia , Lesão Pulmonar Aguda/sangue , Estudos de Casos e Controles , Centrifugação com Gradiente de Concentração , Proteínas Ligadas por GPI/análise , Perfilação da Expressão Gênica , Humanos , Técnicas Analíticas Microfluídicas , RNA/isolamento & purificação , Síndrome do Desconforto Respiratório/sangue
11.
Lab Invest ; 91(10): 1540-50, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21808234

RESUMO

Diffuse alveolar hemorrhage is an uncommon, yet often fatal, complication of systemic lupus erythematosus (SLE). Advances in the treatment of alveolar hemorrhage have been hampered because of the heterogeneity of clinical findings and the lack of suitable animal models. A single intraperitoneal injection of pristane induces a lupus-like syndrome characterized by lupus-related autoantibodies and glomerulonephritis in non-autoimmune-prone strains of mice. In addition, C57BL/6 (B6) mice frequently develop alveolar hemorrhage within a few weeks of pristane injection. Immunopathogenesis of pristane-induced alveolar hemorrhage was investigated in the present study. Early (2-4 weeks after injection) mortality due to hemorrhage was unique to C57BL/6 and C57BL/10 strains of mice. Recruitment of the macrophages and neutrophils preceded the hemorrhage by several days, and hemorrhage started 3-7 days after pristane injection in some mice, peaked at 2 weeks (84% in B6) and then resolved by 4 weeks in a majority of mice. Alveolar hemorrhage was independent of MyD88 (myeloid differentiation factor 88), or TLR7 pathways, in contrast to autoantibody production and glomerulonephritis, and was also independent of FcγR or Fas. Rag1(-/-) mice had a reduced prevalence of alveolar hemorrhage compared with B6 (P=0.01) congenics. However, T-cell receptor-deficient mice developed alveolar hemorrhage at a rate comparable to wild-type controls, whereas B6 Igµ(-/-) mice surprisingly had a strikingly reduced prevalence (7% vs 84% in B6, P<0.0001). Reconstitution of B6 Igµ(-/-) mice with wild-type B cells increased the prevalence to 50% (P=0.028). Pristane-induced alveolar hemorrhage is a useful model to study the pathogenesis and develop new therapy for this underappreciated and often life-threatening complication of SLE.


Assuntos
Linfócitos B , Hemorragia/induzido quimicamente , Pneumopatias/induzido quimicamente , Alvéolos Pulmonares , Terpenos , Animais , Linfócitos B/patologia , Linhagem Celular , Hemorragia/patologia , Proteínas de Homeodomínio/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pneumopatias/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Especificidade da Espécie , Especificidade por Substrato
12.
J Exp Med ; 208(8): 1673-82, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21746813

RESUMO

Microbes activate pattern recognition receptors to initiate adaptive immunity. T cells affect early innate inflammatory responses to viral infection, but both activation and suppression have been demonstrated. We identify a novel role for B cells in the early innate immune response during bacterial sepsis. We demonstrate that Rag1(-/-) mice display deficient early inflammatory responses and reduced survival during sepsis. Interestingly, B cell-deficient or anti-CD20 B cell-depleted mice, but not α/ß T cell-deficient mice, display decreased inflammatory cytokine and chemokine production and reduced survival after sepsis. Both treatment of B cell-deficient mice with serum from wild-type (WT) mice and repletion of Rag1(-/-) mice with B cells improves sepsis survival, suggesting antibody-independent and antibody-dependent roles for B cells in the outcome to sepsis. During sepsis, marginal zone and follicular B cells are activated through type I interferon (IFN-I) receptor (IFN-α/ß receptor [IFNAR]), and repleting Rag1(-/-) mice with WT, but not IFNAR(-/-), B cells improves IFN-I-dependent and -independent early cytokine responses. Repleting B cell-deficient mice with the IFN-I-dependent chemokine, CXCL10 was also sufficient to improve sepsis survival. This study identifies a novel role for IFN-I-activated B cells in protective early innate immune responses during bacterial sepsis.


Assuntos
Linfócitos B/imunologia , Imunidade Inata/imunologia , Sepse/imunologia , Análise de Variância , Animais , Linfócitos B/metabolismo , Quimiocina CXCL10/imunologia , Contagem de Colônia Microbiana , Citometria de Fluxo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Camundongos , Camundongos Knockout , Receptor de Interferon alfa e beta/imunologia , Receptores de Reconhecimento de Padrão/imunologia
13.
J Immunol ; 187(2): 911-8, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21690321

RESUMO

Neutrophils are essential for successful host eradication of bacterial pathogens and for survival to polymicrobial sepsis. During inflammation, the bone marrow provides a large reserve of neutrophils that are released into the peripheral circulation where they traverse to sites of infection. Although neutrophils are essential for survival, few studies have investigated the mechanisms responsible for neutrophil mobilization from the bone marrow during polymicrobial sepsis. Using a cecal ligation and puncture model of polymicrobial sepsis, we demonstrated that neutrophil mobilization from the bone marrow is not dependent on TLR4, MyD88, TRIF, IFNARα/ß, or CXCR2 pathway signaling during sepsis. In contrast, we observed that bone marrow CXCL12 mRNA abundance and specific CXCL12 levels are sharply reduced, whereas splenic CXCR4 mRNA and cell surface expression are increased during sepsis. Blocking CXCL12 activity significantly reduced blood neutrophilia by inhibiting bone marrow release of granulocytes during sepsis. However, CXCL12 inhibition had no impact on the expansion of bone marrow neutrophil precursors and hematopoietic progenitors. Bone marrow neutrophil retention by CXCL12 blockade prevented blood neutrophilia, inhibited peritoneal neutrophil accumulation, allowed significant peritoneal bacterial invasion, and increased polymicrobial sepsis mortality. We concluded that changes in the pattern of CXCL12 signaling during sepsis are essential for neutrophil bone marrow mobilization and host survival but have little impact on bone marrow granulopoiesis.


Assuntos
Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Quimiocina CXCL12/fisiologia , Infiltração de Neutrófilos/imunologia , Sepse/imunologia , Sepse/microbiologia , Transdução de Sinais/imunologia , Doença Aguda , Animais , Células da Medula Óssea/microbiologia , Quimiocina CXCL12/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mielopoese/genética , Mielopoese/imunologia , Infiltração de Neutrófilos/genética , Sepse/mortalidade , Transdução de Sinais/genética , Análise de Sobrevida
14.
Infect Immun ; 79(7): 2746-54, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21518789

RESUMO

Previous studies have suggested that neonates rely heavily on innate immunity for their antimicrobial response to bacterial infections. However, the innate immune response by neonates to bacterial infection remains poorly characterized. Here, we show that in a murine model of neonatal polymicrobial sepsis, CXC ligand 10 (CXCL10) concentrations increase in the blood and peritoneum concordant with the peritoneal recruitment of granulocytes and macrophages. Additionally, CXC receptor 3 (CXCR3) expression on elicited peritoneal macrophages and granulocytes increases following sepsis. Blockade of CXCL10 worsens not only recruitment and phagocytic function of peritoneal granulocytes and macrophages but also survival. Deletion of CXCR3 also significantly increases mortality to a septic challenge. Finally, we demonstrate that the protective adjuvant effect of pretreatment with a Toll-like receptor 4 agonist to neonatal sepsis is dependent on an endogenous CXCL10 response and that pretreatment of neonates with CXCL10 can also significantly improve macrophage and granulocyte function and modestly improve outcome to polymicrobial sepsis. Together, these data suggest a critical role for CXCL10 signaling during neonatal sepsis.


Assuntos
Infecções Bacterianas/imunologia , Quimiocina CXCL10/imunologia , Receptores CXCR3/metabolismo , Sepse/imunologia , Transdução de Sinais , Animais , Animais Recém-Nascidos , Infecções Bacterianas/metabolismo , Quimiocina CXCL10/antagonistas & inibidores , Quimiocina CXCL10/sangue , Feminino , Citometria de Fluxo , Granulócitos/imunologia , Granulócitos/metabolismo , Imunidade Inata , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Receptores CXCR3/sangue , Receptores CXCR3/deficiência , Receptores CXCR3/genética , Sepse/metabolismo , Sepse/microbiologia , Receptor 4 Toll-Like/imunologia
15.
Mol Med ; 17(3-4): 281-92, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21085745

RESUMO

Myeloid-derived suppressor cells (MDSCs) are a heterogenous population of immature myeloid cells whose numbers dramatically increase in chronic and acute inflammatory diseases, including cancer, autoimmune disease, trauma, burns and sepsis. Studied originally in cancer, these cells are potently immunosuppressive, particularly in their ability to suppress antigen-specific CD8(+) and CD4(+) T-cell activation through multiple mechanisms, including depletion of extracellular arginine, nitrosylation of regulatory proteins, and secretion of interleukin 10, prostaglandins and other immunosuppressive mediators. However, additional properties of these cells, including increased reactive oxygen species and inflammatory cytokine production, as well as their universal expansion in nearly all inflammatory conditions, suggest that MDSCs may be more of a normal component of the inflammatory response ("emergency myelopoiesis") than simply a pathological response to a growing tumor. Recent evocative data even suggest that the expansion of MDSCs in acute inflammatory processes, such as burns and sepsis, plays a beneficial role in the host by increasing immune surveillance and innate immune responses. Although clinical efforts are currently underway to suppress MDSC numbers and function in cancer to improve antineoplastic responses, such approaches may not be desirable or beneficial in other clinical conditions in which immune surveillance and antimicrobial activities are required.


Assuntos
Ativação Linfocitária/imunologia , Células Mieloides/imunologia , Sepse/imunologia , Ferimentos e Lesões/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Humanos , Modelos Imunológicos
16.
J Immunol ; 186(1): 195-202, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21106855

RESUMO

Sepsis, the systemic inflammatory response to microbial infection, induces changes in both innate and adaptive immunity that presumably lead to increased susceptibility to secondary infections, multiorgan failure, and death. Using a model of murine polymicrobial sepsis whose severity approximates human sepsis, we examined outcomes and defined requirements for survival after secondary Pseudomonas aeruginosa pneumonia or disseminated Listeria monocytogenes infection. We demonstrate that early after sepsis neutrophil numbers and function are decreased, whereas monocyte recruitment through the CCR2/MCP-1 pathway and function are enhanced. Consequently, lethality to Pseudomonas pneumonia is increased early but not late after induction of sepsis. In contrast, lethality to listeriosis, whose eradication is dependent upon monocyte/macrophage phagocytosis, is actually decreased both early and late after sepsis. Adaptive immunity plays little role in these secondary infectious responses. This study demonstrates that sepsis promotes selective early, impaired innate immune responses, primarily in neutrophils, that lead to a pathogen-specific, increased susceptibility to secondary infections.


Assuntos
Bacteriemia/imunologia , Bacteriemia/mortalidade , Imunidade Inata , Sepse/imunologia , Sepse/mortalidade , Animais , Bacteriemia/patologia , Ceco , Modelos Animais de Doenças , Predisposição Genética para Doença , Imunidade Inata/genética , Ligadura , Listeriose/imunologia , Listeriose/mortalidade , Listeriose/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/patologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/mortalidade , Pneumonia Bacteriana/patologia , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/mortalidade , Infecções por Pseudomonas/patologia , Punções , Sepse/patologia , Fatores de Tempo
17.
Curr Protoc Immunol ; Chapter 19: Unit 19.13, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21053304

RESUMO

The cecum contains a high concentration of microbes, which are a combination of Gram-negative and Gram-positive flora. These bacteria range from anaerobic to facultative aerobic to aerobic organisms. In the procedure described in this unit, the ligation of the cecum produces a source of ischemic tissue as well as polymicrobial infection. This combination of ischemic/necrotic tissue and microbial infection distinguishes this multifactorial model from a number of other bacterial sepsis models, including but not limited to: bacteremia secondary to intravenous or intraperitoneal administration; fecal administration or intraperitoneal administration of fecal or bacterial plugs; colonic stents; and bacterial abscess formation.


Assuntos
Ceco/lesões , Ceco/microbiologia , Modelos Animais de Doenças , Sepse/etiologia , Sepse/microbiologia , Animais , Humanos , Isquemia/etiologia , Isquemia/microbiologia , Ligadura/métodos , Camundongos , Peritonite/etiologia , Peritonite/microbiologia , Punções/métodos , Ferimentos e Lesões/etiologia , Ferimentos e Lesões/microbiologia
18.
J Immunol ; 184(5): 2247-51, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20130216

RESUMO

Bone marrow (BM) hematopoietic stem and progenitor cells (HSPCs) can be activated by type I IFNs, TLR agonists, viruses, and bacteria to increase hematopoiesis. In this study, we report that endotoxin treatment in vivo induces TLR4, MyD88, and Toll/IL-1 resistance domain-containing adaptor-inducing IFN-beta (TRIF)-dependent expansion of BM HSPCs. Bacterial infection by Staphylococcus aureus or cecal ligation and puncture also induces HSPC expansion, but MyD88, TRIF, type I IFN, cytokine, PG, or oxidative stress pathways are not required for their expansion. S. aureus-induced HSPC expansion in MyD88(-/-)TRIF(-/-) mice is also normal, but is associated with BM remodeling as granulocyte stores are released peripherally. Importantly, reduction in BM cellularity alone can reproduce HSPC expansion. These data show in vivo HSPC responses to bacterial infection are complex and not absolutely dependent upon key inflammatory signaling pathways.


Assuntos
Infecções Bacterianas/imunologia , Células-Tronco Hematopoéticas/imunologia , Transdução de Sinais/imunologia , Células-Tronco/imunologia , Receptor 4 Toll-Like/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Animais , Infecções Bacterianas/etiologia , Infecções Bacterianas/microbiologia , Medula Óssea/imunologia , Medula Óssea/metabolismo , Medula Óssea/microbiologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Ceco/cirurgia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Interleucina-6/genética , Interleucina-6/imunologia , Ligadura/efeitos adversos , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/imunologia , Transdução de Sinais/efeitos dos fármacos , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Células-Tronco/citologia , Receptor 4 Toll-Like/genética
19.
J Exp Med ; 207(2): 319-26, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20071504

RESUMO

Type I interferon (IFN) alpha/beta is critical for host defense. During endotoxicosis or highly lethal bacterial infections where systemic inflammation predominates, mice deficient in IFN-alpha/beta receptor (IFNAR) display decreased systemic inflammation and improved outcome. However, human sepsis mortality often occurs during a prolonged period of immunosuppression and not from exaggerated inflammation. We used a low lethality cecal ligation and puncture (CLP) model of sepsis to determine the role of type I IFNs in host defense during sepsis. Despite increased endotoxin resistance, IFNAR(-/-) and chimeric mice lacking IFNAR in hematopoietic cells display increased mortality to CLP. This was not associated with an altered early systemic inflammatory response, except for decreased CXCL10 production. IFNAR(-/-) mice display persistently elevated peritoneal bacterial counts compared with wild-type mice, reduced peritoneal neutrophil recruitment, and recruitment of neutrophils with poor phagocytic function despite normal to enhanced adaptive immune function during sepsis. Importantly, CXCL10 treatment of IFNAR(-/-) mice improves survival and decreases peritoneal bacterial loads, and CXCL10 increases mouse and human neutrophil phagocytosis. Using a low lethality sepsis model, we identify a critical role of type I IFN-dependent CXCL10 in host defense during polymicrobial sepsis by increasing neutrophil recruitment and function.


Assuntos
Quimiocina CXCL10/imunologia , Interferon Tipo I/imunologia , Infiltração de Neutrófilos/imunologia , Sepse/imunologia , Transdução de Sinais , Imunidade Adaptativa , Animais , Ceco/patologia , Quimiocina CXCL10/metabolismo , Modelos Animais de Doenças , Hematopoese/imunologia , Humanos , Interferon Tipo I/metabolismo , Camundongos , Neutrófilos/imunologia , Fagocitose/imunologia , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/imunologia , Sepse/microbiologia , Sepse/mortalidade , Sepse/fisiopatologia
20.
J Immunol ; 182(7): 4226-36, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19299721

RESUMO

Intraperitoneal exposure of nonautoimmune mice to 2,6,10,14-tetramethylpentadecane (TMPD) causes lupus and the formation of ectopic lymphoid tissue. Although associated with humoral autoimmunity, it is not known whether Ab responses develop within ectopic lymphoid tissue or if B cells only secondarily migrate there. We show that ectopic lymphoid tissue induced by TMPD not only resembles secondary lymphoid tissue morphologically, but it also displays characteristics of germinal center reactions. Proliferating T and B lymphocytes were found within ectopic lymphoid tissue, activation-induced cytidine deaminase was expressed, and class-switched B cells were present. The presence of circular DNA intermediates, a hallmark of active class switch recombination, suggested that class switching occurs within the ectopic lymphoid tissue. Individual collections of ectopic lymphoid tissue ("lipogranulomas") from the same mouse contained different B cell repertoires, consistent with local germinal center-like reactions. Class-switched anti-RNP autoantibody-producing cells were also found in the lipogranulomas. Somatic hypermutation in the lipogranulomas was T cell-dependent, as was the production of isotype-switched anti-Sm/RNP autoantibodies. Thus, ectopic lymphoid tissue induced by TMPD recapitulates many of the functional characteristics of secondary lymphoid tissue and contains autoantibody-secreting cells, which may escape from normal censoring mechanisms in this location.


Assuntos
Tecido Adiposo/imunologia , Tecido Adiposo/patologia , Linfócitos B/imunologia , Coristoma/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Tecido Linfoide , Animais , Autoanticorpos/biossíntese , Autoantígenos/imunologia , Proliferação de Células , Coristoma/patologia , Ensaio de Imunoadsorção Enzimática , Feminino , Imunofluorescência , Centro Germinativo/imunologia , Centro Germinativo/patologia , Switching de Imunoglobulina/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulina M/sangue , Imunoglobulina M/imunologia , Imuno-Histoquímica , Imunossupressores/toxicidade , Lúpus Eritematoso Sistêmico/induzido quimicamente , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleoproteínas Nucleares Pequenas/imunologia , Hipermutação Somática de Imunoglobulina/imunologia , Linfócitos T/imunologia , Terpenos/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...