Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell ; 170(5): 973-985.e10, 2017 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-28841420

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/metabolismo , Modelos Animales de Enfermedad , Glucolípidos/metabolismo , Lepra/microbiología , Lepra/patología , Macrófagos/inmunología , Mycobacterium leprae/fisiología , Animales , Axones/metabolismo , Axones/patología , Enfermedades Desmielinizantes , Larva/crecimiento & desarrollo , Lepra/inmunología , Mycobacterium marinum/metabolismo , Vaina de Mielina/química , Vaina de Mielina/metabolismo , Vaina de Mielina/ultraestructura , Neuroglía/metabolismo , Neuroglía/patología , Óxido Nítrico/metabolismo , Pez Cebra
2.
PLoS Pathog ; 13(7): e1006496, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28704551

RESUMEN

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.


Asunto(s)
Citosol/inmunología , Proteínas de la Membrana/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/fisiología , Receptores Toll-Like/inmunología , Animales , Citosol/microbiología , ADN , ADN Bacteriano/genética , ADN Bacteriano/inmunología , Femenino , Humanos , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/inmunología , Transducción de Señal , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/inmunología , Receptores Toll-Like/genética
3.
PLoS Pathog ; 12(6): e1005705, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27355424

RESUMEN

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.


Asunto(s)
Lepra/inmunología , Macrófagos/inmunología , Proteína S100A12/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Macrófagos/microbiología , Infecciones por Mycobacterium/inmunología , Mycobacterium leprae/inmunología , Mycobacterium tuberculosis/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma
4.
J Immunol ; 194(3): 1169-77, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25548220

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Infecciones por Bacterias Gramnegativas/genética , Infecciones por Bacterias Gramnegativas/inmunología , Inmunidad Innata , Sepsis/genética , Sepsis/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Animales Recién Nacidos , Quimiocina CXCL10/metabolismo , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Escherichia coli/inmunología , Femenino , Predisposición Genética a la Enfermedad , Infecciones por Bacterias Gramnegativas/metabolismo , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Gramnegativas/mortalidad , Granulocitos/inmunología , Granulocitos/metabolismo , Interferón Tipo I/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/metabolismo , Fagocitosis/genética , Fagocitosis/inmunología , Especies Reactivas de Oxígeno/metabolismo , Sepsis/metabolismo , Sepsis/microbiología , Sepsis/mortalidad , Receptores Toll-Like/metabolismo
5.
J Immunol ; 192(12): 6111-9, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24829407

RESUMEN

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.


Asunto(s)
Caquexia/inmunología , Tolerancia Inmunológica , Células Mieloides/inmunología , Neoplasias Experimentales/inmunología , Animales , Caquexia/etiología , Línea Celular Tumoral , Femenino , Ratones , Ratones Endogámicos BALB C , Células Mieloides/patología , Neoplasias Experimentales/patología
6.
J Immunol ; 190(8): 3916-27, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23509349

RESUMEN

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.


Asunto(s)
Autoanticuerpos/biosíntesis , Subgrupos de Linfocitos B/inmunología , Células de la Médula Ósea/inmunología , Memoria Inmunológica , Tejido Linfoide/inmunología , Células Plasmáticas/inmunología , Ribonucleoproteínas Nucleares Pequeñas/inmunología , Animales , Autoanticuerpos/metabolismo , Subgrupos de Linfocitos B/citología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Coristoma/inmunología , Femenino , Granuloma/sangre , Granuloma/inmunología , Granuloma/patología , Tejido Linfoide/citología , Tejido Linfoide/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Plasmáticas/metabolismo
7.
iScience ; 27(9): 110676, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39262784

RESUMEN

Classic Ehlers-Danlos syndrome (cEDS) is a genetic disorder of the connective tissue that is characterized by mutations in genes coding type V collagen. Wound healing defects are characteristic of cEDS and no therapeutic strategies exist. Herein we describe a murine model of cEDS that phenocopies wound healing defects seen in humans. Our model features mice with conditional loss of Col5a1 in Col1a2 + fibroblasts (Col5a1CKO). This model shows that an abnormal extracellular matrix (ECM) characterized by fibrillar disarray, altered mechanical properties, and decreased collagen deposition contribute to the wound healing defect. The cEDS animals exhibit decreased expression of epidermal genes and increased inflammation. Finally, we demonstrate that inhibiting mechanosensitive integrin signaling or by injecting wild-type (WT) fibroblasts into cEDS animals enhances epidermal gene expression, decreases inflammation, and augments wound closure. These findings suggest that cell delivery and/or blocking integrin signaling are potentially therapeutic strategies to rescue wound healing defects in cEDS.

8.
J Exp Med ; 204(6): 1463-74, 2007 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-17548519

RESUMEN

Polymicrobial sepsis alters the adaptive immune response and induces T cell suppression and Th2 immune polarization. We identify a GR-1(+)CD11b(+) population whose numbers dramatically increase and remain elevated in the spleen, lymph nodes, and bone marrow during polymicrobial sepsis. Phenotypically, these cells are heterogeneous, immature, predominantly myeloid progenitors that express interleukin 10 and several other cytokines and chemokines. Splenic GR-1(+) cells effectively suppress antigen-specific CD8(+) T cell interferon (IFN) gamma production but only modestly suppress antigen-specific and nonspecific CD4(+) T cell proliferation. GR-1(+) cell depletion in vivo prevents both the sepsis-induced augmentation of Th2 cell-dependent and depression of Th1 cell-dependent antibody production. Signaling through MyD88, but not Toll-like receptor 4, TIR domain-containing adaptor-inducing IFN-beta, or the IFN-alpha/beta receptor, is required for complete GR-1(+)CD11b(+) expansion. GR-1(+)CD11b(+) cells contribute to sepsis-induced T cell suppression and preferential Th2 polarization.


Asunto(s)
Antígeno CD11b/metabolismo , Tolerancia Inmunológica/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Células Progenitoras Mieloides/inmunología , Receptores de Quimiocina/metabolismo , Sepsis/inmunología , Células Th2/inmunología , Animales , Proliferación Celular , Ensayo de Unidades Formadoras de Colonias , Citocinas/metabolismo , Citometría de Flujo , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Ratones , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/metabolismo , Transducción de Señal/inmunología
9.
J Immunol ; 186(1): 195-202, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21106855

RESUMEN

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.


Asunto(s)
Bacteriemia/inmunología , Bacteriemia/mortalidad , Inmunidad Innata , Sepsis/inmunología , Sepsis/mortalidad , Animales , Bacteriemia/patología , Ciego , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Inmunidad Innata/genética , Ligadura , Listeriosis/inmunología , Listeriosis/mortalidad , Listeriosis/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/patología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/mortalidad , Neumonía Bacteriana/patología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/mortalidad , Infecciones por Pseudomonas/patología , Punciones , Sepsis/patología , Factores de Tiempo
10.
J Immunol ; 187(2): 911-8, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21690321

RESUMEN

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.


Asunto(s)
Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Quimiocina CXCL12/fisiología , Infiltración Neutrófila/inmunología , Sepsis/inmunología , Sepsis/microbiología , Transducción de Señal/inmunología , Enfermedad Aguda , Animales , Células de la Médula Ósea/microbiología , Quimiocina CXCL12/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mielopoyesis/genética , Mielopoyesis/inmunología , Infiltración Neutrófila/genética , Sepsis/mortalidad , Transducción de Señal/genética , Análisis de Supervivencia
11.
Adv Sci (Weinh) ; 10(31): e2302248, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37750461

RESUMEN

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.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Orthomyxoviridae , Animales , Ratones , Humanos , Inmunidad Humoral , Porosidad , Anticuerpos Antivirales , Antígenos
12.
J Immunol ; 184(5): 2247-51, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20130216

RESUMEN

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.


Asunto(s)
Infecciones Bacterianas/inmunología , Células Madre Hematopoyéticas/inmunología , Transducción de Señal/inmunología , Células Madre/inmunología , Receptor Toll-Like 4/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Animales , Infecciones Bacterianas/etiología , Infecciones Bacterianas/microbiología , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/microbiología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Ciego/cirugía , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Interleucina-6/genética , Interleucina-6/inmunología , Ligadura/efectos adversos , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/inmunología , Transducción de Señal/efectos de los fármacos , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Células Madre/citología , Receptor Toll-Like 4/genética
13.
Infect Immun ; 79(7): 2746-54, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21518789

RESUMEN

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.


Asunto(s)
Infecciones Bacterianas/inmunología , Quimiocina CXCL10/inmunología , Receptores CXCR3/metabolismo , Sepsis/inmunología , Transducción de Señal , Animales , Animales Recién Nacidos , Infecciones Bacterianas/metabolismo , Quimiocina CXCL10/antagonistas & inhibidores , Quimiocina CXCL10/sangre , Femenino , Citometría de Flujo , Granulocitos/inmunología , Granulocitos/metabolismo , Inmunidad Innata , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis , Receptores CXCR3/sangre , Receptores CXCR3/deficiencia , Receptores CXCR3/genética , Sepsis/metabolismo , Sepsis/microbiología , Receptor Toll-Like 4/inmunología
14.
Lab Invest ; 91(10): 1540-50, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21808234

RESUMEN

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.


Asunto(s)
Linfocitos B , Hemorragia/inducido químicamente , Enfermedades Pulmonares/inducido químicamente , Alveolos Pulmonares , Terpenos , Animales , Linfocitos B/patología , Línea Celular , Hemorragia/patología , Proteínas de Homeodominio/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular , Pulmón/efectos de los fármacos , Pulmón/patología , Enfermedades Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología , Especificidad de la Especie , Especificidad por Sustrato
15.
Lab Invest ; 91(12): 1787-95, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21931299

RESUMEN

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.


Asunto(s)
Lesión Pulmonar Aguda/patología , Antígenos CD/análisis , Líquido del Lavado Bronquioalveolar/citología , Moléculas de Adhesión Celular/análisis , Neutrófilos/patología , Síndrome de Dificultad Respiratoria/patología , Lesión Pulmonar Aguda/sangre , Estudios de Casos y Controles , Centrifugación por Gradiente de Densidad , Proteínas Ligadas a GPI/análisis , Perfilación de la Expresión Génica , Humanos , Técnicas Analíticas Microfluídicas , ARN/aislamiento & purificación , Síndrome de Dificultad Respiratoria/sangre
16.
Mol Med ; 17(3-4): 281-92, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21085745

RESUMEN

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.


Asunto(s)
Activación de Linfocitos/inmunología , Células Mieloides/inmunología , Sepsis/inmunología , Heridas y Lesiones/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Humanos , Modelos Inmunológicos
17.
J Immunol ; 182(7): 4226-36, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19299721

RESUMEN

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.


Asunto(s)
Tejido Adiposo/inmunología , Tejido Adiposo/patología , Linfocitos B/inmunología , Coristoma/inmunología , Lupus Eritematoso Sistémico/inmunología , Tejido Linfoide , Animales , Autoanticuerpos/biosíntesis , Autoantígenos/inmunología , Proliferación Celular , Coristoma/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Centro Germinal/inmunología , Centro Germinal/patología , Cambio de Clase de Inmunoglobulina/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Inmunohistoquímica , Inmunosupresores/toxicidad , Lupus Eritematoso Sistémico/inducido químicamente , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleoproteínas Nucleares Pequeñas/inmunología , Hipermutación Somática de Inmunoglobulina/inmunología , Linfocitos T/inmunología , Terpenos/toxicidad
18.
J Immunol ; 181(5): 3259-67, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18713997

RESUMEN

Chronic inflammation promotes the formation of ectopic lymphoid tissue morphologically resembling secondary lymphoid tissues, though it is unclear whether this is a location where Ag-specific immune responses develop or merely a site of lymphocyte accumulation. Ectopic lymphoid tissue formation is associated with many humoral autoimmune diseases, including lupus induced by tetramethylpecadentane in mice. We examined whether an immune response to 4-hydroxy-3-nitrophenyl acetyl-keyhole limpet hemocyanin (NP-KLH) and NP-OVA develops within ectopic lymphoid tissue ("lipogranulomas") induced by tetramethylpecadentane in C57BL/6 mice. Following primary immunization, NP-specific B cells bearing V186.2 and related heavy chains as well as lambda-light chains accumulated within ectopic lymphoid tissue. The number of anti-NP-secreting B cells in the ectopic lymphoid tissue was greatly enhanced by immunization with NP-KLH. Remarkably, the H chain sequences isolated from individual lipogranulomas from these mice were diverse before immunization, whereas individual lipogranulomas from single immunized mice had unique oligo- or monoclonal populations of presumptive NP-specific B cells. H chain CDR sequences bore numerous replacement mutations, consistent with an Ag-driven and T cell-mediated response. In mice adoptively transferred with OT-II or DO11 T cells, there was a striking accumulation of OVA-specific T cells in lipogranulomas after s.c. immunization with NP-OVA. The selective colocalization of proliferating, Ag-specific T and B lymphocytes in lipogranulomas from tetramethylpecadentane-treated mice undergoing primary immunization implicates ectopic lymphoid tissue as a site where Ag-specific humoral immune responses can develop. This has implications for understanding the strong association of humoral autoimmunity with lymphoid neogenesis, which may be associated with deficient censoring of autoreactive cells.


Asunto(s)
Antígenos/inmunología , Linfocitos B/inmunología , Coristoma/patología , Tejido Linfoide/patología , Linfocitos T/inmunología , Animales , Formación de Anticuerpos , Autoinmunidad , Femenino , Inmunización , Cadenas Pesadas de Inmunoglobulina , Cadenas Ligeras de Inmunoglobulina , Ratones
19.
iScience ; 23(5): 101050, 2020 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-32339990

RESUMEN

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.

20.
J Invest Dermatol ; 135(10): 2410-2417, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26030183

RESUMEN

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.


Asunto(s)
Interferón beta/farmacología , Interleucina-10/metabolismo , Interleucina-27/metabolismo , Lepra Lepromatosa/tratamiento farmacológico , Lepra Lepromatosa/metabolismo , Mycobacterium leprae/metabolismo , Animales , Biomarcadores/metabolismo , Células Cultivadas , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunosupresores/farmacología , Interleucina-27/farmacología , Lepra Lepromatosa/patología , Ratones , Microscopía Confocal , Modelos Animales , Monocitos/citología , Monocitos/efectos de los fármacos , Mycobacterium leprae/patogenicidad , Pronóstico , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Muestreo , Sensibilidad y Especificidad , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA