Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Immunity ; 50(3): 591-599.e6, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30893587

RESUMEN

Immune suppression is a crucial component of immunoregulation and a subgroup of nucleotide-binding domain (NBD), leucine-rich repeat (LRR)-containing proteins (NLRs) attenuate innate immunity. How this inhibitory function is controlled is unknown. A key question is whether microbial ligands can regulate this inhibition. NLRC3 is a negative regulator that attenuates type I interferon (IFN-I) response by sequestering and attenuating stimulator of interferon genes (STING) activation. Here, we report that NLRC3 binds viral DNA and other nucleic acids through its LRR domain. DNA binding to NLRC3 increases its ATPase activity, and ATP-binding by NLRC3 diminishes its interaction with STING, thus licensing an IFN-I response. This work uncovers a mechanism wherein viral nucleic acid binding releases an inhibitory innate receptor from its target.


Asunto(s)
ADN Viral/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interferón Tipo I/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata/inmunología , Ratones , Ratones Endogámicos C57BL , Ácidos Nucleicos/metabolismo , Unión Proteica/inmunología
2.
J Immunol ; 206(9): 2015-2028, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33820855

RESUMEN

The cGAS-cyclic GMP-AMP (cGAMP)-stimulator of IFN genes (STING) pathway induces a powerful type I IFN (IFN-I) response and is a prime candidate for augmenting immunity in cancer immunotherapy and vaccines. IFN-I also has immune-regulatory functions manifested in several autoimmune diseases and is a first-line therapy for relapsing-remitting multiple sclerosis. However, it is only moderately effective and can induce adverse effects and neutralizing Abs in recipients. Targeting cGAMP in autoimmunity is unexplored and represents a challenge because of the intracellular location of its receptor, STING. We used microparticle (MP)-encapsulated cGAMP to increase cellular delivery, achieve dose sparing, and reduce potential toxicity. In the C57BL/6 experimental allergic encephalomyelitis (EAE) model, cGAMP encapsulated in MPs (cGAMP MPs) administered therapeutically protected mice from EAE in a STING-dependent fashion, whereas soluble cGAMP was ineffective. Protection was also observed in a relapsing-remitting model. Importantly, cGAMP MPs protected against EAE at the peak of disease and were more effective than rIFN-ß. Mechanistically, cGAMP MPs showed both IFN-I-dependent and -independent immunosuppressive effects. Furthermore, it induced the immunosuppressive cytokine IL-27 without requiring IFN-I. This augmented IL-10 expression through activated ERK and CREB. IL-27 and subsequent IL-10 were the most important cytokines to mitigate autoreactivity. Critically, cGAMP MPs promoted IFN-I as well as the immunoregulatory cytokines IL-27 and IL-10 in PBMCs from relapsing-remitting multiple sclerosis patients. Collectively, this study reveals a previously unappreciated immune-regulatory effect of cGAMP that can be harnessed to restrain T cell autoreactivity.


Asunto(s)
Micropartículas Derivadas de Células/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Interferón Tipo I/inmunología , Proteínas de la Membrana/inmunología , Nucleótidos Cíclicos/inmunología , Transducción de Señal/inmunología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Micropartículas Derivadas de Células/metabolismo , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Humanos , Interferón Tipo I/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Nucleótidos Cíclicos/administración & dosificación , Nucleótidos Cíclicos/metabolismo , Transducción de Señal/efectos de los fármacos
3.
Infect Immun ; 90(4): e0007022, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35311578

RESUMEN

Persistent infections generally involve a complex balance between protective immunity and immunopathology. We used a murine model to investigate the role of inflammatory monocytes in immunity and host defense against persistent salmonellosis. Mice exhibit increased susceptibility to persistent infection when inflammatory monocytes cannot be recruited into tissues or when they are depleted at specific stages of persistent infection. Inflammatory monocytes contribute to the pathology of persistent salmonellosis and cluster with other cells in pathogen-containing granulomas. Depletion of inflammatory monocytes during the chronic phase of persistent salmonellosis causes regression of already established granulomas with resultant pathogen growth and spread in tissues. Thus, inflammatory monocytes promote granuloma-mediated control of persistent salmonellosis and may be key to uncovering new therapies for granulomatous diseases.


Asunto(s)
Monocitos , Infecciones por Salmonella , Animales , Granuloma , Ratones , Receptores CCR2
4.
PLoS Pathog ; 15(7): e1007847, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31306468

RESUMEN

Salmonella exploit host-derived nitrate for growth in the lumen of the inflamed intestine. The generation of host-derived nitrate is dependent on Nos2, which encodes inducible nitric oxide synthase (iNOS), an enzyme that catalyzes nitric oxide (NO) production. However, the cellular sources of iNOS and, therefore, NO-derived nitrate used by Salmonella for growth in the lumen of the inflamed intestine remain unidentified. Here, we show that iNOS-producing inflammatory monocytes infiltrate ceca of mice infected with Salmonella. In addition, we show that inactivation of type-three secretion system (T3SS)-1 and T3SS-2 renders Salmonella unable to induce CC- chemokine receptor-2- and CC-chemokine ligand-2-dependent inflammatory monocyte recruitment. Furthermore, we show that the severity of the pathology of Salmonella- induced colitis as well as the nitrate-dependent growth of Salmonella in the lumen of the inflamed intestine are reduced in mice that lack Ccr2 and, therefore, inflammatory monocytes in the tissues. Thus, inflammatory monocytes provide a niche for Salmonella expansion in the lumen of the inflamed intestine.


Asunto(s)
Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Monocitos/metabolismo , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidad , Animales , Quimiocina CCL2/deficiencia , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Femenino , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/fisiología , Humanos , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Monocitos/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores CCR2/deficiencia , Receptores CCR2/genética , Receptores CCR2/metabolismo , Salmonelosis Animal/metabolismo , Salmonelosis Animal/microbiología , Salmonelosis Animal/patología , Salmonella typhimurium/genética , Sistemas de Secreción Tipo III/metabolismo
5.
PLoS Pathog ; 11(10): e1005167, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26468944

RESUMEN

During Yersinia pseudotuberculosis infection of C57BL/6 mice, an exceptionally large CD8+ T cell response to a protective epitope in the type III secretion system effector YopE is produced. At the peak of the response, up to 50% of splenic CD8+ T cells recognize the epitope YopE69-77. The features of the interaction between pathogen and host that result in this large CD8+ T cell response are unknown. Here, we used Y. pseudotuberculosis strains defective for production, secretion and/or translocation of YopE to infect wild-type or mutant mice deficient in specific dendritic cells (DCs). Bacterial colonization of organs and translocation of YopE into spleen cells was measured, and flow cytometry and tetramer staining were used to characterize the cellular immune response. We show that the splenic YopE69-77-specific CD8+ T cells generated during the large response are polyclonal and are produced by a "translocation-dependent" pathway that requires injection of YopE into host cell cytosol. Additionally, a smaller YopE69-77-specific CD8+ T cell response (~10% of the large expansion) can be generated in a "translocation-independent" pathway in which CD8α+ DCs cross present secreted YopE. CCR2-expressing inflammatory DCs were required for the large YopE69-77-specific CD8+ T cell expansion because this response was significantly reduced in Ccr2-/- mice, YopE was translocated into inflammatory DCs in vivo, inflammatory DCs purified from infected spleens activated YopE69-77-specific CD8+ T cells ex vivo and promoted the expansion of YopE69-77-specific CD8+ T cells in infected Ccr2-/- mice after adoptive transfer. A requirement for inflammatory DCs in producing a protective CD8+ T cell response to a bacterial antigen has not previously been demonstrated. Therefore, the production of YopE69-77-specific CD8+ T cells by inflammatory DCs that are injected with YopE during Y. pseudotuberculosis infection represents a novel mechanism for generating a massive and protective adaptive immune response.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Sistemas de Secreción Tipo III/inmunología , Infecciones por Yersinia pseudotuberculosis/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Femenino , Citometría de Flujo , Immunoblotting , Inflamación/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Transporte de Proteínas/inmunología , Receptores CCR2/inmunología , Factores de Virulencia/inmunología , Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/patogenicidad
6.
Infect Immun ; 82(6): 2606-14, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24711563

RESUMEN

Immature myeloid cells in bone marrow are a heterogeneous population of cells that, under normal conditions, provide tissues with protective cell types such as granulocytes and macrophages. Under certain pathological conditions, myeloid cell homeostasis is altered and immature forms of these cells appear in tissues. Murine immature myeloid cells that express CD11b and Ly6C or Ly6G (two isoforms of Gr-1) have been associated with immunosuppression in cancer (in the form of myeloid-derived suppressor cells) and, more recently, infection. Here, we found that CD11b(+) Ly6C(hi) Ly6G(-) and CD11b(+) Ly6C(int) Ly6G(+) cells accumulated and persisted in tissues of mice infected with Salmonella enterica serovar Typhimurium (S. Typhimurium). Recruitment of CD11b(+) Ly6C(hi) Ly6G(-) but not CD11b(+) Ly6C(int) Ly6G(+) cells from bone marrow into infected tissues depended on chemokine receptor CCR2. The CD11b(+) Ly6C(hi) Ly6G(-) cells exhibited a mononuclear morphology, whereas the CD11b(+) Ly6C(int) Ly6G(+) cells exhibited a polymorphonuclear or band-shaped nuclear morphology. The CD11b(+) Ly6C(hi) Ly6G(-) cells differentiated into macrophage-like cells following ex vivo culture and could present antigen to T cells in vitro. However, significant proliferation of T cells was observed only when the ability of the CD11b(+) Ly6C(hi) Ly6G(-) cells to produce nitric oxide was blocked. CD11b(+) Ly6C(hi) Ly6G(-) cells recruited in response to S. Typhimurium infection could also present antigen to T cells in vivo, but increasing their numbers by adoptive transfer did not cause a corresponding increase in T cell response. Thus, CD11b(+) Ly6C(hi) Ly6G(-) immature myeloid cells recruited in response to S. Typhimurium infection exhibit protective and immunosuppressive properties that may influence the outcome of infection.


Asunto(s)
Antígenos Ly/inmunología , Antígeno CD11b/inmunología , Células Mieloides/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Análisis de Varianza , Animales , Diferenciación Celular/fisiología , Proliferación Celular , Inmunidad Innata/fisiología , Terapia de Inmunosupresión , Hígado/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores CCR2/fisiología , Bazo/citología , Linfocitos T/inmunología
7.
Infect Immun ; 80(7): 2371-81, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22526678

RESUMEN

Recent studies have linked accumulation of the Gr-1⁺ CD11b⁺ cell phenotype with functional immunosuppression in diverse pathological conditions, including bacterial and parasitic infections and cancer. Gr-1⁺ CD11b⁺ cells were the largest population of cells present in the spleens of mice infected with sublethal doses of the Francisella tularensis live vaccine strain (LVS). In contrast, the number of T cells present in the spleens of these mice did not increase during early infection. There was a significant delay in the kinetics of accumulation of Gr-1⁺ CD11b⁺ cells in the spleens of B-cell-deficient mice, indicating that B cells play a role in recruitment and maintenance of this population in the spleens of mice infected with F. tularensis. The splenic Gr-1⁺ CD11b⁺ cells in tularemia were a heterogeneous population that could be further subdivided into monocytic (mononuclear) and granulocytic (polymorphonuclear) cells using the Ly6C and Ly6G markers and differentiated into antigen-presenting cells following ex vivo culture. Monocytic, CD11b⁺ Ly6C(hi) Ly6G⁻ cells but not granulocytic, CD11b⁺ Ly6C(int) Ly6G⁺ cells purified from the spleens of mice infected with F. tularensis suppressed polyclonal T-cell proliferation via a nitric oxide-dependent pathway. Although the monocytic, CD11b⁺ Ly6C(hi) Ly6G⁻ cells were able to suppress the proliferation of T cells, the large presence of Gr-1⁺ CD11b⁺ cells in mice that survived F. tularensis infection also suggests a potential role for these cells in the protective host response to tularemia.


Asunto(s)
Francisella tularensis/patogenicidad , Células Mieloides/citología , Células Mieloides/fisiología , Bazo/inmunología , Bazo/patología , Tularemia/inmunología , Tularemia/patología , Animales , Linfocitos B/inmunología , Antígeno CD11b/análisis , Modelos Animales de Enfermedad , Femenino , Inmunofenotipificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Receptores de Quimiocina/análisis , Linfocitos T/inmunología
8.
Nat Commun ; 11(1): 2193, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32366851

RESUMEN

Innate immunity to nucleic acids forms the backbone for anti-viral immunity and several inflammatory diseases. Upon sensing cytosolic viral RNA, retinoic acid-inducible gene-I-like receptors (RLRs) interact with the mitochondrial antiviral signaling protein (MAVS) and activate TANK-binding kinase 1 (TBK1) to induce type I interferon (IFN-I). TRAF3-interacting protein 3 (TRAF3IP3, T3JAM) is essential for T and B cell development. It is also well-expressed by myeloid cells, where its role is unknown. Here we report that TRAF3IP3 suppresses cytosolic poly(I:C), 5'ppp-dsRNA, and vesicular stomatitis virus (VSV) triggers IFN-I expression in overexpression systems and Traf3ip3-/- primary myeloid cells. The mechanism of action is through the interaction of TRAF3IP3 with endogenous TRAF3 and TBK1. This leads to the degradative K48 ubiquitination of TBK1 via its K372 residue in a DTX4-dependent fashion. Mice with myeloid-specific gene deletion of Traf3ip3 have increased RNA virus-triggered IFN-I production and reduced susceptibility to virus. These results identify a function of TRAF3IP3 in the regulation of the host response to cytosolic viral RNA in myeloid cells.


Asunto(s)
Proteínas Portadoras/genética , Regulación de la Expresión Génica , Interferón Tipo I/genética , Proteínas de la Membrana/genética , Células Mieloides/metabolismo , Proteínas Serina-Treonina Quinasas/genética , ARN Viral/genética , Animales , Proteínas Portadoras/metabolismo , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Citosol/metabolismo , Citosol/virología , Células HEK293 , Células HeLa , Humanos , Interferón Tipo I/metabolismo , Células Jurkat , Lisina/genética , Lisina/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/virología , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Viral/metabolismo , Células THP-1 , Ubiquitinación , Células Vero , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/fisiología
9.
Science ; 370(6516)2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-33122357

RESUMEN

Ionizing radiation causes acute radiation syndrome, which leads to hematopoietic, gastrointestinal, and cerebrovascular injuries. We investigated a population of mice that recovered from high-dose radiation to live normal life spans. These "elite-survivors" harbored distinct gut microbiota that developed after radiation and protected against radiation-induced damage and death in both germ-free and conventionally housed recipients. Elevated abundances of members of the bacterial taxa Lachnospiraceae and Enterococcaceae were associated with postradiation restoration of hematopoiesis and gastrointestinal repair. These bacteria were also found to be more abundant in leukemia patients undergoing radiotherapy, who also displayed milder gastrointestinal dysfunction. In our study in mice, metabolomics revealed increased fecal concentrations of microbially derived propionate and tryptophan metabolites in elite-survivors. The administration of these metabolites caused long-term radioprotection, mitigation of hematopoietic and gastrointestinal syndromes, and a reduction in proinflammatory responses.


Asunto(s)
Síndrome de Radiación Aguda/microbiología , Clostridiales/metabolismo , Enterococcaceae/metabolismo , Ácidos Grasos Volátiles/metabolismo , Microbioma Gastrointestinal , Protección Radiológica , Triptófano/metabolismo , Síndrome de Radiación Aguda/prevención & control , Síndrome de Radiación Aguda/terapia , Animales , Ácidos Grasos Volátiles/uso terapéutico , Humanos , Metabolómica , Ratones , Ratones Endogámicos C57BL , Sobrevivientes
10.
J Exp Med ; 216(12): 2838-2853, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31558613

RESUMEN

The role of lipids in inflammasome activation remains underappreciated. The phospholipid, platelet-activating factor (PAF), exerts multiple physiological functions by binding to a G protein-coupled seven-transmembrane receptor (PAFR). PAF is associated with a number of inflammatory disorders, yet the molecular mechanism underlying its proinflammatory function remains to be fully elucidated. We show that multiple PAF isoforms and PAF-like lipids can activate the inflammasome, resulting in IL-1ß and IL-18 maturation. This is dependent on NLRP3, ASC, caspase-1, and NEK7, but not on NLRC4, NLRP1, NLRP6, AIM2, caspase-11, or GSDMD. Inflammasome activation by PAF also requires potassium efflux and calcium influx but not lysosomal cathepsin or mitochondrial reactive oxygen species. PAF exacerbates peritonitis partly through inflammasome activation, but PAFR is dispensable for PAF-induced inflammasome activation in vivo or in vitro. These findings reveal that PAF represents a damage-associated signal that activates the canonical inflammasome independently of PAFR and provides an explanation for the ineffectiveness of PAFR antagonist in blocking PAF-mediated inflammation in the clinic.


Asunto(s)
Inflamasomas/metabolismo , Quinasas Relacionadas con NIMA/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factor de Activación Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Calcio/metabolismo , Caspasa 1/metabolismo , Furanos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos , Humanos , Indenos , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteínas de Unión a Fosfato/metabolismo , Potasio/metabolismo , Sulfonamidas/farmacología , Sulfonas
11.
Cell Host Microbe ; 25(4): 602-616.e7, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30902577

RESUMEN

Establishing the balance between positive and negative innate immune mechanisms is crucial for maintaining homeostasis. Here we uncover the regulatory crosstalk between two previously unlinked innate immune receptor families: RIG-I, an anti-viral cytosolic receptor activated type I interferon production, and NLR (nucleotide-binding domain, leucine repeat domain-containing protein). We show that NLRP12 dampens RIG-I-mediated immune signaling against RNA viruses by controlling RIG-I's association with its adaptor MAVS. The nucleotide-binding domain of NLRP12 interacts with the ubiquitin ligase TRIM25 to prevent TRIM25-mediated, Lys63-linked ubiquitination and activation of RIG-I. NLRP12 also enhances RNF125-mediated, Lys48-linked degradative ubiquitination of RIG-I. Vesicular stomatitis virus (VSV) infection downregulates NLRP12 expression to allow RIG-I activation. Myeloid-cell-specific Nlrp12-deficient mice display a heightened interferon and TNF response and are more resistant to VSV infection. These results indicate that NLRP12 functions as a checkpoint for anti-viral RIG-I activation.


Asunto(s)
Proteína 58 DEAD Box/inmunología , Proteínas de Unión al ADN/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Infecciones por Virus ARN/inmunología , Virus ARN/fisiología , Factores de Transcripción/inmunología , Animales , Proteína 58 DEAD Box/genética , Proteínas de Unión al ADN/genética , Femenino , Humanos , Interferones/genética , Interferones/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Infecciones por Virus ARN/genética , Infecciones por Virus ARN/virología , Virus ARN/genética , Factores de Transcripción/genética , Ubiquitinación
12.
Cell Host Microbe ; 24(3): 364-378.e6, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30212649

RESUMEN

In addition to high-fat diet (HFD) and inactivity, inflammation and microbiota composition contribute to obesity. Inhibitory immune receptors, such as NLRP12, dampen inflammation and are important for resolving inflammation, but their role in obesity is unknown. We show that obesity in humans correlates with reduced expression of adipose tissue NLRP12. Similarly, Nlrp12-/- mice show increased weight gain, adipose deposition, blood glucose, NF-κB/MAPK activation, and M1-macrophage polarization. Additionally, NLRP12 is required to mitigate HFD-induced inflammasome activation. Co-housing with wild-type animals, antibiotic treatment, or germ-free condition was sufficient to restrain inflammation, obesity, and insulin tolerance in Nlrp12-/- mice, implicating the microbiota. HFD-fed Nlrp12-/- mice display dysbiosis marked by increased obesity-associated Erysipelotrichaceae, but reduced Lachnospiraceae family and the associated enzymes required for short-chain fatty acid (SCFA) synthesis. Lachnospiraceae or SCFA administration attenuates obesity, inflammation, and dysbiosis. These findings reveal that Nlrp12 reduces HFD-induced obesity by maintaining beneficial microbiota.


Asunto(s)
Microbioma Gastrointestinal , Péptidos y Proteínas de Señalización Intracelular/inmunología , Obesidad/inmunología , Obesidad/microbiología , Tejido Adiposo/inmunología , Adulto , Anciano , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Femenino , Homeostasis , Humanos , Inmunidad Innata , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Obesidad/genética , Obesidad/metabolismo
13.
PLoS One ; 9(4): e92807, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24717768

RESUMEN

Pancreatitis, a known risk factor for the development of pancreatic ductal adenocarcinoma, is a serious, widespread medical condition usually caused by alcohol abuse or gallstone-mediated ductal obstruction. However, many cases of pancreatitis are of an unknown etiology. Pancreatitis has been linked to bacterial infection, but causality has yet to be established. Here, we found that persistent infection of mice with the bacterial pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) was sufficient to induce pancreatitis reminiscent of the human disease. Specifically, we found that pancreatitis induced by persistent S. Typhimurium infection was characterized by a loss of pancreatic acinar cells, acinar-to-ductal metaplasia, fibrosis and accumulation of inflammatory cells, including CD11b+ F4/80+, CD11b+ Ly6Cint Ly6G+ and CD11b+ Ly6Chi Ly6G- cells. Furthermore, we found that S. Typhimurium colonized and persisted in the pancreas, associated with pancreatic acinar cells in vivo, and could invade cultured pancreatic acinar cells in vitro. Thus, persistent infection of mice with S. Typhimurium may serve as a useful model for the study of pancreatitis as it relates to bacterial infection. Increased knowledge of how pathogenic bacteria can cause pancreatitis will provide a more integrated picture of the etiology of the disease and could lead to the development of new therapeutic approaches for treatment and prevention of pancreatitis and pancreatic ductal adenocarcinoma.


Asunto(s)
Pancreatitis/etiología , Salmonelosis Animal/complicaciones , Células Acinares/microbiología , Células Acinares/patología , Animales , Modelos Animales de Enfermedad , Humanos , Lipopolisacáridos , Ratones Endogámicos C57BL , Páncreas/microbiología , Páncreas/patología , Pancreatitis/patología , Salmonelosis Animal/patología , Salmonella typhimurium/fisiología
14.
Cell Host Microbe ; 12(6): 791-8, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23245323

RESUMEN

Salmonella enterica serovar Typhimurium avoids clearance by the host immune system by suppressing T cell responses; however, the mechanisms that mediate this immunosuppression remain unknown. We show that S. Typhimurium inhibit T cell responses by producing L-Asparaginase II, which catalyzes the hydrolysis of L-asparagine to aspartic acid and ammonia. L-Asparaginase II is necessary and sufficient to suppress T cell blastogenesis, cytokine production, and proliferation and to downmodulate expression of the T cell receptor. Furthermore, S. Typhimurium-induced inhibition of T cells in vitro is prevented upon addition of L-asparagine. S. Typhimurium lacking the L-Asparaginase II gene (STM3106) are unable to inhibit T cell responses and exhibit attenuated virulence in vivo. L-Asparaginases are used to treat acute lymphoblastic leukemia through mechanisms that likely involve amino acid starvation of leukemic cells, and these findings indicate that pathogens similarly use L-asparagine deprivation to limit T cell responses.


Asunto(s)
Asparaginasa/metabolismo , Evasión Inmune , Salmonella typhimurium/enzimología , Salmonella typhimurium/patogenicidad , Linfocitos T/inmunología , Factores de Virulencia/metabolismo , Amoníaco/metabolismo , Animales , Asparaginasa/genética , Asparagina/metabolismo , Ácido Aspártico/metabolismo , Proliferación Celular , Citocinas/metabolismo , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL , Salmonella typhimurium/inmunología , Virulencia , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda