Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Nat Immunol ; 24(6): 1036-1048, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37106040

RESUMEN

Allergic diseases are a major global health issue. Interleukin (IL)-9-producing helper T (TH9) cells promote allergic inflammation, yet TH9 cell effector functions are incompletely understood because their lineage instability makes them challenging to study. Here we found that resting TH9 cells produced IL-9 independently of T cell receptor (TCR) restimulation, due to STAT5- and STAT6-dependent bystander activation. This mechanism was seen in circulating cells from allergic patients and was restricted to recently activated cells. STAT5-dependent Il9/IL9 regulatory elements underwent remodeling over time, inactivating the locus. A broader 'allergic TH9' transcriptomic and epigenomic program was also unstable. In vivo, TH9 cells induced airway inflammation via TCR-independent, STAT-dependent mechanisms. In allergic patients, TH9 cell expansion was associated with responsiveness to JAK inhibitors. These findings suggest that TH9 cell instability is a negative checkpoint on bystander activation that breaks down in allergy and that JAK inhibitors should be considered for allergic patients with TH9 cell expansion.


Asunto(s)
Hipersensibilidad , Inhibidores de las Cinasas Janus , Humanos , Interleucina-9/genética , Linfocitos T Colaboradores-Inductores , Factor de Transcripción STAT5/genética , Cromatina/genética , Inflamación , Hipersensibilidad/genética , Diferenciación Celular , Factor de Transcripción STAT6
3.
Proc Natl Acad Sci U S A ; 120(40): e2311557120, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37748059

RESUMEN

Plasmodium parasites cause malaria with disease outcomes ranging from mild illness to deadly complications such as severe malarial anemia (SMA), pulmonary edema, acute renal failure, and cerebral malaria. In young children, SMA often requires blood transfusion and is a major cause of hospitalization. Malaria parasite infection leads to the destruction of infected and noninfected erythrocytes as well as dyserythropoiesis; however, the mechanism of dyserythropoiesis accompanied by splenomegaly is not completely understood. Using Plasmodium yoelii yoelii 17XNL as a model, we show that both a defect in erythroblastic island (EBI) macrophages in supporting red blood cell (RBC) maturation and the destruction of reticulocytes/RBCs by the parasites contribute to SMA and splenomegaly. After malaria parasite infection, the destruction of both infected and noninfected RBCs stimulates extramedullary erythropoiesis in mice. The continuous decline of RBCs stimulates active erythropoiesis and drives the expansion of EBIs in the spleen, contributing to splenomegaly. Phagocytosis of malaria parasites by macrophages in the bone marrow and spleen may alter their functional properties and abilities to support erythropoiesis, including reduced expression of the adherence molecule CD169 and inability to support erythroblast differentiation, particularly RBC maturation in vitro and in vivo. Therefore, macrophage dysfunction is a key mechanism contributing to SMA. Mitigating and/or alleviating the inhibition of RBC maturation may provide a treatment strategy for SMA.


Asunto(s)
Anemia , Malaria Cerebral , Plasmodium yoelii , Niño , Humanos , Animales , Ratones , Preescolar , Eritropoyesis , Esplenomegalia , Eritrocitos , Macrófagos
4.
Immunity ; 44(6): 1365-78, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27261276

RESUMEN

Receptor CD300b is implicated in regulating the immune response to bacterial infection by an unknown mechanism. Here, we identified CD300b as a lipopolysaccharide (LPS)-binding receptor and determined the mechanism underlying CD300b augmentation of septic shock. In vivo depletion and adoptive transfer studies identified CD300b-expressing macrophages as the key cell type augmenting sepsis. We showed that CD300b, and its adaptor DAP12, associated with Toll-like receptor 4 (TLR4) upon LPS binding, thereby enhancing TLR4-adaptor MyD88- and TRIF-dependent signaling that resulted in an elevated pro-inflammatory cytokine storm. LPS engagement of the CD300b-TLR4 complex led to the recruitment and activation of spleen tyrosine kinase (Syk) and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K). This resulted in an inhibition of the ERK1/2 protein kinase- and NF-κB transcription factor-mediated signaling pathways, which subsequently led to a reduced interleukin-10 (IL-10) production. Collectively, our data describe a mechanism of TLR4 signaling regulated by CD300b in myeloid cells in response to LPS.


Asunto(s)
Interleucina-10/metabolismo , Macrófagos/inmunología , Peritonitis/inmunología , Receptores Inmunológicos/metabolismo , Sepsis/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Células HEK293 , Humanos , Interleucina-10/genética , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Receptores Inmunológicos/genética , Transducción de Señal , Quinasa Syk/metabolismo , Receptor Toll-Like 4/metabolismo
5.
Proc Natl Acad Sci U S A ; 117(32): 19465-19474, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32709745

RESUMEN

Infection by malaria parasites triggers dynamic immune responses leading to diverse symptoms and pathologies; however, the molecular mechanisms responsible for these reactions are largely unknown. We performed Trans-species Expression Quantitative Trait Locus analysis to identify a large number of host genes that respond to malaria parasite infections. Here we functionally characterize one of the host genes called receptor transporter protein 4 (RTP4) in responses to malaria parasite and virus infections. RTP4 is induced by type I IFN (IFN-I) and binds to the TANK-binding kinase (TBK1) complex where it negatively regulates TBK1 signaling by interfering with expression and phosphorylation of both TBK1 and IFN regulatory factor 3. Rtp4-/- mice were generated and infected with malaria parasite Plasmodiun berghei ANKA. Significantly higher levels of IFN-I response in microglia, lower parasitemia, fewer neurologic symptoms, and better survival rates were observed in Rtp4-/- than in wild-type mice. Similarly, RTP4 deficiency significantly reduced West Nile virus titers in the brain, but not in the heart and the spleen, of infected mice, suggesting a specific role for RTP4 in brain infection and pathology. This study reveals functions of RTP4 in IFN-I response and a potential target for therapy in diseases with neuropathology.


Asunto(s)
Encéfalo/patología , Interferón Tipo I/metabolismo , Malaria Cerebral/patología , Chaperonas Moleculares/metabolismo , Animales , Encéfalo/parasitología , Encéfalo/virología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Factor 3 Regulador del Interferón , Malaria Cerebral/metabolismo , Malaria Cerebral/parasitología , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Chaperonas Moleculares/genética , Fosforilación , Plasmodium berghei/fisiología , Plasmodium yoelii/fisiología , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Fiebre del Nilo Occidental/metabolismo , Fiebre del Nilo Occidental/patología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/fisiología
6.
Proc Natl Acad Sci U S A ; 117(28): 16567-16578, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32606244

RESUMEN

Malaria infection induces complex and diverse immune responses. To elucidate the mechanisms underlying host-parasite interaction, we performed a genetic screen during early (24 h) Plasmodium yoelii infection in mice and identified a large number of interacting host and parasite genes/loci after transspecies expression quantitative trait locus (Ts-eQTL) analysis. We next investigated a host E3 ubiquitin ligase gene (March1) that was clustered with interferon (IFN)-stimulated genes (ISGs) based on the similarity of the genome-wide pattern of logarithm of the odds (LOD) scores (GPLS). March1 inhibits MAVS/STING/TRIF-induced type I IFN (IFN-I) signaling in vitro and in vivo. However, in malaria-infected hosts, deficiency of March1 reduces IFN-I production by activating inhibitors such as SOCS1, USP18, and TRIM24 and by altering immune cell populations. March1 deficiency increases CD86+DC (dendritic cell) populations and levels of IFN-γ and interleukin 10 (IL-10) at day 4 post infection, leading to improved host survival. T cell depletion reduces IFN-γ level and reverse the protective effects of March1 deficiency, which can also be achieved by antibody neutralization of IFN-γ. This study reveals functions of MARCH1 (membrane-associated ring-CH-type finger 1) in innate immune responses and provides potential avenues for activating antimalaria immunity and enhancing vaccine efficacy.


Asunto(s)
Malaria/inmunología , Plasmodium yoelii/fisiología , Linfocitos T/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Parásitos , Humanos , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Malaria/enzimología , Malaria/genética , Malaria/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium yoelii/inmunología , Ubiquitina-Proteína Ligasas/genética
7.
Proc Natl Acad Sci U S A ; 116(19): 9511-9520, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31000603

RESUMEN

The IRF and Ets families of transcription factors regulate the expression of a range of genes involved in immune cell development and function. However, the understanding of the molecular mechanisms of each family member has been limited due to their redundancy and broad effects on multiple lineages of cells. Here, we report that double deletion of floxed Irf8 and Spi1 (encoding PU.1) by Mb1-Cre (designated DKO mice) in the B cell lineage resulted in severe defects in the development of follicular and germinal center (GC) B cells. Class-switch recombination and antibody affinity maturation were also compromised in DKO mice. RNA-seq (sequencing) and ChIP-seq analyses revealed distinct IRF8 and PU.1 target genes in follicular and activated B cells. DKO B cells had diminished expression of target genes vital for maintaining follicular B cell identity and GC development. Moreover, our findings reveal that expression of B-cell lymphoma protein 6 (BCL6), which is critical for development of germinal center B cells, is dependent on IRF8 and PU.1 in vivo, providing a mechanism for the critical role for IRF8 and PU.1 in the development of GC B cells.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Factores Reguladores del Interferón/inmunología , Proteínas Proto-Oncogénicas c-bcl-6/inmunología , Proteínas Proto-Oncogénicas/inmunología , Transactivadores/inmunología , Animales , Linfocitos B/citología , Centro Germinal/citología , Cambio de Clase de Inmunoglobulina/inmunología , Factores Reguladores del Interferón/genética , Activación de Linfocitos/genética , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-6/genética , Transactivadores/genética
8.
J Immunol ; 202(2): 428-440, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30559322

RESUMEN

Engagement of the BCR with Ags triggers signaling pathways for commitment of B lymphocyte responses that can be regulated, in part, by reactive oxygen species. To investigate the functional relevance of reactive oxygen species produced in primary B cells, we focused on the role of the hydrogen peroxide generator Duox1 in stimulated splenic B cells under the influence of the TH2 cytokine IL-4. We found that H2O2 production in wild type (WT) and Nox2-deficient CD19+ B cells was boosted concomitantly with enhanced expression of Duox1 following costimulation with BCR agonists together with IL-4, whereas stimulated Duox1-/- cells showed attenuated H2O2 release. We examined whether Duox1-derived H2O2 contributes to proliferative activity and Ig isotype production in CD19+ cells upon BCR stimulation. Duox1-/- CD19+ B cells showed normal responses of Ig production but a higher rate of proliferation than WT or Nox2-deficient cells. Furthermore, we demonstrated that the H2O2 scavenger catalase mimics the effect of Duox1 deficiency by enhancing proliferation of WT CD19+ B cells in vitro. Results from immunized mice reflected the in vitro observations: T cell-independent Ag induced increased B cell expansion in germinal centers from Duox1-/- mice relative to WT and Nox2-/- mice, whereas immunization with T cell-dependent or -independent Ag elicited normal Ig isotype secretion in the Duox1 mutant mice. These observations, obtained both by in vitro and in vivo approaches, strongly suggest that Duox1-derived hydrogen peroxide negatively regulates proliferative activity but not Ig isotype production in primary splenic CD19+ B cells.


Asunto(s)
Linfocitos B/inmunología , Oxidasas Duales/metabolismo , Centro Germinal/inmunología , Peróxido de Hidrógeno/metabolismo , Interleucina-4/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Animales , Antígenos CD19/metabolismo , Proliferación Celular , Células Cultivadas , Oxidasas Duales/genética , Cambio de Clase de Inmunoglobulina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Regulación hacia Arriba
9.
J Autoimmun ; 102: 50-64, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31080014

RESUMEN

Accumulating evidence suggests granulocyte macrophage-colony stimulating factor (GM-CSF) can function as an inflammatory mediator, but whether GM-CSF-producing CD4+ T cells (TH-GM-CSF) are a distinct T helper cell subset is lacking. Herein we demonstrate that interleukin (IL)-1ß exclusively drives differentiation of naïve CD4+ T cells into TH-GM-CSF cells via inducing ubiquitination of IL-1 receptor-associated kinase 1 (IRAK1) and subsequent activation of the transcription factor NF-kappaB (NF-κB), independent of RAR-related orphan receptor gamma (RORγt) required for TH17 differentiation. In vivo, TH-GM-CSF cells are present in murine Citrobacter Rodentium infections and mediate colitis following adoptive transfer of CD4+ T cells into Rag1-/- mice via GM-CSF-induced macrophage activation. The TH-GM-CSF cell phenotype is stable and distinct from the TH17 genetic program, but IL-1ß can convert pre-formed TH17 cells into TH-GM-CSF cells, thereby accounting for previously reported associations between IL-17 and GM-CSF. Together, our results newly identify IL-1ß/NF-κB-dependent TH-GM-CSF cells as a unique T helper cell subset and highlight the importance of CD4+ T cell-derived GM-CSF induced macrophage activation as a previously undescribed T cell effector mechanism.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-1beta/inmunología , Activación de Macrófagos/inmunología , Células Th17/citología , Células Th17/inmunología , Animales , Diferenciación Celular/inmunología , Citrobacter rodentium/inmunología , Colitis/inmunología , Inflamación/inmunología , Inflamación/patología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Células Th17/patología , Ubiquitinación
10.
J Autoimmun ; 96: 113-122, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30241692

RESUMEN

The follicular helper T cell (TFH) are established regulators of germinal center (GC) B cells, whether TFH have pathogenic potential independent of B cells is unknown. Based on in vitro TFH cell differentiation, in vivo T cell transfer animal colitis model, and intestinal tissues of inflammatory bowel disease (IBD) patients, TFH and its functions in colitis development were analyzed by FACS, ChIP, ChIP-sequencing, WB, ELISA and PCR. Herein we demonstrate that intestinal tissues of patients and colon tissues obtained from Rag1-/- recipients of naïve CD4+ T cells with colitis, each over-express TFH-associated gene products. Adoptive transfer of naïve Bcl6-/- CD4+ T cells into Rag1-/- recipient mice abrogated development of colitis and limited TFH differentiation in vivo, demonstrating a mechanistic link. In contrast, T cell deficiency of interferon regulatory factor 8 (IRF8) resulted in augmentation of TFH induction in vitro and in vivo. Functional studies showed that adoptive transfer of IRF8 deficient CD4+ T cells into Rag1-/- recipients exacerbated colitis development associated with increased gut TFH-related gene expression, while Irf8-/-/Bcl6-/- CD4+ T cells abrogated colitis, together indicating that IRF8-regulated TFH can directly cause colon inflammation. Molecular analyses revealed that IRF8 suppresses TFH differentiation by inhibiting transcription and transactivation of the TF IRF4, which is also known to be essential for TFH induction. Our documentation showed that IRF8-regulated TFH can function as B-cell-independent, pathogenic, mediators of colitis suggests that targeting TFH could be effective for treatment of IBD.


Asunto(s)
Linfocitos B/inmunología , Colitis/inmunología , Colon/metabolismo , Enfermedad de Crohn/inmunología , Centro Germinal/inmunología , Factores Reguladores del Interferón/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Traslado Adoptivo , Animales , Células Cultivadas , Colitis/genética , Colon/patología , Enfermedad de Crohn/genética , Modelos Animales de Enfermedad , Humanos , Factores Reguladores del Interferón/genética , Activación de Linfocitos , Ratones , Ratones Noqueados , Comunicación Paracrina , Proteínas Proto-Oncogénicas c-bcl-6/genética , Linfocitos T Colaboradores-Inductores/trasplante
11.
Eur J Immunol ; 47(11): 1890-1899, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28762497

RESUMEN

The development of vaccines for infectious diseases for which we currently have none, including HIV, will likely require the use of adjuvants that strongly promote germinal center responses and somatic hypermutation to produce broadly neutralizing antibodies. Here we compared the outcome of immunization with the T-cell dependent antigen, NP-conjugated to chicken gamma globulin (NP-CGG) adjuvanted with the toll-like receptor 9 (TLR9) ligands, CpG-A or CpG-B, alone or conjugated with the cationic lipid carrier, DOTAP. We provide evidence that only NP-CGG adjuvanted with DOTAP-CpG-B was an effective vaccine in mice resulting in robust germinal center responses, isotype switching and high affinity NP-specific antibodies. The effectiveness of DOTAP-CpG-B as an adjuvant was dependent on the expression of the TLR9 signaling adaptor MyD88 in immunized mice. These results indicate DOTAP-CpG-B but not DOTAP-CpG-A is an effective adjuvant for T cell-dependent protein antigen-based vaccines.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Ácidos Grasos Monoinsaturados/farmacología , Oligodesoxirribonucleótidos/inmunología , Compuestos de Amonio Cuaternario/farmacología , Linfocitos T/inmunología , Vacunas/inmunología , Animales , Afinidad de Anticuerpos , Ácidos Grasos Monoinsaturados/inmunología , Centro Germinal/inmunología , Ratones , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/farmacología , Compuestos de Amonio Cuaternario/inmunología , Vacunas/farmacología
12.
Proc Natl Acad Sci U S A ; 112(42): 13075-80, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26438846

RESUMEN

The most deadly complication of Plasmodium falciparum infection is cerebral malaria (CM) with a case fatality rate of 15-25% in African children despite effective antimalarial chemotherapy. There are no adjunctive treatments for CM, so there is an urgent need to identify new targets for therapy. Here we show that the glutamine analog 6-diazo-5-oxo-L-norleucine (DON) rescues mice from CM when administered late in the infection a time at which mice already are suffering blood-brain barrier dysfunction, brain swelling, and hemorrhaging accompanied by accumulation of parasite-specific CD8(+) effector T cells and infected red blood cells in the brain. Remarkably, within hours of DON treatment mice showed blood-brain barrier integrity, reduced brain swelling, decreased function of activated effector CD8(+) T cells in the brain, and levels of brain metabolites that resembled those in uninfected mice. These results suggest DON as a strong candidate for an effective adjunctive therapy for CM in African children.


Asunto(s)
Antimaláricos/uso terapéutico , Diazooxonorleucina/uso terapéutico , Glutamina/metabolismo , Malaria Cerebral/tratamiento farmacológico , Malaria Falciparum/tratamiento farmacológico , Animales , Antimaláricos/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Diazooxonorleucina/farmacología , Malaria Cerebral/metabolismo , Malaria Falciparum/metabolismo , Ratones
13.
J Autoimmun ; 81: 13-23, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28325644

RESUMEN

TREX1/DNASE III, the most abundant 3'-5' DNA exonuclease in mammalian cells, is tail-anchored on the endoplasmic reticulum (ER). Mutations at the N-terminus affecting TREX1 DNase activity are associated with autoimmune and inflammatory conditions such as Aicardi-Goutières syndrome (AGS). Mutations in the C-terminus of TREX1 cause loss of localization to the ER and dysregulation of oligosaccharyltransferase (OST) activity, and are associated with retinal vasculopathy with cerebral leukodystrophy (RVCL) and in some cases with systemic lupus erythematosus (SLE). Here we investigate mice with conditional expression of the most common RVCL mutation, V235fs, and another mouse expressing a conditional C-terminal mutation, D272fs, associated with a case of human SLE. Mice homozygous for either mutant allele express the encoded human TREX1 truncations without endogenous mouse TREX1, and both remain DNase active in tissues. The two mouse strains are similar phenotypically without major signs of retinal, cerebral or renal disease but exhibit striking elevations of autoantibodies in the serum. The broad range of autoantibodies is primarily against non-nuclear antigens, in sharp contrast to the predominantly DNA-related autoantibodies produced by a TREX1-D18N mouse that specifically lacks DNase activity. We also found that treatment with an OST inhibitor, aclacinomycin, rapidly suppressed autoantibody production in the TREX1 frame-shift mutant mice. Together, our study presents two new mouse models based on TREX1 frame-shift mutations with a unique set of serologic autoimmune-like phenotypes.


Asunto(s)
Autoinmunidad/genética , Autoinmunidad/inmunología , Exodesoxirribonucleasas/genética , Mutación del Sistema de Lectura , Fosfoproteínas/genética , Aclarubicina/análogos & derivados , Aclarubicina/farmacología , Sustitución de Aminoácidos , Animales , Apoptosis/genética , Apoptosis/inmunología , Autoanticuerpos/inmunología , Autoinmunidad/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Activación Enzimática , Exodesoxirribonucleasas/química , Exodesoxirribonucleasas/metabolismo , Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Ratones , Ratones Transgénicos , Fenotipo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Retina/inmunología , Retina/metabolismo , Retina/patología , Timocitos/inmunología , Timocitos/metabolismo , Transcriptoma
14.
J Immunol ; 195(3): 806-9, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26109646

RESUMEN

Several mouse models of systemic lupus erythematosus, including FcγRIIB-KO and TLR7tg mice, develop an expansion of an atypical NK cell subset with functional similarity to cells referred as IFN-producing killer DCs or pre-mature NKs in other systems. In this study, we show that atypical NKs purified from spleens of systemic lupus erythematosus-prone mice, and identified as NK1.1(+)CD11c(+)CD122(+)MHC-II(+), induce persistent autoimmune disease in an IFN-I- and CD40L-dependent manner when transferred to wild-type mice. A single transfer of 4 × 10(6) NK1.1(+) cells from TLR7tg into wild-type induces a 2-wk-long wave of inflammatory cytokines in the serum; a sustained increase in T cell activation and follicular helper cells for the following months; and a progressive expansion of dendritic cells, monocytes, and granulocytes. Furthermore, IL-15 deficiency, which impedes development of NK cells, ameliorates the autoimmune pathology of TLR7tg mice. These results suggest that cells of the NK lineage can develop into cytokine-producing/APCs that affect the priming and progression of systemic autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Células Dendríticas/citología , Interferón Tipo I/inmunología , Células Asesinas Naturales/inmunología , Traslado Adoptivo , Animales , Ligando de CD40/inmunología , Citocinas/sangre , Células Dendríticas/inmunología , Granulocitos/inmunología , Inflamación/inmunología , Interferón Tipo I/biosíntesis , Interleucina-15/genética , Células Asesinas Naturales/trasplante , Lupus Eritematoso Sistémico/inmunología , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Monocitos/inmunología , Receptores de IgG/genética , Linfocitos T Colaboradores-Inductores/inmunología , Receptor Toll-Like 7/genética
15.
J Immunol ; 194(5): 2369-79, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25646302

RESUMEN

During hematopoiesis, hematopoietic stem cells constantly differentiate into granulocytes and macrophages via a distinct differentiation program that is tightly controlled by myeloid lineage-specific transcription factors. Mice with a null mutation of IFN regulatory factor 8 (IRF8) accumulate CD11b(+)Gr1(+) myeloid cells that phenotypically and functionally resemble tumor-induced myeloid-derived suppressor cells (MDSCs), indicating an essential role of IRF8 in myeloid cell lineage differentiation. However, IRF8 is expressed in various types of immune cells, and whether IRF8 functions intrinsically or extrinsically in regulation of myeloid cell lineage differentiation is not fully understood. In this study, we report an intriguing finding that, although IRF8-deficient mice exhibit deregulated myeloid cell differentiation and resultant accumulation of CD11b(+)Gr1(+) MDSCs, surprisingly, mice with IRF8 deficiency only in myeloid cells exhibit no abnormal myeloid cell lineage differentiation. Instead, mice with IRF8 deficiency only in T cells exhibited deregulated myeloid cell differentiation and MDSC accumulation. We further demonstrated that IRF8-deficient T cells exhibit elevated GM-CSF expression and secretion. Treatment of mice with GM-CSF increased MDSC accumulation, and adoptive transfer of IRF8-deficient T cells, but not GM-CSF-deficient T cells, increased MDSC accumulation in the recipient chimeric mice. Moreover, overexpression of IRF8 decreased GM-CSF expression in T cells. Our data determine that, in addition to its intrinsic function as an apoptosis regulator in myeloid cells, IRF8 also acts extrinsically to repress GM-CSF expression in T cells to control myeloid cell lineage differentiation, revealing a novel mechanism that the adaptive immune component of the immune system regulates the innate immune cell myelopoiesis in vivo.


Asunto(s)
Linaje de la Célula/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factores Reguladores del Interferón/inmunología , Células Mieloides/inmunología , Mielopoyesis/inmunología , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Proliferación Celular , Quimera , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Factores Reguladores del Interferón/deficiencia , Factores Reguladores del Interferón/genética , Ratones , Células Mieloides/citología , Células Mieloides/efectos de los fármacos , Mielopoyesis/efectos de los fármacos , Mielopoyesis/genética , Transducción de Señal , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/trasplante
16.
Proc Natl Acad Sci U S A ; 111(4): E511-20, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-24474800

RESUMEN

Malaria infection triggers vigorous host immune responses; however, the parasite ligands, host receptors, and the signaling pathways responsible for these reactions remain unknown or controversial. Malaria parasites primarily reside within RBCs, thereby hiding themselves from direct contact and recognition by host immune cells. Host responses to malaria infection are very different from those elicited by bacterial and viral infections and the host receptors recognizing parasite ligands have been elusive. Here we investigated mouse genome-wide transcriptional responses to infections with two strains of Plasmodium yoelii (N67 and N67C) and discovered differences in innate response pathways corresponding to strain-specific disease phenotypes. Using in vitro RNAi-based gene knockdown and KO mice, we demonstrated that a strong type I IFN (IFN-I) response triggered by RNA polymerase III and melanoma differentiation-associated protein 5, not Toll-like receptors (TLRs), binding of parasite DNA/RNA contributed to a decline of parasitemia in N67-infected mice. We showed that conventional dendritic cells were the major sources of early IFN-I, and that surface expression of phosphatidylserine on infected RBCs might promote their phagocytic uptake, leading to the release of parasite ligands and the IFN-I response in N67 infection. In contrast, an elevated inflammatory response mediated by CD14/TLR and p38 signaling played a role in disease severity and early host death in N67C-infected mice. In addition to identifying cytosolic DNA/RNA sensors and signaling pathways previously unrecognized in malaria infection, our study demonstrates the importance of parasite genetic backgrounds in malaria pathology and provides important information for studying human malaria pathogenesis.


Asunto(s)
Interacciones Huésped-Parásitos , Inmunidad Innata , Malaria/inmunología , Parasitemia/inmunología , Plasmodium yoelii/fisiología , Transducción de Señal , Anciano , Animales , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Interferón Tipo I/metabolismo , Malaria/mortalidad , Malaria/parasitología , Ratones , Ratones Noqueados , Parasitemia/parasitología , Fagocitosis , Plasmodium yoelii/inmunología
17.
Am J Pathol ; 183(2): 402-12, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23747510

RESUMEN

Although it is clear that T helper (Th)17 cells play a pathologic role in the pathogenesis of several inflammatory diseases, the contribution and regulation of pathogenic Th17 cells in the development of glomerulonephritis are still not fully understood. Herein, we show that IL-10-deficient mice exhibit exacerbation of glomerulonephritis after induction with anti-glomerular basement membrane globulin, with enhanced pathogenic Th17 immune responses. We further demonstrate that Rag1(-/-) mice reconstituted with IL-10(-/-) CD4(+) T cells develop more severe glomerulonephritis after induction of anti-glomerular basement membrane disease, with more infiltration of inflammatory cells into the kidneys. Finally, IL-17 and interferon γ double-positive cells were significantly increased in IL-10(-/-) CD4(+) T-cell cultures under pathogenic Th17 conditions compared with wild-type cell cultures. These findings suggest that T-cell-derived IL-10 plays a critical suppressive role in the control of pathogenic Th17 cell differentiation and highlights the importance of IL-10 as protection against glomerulonephritis development.


Asunto(s)
Glomerulonefritis/inmunología , Interleucina-10/deficiencia , Células Th17/patología , Animales , Autoanticuerpos/toxicidad , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Glomerulonefritis/patología , Proteínas de Homeodominio/metabolismo , Inmunidad Celular/inmunología , Interferón gamma/metabolismo , Interleucina-10/fisiología , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Th17/inmunología
18.
Blood ; 119(2): 465-8, 2012 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-22110250

RESUMEN

The splenic marginal zone (MZ) is comprised of specialized populations of B cells, dendritic cells, and macrophages that are uniquely arrayed outside the white pulp follicles to screen the blood for bacterial and other particulate Ags. Mechanisms responsible for MZ B-cell formation, localization, retention, and function are understood to include antigenic specificity, transcription factors, integrins, and surface receptors for soluble ligands such as S1P. Here, we add to this repertoire by demonstrating that the receptor for CXCL12, CXCR7, is expressed on MZ but not on follicular B cells. Treatment of mice with CXCR7 inhibitors led to disruption of MZ architecture, reduced numbers of MZ B cells, and altered granulocyte homeostasis associated with increasing serum levels of CXCL12. CXCR7 thus appears to function as a scavenger receptor for CXCL12 on MZ B cells.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Quimiocina CXCL12/sangre , Receptores CXCR/metabolismo , Bazo/citología , Bazo/metabolismo , Animales , Linfocitos B/patología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Granulocitos/citología , Granulocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores CXCR/antagonistas & inhibidores , Bazo/inmunología
19.
Methods Cell Biol ; 188: 131-152, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38880521

RESUMEN

Renal injury often occurs as a complication in autoimmune diseases such as systemic lupus erythematosus (SLE). It is estimated that a minimum of 20% SLE patients develop lupus nephritis, a condition that can be fatal when the pathology progresses to end-stage renal disease. Studies in animal models showed that incidence of immune cell infiltrates in the kidney was linked to pathological injury and correlated with severe lupus nephritis. Thus, preventing immune cell infiltration into the kidney is a potential approach to impede the progression to an end-stage disease. A requirement to investigate the role of kidney-infiltrating leukocytes is the development of reproducible and efficient protocols for purification and characterization of immune cells in kidney samples. This chapter describes a detailed methodology that discriminates tissue-resident leukocytes from blood-circulating cells that are found in kidney. Our protocol was designed to maximize cell viability and to reduce variability among samples, with a combination of intravascular staining and magnetic bead separation for leukocyte enrichment. Experiments included as example were performed with FcγRIIb[KO] mice, a well-characterized murine model of SLE. We identified T cells and macrophages as the primary leukocyte subsets infiltrating into the kidney during severe nephritis, and we extensively characterized them phenotypically by flow cytometry.


Asunto(s)
Modelos Animales de Enfermedad , Riñón , Leucocitos , Nefritis Lúpica , Animales , Nefritis Lúpica/patología , Nefritis Lúpica/inmunología , Ratones , Riñón/patología , Leucocitos/inmunología , Leucocitos/patología , Separación Celular/métodos , Ratones Noqueados , Macrófagos/inmunología , Macrófagos/patología , Citometría de Flujo/métodos , Linfocitos T/inmunología , Receptores de IgG/metabolismo
20.
J Immunol ; 186(3): 1458-66, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21178004

RESUMEN

Transcriptional control of marginal zone (MZ) and follicular (FO) B cell development remains incompletely understood. The transcription factor, IFN regulatory factor (IRF)8, is known to play important roles in the differentiation of early B cells. In this article, we demonstrate that IRF8 is also required for normal development of MZ and FO B cells. Mice with a conventional knockout of Irf8 (IRF8(-/-)) or a point mutation in the IRF association domain of IRF8 had increased numbers of MZ B cells. To determine the B cell-intrinsic effects of IRF8 deficiency, we generated mice with a conditional allele of Irf8 crossed with CD19-Cre mice (designated IRF8-conditional knockout [CKO]). These mice had enlarged MZ and increased numbers of MZ and FO B cells compared with controls. The FO B cells of CKO mice exhibited reduced expression of CD23 and moderately increased expression of CD21. Gene-expression profiling showed that increased B cell production in IRF8-CKO mice was associated with changes in expression of genes involved in regulation of transcription, signaling, and inflammation. Functional studies showed that IRF8-CKO mice generated normal Ab responses to T-independent and T-dependent Ags. Thus, IRF8 controls the expansion and maturation of MZ and FO B cells but has little effect on B cell function.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Factores Reguladores del Interferón/fisiología , Bazo/citología , Bazo/inmunología , Animales , Subgrupos de Linfocitos B/patología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Femenino , Eliminación de Gen , Factores Reguladores del Interferón/deficiencia , Factores Reguladores del Interferón/genética , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutagénesis Insercional , Mutación Puntual , Estructura Terciaria de Proteína/genética , Bazo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA