Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Immunity ; 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38901428

RESUMEN

Many infections, including malaria, are associated with an increase in autoantibodies (AAbs). Prior studies have reported an association between genetic markers of susceptibility to autoimmune disease and resistance to malaria, but the underlying mechanisms are unclear. Here, we performed a longitudinal study of children and adults (n = 602) in Mali and found that high levels of plasma AAbs before the malaria season independently predicted a reduced risk of clinical malaria in children during the ensuing malaria season. Baseline AAb seroprevalence increased with age and asymptomatic Plasmodium falciparum infection. We found that AAbs purified from the plasma of protected individuals inhibit the growth of blood-stage parasites and bind P. falciparum proteins that mediate parasite invasion. Protected individuals had higher plasma immunoglobulin G (IgG) reactivity against 33 of the 123 antigens assessed in an autoantigen microarray. This study provides evidence in support of the hypothesis that a propensity toward autoimmunity offers a survival advantage against malaria.

2.
Trends Immunol ; 41(7): 601-613, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32446878

RESUMEN

The mature naïve B cell repertoire consists of three well-defined populations: B1, B2 (follicular B, FOB), and marginal zone B (MZB) cells. FOB cells are the dominant mature B cell population in the secondary lymphoid organs and blood of both humans and mice. The driving forces behind mature B lineage selection have been linked to B cell receptor (BCR) signaling strength and environmental cues, but how these fate-determination factors are transcriptionally regulated remains poorly understood. We summarize emerging data on the role of transcription factors (TFs) - particularly the ETS and IRF families - in regulating MZB and FOB lineage selection. Indeed, genomic analyses have identified four major groups of target genes that are crucial for FOB differentiation, revealing previously unrecognized pathways that ultimately determine biological responses specific to this lineage.


Asunto(s)
Linfocitos B , Diferenciación Celular , Regulación de la Expresión Génica , Bazo , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Receptores de Antígenos de Linfocitos B/inmunología , Bazo/citología , Bazo/inmunología
3.
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
4.
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
5.
Nature ; 521(7552): 357-61, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25799995

RESUMEN

B cells are selected for an intermediate level of B-cell antigen receptor (BCR) signalling strength: attenuation below minimum (for example, non-functional BCR) or hyperactivation above maximum (for example, self-reactive BCR) thresholds of signalling strength causes negative selection. In ∼25% of cases, acute lymphoblastic leukaemia (ALL) cells carry the oncogenic BCR-ABL1 tyrosine kinase (Philadelphia chromosome positive), which mimics constitutively active pre-BCR signalling. Current therapeutic approaches are largely focused on the development of more potent tyrosine kinase inhibitors to suppress oncogenic signalling below a minimum threshold for survival. We tested the hypothesis that targeted hyperactivation--above a maximum threshold--will engage a deletional checkpoint for removal of self-reactive B cells and selectively kill ALL cells. Here we find, by testing various components of proximal pre-BCR signalling in mouse BCR-ABL1 cells, that an incremental increase of Syk tyrosine kinase activity was required and sufficient to induce cell death. Hyperactive Syk was functionally equivalent to acute activation of a self-reactive BCR on ALL cells. Despite oncogenic transformation, this basic mechanism of negative selection was still functional in ALL cells. Unlike normal pre-B cells, patient-derived ALL cells express the inhibitory receptors PECAM1, CD300A and LAIR1 at high levels. Genetic studies revealed that Pecam1, Cd300a and Lair1 are critical to calibrate oncogenic signalling strength through recruitment of the inhibitory phosphatases Ptpn6 (ref. 7) and Inpp5d (ref. 8). Using a novel small-molecule inhibitor of INPP5D (also known as SHIP1), we demonstrated that pharmacological hyperactivation of SYK and engagement of negative B-cell selection represents a promising new strategy to overcome drug resistance in human ALL.


Asunto(s)
Linfocitos B/metabolismo , Linfocitos B/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Transducción de Señal , Secuencias de Aminoácidos/genética , Animales , Antígenos CD/metabolismo , Linfocitos B/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Proteínas de Fusión bcr-abl/genética , Eliminación de Gen , Humanos , Inositol Polifosfato 5-Fosfatasas , Péptidos y Proteínas de Señalización Intracelular/agonistas , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Células Precursoras de Linfocitos B/efectos de los fármacos , Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores de Antígenos de Linfocitos B/deficiencia , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Transducción de Señal/efectos de los fármacos , Quinasa Syk , Tirosina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Trends Immunol ; 38(5): 373-382, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28274696

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by a breakdown of self-tolerance in B cells and the production of antibodies against nuclear self-antigens. Increasing evidence supports the notion that additional cellular contributors beyond B cells are important for lupus pathogenesis. In this review we consider recent advances regarding both the pathogenic and the regulatory role of lymphocytes in SLE beyond the production of IgG autoantibodies. We also discuss various inflammatory effector cell types involved in cytokine production, removal of self-antigens, and responses to autoreactive IgE antibodies. We aim to integrate these ideas to expand the current understanding of the cellular components that contribute to disease progression and ultimately help in the design of novel, targeted therapeutics.


Asunto(s)
Autoanticuerpos/inmunología , Linfocitos B/inmunología , Inmunoglobulina G/inmunología , Lupus Eritematoso Sistémico/inmunología , Autotolerancia/inmunología , Autoanticuerpos/metabolismo , Linfocitos B/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunoglobulina G/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Modelos Inmunológicos
7.
J Immunol ; 199(3): 931-940, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28652397

RESUMEN

B cells express the innate receptor, TLR9, which signals in response to unmethylated CpG sequences in microbial DNA. Of the two major classes of CpG-containing oligonucleotides, CpG-A appears restricted to inducing type 1 IFN in innate immune cells and CpG-B to activating B cells to proliferate and produce Abs and inflammatory cytokines. Although CpGs are candidates for adjuvants to boost innate and adaptive immunity, our understanding of the effect of CpG-A and CpG-B on B cell responses is incomplete. In this study we show that both CpG-B and CpG-A activated B cells in vitro to proliferate, secrete Abs and IL-6, and that neither CpG-B nor CpG-A alone induced type 1 IFN production. However, when incorporated into the cationic lipid, DOTAP, CpG-A, but not CpG-B, induced a type 1 IFN response in B cells in vitro and in vivo. We provide evidence that differences in the function of CpG-A and CpG-B may be related to their intracellular trafficking in B cells. These findings fill an important gap in our understanding of the B cell response to CpGs, with implications for the use of CpG-A and CpG-B as immunomodulators.


Asunto(s)
Linfocitos B/inmunología , Interferón Tipo I/biosíntesis , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/inmunología , Animales , Formación de Anticuerpos , Linfocitos B/efectos de los fármacos , Cationes/inmunología , Citocinas/genética , Citocinas/inmunología , Inmunidad Innata , Factores Inmunológicos/metabolismo , Interferón Tipo I/inmunología , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Lípidos/administración & dosificación , Lípidos/química , Lípidos/farmacología , Activación de Linfocitos , Ratones , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/farmacología , Receptor Toll-Like 9/agonistas
8.
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
9.
PLoS Pathog ; 12(10): e1005930, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27716849

RESUMEN

Both type I interferon (IFN-I) and CD40 play a significant role in various infectious diseases, including malaria and autoimmune disorders. CD40 is mostly known to function in adaptive immunity, but previous observations of elevated CD40 levels early after malaria infection of mice led us to investigate its roles in innate IFN-I responses and disease control. Using a Plasmodium yoelii nigeriensis N67 and C57BL/6 mouse model, we showed that infected CD40-/- mice had reduced STING and serum IFN-ß levels day-2 post infection, higher day-4 parasitemia, and earlier deaths. CD40 could greatly enhance STING-stimulated luciferase signals driven by the IFN-ß promoter in vitro, which was mediated by increased STING protein levels. The ability of CD40 to influence STING expression was confirmed in CD40-/- mice after malaria infection. Substitutions at CD40 TRAF binding domains significantly decreased the IFN-ß signals and STING protein level, which was likely mediated by changes in STING ubiquitination and degradation. Increased levels of CD40, STING, and ISRE driven luciferase signal in RAW Lucia were observed after phagocytosis of N67-infected red blood cells (iRBCs), stimulation with parasite DNA/RNA, or with selected TLR ligands [LPS, poly(I:C), and Pam3CSK4]. The results suggest stimulation of CD40 expression by parasite materials through TLR signaling pathways, which was further confirmed in bone marrow derived dendritic cells/macrophages (BMDCs/BMDMs) and splenic DCs from CD40-/-, TLR3-/- TLR4-/-, TRIF-/-, and MyD88-/- mice after iRBC stimulation or parasite infection. Our data connect several signaling pathways consisting of phagocytosis of iRBCs, recognition of parasite DNA/RNA (possibly GPI) by TLRs, elevated levels of CD40 and STING proteins, increased IFN-I production, and longer host survival time. This study reveals previously unrecognized CD40 function in innate IFN-I responses and protective pathways in infections with malaria strains that induce a strong IFN-I response, which may provide important information for better understanding and management of malaria.


Asunto(s)
Antígenos CD40/inmunología , Interacciones Huésped-Parásitos/inmunología , Interferón Tipo I/inmunología , Malaria/inmunología , Proteínas de la Membrana/inmunología , Animales , Western Blotting , Antígenos CD40/biosíntesis , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium yoelii/inmunología
10.
Immunity ; 29(5): 667-9, 2008 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-19006689

RESUMEN

Self-reactive B cells are eliminated during development by antibody-affinity selection and receptor-editing mechanisms. Work by Isnardi et al. (2008) in this issue of Immunity suggests that removal of autoreactivity from the immature B cell pool also requires innate immunity pathways.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos B/inmunología , Inmunidad Innata , Autotolerancia/inmunología , Autoinmunidad/genética , Linfocitos B/metabolismo , Diferenciación Celular , Supresión Clonal , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/deficiencia , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo
11.
J Immunol ; 194(4): 1503-13, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25595787

RESUMEN

Chronic inflammatory conditions, such as in autoimmune disease, can disturb immune cell homeostasis and induce the expansion of normally rare cell populations. In our analysis of various murine models of lupus, we detect increased frequency of an uncommon subset identified as NK1.1(+)CD11c(+)CD122(+)MHC class II(+). These cells share characteristics with the NK cell lineage and with cells previously described as IFN-producing killer dendritic cells: 1) they depend on IL-15 and express E4BP4; 2) they are cytotoxic and produce type I and type II IFN upon activation; and 3) they are efficient APCs both through MHC class II expression and in cross-presentation to CD8s. These atypical NK cells are responsive to TLR stimulation and thus are most abundant in mice with high copy number of the Tlr7 gene. They are highly proliferative as assessed by in vivo BrdU incorporation. In adoptive transfer experiments they persist in high numbers for months and maintain their surface marker profile, indicating that this population is developmentally stable. Gene expression analyses on both mRNA and microRNAs show a modified cell cycle program in which various miR-15/16 family members are upregulated, presumably as a consequence of the proliferative signal mediated by the increased level of growth factors, Ras and E2F activity. Alternatively, low expression of miR-150, miR-181, and miR-744 in these cells implies a reduction in their differentiation capacity. These results suggest that cells of the NK lineage that undergo TLR stimulation might turn on a proliferative program in detriment of their full differentiation into mature NK cells.


Asunto(s)
Células Asesinas Naturales/inmunología , Lupus Eritematoso Sistémico/inmunología , Activación de Linfocitos/inmunología , Subgrupos Linfocitarios/inmunología , Traslado Adoptivo , Animales , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica , Células Asesinas Naturales/citología , Subgrupos Linfocitarios/citología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Quimera por Trasplante
12.
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
14.
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
15.
Nature ; 465(7300): 937-41, 2010 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-20559388

RESUMEN

Glucocorticoids are widely used to treat patients with autoimmune diseases such as systemic lupus erythematosus (SLE). However, regimens used to treat many such conditions cannot maintain disease control in the majority of SLE patients and more aggressive approaches such as high-dose methylprednisolone pulse therapy are used to provide transient reductions in disease activity. The primary anti-inflammatory mechanism of glucocorticoids is thought to be NF-kappaB inhibition. Recognition of self nucleic acids by toll-like receptors TLR7 and TLR9 on B cells and plasmacytoid dendritic cells (PDCs) is an important step in the pathogenesis of SLE, promoting anti-nuclear antibodies and the production of type I interferon (IFN), both correlated with the severity of disease. Following their activation by self-nucleic acid-associated immune complexes, PDCs migrate to the tissues. We demonstrate, in vitro and in vivo, that stimulation of PDCs through TLR7 and 9 can account for the reduced activity of glucocorticoids to inhibit the IFN pathway in SLE patients and in two lupus-prone mouse strains. The triggering of PDCs through TLR7 and 9 by nucleic acid-containing immune complexes or by synthetic ligands activates the NF-kappaB pathway essential for PDC survival. Glucocorticoids do not affect NF-kappaB activation in PDCs, preventing glucocorticoid induction of PDC death and the consequent reduction of systemic IFN-alpha levels. These findings unveil a new role for self nucleic acid recognition by TLRs and indicate that inhibitors of TLR7 and 9 signalling could prove to be effective corticosteroid-sparing drugs.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Glucocorticoides/farmacología , Lupus Eritematoso Sistémico/fisiopatología , Ácidos Nucleicos/inmunología , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/inmunología , Adolescente , Animales , Autoanticuerpos/inmunología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Niño , Modelos Animales de Enfermedad , Femenino , Humanos , Interferón-alfa/inmunología , Interferones/inmunología , Masculino , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/inmunología , Regulación hacia Arriba
16.
J Immunol ; 190(6): 2536-43, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23382559

RESUMEN

TLR7 activation is implicated in the pathogenesis of systemic lupus erythematosus. Mice that overexpress TLR7 develop a lupus-like disease with autoantibodies and glomerulonephritis and early death. To determine whether degradation of the TLR7 ligand RNA would alter the course of disease, we created RNase A transgenic (Tg) mice. We then crossed the RNase Tg to TLR7 Tg mice to create TLR7 × RNase double Tg (DTg) mice. DTg mice had a significantly increased survival associated with reduced activation of T and B lymphocytes and reduced kidney deposition of IgG and C3. We observed massive hepatic inflammation and cell death in TLR7 Tg mice. In contrast, hepatic inflammation and necrosis were strikingly reduced in DTg mice. These findings indicate that high concentrations of serum RNase protect against immune activation and inflammation associated with TLR7 stimulation and that RNase may be a useful therapeutic strategy in the prevention or treatment of inflammation in systemic lupus erythematosus and, possibly, liver diseases.


Asunto(s)
Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Ribonucleasa Pancreática/genética , Receptor Toll-Like 7/biosíntesis , Receptor Toll-Like 7/genética , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Animales , Bovinos , Células Cultivadas , Células Madre Embrionarias , Hepatitis/enzimología , Hepatitis/inmunología , Hepatitis/patología , Humanos , Inflamación/enzimología , Inflamación/inmunología , Inflamación/prevención & control , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/mortalidad , Lupus Eritematoso Sistémico/prevención & control , Masculino , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Ribonucleasa Pancreática/sangre , Ribonucleasa Pancreática/fisiología , Bazo/enzimología , Bazo/inmunología , Bazo/patología , Análisis de Supervivencia , Receptor Toll-Like 7/fisiología
17.
Proc Natl Acad Sci U S A ; 109(40): 16276-81, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-22988104

RESUMEN

Toll-like receptor 7 (Tlr7) has been linked to systemic lupus disease incidence in humans and mice, but how TLR7 potentiates autoimmunity is unclear. We used a Tlr7 transgenic (tg) mouse model to investigate the cellular and molecular events required to induce spontaneous autoimmunity through increased TLR7 activity. We determined that Tlr7 exerts B-cell-intrinsic effects in promoting spontaneous germinal center (GC) and plasmablast B-cell development, and that these B-cell subsets are dependent on T-cell-derived signals through CD40L and SLAM-associated protein (SAP), but not IL-17. Antigen specificity also factored into TLR7-induced disease, as both a restricted T cell receptor (TCR) specificity and MHC haplotype H2(k/k) protected Tlr7tg mice from spontaneous lymphocyte activation and autoantibody production. Inflammatory myeloid cell expansion and autoimmunity did not develop in Tlr7tgIgH(-/-) mice, suggesting either that spontaneous TLR7 activation does not occur in dendritic cells, or, if it does occur, cannot drive these events in the absence of B-cell aid. These data indicate that autoimmune disease in Tlr7tg mice is contingent upon B cells receiving stimulation both through innate pathways and T-cell-derived signals and suggest a codependent relationship between B cells and T cells in the development of autoimmunity.


Asunto(s)
Inmunidad Adaptativa/inmunología , Autoinmunidad/inmunología , Linfocitos B/inmunología , Inmunidad Innata/inmunología , Lupus Eritematoso Sistémico/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 7/inmunología , Análisis de Varianza , Animales , Ligando de CD40/metabolismo , Citometría de Flujo , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Antígenos de Linfocitos T/metabolismo , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria
18.
Eur J Immunol ; 43(2): 371-81, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23135975

RESUMEN

The strength of the Ag receptor signal influences development and negative selection of B cells, and it might also affect B-cell survival and selection in the GC. Here, we have used mice with B-cell-specific deletion of the 5'-inositol phosphatase SHIP as a model to study affinity selection in cells that are hyperresponsive to Ag and cytokine receptor stimulation. In the absence of SHIP, B cells have lower thresholds for Ag- and interferon (IFN)-induced activation, resulting in augmented negative selection in the BM and enhanced B-cell maturation in the periphery. Despite a tendency to spontaneously downregulate surface IgM expression, SHIP deficiency does not alter anergy induction in response to soluble hen-egg lysozyme Ag in the MDA4 transgenic model. SHIP-deficient B cells spontaneously produce isotype-switched antibodies; however, they are poor responders in immunization and infection models. While SHIP-deficient B cells form GCs and undergo mutation, they are not properly selected for high-affinity antibodies. These results illustrate the importance of negative regulation of B-cell responses, as lower thresholds for B-cell activation promote survival of low affinity and deleterious receptors to the detriment of optimal Ab affinity maturation.


Asunto(s)
Linfocitos B/inmunología , Monoéster Fosfórico Hidrolasas/inmunología , Animales , Afinidad de Anticuerpos , Antígenos/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Inositol Polifosfato 5-Fosfatasas , Interferones/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos/inmunología , Linfocitos T/inmunología
19.
J Immunol ; 188(3): 1451-9, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22205024

RESUMEN

Type I IFNs (IFN-I) are normally produced during antiviral responses, yet high levels of chronic IFN-I expression correlate with autoimmune disease. A variety of viral sensors generate IFN-I in their response, but other than TLRs, it is not fully known which pathways are directly involved in the development of spontaneous immune pathologies. To further explore the link between IFN-I induced by viral pathways and autoimmunity, we generated a new transgenic mouse line containing multiple copies of Ifih1, a gene encoding the cytoplasmic dsRNA sensor MDA5 with proven linkage to diabetes and lupus. We show that MDA5 overexpression led to a chronic IFN-I state characterized by resistance to a lethal viral infection through rapid clearance of virus in the absence of a CD8(+) or Ab response. Spontaneous MDA5 activation was not sufficient to initiate autoimmune or inflammatory pathology by itself, even though every immune cell population had signs of IFN activation. When combined with the lupus-susceptible background of the FcγR2B deficiency, MDA5 overexpression did accelerate the production of switched autoantibodies, the incidence of glomerulonephritis, and early lethality. Thus, MDA5 transgenic mice provide evidence that chronic elevated levels of IFN-I are not sufficient to initiate autoimmunity or inflammation although they might exacerbate an ongoing autoimmune pathology.


Asunto(s)
Autoinmunidad/genética , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , Dosificación de Gen , Interferón Tipo I/genética , Virosis/inmunología , Animales , Línea Celular , Inflamación/etiología , Helicasa Inducida por Interferón IFIH1 , Ratones , Ratones Transgénicos , Virus/inmunología
20.
Proc Natl Acad Sci U S A ; 108(3): 1122-7, 2011 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21187399

RESUMEN

Plasmodium falciparum has exerted tremendous selective pressure on genes that improve survival in severe malarial infections. Systemic lupus erythematosus (SLE) is an autoimmune disease that is six to eight times more prevalent in women of African descent than in women of European descent. Here we provide evidence that a genetic susceptibility to SLE protects against cerebral malaria. Mice that are prone to SLE because of a deficiency in FcγRIIB or overexpression of Toll-like receptor 7 are protected from death caused by cerebral malaria. Protection appears to be by immune mechanisms that allow SLE-prone mice better to control their overall inflammatory responses to parasite infections. These findings suggest that the high prevalence of SLE in women of African descent living outside of Africa may result from the inheritance of genes that are beneficial in the immune control of cerebral malaria but that, in the absence of malaria, contribute to autoimmune disease.


Asunto(s)
Población Negra/genética , Predisposición Genética a la Enfermedad/genética , Lupus Eritematoso Sistémico/genética , Malaria Cerebral/genética , Plasmodium berghei/inmunología , Receptores de IgG/deficiencia , Receptor Toll-Like 7/metabolismo , Animales , Encéfalo/inmunología , Encéfalo/patología , Citocinas/sangre , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/parasitología , Femenino , Citometría de Flujo , Humanos , Lupus Eritematoso Sistémico/etnología , Malaria Cerebral/inmunología , Malaria Cerebral/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Receptores de IgG/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/fisiología , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA