Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Leukemia ; 36(10): 2430-2442, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36042317

RESUMO

Activation-induced cytidine deaminase (AID) has been implicated as both a positive and a negative factor in the progression of B cell chronic lymphocytic leukemia (CLL), but the role that it plays in the development and progression of this disease is still unclear. We generated an AID knockout CLL mouse model, AID-/-/Eµ-TCL1, and found that these mice die significantly earlier than their AID-proficient counterparts. AID-deficient CLL cells exhibit a higher ER stress response compared to Eµ-TCL1 controls, particularly through activation of the IRE1/XBP1s pathway. The increased production of secretory IgM in AID-deficient CLL cells contributes to their elevated expression levels of XBP1s, while secretory IgM-deficient CLL cells express less XBP1s. This increase in XBP1s in turn leads AID-deficient CLL cells to exhibit higher levels of B cell receptor signaling, supporting leukemic growth and survival. Further, AID-/-/Eµ-TCL1 CLL cells downregulate the tumor suppressive SMAD1/S1PR2 pathway and have altered homing to non-lymphoid organs. Notably, CLL cells from patients with IgHV-unmutated disease express higher levels of XBP1s mRNA compared to those from patients with IgHV-mutated CLL. Our studies thus reveal novel mechanisms by which the loss of AID leads to worsened CLL and may explain why unmutated CLL is more aggressive than mutated CLL.


Assuntos
Leucemia Linfocítica Crônica de Células B , Animais , Citidina Desaminase/genética , Leucemia Linfocítica Crônica de Células B/patologia , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases , RNA Mensageiro/genética , Receptores de Antígenos de Linfócitos B/genética
2.
J Exp Med ; 218(5)2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33724364

RESUMO

The spleen contains a myriad of conventional dendritic cell (cDC) subsets that protect against systemic pathogen dissemination by bridging antigen detection to the induction of adaptive immunity. How cDC subsets differentiate in the splenic environment is poorly understood. Here, we report that LTα1ß2-expressing Rorgt+ ILC3s, together with B cells, control the splenic cDC niche size and the terminal differentiation of Sirpα+CD4+Esam+ cDC2s, independently of the microbiota and of bone marrow pre-cDC output. Whereas the size of the splenic cDC niche depended on lymphotoxin signaling only during a restricted time frame, the homeostasis of Sirpα+CD4+Esam+ cDC2s required continuous lymphotoxin input. This latter property made Sirpα+CD4+Esam+ cDC2s uniquely susceptible to pharmacological interventions with LTßR agonists and antagonists and to ILC reconstitution strategies. Together, our findings demonstrate that LTα1ß2-expressing Rorgt+ ILC3s drive splenic cDC differentiation and highlight the critical role of ILC3s as perpetual regulators of lymphoid tissue homeostasis.


Assuntos
Células Dendríticas/imunologia , Imunidade Inata , Tecido Linfoide/imunologia , Linfotoxina-alfa/imunologia , Transdução de Sinais/imunologia , Baço/imunologia , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Moléculas de Adesão Celular/metabolismo , Células Dendríticas/metabolismo , Feminino , Tecido Linfoide/citologia , Tecido Linfoide/metabolismo , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/imunologia , Receptor beta de Linfotoxina/metabolismo , Linfotoxina-alfa/genética , Linfotoxina-alfa/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Transdução de Sinais/genética , Baço/citologia , Baço/metabolismo
3.
Oncogene ; 39(37): 6009-6023, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32772043

RESUMO

Aurora B kinase plays essential roles in mitosis. Its protein levels increase before the onset of mitosis and sharply decrease during mitosis exit. The latter decrease is due to a balance between the actions of the E3 ubiquitin ligase anaphase-promoting complex or cyclosome (activated by the Cdh1 adapter), and the deubiquitinating enzyme USP35. Aurora B also executes important functions in interphase. Abnormal modulation of Aurora B in interphase leads to cell cycle defects often linked to aberrant chromosomal condensation and segregation. Very little is however known about how Aurora B levels are regulated in interphase. Here we found that USP13-associates with and stabilizes Aurora B in cells, especially before their entry into mitosis. In order for USP13 to exert its stabilizing effect on Aurora B, their association is promoted by the Aurora B-mediated phosphorylation of USP13 at Serine 114. We also present evidence that USP13 instigates Aurora B deubiquitination and/or protect it from degradation in a non-catalytic manner. In addition, we report that genetic or chemical modulation of the cellular levels/activity of USP13 affects unperturbed cell-cycle progression. Overall our study unveils the molecular and cellular connections of the USP13-Aurora B axis, which potentially participates in the rewiring of the cell cycle happening in cancer cells.


Assuntos
Aurora Quinase B/metabolismo , Ciclo Celular/genética , Endopeptidases/genética , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Progressão da Doença , Endopeptidases/metabolismo , Estabilidade Enzimática , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Fosforilação , Ligação Proteica , Serina/metabolismo , Proteases Específicas de Ubiquitina
4.
PLoS Negl Trop Dis ; 14(6): e0008358, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32589656

RESUMO

Antibody-mediated parasite killing is considered the most effective host immune response against extracellular trypanosome parasites. However, due to host-parasite co-evolution pressure, these parasites have "learned" how to hijack the host immune system via the development of immune evasion strategies. Hereby they prevent elimination and promote transmission. In the past, our group has shown that African trypanosome parasites are able to "shut down" the host B cell compartment, via the abolishment of the homeostatic B cell compartment. In line with this, we have reported that trypanosome infections result in detrimental outcomes on auto-reactive and cancer B cells. To unravel the immune mechanisms involved in these processes we adopted here a well-defined B cell vaccine model, i.e. the thymo-dependent hapten-carrier NP-CGG (4-Hydroxy-3-nitrophenylacetyl-Chicken Gamma Globulin) emulsified in Alum adjuvant. Results show that T. brucei infections abrogate the circulating titres of vaccine-induced CGG-specific as well as NP-specific IgG1+ antibodies, a hallmark of memory B cell responses in this model. This happens independently of their affinity and IFNÉ£ signalling. Next, we demonstrate that T. brucei infections also induce a decrease of anti-NP IgG3+ antibodies induced by the administration of NP coupled to Ficoll, a thymo-independent antigen. Confirming the non-specificity of the infection-associated immunopathology, this report also shows that trypanosome infections abolish vaccine-induced memory response against malaria parasite in BALB/c mice. Together, these data indicates that T. brucei infections impair every stages of B cell development, including effector plasma B cells, independently of their specificity and affinity as well as the host genetic background.


Assuntos
Subpopulações de Linfócitos B/imunologia , Plasmócitos/imunologia , Tripanossomíase Africana/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Linfócitos B/imunologia , Modelos Animais de Doenças , Feminino , Patrimônio Genético , Interações Hospedeiro-Parasita/imunologia , Evasão da Resposta Imune , Imunidade Humoral , Imunoglobulina G , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vacinas Protozoárias/imunologia , Trypanosoma brucei brucei
5.
PLoS Pathog ; 16(2): e1008170, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32012211

RESUMO

Bovine African Trypanosomosis is an infectious parasitic disease affecting livestock productivity and thereby impairing the economic development of Sub-Saharan Africa. The most important trypanosome species implicated is T. congolense, causing anemia as most important pathological feature. Using murine models, it was shown that due to the parasite's efficient immune evasion mechanisms, including (i) antigenic variation of the variable surface glycoprotein (VSG) coat, (ii) induction of polyclonal B cell activation, (iii) loss of B cell memory and (iv) T cell mediated immunosuppression, disease prevention through vaccination has so far been impossible. In trypanotolerant models a strong, early pro-inflammatory immune response involving IFN-γ, TNF and NO, combined with a strong humoral anti-VSG response, ensures early parasitemia control. This potent protective inflammatory response is counterbalanced by the production of the anti-inflammatory cytokine IL-10, which in turn prevents early death of the host from uncontrolled hyper-inflammation-mediated immunopathologies. Though at this stage different hematopoietic cells, such as NK cells, T cells and B cells as well as myeloid cells (i.e. alternatively activated myeloid cells (M2) or Ly6c- monocytes), were found to produce IL-10, the contribution of non-hematopoietic cells as potential IL-10 source during experimental T. congolense infection has not been addressed. Here, we report for the first time that during the chronic stage of T. congolense infection non-hematopoietic cells constitute an important source of IL-10. Our data shows that hepatocyte-derived IL-10 is mandatory for host survival and is crucial for the control of trypanosomosis-induced inflammation and associated immunopathologies such as anemia, hepatosplenomegaly and excessive tissue injury.


Assuntos
Hepatócitos , Evasão da Resposta Imune , Interleucina-10/imunologia , Trypanosoma congolense , Tripanossomíase Africana , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Doença Crônica , Modelos Animais de Doenças , Feminino , Hepatócitos/imunologia , Hepatócitos/parasitologia , Hepatócitos/patologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Ativação Linfocitária , Camundongos , Monócitos/imunologia , Monócitos/patologia , Linfócitos T/imunologia , Linfócitos T/patologia , Trypanosoma congolense/imunologia , Trypanosoma congolense/patogenicidade , Tripanossomíase Africana/imunologia , Tripanossomíase Africana/patologia
6.
Sci Rep ; 8(1): 11203, 2018 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-30046157

RESUMO

In this study, Trypanosoma brucei was naturally transmitted to mice through the bites of infected Glossina morsitans tsetse flies. Neutrophils were recruited rapidly to the bite site, whereas monocytes were attracted more gradually. Expression of inflammatory cytokines (il1b, il6), il10 and neutrophil chemokines (cxcl1, cxcl5) was transiently up-regulated at the site of parasite inoculation. Then, a second influx of neutrophils occurred that coincided with the previously described parasite retention and expansion in the ear dermis. Congenital and experimental neutropenia models, combined with bioluminescent imaging, indicate that neutrophils do not significantly contribute to dermal parasite control and elicit higher systemic parasitemia levels during the infection onset. Engulfment of parasites by neutrophils in the skin was rarely observed and was restricted to parasites with reduced motility/viability, whereas live parasites escaped phagocytosis. To our knowledge, this study represents the first description of a trypanosome infection promoting role of early innate immunological reactions following an infective tsetse fly bite. Our data indicate that the trypanosome is not hindered in its early development and benefits from the host innate responses with the neutrophils being important regulators of the early infection, as already demonstrated for the sand fly transmitted Leishmania parasite.


Assuntos
Derme/parasitologia , Neutrófilos/parasitologia , Trypanosoma brucei brucei/genética , Tripanossomíase Africana/genética , Animais , Quimiocina CXCL1/genética , Quimiocina CXCL5/genética , Derme/metabolismo , Regulação da Expressão Gênica , Mordeduras e Picadas de Insetos/parasitologia , Insetos Vetores/genética , Insetos Vetores/parasitologia , Interleucina-10/genética , Interleucina-1beta/genética , Interleucina-6/genética , Medições Luminescentes , Camundongos , Neutrófilos/metabolismo , Neutrófilos/patologia , Trypanosoma brucei brucei/patogenicidade , Tripanossomíase Africana/parasitologia , Tripanossomíase Africana/transmissão , Moscas Tsé-Tsé/parasitologia , Moscas Tsé-Tsé/patogenicidade
8.
Oncotarget ; 8(32): 52016-52025, 2017 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-28881710

RESUMO

Multiple myeloma (MM) is characterized by the accumulation of malignant plasma cells in the bone marrow (BM). Recently, several studies have highlighted the role of pathogens in either promoting or dampening malignancies of unrelated origin. Trypanosoma brucei is an extracellular protozoan parasite which causes sleeping sickness. Our group has previously demonstrated that trypanosome infection affects effector plasma B cells. Therefore, we hypothesized that T. brucei infection could have an impact on MM development. Using the immunocompetent 5T33MM model, we demonstrated a significant reduction in BM-plasmacytosis and M-protein levels in mice infected with T. brucei, resulting in an increased survival of these mice. Blocking IFNγ could only partially abrogate these effects, suggesting that other mechanisms are involved in the destruction of malignant plasma cells. We found that T. brucei induces intrinsic apoptosis of 5T33MM cells in vivo, and that this was associated with reduced endogenous unfolded protein response (UPR) activation. Interestingly, pharmacological inhibition of IRE1α and PERK was sufficient to induce apoptosis in these cells. Together, these results demonstrate that trypanosome infections can interfere with MM development by suppressing endogenous UPR activation and promoting intrinsic apoptosis.

9.
Infect Immun ; 85(11)2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28808159

RESUMO

The spleen is known as an important filter for blood-borne pathogens that are trapped by specialized macrophages in the marginal zone (MZ): the CD209+ MZ macrophages (MZMs) and the CD169+ marginal metallophilic macrophages (MMMs). Acute systemic infection strongly impacts MZ populations and the location of T and B lymphocytes. This phenomenon has been linked to reduced chemokine secretion by stromal cells. Brucella spp. are the causative agent of brucellosis, a widespread zoonotic disease. Here, we used Brucella melitensis infection as a model to investigate the impact of chronic stealth infection on splenic MZ macrophage populations. During the late phase of Brucella infection, we observed a loss of both MZMs and MMMs, with a durable disappearance of MZMs, leading to a reduction of the ability of the spleen to take up soluble antigens, beads, and unrelated bacteria. This effect appears to be selective as every other lymphoid and myeloid population analyzed increased during infection, which was also observed following Brucella abortus and Brucella suis infection. Comparison of wild-type and deficient mice suggested that MZ macrophage population loss is dependent on interferon gamma (IFN-γ) receptor but independent of T cells or tumor necrosis factor alpha receptor 1 (TNF-αR1) signaling pathways and is not correlated to an alteration of CCL19, CCL21, and CXCL13 chemokine mRNA expression. Our results suggest that MZ macrophage populations are particularly sensitive to persistent low-level IFN-γ-mediated inflammation and that Brucella infection could reduce the ability of the spleen to perform certain MZM- and MMM-dependent tasks, such as antigen delivery to lymphocytes and control of systemic infection.


Assuntos
Brucelose/imunologia , Interações Hospedeiro-Patógeno , Interferon gama/imunologia , Macrófagos/imunologia , Receptores de Interferon/imunologia , Baço/imunologia , Animais , Antibacterianos/farmacologia , Linfócitos B/imunologia , Linfócitos B/microbiologia , Brucella abortus/efeitos dos fármacos , Brucella abortus/imunologia , Brucella abortus/patogenicidade , Brucella melitensis/efeitos dos fármacos , Brucella melitensis/imunologia , Brucella melitensis/patogenicidade , Brucella suis/efeitos dos fármacos , Brucella suis/imunologia , Brucella suis/patogenicidade , Brucelose/tratamento farmacológico , Brucelose/genética , Brucelose/microbiologia , Quimiocina CCL19/genética , Quimiocina CCL19/imunologia , Quimiocina CCL21/genética , Quimiocina CCL21/imunologia , Quimiocina CXCL13/genética , Quimiocina CXCL13/imunologia , Doença Crônica , Regulação da Expressão Gênica , Interferon gama/genética , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Rifampina/farmacologia , Transdução de Sinais , Baço/microbiologia , Estreptomicina/farmacologia , Linfócitos T/imunologia , Linfócitos T/microbiologia , Receptor de Interferon gama
10.
Immunobiology ; 222(6): 858-867, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28552269

RESUMO

Macrophage migration inhibitory factor (MIF) was first described as a cytokine 50 years ago, and emerged in mammals as a pleiotropic protein with pro-inflammatory, chemotactic, and growth-promoting activities. In addition, MIF has gained substantial attention as a pivotal upstream mediator of innate and adaptive immune responses and with pathologic roles in several diseases. Of less importance in mammals is an intrinsic but non-physiologic enzymatic activity that points to MIF's evolution from an ancient defense molecule. Therefore, it is not surprising that mif-like genes also have been found across a range of different organisms including bacteria, plants, protozoa, helminths, molluscs, arthropods, fish, amphibians and birds. While Genebank analysis identifying mif-like genes across species is extensive, contained herein is an overview of the non-mammalian MIF-like proteins that have been most well studied experimentally. For many of these organisms, MIF contributes to an innate defense system or plays a role in development. For parasitic organisms however, MIF appears to function as a virulence factor aiding in the establishment or persistence of infection by modulating the host immune response. Consequently, a combined targeting of both parasitic and host MIF could lead to more effective treatment strategies for parasitic diseases of socioeconomic importance.


Assuntos
Bactérias/imunologia , Imunidade Inata , Infecções/imunologia , Fatores Inibidores da Migração de Macrófagos/metabolismo , Macrófagos/imunologia , Fatores de Virulência , Animais , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune
11.
PLoS Pathog ; 12(9): e1005862, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27632207

RESUMO

Animal African trypanosomosis is a major threat to the economic development and human health in sub-Saharan Africa. Trypanosoma congolense infections represent the major constraint in livestock production, with anemia as the major pathogenic lethal feature. The mechanisms underlying anemia development are ill defined, which hampers the development of an effective therapy. Here, the contribution of the erythropoietic and erythrophagocytic potential as well as of hemodilution to the development of T. congolense-induced anemia were addressed in a mouse model of low virulence relevant for bovine trypanosomosis. We show that in infected mice, splenic extramedullary erythropoiesis could compensate for the chronic low-grade type I inflammation-induced phagocytosis of senescent red blood cells (RBCs) in spleen and liver myeloid cells, as well as for the impaired maturation of RBCs occurring in the bone marrow and spleen. Rather, anemia resulted from hemodilution. Our data also suggest that the heme catabolism subsequent to sustained erythrophagocytosis resulted in iron accumulation in tissue and hyperbilirubinemia. Moreover, hypoalbuminemia, potentially resulting from hemodilution and liver injury in infected mice, impaired the elimination of toxic circulating molecules like bilirubin. Hemodilutional thrombocytopenia also coincided with impaired coagulation. Combined, these effects could elicit multiple organ failure and uncontrolled bleeding thus reduce the survival of infected mice. MIF (macrophage migrating inhibitory factor), a potential pathogenic molecule in African trypanosomosis, was found herein to promote erythrophagocytosis, to block extramedullary erythropoiesis and RBC maturation, and to trigger hemodilution. Hence, these data prompt considering MIF as a potential target for treatment of natural bovine trypanosomosis.


Assuntos
Anemia/metabolismo , Eritropoese , Hematopoese Extramedular , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Trypanosoma congolense/metabolismo , Tripanossomíase Africana/metabolismo , Anemia/genética , Anemia/parasitologia , Anemia/patologia , Animais , Medula Óssea/metabolismo , Medula Óssea/parasitologia , Medula Óssea/patologia , Bovinos , Modelos Animais de Doenças , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Eritrócitos/patologia , Hemodiluição , Humanos , Oxirredutases Intramoleculares/genética , Fatores Inibidores da Migração de Macrófagos/genética , Camundongos , Camundongos Knockout , Baço/metabolismo , Baço/parasitologia , Baço/patologia , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombocitopenia/parasitologia , Trombocitopenia/patologia , Tripanossomíase Africana/genética , Tripanossomíase Africana/patologia
12.
PLoS One ; 10(9): e0137835, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26376185

RESUMO

Brucella are facultative intracellular Gram-negative coccobacilli that chronically infect humans as well as domestic and wild-type mammals, and cause brucellosis. Alternatively activated macrophages (M2a) induced by IL-4/IL-13 via STAT6 signaling pathways have been frequently described as a favorable niche for long-term persistence of intracellular pathogens. Based on the observation that M2a-like macrophages are induced in the spleen during the chronic phase of B. abortus infection in mice and are strongly infected in vitro, it has been suggested that M2a macrophages could be a potential in vivo niche for Brucella. In order to test this hypothesis, we used a model in which infected cells can be observed directly in situ and where the differentiation of M2a macrophages is favored by the absence of an IL-12-dependent Th1 response. We performed an in situ analysis by fluorescent microscopy of the phenotype of B. melitensis infected spleen cells from intranasally infected IL-12p40-/- BALB/c mice and the impact of STAT6 deficiency on this phenotype. Most of the infected spleen cells contained high levels of lipids and expressed CD11c and CD205 dendritic cell markers and Arginase1, but were negative for the M2a markers Fizz1 or CD301. Furthermore, STAT6 deficiency had no effect on bacterial growth or the reservoir cell phenotype in vivo, leading us to conclude that, in our model, the infected cells were not Th2-induced M2a macrophages. This characterization of B. melitensis reservoir cells could provide a better understanding of Brucella persistence in the host and lead to the design of more efficient therapeutic strategies.


Assuntos
Brucella melitensis/patogenicidade , Brucelose/microbiologia , Interleucina-12/fisiologia , Macrófagos/microbiologia , Fator de Transcrição STAT6/fisiologia , Baço/microbiologia , Animais , Brucelose/patologia , Células Cultivadas , Doença Crônica , Feminino , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Microscopia Confocal , Microscopia de Fluorescência , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/patologia
13.
Immunobiology ; 217(12): 1250-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22795648

RESUMO

Dendritic cells (DCs) play a fundamental function, either positive or detrimental, in regulating immune responses. Numerous specialized DC subsets exist in different organs. However, the trophic factors regulating their origin, location, homeostasis and functions remains to be fully understood. Recent evidence indicates that signaling via the lymphotoxin ß receptor (LTßR) can function as a trophic signaling system for specific DCs. LTßR is part of a complex signaling network that provides both positive and inhibitory signals to DC subsets. In this review, we focus on the role of LTßR expressed in DC subsets and its associated signaling pathways that regulate DC homeostasis and function. Therapeutically targeting the LTßR signaling pathway could support the development of a beneficial immune response for the host.


Assuntos
Células Dendríticas/fisiologia , Receptor beta de Linfotoxina/biossíntese , Receptor beta de Linfotoxina/genética , Transdução de Sinais/imunologia , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Homeostase/fisiologia , Receptor beta de Linfotoxina/imunologia , Receptor beta de Linfotoxina/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
14.
PLoS Pathog ; 8(3): e1002575, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22479178

RESUMO

Brucella are facultative intracellular bacteria that chronically infect humans and animals causing brucellosis. Brucella are able to invade and replicate in a broad range of cell lines in vitro, however the cells supporting bacterial growth in vivo are largely unknown. In order to identify these, we used a Brucella melitensis strain stably expressing mCherry fluorescent protein to determine the phenotype of infected cells in spleen and liver, two major sites of B. melitensis growth in mice. In both tissues, the majority of primary infected cells expressed the F4/80 myeloid marker. The peak of infection correlated with granuloma development. These structures were mainly composed of CD11b⁺ F4/80⁺ MHC-II⁺ cells expressing iNOS/NOS2 enzyme. A fraction of these cells also expressed CD11c marker and appeared similar to inflammatory dendritic cells (DCs). Analysis of genetically deficient mice revealed that differentiation of iNOS⁺ inflammatory DC, granuloma formation and control of bacterial growth were deeply affected by the absence of MyD88, IL-12p35 and IFN-γ molecules. During chronic phase of infection in susceptible mice, we identified a particular subset of DC expressing both CD11c and CD205, serving as a reservoir for the bacteria. Taken together, our results describe the cellular nature of immune effectors involved during Brucella infection and reveal a previously unappreciated role for DC subsets, both as effectors and reservoir cells, in the pathogenesis of brucellosis.


Assuntos
Brucella/imunologia , Brucelose/imunologia , Células Dendríticas/imunologia , Imunidade Inata , Pneumopatias/imunologia , Animais , Biomarcadores/metabolismo , Brucella/patogenicidade , Brucelose/microbiologia , Brucelose/patologia , Separação Celular , Células Dendríticas/microbiologia , Células Dendríticas/patologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Fígado/imunologia , Fígado/microbiologia , Fígado/patologia , Pneumopatias/microbiologia , Pneumopatias/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Fenótipo , Baço/imunologia , Baço/microbiologia , Baço/patologia
15.
Cytokine Growth Factor Rev ; 19(3-4): 277-84, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18511331

RESUMO

Dendritic cells (DC) constitute the most potent antigen presenting cells of the immune system, playing a key role bridging innate and adaptive immune responses. Specialized DC subsets differ depending on their origin, tissue location and the influence of trophic factors, the latter remain to be fully understood. Myeloid-associated lymphotoxin-beta receptor (LTbetaR) signaling is required for the local proliferation of lymphoid tissue DC. This review focuses on the LTbetaR signaling cascade as a crucial positive trophic signal in the homeostasis of DC subsets. The noncanonical coreceptor pathway comprised of the immunoglobulin (Ig) superfamily member, B and T lymphocyte attenuator (BTLA) and TNFR superfamily member, herpesvirus entry mediator (HVEM) counter regulates the trophic signaling by LTbetaR. Together both pathways form an integrated signaling circuit achieving homeostasis of DC subsets.


Assuntos
Células Dendríticas/imunologia , Receptor beta de Linfotoxina/metabolismo , Receptores Imunológicos/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Animais , Homeostase , Camundongos , NF-kappa B/metabolismo
16.
Cell Host Microbe ; 3(2): 67-76, 2008 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-18312841

RESUMO

Toll-like receptor (TLR)-dependent pathways control the production of IFNalphabeta, a key cytokine in innate immune control of viruses including mouse cytomegalovirus (MCMV). The lymphotoxin (LT) alphabeta-LTbeta receptor signaling pathway is also critical for defense against MCMV and thought to aid in the IFNbeta response. We find that upon MCMV infection, mice deficient for lymphotoxin (LT)alphabeta signaling cannot mount the initial part of a biphasic IFNalphabeta response, but show normal levels of IFNalphabeta during the sustained phase of infection. Significantly, the LTalphabeta-dependent, IFNalphabeta response is independent of TLR signaling. B, but not T, cells expressing LTbeta are essential for promoting the initial IFNalphabeta response. LTbetaR expression is required strictly in splenic stromal cells for initial IFNalphabeta production to MCMV and is dependent upon the NF-kappaB-inducing kinase (NIK). These results reveal a TLR-independent innate host defense strategy directed by B cells in communication with stromal cells via the LTalphabeta cytokine system.


Assuntos
Infecções por Herpesviridae/imunologia , Interferon Tipo I/metabolismo , Heterotrímero de Linfotoxina alfa1 e beta2/metabolismo , Receptor beta de Linfotoxina/metabolismo , Muromegalovirus/imunologia , Baço/imunologia , Células Estromais/imunologia , Animais , Linfócitos B/metabolismo , Imunidade Inata , Heterotrímero de Linfotoxina alfa1 e beta2/deficiência , Heterotrímero de Linfotoxina alfa1 e beta2/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Serina-Treonina Quinases/metabolismo , Receptor Cross-Talk , Receptores do Fator de Necrose Tumoral , Transdução de Sinais , Células Estromais/metabolismo , Quinase Induzida por NF-kappaB
17.
J Immunol ; 180(1): 238-48, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18097025

RESUMO

Proliferation of dendritic cells (DC) in the spleen is regulated by positive growth signals through the lymphotoxin (LT)-beta receptor; however, the countering inhibitory signals that achieve homeostatic control are unresolved. Mice deficient in LTalpha, LTbeta, LTbetaR, and the NFkappaB inducing kinase show a specific loss of CD8- DC subsets. In contrast, the CD8alpha- DC subsets were overpopulated in mice deficient in the herpesvirus entry mediator (HVEM) or B and T lymphocyte attenuator (BTLA). HVEM- and BTLA-deficient DC subsets displayed a specific growth advantage in repopulating the spleen in competitive replacement bone marrow chimeric mice. Expression of HVEM and BTLA were required in DC and in the surrounding microenvironment, although DC expression of LTbetaR was necessary to maintain homeostasis. Moreover, enforced activation of the LTbetaR with an agonist Ab drove expansion of CD8alpha- DC subsets, overriding regulation by the HVEM-BTLA pathway. These results indicate the HVEM-BTLA pathway provides an inhibitory checkpoint for DC homeostasis in lymphoid tissue. Together, the LTbetaR and HVEM-BTLA pathways form an integrated signaling network regulating DC homeostasis.


Assuntos
Células Dendríticas/imunologia , Receptor beta de Linfotoxina/metabolismo , Receptores Imunológicos/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Animais , Antígenos CD8/análise , Proliferação de Células , Homeostase , Receptor beta de Linfotoxina/genética , Linfotoxina-alfa/genética , Linfotoxina-alfa/metabolismo , Linfotoxina-beta/genética , Linfotoxina-beta/metabolismo , Camundongos , Camundongos Mutantes , Receptores Imunológicos/genética , Membro 14 de Receptores do Fator de Necrose Tumoral/genética
18.
J Immunol ; 179(4): 2195-202, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17675479

RESUMO

The mechanisms that regulate CMV-specific T cell responses in vivo are poorly understood. During murine CMV infection of B6 mice, primary responses in the spleen are dominated by CD8 T cells reactive with antigenic epitopes in M45, M57, and m139 murine CMV gene products. However, during the later persistent phase of infection, CD8 T cell responses to epitopes in m139 and M38 viral gene products predominate. The basis for this shift in CD8 T populations is unknown. In this study, we demonstrate that OX40, a TNFR superfamily member, specifically regulates the accumulation of CD8 T cells reactive with the persistent-phase epitopes. Defective CD8 T cell responses in OX40(-/-) mice were replicated in MHC class II(-/-) mice implying that CD4 T cells in part controlled the differentiation of the CD8 T cell clones responsive to these epitopes during persistent infection. Furthermore, treatment of infected mice with an agonist OX40 Ab induced expansion of protective primary virus-specific CD8 T cells independent of CD4 T cell help, but CD4 T cells were crucial for anti-OX40 to promote CD8 T cells reactive to the persistent dominant epitopes. Collectively, these results indicate manipulation of OX40 may be useful in improving cellular immunotherapy regimes for treatment of persistent virus infections.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Infecções por Herpesviridae/imunologia , Muromegalovirus/imunologia , Receptores OX40/imunologia , Proteínas Virais/imunologia , Transferência Adotiva , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Proliferação de Células , Infecções por Herpesviridae/terapia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Camundongos , Camundongos Knockout , Receptores OX40/deficiência
19.
J Exp Med ; 204(5): 1217-25, 2007 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-17485516

RESUMO

The salivary glands represent a major site of cytomegalovirus replication and transmission to other hosts. Despite control of viral infection by strong T cell responses in visceral organs cytomegalovirus replication continues in the salivary glands of mice, suggesting that the virus exploits the mucosal microenvironment. Here, we show that T cell immunity in the salivary glands is limited by the induction of CD4 T cells expressing the regulatory cytokine interleukin (IL)-10. Blockade of IL-10 receptor (IL-10R) with an antagonist antibody dramatically reduced viral load in the salivary glands, but not in the spleen. The mucosa-specific protection afforded by IL-10R blockade was associated with an increased accumulation of CD4 T cells expressing interferon gamma, suggesting that IL-10R signaling limits effector T cell differentiation. Consistent with this, an agonist antibody targeting the tumor necrosis factor receptor superfamily member OX40 (TNFRSF4) enhanced effector T cell differentiation and increased the number of interferon gamma-producing T cells, thus limiting virus replication in the salivary glands. Collectively, the results indicate that modulating effector T cell differentiation can counteract pathogen exploitation of the mucosa, thus limiting persistent virus replication and transmission.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/imunologia , Infecções por Herpesviridae/imunologia , Interleucina-10/metabolismo , Muromegalovirus/fisiologia , Glândulas Salivares/imunologia , Replicação Viral/fisiologia , Animais , Anticorpos/farmacologia , Linfócitos T CD4-Positivos/imunologia , Primers do DNA , Feminino , Citometria de Fluxo , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Muromegalovirus/imunologia , Receptores de Interleucina-10/antagonistas & inibidores , Receptores OX40/agonistas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Glândulas Salivares/virologia
20.
PLoS Pathog ; 2(3): e16, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16518465

RESUMO

Efficient immune defenses are facilitated by the organized microarchitecture of lymphoid organs, and this organization is regulated by the compartmentalized expression of lymphoid tissue chemokines. Mouse cytomegalovirus (MCMV) infection induces significant remodeling of splenic microarchitecture, including loss of marginal zone macrophage populations and dissolution of T and B cell compartmentalization. MCMV preferentially infected the splenic stroma, targeting endothelial cells (EC) as revealed using MCMV-expressing green fluorescent protein. MCMV infection caused a specific, but transient transcriptional suppression of secondary lymphoid chemokine (CCL21). The loss of CCL21 was associated with the failure of T lymphocytes to locate within the T cell zone, although trafficking to the spleen was unaltered. Expression of CCL21 in lymphotoxin (LT)-alpha-deficient mice is dramatically reduced, however MCMV infection further reduced CCL21 levels, suggesting that viral modulation of CCL21 was independent of LTalpha signaling. Activation of LTbeta-receptor signaling with an agonistic antibody partially restored CCL21 mRNA expression and redirected transferred T cells to the splenic T cell zone in MCMV-infected mice. These results indicate that virus-induced alterations in lymphoid tissues can occur through an LT-independent modulation of chemokine transcription, and targeting of the LT cytokine system can counteract lymphoid tissue remodeling by MCMV.


Assuntos
Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Baço/patologia , Baço/virologia , Animais , Quimiocina CCL21 , Quimiocinas CC/antagonistas & inibidores , Quimiocinas CC/genética , Infecções por Citomegalovirus/metabolismo , Receptor beta de Linfotoxina , Linfotoxina-alfa/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Receptores do Fator de Necrose Tumoral/agonistas , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Baço/metabolismo , Linfócitos T/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA