Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 130, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38167258

RESUMO

TBK1 and IKKε regulate multiple cellular processes including anti-viral type-I interferon responses, metabolism and TNF receptor signaling. However, the relative contributions and potentially redundant functions of IKKε and TBK1 in cell death, inflammation and tissue homeostasis remain poorly understood. Here we show that IKKε compensates for the loss of TBK1 kinase activity to prevent RIPK1-dependent and -independent inflammation in mice. Combined inhibition of IKKε and TBK1 kinase activities caused embryonic lethality that was rescued by heterozygous expression of kinase-inactive RIPK1. Adult mice expressing kinase-inactive versions of IKKε and TBK1 developed systemic inflammation that was induced by both RIPK1-dependent and -independent mechanisms. Combined inhibition of IKKε and TBK1 kinase activities in myeloid cells induced RIPK1-dependent cell death and systemic inflammation mediated by IL-1 family cytokines. Tissue-specific studies showed that IKKε and TBK1 were required to prevent cell death and inflammation in the intestine but were dispensable for liver and skin homeostasis. Together, these findings revealed that IKKε and TBK1 exhibit tissue-specific functions that are important to prevent cell death and inflammation and maintain tissue homeostasis.


Assuntos
Quinase I-kappa B , Proteínas Serina-Treonina Quinases , Animais , Camundongos , Quinase I-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fosforilação , Citocinas/metabolismo , Inflamação
2.
Front Cell Infect Microbiol ; 12: 944819, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36034693

RESUMO

Inducible nitric oxide synthase (iNOS) is essential to the production of nitric oxide (NO), an efficient effector molecule against intracellular human pathogens such as Leishmania protozoan parasites. Some strains of Leishmania are known to bear a viral endosymbiont termed Leishmania RNA virus 1 (LRV1). Recognition of LRV1 by the innate immune sensor Toll-like receptor-3 (TLR3) leads to conditions worsening the disease severity in mice. This process is governed by type I interferon (type I IFNs) arising downstream of TLR3 stimulation and favoring the formation of secondary metastatic lesions. The formation of these lesions is mediated by the inflammatory cytokine IL-17A and occurs in the absence, or low level of, protective cytokine IFN-γ. Here, we described that the presence of LRV1 led to the initial expression of iNOS and low production of NO that failed to control infection. We subsequently showed that LRV1-triggered type I IFN was essential but insufficient to induce robust iNOS induction, which requires strong activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Leishmania guyanensis carrying LRV1 (LgyLRV1+) parasites mitigated strong iNOS production by limiting NF-kB activation via the induction of tumor necrosis factor-alpha-induced protein 3 (TNFAIP3), also known as A20. Moreover, our data suggested that production of LRV1-induced iNOS could be correlated with parasite dissemination and metastasis via elevated secretion of IL-17A in the draining lymph nodes. Our findings support an additional strategy by which LRV1-bearing Leishmania guyanensis evaded killing by nitric oxide and suggest that low levels of LRV1-induced NO might contribute to parasite metastasis.


Assuntos
Leishmania guyanensis , Leishmania , Óxido Nítrico Sintase Tipo II , Animais , Citocinas , Humanos , Interleucina-17 , Leishmania guyanensis/virologia , Leishmaniavirus , Camundongos , NF-kappa B , Óxido Nítrico , Óxido Nítrico Sintase Tipo II/metabolismo , Receptor 3 Toll-Like
3.
PLoS Pathog ; 17(3): e1009422, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33765083

RESUMO

The oxidative burst generated by the host immune system can restrict intracellular parasite entry and growth. While this burst leads to the induction of antioxidative enzymes, the molecular mechanisms and the consequences of this counter-response on the life of intracellular human parasites are largely unknown. The transcription factor NF-E2-related factor (NRF2) could be a key mediator of antioxidant signaling during infection due to the entry of parasites. Here, we showed that NRF2 was strongly upregulated in infection with the human Leishmania protozoan parasites, its activation was dependent on a NADPH oxidase 2 (NOX2) and SRC family of protein tyrosine kinases (SFKs) signaling pathway and it reprogrammed host cell metabolism. In inflammatory leishmaniasis caused by a viral endosymbiont inducing TNF-α in chronic leishmaniasis, NRF2 activation promoted parasite persistence but limited TNF-α production and tissue destruction. These data provided evidence of the dual role of NRF2 in protecting both the invading pathogen from reactive oxygen species and the host from an excess of the TNF-α destructive pro-inflammatory cytokine.


Assuntos
Interações Hospedeiro-Parasita/fisiologia , Leishmania/metabolismo , Leishmaniose/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/fisiologia , Animais , Inflamação/imunologia , Inflamação/metabolismo , Leishmania/imunologia , Leishmaniose/imunologia , Camundongos , Fator 2 Relacionado a NF-E2/imunologia , Transdução de Sinais/imunologia
4.
Nature ; 580(7803): 391-395, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32296175

RESUMO

The biological function of Z-DNA and Z-RNA, nucleic acid structures with a left-handed double helix, is poorly understood1-3. Z-DNA-binding protein 1 (ZBP1; also known as DAI or DLM-1) is a nucleic acid sensor that contains two Zα domains that bind Z-DNA4,5 and Z-RNA6-8. ZBP1 mediates host defence against some viruses6,7,9-14 by sensing viral nucleic acids6,7,10. RIPK1 deficiency, or mutation of its RIP homotypic interaction motif (RHIM), triggers ZBP1-dependent necroptosis and inflammation in mice15,16. However, the mechanisms that induce ZBP1 activation in the absence of viral infection remain unknown. Here we show that Zα-dependent sensing of endogenous ligands induces ZBP1-mediated perinatal lethality in mice expressing RIPK1 with mutated RHIM (Ripk1mR/mR), skin inflammation in mice with epidermis-specific RIPK1 deficiency (RIPK1E-KO) and colitis in mice with intestinal epithelial-specific FADD deficiency (FADDIEC-KO). Consistently, functional Zα domains were required for ZBP1-induced necroptosis in fibroblasts that were treated with caspase inhibitors or express RIPK1 with mutated RHIM. Inhibition of nuclear export triggered the Zα-dependent activation of RIPK3 in the nucleus resulting in cell death, which suggests that ZBP1 may recognize nuclear Z-form nucleic acids. We found that ZBP1 constitutively bound cellular double-stranded RNA in a Zα-dependent manner. Complementary reads derived from endogenous retroelements were detected in epidermal RNA, which suggests that double-stranded RNA derived from these retroelements may act as a Zα-domain ligand that triggers the activation of ZBP1. Collectively, our results provide evidence that the sensing of endogenous Z-form nucleic acids by ZBP1 triggers RIPK3-dependent necroptosis and inflammation, which could underlie the development of chronic inflammatory conditions-particularly in individuals with mutations in RIPK1 and CASP817-20.


Assuntos
Inflamação/metabolismo , Necroptose , Proteínas de Ligação a RNA/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Caspase 8/metabolismo , Feminino , Inflamação/genética , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ácidos Nucleicos/metabolismo , RNA de Cadeia Dupla/metabolismo , Proteínas de Ligação a RNA/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Dermatopatias/genética , Dermatopatias/metabolismo , Dermatopatias/patologia
5.
Nature ; 580(7804): E10, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32322058

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
Nat Cell Biol ; 21(6): 731-742, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31086261

RESUMO

Deficiency in the deubiquitinating enzyme A20 causes severe inflammation in mice, and impaired A20 function is associated with human inflammatory diseases. A20 has been implicated in negatively regulating NF-κB signalling, cell death and inflammasome activation; however, the mechanisms by which A20 inhibits inflammation in vivo remain poorly understood. Genetic studies in mice revealed that its deubiquitinase activity is not essential for A20 anti-inflammatory function. Here we show that A20 prevents inflammasome-dependent arthritis by inhibiting macrophage necroptosis and that this function depends on its zinc finger 7 (ZnF7). We provide genetic evidence that RIPK1 kinase-dependent, RIPK3-MLKL-mediated necroptosis drives inflammasome activation in A20-deficient macrophages and causes inflammatory arthritis in mice. Single-cell imaging revealed that RIPK3-dependent death caused inflammasome-dependent IL-1ß release from lipopolysaccharide-stimulated A20-deficient macrophages. Importantly, mutation of the A20 ZnF7 ubiquitin binding domain caused arthritis in mice, arguing that ZnF7-dependent inhibition of necroptosis is critical for A20 anti-inflammatory function in vivo.


Assuntos
Artrite/genética , Inflamação/genética , Fatores de Transcrição Kruppel-Like/genética , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética , Animais , Artrite/induzido quimicamente , Artrite/patologia , Humanos , Inflamassomos/genética , Inflamassomos/metabolismo , Inflamação/induzido quimicamente , Inflamação/patologia , Interleucina-1beta/genética , Lipopolissacarídeos/toxicidade , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Mutação , NF-kappa B/genética , Necrose/genética , Necrose/patologia , Ligação Proteica , Proteínas Quinases/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Ubiquitina/genética
7.
Front Biosci (Elite Ed) ; 10(2): 242-253, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28930616

RESUMO

We previously reported that the innate sensing of the endosymbiont Leishmania RNA virus 1 (LRV1) within Leishmania (Viannia) guyanensis through Toll-like receptor 3, worsens the pathogenesis of parasite infection in mice. The presence of LRV1 has been associated with the failure of first-line treatment in patients infected with LRV1 containing -L. guyanensis and -L. braziliensis parasites. Here, we established a semi-automated image-based high-throughput drug screening (HTDS) protocol to measure parasiticidal activity of the Prestwick chemical library in primary murine macrophages infected with LRV1-containing L. guyanensis. The two-independent screens generated 14 hit compounds with over sixty-nine percent reduction in parasite growth compared to control, at a single dose in both screens. Our screening strategy offers great potential in the search for new drugs and accelerates the discovery rate in the field of drug repurposing against Leishmania. Moreover, this technique allows the concomitant assessment of the effect of drug toxicity on host cell number.


Assuntos
Automação , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala , Animais , Antiprotozoários/farmacologia , Leishmania/efeitos dos fármacos , Macrófagos/parasitologia , Camundongos , Camundongos Endogâmicos C57BL
8.
Proc Natl Acad Sci U S A ; 114(19): 4987-4992, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28439019

RESUMO

The presence of the endogenous Leishmania RNA virus 1 (LRV1) replicating stably within some parasite species has been associated with the development of more severe forms of leishmaniasis and relapses after drug treatment in humans. Here, we show that the disease-exacerbatory role of LRV1 relies on type I IFN (type I IFNs) production by macrophages and signaling in vivo. Moreover, infecting mice with the LRV1-cured Leishmania guyanensis (LgyLRV1- ) strain of parasites followed by type I IFN treatment increased lesion size and parasite burden, quantitatively reproducing the LRV1-bearing (LgyLRV1+ ) infection phenotype. This finding suggested the possibility that exogenous viral infections could likewise increase pathogenicity, which was tested by coinfecting mice with L. guyanensis and lymphocytic choriomeningitis virus (LCMV), or the sand fly-transmitted arbovirus Toscana virus (TOSV). The type I IFN antiviral response increased the pathology of L. guyanensis infection, accompanied by down-regulation of the IFN-γ receptor normally required for antileishmanial control. Further, LCMV coinfection of IFN-γ-deficient mice promoted parasite dissemination to secondary sites, reproducing the LgyLRV1+ metastatic phenotype. Remarkably, LCMV coinfection of mice that had healed from L. guyanensis infection induced reactivation of disease pathology, overriding the protective adaptive immune response. Our findings establish that type I IFN-dependent responses, arising from endogenous viral elements (dsRNA/LRV1), or exogenous coinfection with IFN-inducing viruses, are able to synergize with New World Leishmania parasites in both primary and relapse infections. Thus, viral infections likely represent a significant risk factor along with parasite and host factors, thereby contributing to the pathological spectrum of human leishmaniasis.


Assuntos
Interferon Tipo I/imunologia , Leishmania guyanensis , Leishmaniose Mucocutânea/imunologia , Leishmaniavirus/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Febre por Flebótomos/imunologia , Vírus da Febre do Flebótomo Napolitano/imunologia , Animais , Coinfecção , Interferon Tipo I/genética , Leishmania guyanensis/imunologia , Leishmania guyanensis/virologia , Leishmaniose Mucocutânea/genética , Leishmaniose Mucocutânea/patologia , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/patologia , Camundongos , Camundongos Knockout , Febre por Flebótomos/genética , Febre por Flebótomos/patologia
9.
PLoS Negl Trop Dis ; 11(1): e0005240, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28099431

RESUMO

Recent studies have shown that a cytoplasmic virus called Leishmaniavirus (LRV) is present in some Leishmania species and acts as a potent innate immunogen, aggravating lesional inflammation and development in mice. In humans, the presence of LRV in Leishmania guyanensis and in L. braziliensis was significantly correlated with poor treatment response and symptomatic relapse. So far, no clinical effort has used LRV for prophylactic purposes. In this context, we designed an original vaccine strategy that targeted LRV nested in Leishmania parasites to prevent virus-related complications. To this end, C57BL/6 mice were immunized with a recombinant LRV1 Leishmania guyanensis viral capsid polypeptide formulated with a T helper 1-polarizing adjuvant. LRV1-vaccinated mice had significant reduction in lesion size and parasite load when subsequently challenged with LRV1+ Leishmania guyanensis parasites. The protection conferred by this immunization could be reproduced in naïve mice via T-cell transfer from vaccinated mice but not by serum transfer. The induction of LRV1 specific T cells secreting IFN-γ was confirmed in vaccinated mice and provided strong evidence that LRV1-specific protection arose via a cell mediated immune response against the LRV1 capsid. Our studies suggest that immunization with LRV1 capsid could be of a preventive benefit in mitigating the elevated pathology associated with LRV1 bearing Leishmania infections and possibly avoiding symptomatic relapses after an initial treatment. This novel anti-endosymbiotic vaccine strategy could be exploited to control other infectious diseases, as similar viral infections are largely prevalent across pathogenic pathogens and could consequently open new vaccine opportunities.


Assuntos
Proteínas do Capsídeo/imunologia , Leishmania guyanensis/virologia , Leishmaniose/prevenção & controle , Leishmaniavirus/imunologia , Animais , Proteínas do Capsídeo/administração & dosagem , Proteínas do Capsídeo/genética , Feminino , Humanos , Imunidade Celular , Leishmania guyanensis/genética , Leishmania guyanensis/imunologia , Leishmania guyanensis/fisiologia , Leishmaniose/imunologia , Leishmaniose/parasitologia , Leishmaniavirus/genética , Leishmaniavirus/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Simbiose , Linfócitos T/imunologia , Vacinação
10.
Bio Protoc ; 7(16): e2509, 2017 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34541172

RESUMO

Macrophages maintain tissue homoeostasis by regulating inflammation and tissue repair mechanisms. Thus, the fate of macrophages has an impact on the state of the tissue. The aim of this protocol is to quantify macrophage survival using high content microscopy and image processing software. Here, we describe a high-content image based protocol to assess the effect of diverse stimuli in combination with pharmacological treatments on macrophage survival in a quantitative, unbiased and high-throughput manner.

11.
Cell Host Microbe ; 20(3): 318-328, 2016 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-27593513

RESUMO

Some strains of the protozoan parasite Leishmania guyanensis (L.g) harbor a viral endosymbiont called Leishmania RNA virus 1 (LRV1). LRV1 recognition by TLR-3 increases parasite burden and lesion swelling in vivo. However, the mechanisms by which anti-viral innate immune responses affect parasitic infection are largely unknown. Upon investigating the mammalian host's response to LRV1, we found that miR-155 was singularly and strongly upregulated in macrophages infected with LRV1+ L.g when compared to LRV1- L.g. LRV1-driven miR-155 expression was dependent on TLR-3/TRIF signaling. Furthermore, LRV1-induced TLR-3 activation promoted parasite persistence by enhancing macrophage survival through Akt activation in a manner partially dependent on miR-155. Pharmacological inhibition of Akt resulted in a decrease in LRV1-mediated macrophage survival and consequently decreased parasite persistence. Consistent with these data, miR-155-deficient mice showed a drastic decrease in LRV1-induced disease severity, and lesional macrophages from these mice displayed reduced levels of Akt phosphorylation.


Assuntos
Imunidade Inata , Leishmania guyanensis/virologia , Leishmaniavirus/imunologia , Macrófagos/parasitologia , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor 3 Toll-Like/metabolismo , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Leishmania guyanensis/patogenicidade , Leishmania guyanensis/fisiologia , Leishmaniose Mucocutânea/parasitologia , Leishmaniose Mucocutânea/patologia , Macrófagos/imunologia , Camundongos , Camundongos Knockout
12.
PLoS Pathog ; 12(9): e1005852, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27658195

RESUMO

Cutaneous leishmaniasis has various outcomes, ranging from self-healing reddened papules to extensive open ulcerations that metastasise to secondary sites and are often resistant to standard therapies. In the case of L. guyanensis (L.g), about 5-10% of all infections result in metastatic complications. We recently showed that a cytoplasmic virus within L.g parasites (LRV1) is able to act as a potent innate immunogen, worsening disease outcome in a murine model. In this study, we investigated the immunophenotype of human patients infected by L.g and found a significant association between the inflammatory cytokine IL-17A, the presence of LRV1 and disease chronicity. Further, IL-17A was inversely correlated to the protective cytokine IFN-γ. These findings were experimentally corroborated in our murine model, where IL-17A produced in LRV1+ L.g infection contributed to parasite virulence and dissemination in the absence of IFN-γ. Additionally, IL-17A inhibition in mice using digoxin or SR1001, showed therapeutic promise in limiting parasite virulence. Thus, this murine model of LRV1-dependent infectious metastasis validated markers of disease chronicity in humans and elucidated the immunologic mechanism for the dissemination of Leishmania parasites to secondary sites. Moreover, it confirms the prognostic value of LRV1 and IL-17A detection to prevent metastatic leishmaniasis in human patients.

13.
PLoS One ; 9(1): e86844, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24489792

RESUMO

Immune responses against intestinal microbiota contribute to the pathogenesis of inflammatory bowel diseases (IBD) and involve CD4(+) T cells, which are activated by major histocompatibility complex class II (MHCII) molecules on antigen-presenting cells (APCs). However, it is largely unexplored how inflammation-induced MHCII expression by intestinal epithelial cells (IEC) affects CD4(+) T cell-mediated immunity or tolerance induction in vivo. Here, we investigated how epithelial MHCII expression is induced and how a deficiency in inducible epithelial MHCII expression alters susceptibility to colitis and the outcome of colon-specific immune responses. Colitis was induced in mice that lacked inducible expression of MHCII molecules on all nonhematopoietic cells, or specifically on IECs, by continuous infection with Helicobacter hepaticus and administration of interleukin (IL)-10 receptor-blocking antibodies (anti-IL10R mAb). To assess the role of interferon (IFN)-γ in inducing epithelial MHCII expression, the T cell adoptive transfer model of colitis was used. Abrogation of MHCII expression by nonhematopoietic cells or IECs induces colitis associated with increased colonic frequencies of innate immune cells and expression of proinflammatory cytokines. CD4(+) T-helper type (Th)1 cells - but not group 3 innate lymphoid cells (ILCs) or Th17 cells - are elevated, resulting in an unfavourably altered ratio between CD4(+) T cells and forkhead box P3 (FoxP3)(+) regulatory T (Treg) cells. IFN-γ produced mainly by CD4(+) T cells is required to upregulate MHCII expression by IECs. These results suggest that, in addition to its proinflammatory roles, IFN-γ exerts a critical anti-inflammatory function in the intestine which protects against colitis by inducing MHCII expression on IECs. This may explain the failure of anti-IFN-γ treatment to induce remission in IBD patients, despite the association of elevated IFN-γ and IBD.


Assuntos
Colite/imunologia , Colite/prevenção & controle , Enterócitos/metabolismo , Interferon gama/metabolismo , Proteínas Nucleares/metabolismo , Transativadores/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Quimiocinas/metabolismo , Colite/patologia , Colo/efeitos dos fármacos , Colo/imunologia , Colo/patologia , Enterócitos/efeitos dos fármacos , Enterócitos/patologia , Fatores de Transcrição Forkhead/metabolismo , Helicobacter/efeitos dos fármacos , Helicobacter/fisiologia , Infecções por Helicobacter/imunologia , Imunidade Inata/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Queratina-14/genética , Contagem de Linfócitos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , Receptores de Interleucina-10/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Transativadores/genética , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...