Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Science ; 384(6692): eadk6200, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38574174

ABSTRACT

Males and females exhibit profound differences in immune responses and disease susceptibility. However, the factors responsible for sex differences in tissue immunity remain poorly understood. Here, we uncovered a dominant role for type 2 innate lymphoid cells (ILC2s) in shaping sexual immune dimorphism within the skin. Mechanistically, negative regulation of ILC2s by androgens leads to a reduction in dendritic cell accumulation and activation in males, along with reduced tissue immunity. Collectively, our results reveal a role for the androgen-ILC2-dendritic cell axis in controlling sexual immune dimorphism. Moreover, this work proposes that tissue immune set points are defined by the dual action of sex hormones and the microbiota, with sex hormones controlling the strength of local immunity and microbiota calibrating its tone.


Subject(s)
Androgens , Dendritic Cells , Immunity, Innate , Lymphocytes , Sex Characteristics , Skin , Female , Male , Androgens/metabolism , Dendritic Cells/immunology , Gonadal Steroid Hormones/metabolism , Lymphocytes/immunology , Skin/immunology , Animals , Mice , Mice, Inbred C57BL , Microbiota
2.
Sci Immunol ; 8(80): eadd4132, 2023 02 24.
Article in English | MEDLINE | ID: mdl-36827419

ABSTRACT

Plasmacytoid dendritic cells (pDCs) have been shown to play an important role during immune responses, ranging from initial viral control through the production of type I interferons to antigen presentation. However, recent studies uncovered unexpected heterogeneity among pDCs. We identified a previously uncharacterized immune subset, referred to as pDC-like cells, that not only resembles pDCs but also shares conventional DC (cDC) features. We show that this subset is a circulating precursor distinct from common DC progenitors, with prominent cDC2 potential. Our findings from human CD2-iCre and CD300c-iCre lineage tracing mouse models suggest that a substantial fraction of cDC2s originates from pDC-like cells, which can therefore be referred to as pre-DC2. This precursor subset responds to homeostatic cytokines, such as macrophage colony stimulating factor, by expanding and differentiating into cDC2 that efficiently prime T helper 17 (TH17) cells. Development of pre-DC2 into CX3CR1+ ESAM- cDC2b but not CX3CR1- ESAM+ cDC2a requires the transcription factor KLF4. Last, we show that, under homeostatic conditions, this developmental pathway regulates the immune threshold at barrier sites by controlling the pool of TH17 cells within skin-draining lymph nodes.


Subject(s)
CD4-Positive T-Lymphocytes , Gene Expression Regulation , Mice , Animals , Humans , CD4-Positive T-Lymphocytes/metabolism , Antigen Presentation , Th17 Cells/metabolism , Cells, Cultured , Dendritic Cells , Antigens, Surface , Membrane Glycoproteins
3.
Proc Natl Acad Sci U S A ; 119(26): e2200348119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35727974

ABSTRACT

Immune checkpoint inhibitors (ICIs) are essential components of the cancer therapeutic armamentarium. While ICIs have demonstrated remarkable clinical responses, they can be accompanied by immune-related adverse events (irAEs). These inflammatory side effects are of unclear etiology and impact virtually all organ systems, with the most common being sites colonized by the microbiota such as the skin and gastrointestinal tract. Here, we establish a mouse model of commensal bacteria-driven skin irAEs and demonstrate that immune checkpoint inhibition unleashes commensal-specific inflammatory T cell responses. These aberrant responses were dependent on production of IL-17 by commensal-specific T cells and induced pathology that recapitulated the cutaneous inflammation seen in patients treated with ICIs. Importantly, aberrant T cell responses unleashed by ICIs were sufficient to perpetuate inflammatory memory responses to the microbiota months following the cessation of treatment. Altogether, we have established a mouse model of skin irAEs and reveal that ICIs unleash aberrant immune responses against skin commensals, with long-lasting inflammatory consequences.


Subject(s)
Dermatitis , Immune Checkpoint Inhibitors , Microbiota , Animals , Dermatitis/immunology , Dermatitis/microbiology , Disease Models, Animal , Immune Checkpoint Inhibitors/adverse effects , Immunity/drug effects , Interleukin-17/metabolism , Mice , Microbiota/drug effects , Microbiota/immunology , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/immunology , Symbiosis/drug effects , T-Lymphocytes/immunology
4.
Science ; 373(6558)2021 08 27.
Article in English | MEDLINE | ID: mdl-34446580

ABSTRACT

The immune system has evolved in the face of microbial exposure. How maternal infection experienced at distinct developmental stages shapes the offspring immune system remains poorly understood. Here, we show that during pregnancy, maternally restricted infection can have permanent and tissue-specific impacts on offspring immunity. Mechanistically, maternal interleukin-6 produced in response to infection can directly impose epigenetic changes on fetal intestinal epithelial stem cells, leading to long-lasting impacts on intestinal immune homeostasis. As a result, offspring of previously infected dams develop enhanced protective immunity to gut infection and increased inflammation in the context of colitis. Thus, maternal infection can be coopted by the fetus to promote long-term, tissue-specific fitness, a phenomenon that may come at the cost of predisposition to inflammatory disorders.


Subject(s)
Colitis/immunology , Immunity , Interleukin-6/immunology , Intestines/immunology , Pregnancy Complications, Infectious/immunology , Th17 Cells/immunology , Yersinia pseudotuberculosis Infections/immunology , Animals , Candidiasis/immunology , Chromatin/metabolism , Epigenesis, Genetic , Epigenome , Female , Fetal Development , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Interleukin-6/blood , Interleukin-6/pharmacology , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestinal Mucosa/immunology , Intestines/embryology , Intestines/microbiology , Mice , Pregnancy , Prenatal Exposure Delayed Effects , Salmonella Infections, Animal/immunology , Stem Cells/immunology , Stem Cells/physiology , T-Lymphocyte Subsets/immunology
5.
Cell ; 184(14): 3794-3811.e19, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34166614

ABSTRACT

The microbiota plays a fundamental role in regulating host immunity. However, the processes involved in the initiation and regulation of immunity to the microbiota remain largely unknown. Here, we show that the skin microbiota promotes the discrete expression of defined endogenous retroviruses (ERVs). Keratinocyte-intrinsic responses to ERVs depended on cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes protein (STING) signaling and promoted the induction of commensal-specific T cells. Inhibition of ERV reverse transcription significantly impacted these responses, resulting in impaired immunity to the microbiota and its associated tissue repair function. Conversely, a lipid-enriched diet primed the skin for heightened ERV- expression in response to commensal colonization, leading to increased immune responses and tissue inflammation. Together, our results support the idea that the host may have co-opted its endogenous virome as a means to communicate with the exogenous microbiota, resulting in a multi-kingdom dialog that controls both tissue homeostasis and inflammation.


Subject(s)
Endogenous Retroviruses/physiology , Homeostasis , Inflammation/microbiology , Inflammation/pathology , Microbiota , Animals , Bacteria/metabolism , Chromosomes, Bacterial/genetics , Diet, High-Fat , Inflammation/immunology , Inflammation/virology , Interferon Type I/metabolism , Keratinocytes/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , Nucleotidyltransferases/metabolism , Retroelements/genetics , Signal Transduction , Skin/immunology , Skin/microbiology , T-Lymphocytes/immunology , Transcription, Genetic
6.
iScience ; 24(1): 102004, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33490912

ABSTRACT

New World species of the intracellular protozoan parasites of the Leishmania genus can cause mucocutaneous leishmaniases. The presence of an endosymbiotic Leishmania RNA virus (LRV) in Leishmania guyanensis (L.g.) promotes disease exacerbation and the development of mucocutaneous disease. It was previously reported that LRV blocks the NLRP3 inflammasome, but additional mechanisms remain unclear. Here, we investigated whether LRV interferes with the inflammasome via caspase-11, which induces non-canonical NLRP3 activation and was reported to be activated by Leishmania. By using macrophages and mice, we found that LRV inhibits caspase-11 activation and IL-1ß release by L.g. in a TLR3- and ATG5-dependent manner. Moreover, LRV exacerbates disease in C57BL/6 mice but not in Casp11 -/- , Nlrp3 -/- , and 129 mice, a mouse strain that is naturally mutant for caspase-11. These results demonstrate that LRV interferes with caspase-11 activation by Leishmania, expanding our understanding about the mechanisms by which LRV promotes disease exacerbation.

8.
Nat Commun ; 10(1): 5273, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31754185

ABSTRACT

Leishmania RNA virus (LRV) is an important virulence factor associated with the development of mucocutaneous Leishmaniasis, a severe form of the disease. LRV-mediated disease exacerbation relies on TLR3 activation, but downstream mechanisms remain largely unexplored. Here, we combine human and mouse data to demonstrate that LRV triggers TLR3 and TRIF to induce type I IFN production, which induces autophagy. This process results in ATG5-mediated degradation of NLRP3 and ASC, thereby limiting NLRP3 inflammasome activation in macrophages. Consistent with the known restricting role of NLRP3 for Leishmania replication, the signaling pathway triggered by LRV results in increased parasite survival and disease progression. In support of this data, we find that lesions in patients infected with LRV+ Leishmania are associated with reduced inflammasome activation and the development of mucocutaneous disease. Our findings reveal the mechanisms triggered by LRV that contribute to the development of the debilitating mucocutaneous form of Leishmaniasis.


Subject(s)
Immunity, Innate/immunology , Inflammasomes/immunology , Leishmania/immunology , Leishmaniasis, Mucocutaneous/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , RNA Viruses/immunology , Toll-Like Receptor 3/immunology , Animals , Autophagy/immunology , Humans , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Leishmania/physiology , Leishmania/virology , Leishmaniasis, Mucocutaneous/parasitology , Leishmaniasis, Mucocutaneous/virology , Macrophages/immunology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , RNA Viruses/physiology , Signal Transduction/immunology , Toll-Like Receptor 3/metabolism
9.
J Infect Dis ; 219(12): 2015-2025, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30715407

ABSTRACT

Rocio virus (ROCV) is a highly neuropathogenic mosquito-transmitted flavivirus responsible for an unprecedented outbreak of human encephalitis during 1975-1976 in Sao Paulo State, Brazil. Previous studies have shown an increased number of inflammatory macrophages in the central nervous system (CNS) of ROCV-infected mice, implying a role for macrophages in the pathogenesis of ROCV. Here, we show that ROCV infection results in increased expression of CCL2 in the blood and in infiltration of macrophages into the brain. Moreover, we show, using CCR2 knockout mice, that CCR2 expression is essential for macrophage infiltration in the brain during ROCV infection and that the lack of CCR2 results in increased disease severity and mortality. Thus, our findings show the protective role of CCR2-mediated infiltration of macrophages in the brain during ROCV infection.


Subject(s)
Encephalitis/metabolism , Flavivirus Infections/metabolism , Flavivirus/pathogenicity , Macrophages/metabolism , Receptors, CCR2/metabolism , Animals , Brain , Brazil , Encephalitis/virology , Female , Flavivirus Infections/virology , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Cell Rep ; 26(2): 429-437.e5, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30625325

ABSTRACT

Activation of the NLRP3 inflammasome by Leishmania parasites is critical for the outcome of leishmaniasis, a disease that affects millions of people worldwide. We investigate the mechanisms involved in NLRP3 activation and demonstrate that caspase-11 (CASP11) is activated in response to infection by Leishmania species and triggers the non-canonical activation of NLRP3. This process accounts for host resistance to infection in macrophages and in vivo. We identify the parasite membrane glycoconjugate lipophosphoglycan (LPG) as the molecule involved in CASP11 activation. Cytosolic delivery of LPG in macrophages triggers CASP11 activation, and infections performed with Lpg1-/- parasites reduce CASP11/NLRP3 activation. Unlike bacterial LPS, purified LPG does not activate mouse CASP11 (or human Casp4) in vitro, suggesting the participation of additional molecules for LPG-mediated CASP11 activation. Our data identify a parasite molecule involved in CASP11 activation, thereby establishing the mechanisms underlying inflammasome activation in response to Leishmania species.


Subject(s)
Caspases, Initiator/metabolism , Glycosphingolipids/metabolism , Inflammasomes/metabolism , Leishmania/metabolism , Leishmania/pathogenicity , Leishmaniasis/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Leishmaniasis/parasitology , Macrophages/metabolism , Macrophages/parasitology , Mice , Mice, Inbred C57BL
11.
PLoS One ; 13(6): e0199071, 2018.
Article in English | MEDLINE | ID: mdl-29897990

ABSTRACT

Rocio virus (ROCV) was the causative agent of an unprecedented outbreak of encephalitis during the 1970s in the Vale do Ribeira, Sao Paulo State, in the Southeast region of Brazil. Surprisingly, no further cases of ROCV infection were identified after this outbreak; however, serological surveys have suggested the circulation of ROCV among humans and animals in different regions of Brazil. Cross-protective immunity among flaviviruses is well documented; consequently, immunity induced by infections with other flaviviruses endemic to Brazil could potentially be responsible for the lack of ROCV infections. Herein, we evaluated the cross-protection mediated by other flaviviruses against ROCV infection using an experimental C57BL/6 mouse model. Cross-protection against ROCV infection was observed when animals had prior exposure to Ilheus virus or Saint Louis encephalitis virus, suggesting that cross-reactive anti-flavivirus antibodies may limit ROCV disease outbreaks.


Subject(s)
Encephalitis Virus, St. Louis/immunology , Flavivirus Infections/prevention & control , Flavivirus/pathogenicity , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Disease Susceptibility , Encephalitis Virus, St. Louis/pathogenicity , Evolution, Molecular , Female , Flavivirus Infections/immunology , Flavivirus Infections/mortality , Flavivirus Infections/veterinary , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Survival Rate
12.
J Immunol ; 199(6): 2055-2068, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28784846

ABSTRACT

Protozoan parasites of the genus Leishmania are the causative agents of Leishmaniasis, a disease that can be lethal and affects 12 million people worldwide. Leishmania replicates intracellularly in macrophages, a process that is essential for disease progression. Although the production of reactive oxygen species (ROS) accounts for restriction of parasite replication, Leishmania is known to induce ROS upon macrophage infection. We have recently demonstrated NLRP3 inflammasome activation in infected macrophages, a process that is important for the outcome of infection. However, the molecular mechanisms responsible for inflammasome activation are unknown. In this article, we demonstrate that ROS induced via NADPH oxidase during the early stages of L. amazonensis infection is critical for inflammasome activation in macrophages. We identified that ROS production during L. amazonensis infection occurs upon engagement of Dectin-1, a C-type lectin receptor that signals via spleen tyrosine kinase (Syk) to induce ROS. Accordingly, inflammasome activation in response to L. amazonensis is impaired by inhibitors of NADPH oxidase, Syk, focal adhesion kinase, and proline-rich tyrosine kinase 2, and in the absence of Dectin-1. Experiments performed with Clec7a-/- mice support the critical role of Dectin-1 for inflammasome activation, restriction of parasite replication in macrophages, and mouse resistance to L. amazonensis infection in vivo. Thus, we reported that activation of the Dectin-1/Syk/ROS/NLRP3 pathway during L. amazonensis phagocytosis is important for macrophage restriction of the parasite replication and effectively accounts for host resistance to Leishmania infection.


Subject(s)
DNA, Protozoan/genetics , Inflammasomes/metabolism , Lectins, C-Type/metabolism , Leishmania/physiology , Leishmaniasis/immunology , Macrophages/immunology , NADPH Oxidases/metabolism , Animals , Cells, Cultured , DNA Replication , Female , Lectins, C-Type/genetics , Macrophages/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phagocytosis , Reactive Oxygen Species/metabolism , Syk Kinase/metabolism
13.
PLoS Negl Trop Dis ; 11(3): e0005486, 2017 03.
Article in English | MEDLINE | ID: mdl-28355218

ABSTRACT

BACKGROUND: CD163, receptor for the haptoglobin-hemoglobin complex, is expressed on monocytes/macrophages and neutrophils. A soluble form of CD163 (sCD163) has been associated with the M2 macrophage phenotype, and M2 macrophages have been shown to down-modulate inflammatory responses. In particular, previous studies have shown that M2 is closely associated with the most severe clinical presentation of leprosy (i.e. lepromatous leprosy (LL)), as well as tuberculosis. We hypothesized that sCD163 correlates with severity of diseases caused by intracellular pathogens. METHODOLOGY/PRINCIPAL FINDINGS: To assess this hypothesis, sCD163 levels were measured in the serum of leprosy and visceral leishmaniasis (VL) patients stratified by severity of the clinical presentation. sCD163 levels were significantly higher in patients with these diseases than those observed in healthy control individuals. Further analyses on infection and disease status of leprosy and VL patients revealed a clear association of sCD163 levels with clinical parameters of disease severity. In vitro culture assays revealed that Leishmania infection induced CD163 expression on the surface of both monocyte/macrophages and neutrophils, suggesting these cells as possible sources of sCD163. FACS analyses shows that the cells expressing CD163 produces both TNF-α and IL-4. CONCLUSIONS/SIGNIFICANCE: Taken together, our results reveal sCD163 as a potential biomarker of severity of diseases caused by intracellular pathogens M. leprae and Leishmania spp. and have a modulatory role, with a mix of an inflammatory property induced by TNF-α release, but that potentially induces an anti-inflammatory T cell response, related to IL-4 release.


Subject(s)
Antigens, CD/blood , Antigens, Differentiation, Myelomonocytic/blood , Biomarkers/blood , Leishmaniasis, Visceral/diagnosis , Leishmaniasis, Visceral/pathology , Leprosy/diagnosis , Leprosy/pathology , Receptors, Cell Surface/blood , Humans , Serum/chemistry , Severity of Illness Index
14.
Sci Rep ; 6: 29289, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27377650

ABSTRACT

Neospora caninum is an apicomplexan parasite responsible for major economic losses due to abortions in cattle. Innate immune responses are crucial for host resistance against the infection, however the molecules involved in parasite recognition are still poorly understood. Nod2 is a cytosolic receptor that recognizes several pathogens and its role during N. caninum infection has not yet been described. In that sense, we evaluated the role of Nod2 in host response against this parasite. We found that infection of macrophages induced increased expression of Nod2, which colocalized with the parasites' vacuoles. Nod2-deficient macrophages showed an impaired induction of pro-inflammatory cytokines, increased production of modulatory molecules, and failure to restrict parasite replication. In vivo, Nod2-knockout mice showed a reduction of MAPK phosphorylation and proinflammatory cytokines, followed by decreased inflammation in target organs and increment in parasite burden. Surprisingly, these mice were partially resistant to lethal doses of tachyzoites. In addition, these phenomena were not observed in Rip2-/- mice. In conclusion, our study indicates that Nod2-dependent responses account for N. caninum elimination. On the other hand, the inflammatory milieu induced by this innate receptor provoked pathogenesis and death in severe experimental neosporosis.


Subject(s)
Coccidiosis/pathology , Host-Pathogen Interactions , Inflammation/pathology , Macrophages/immunology , Macrophages/parasitology , Neospora/immunology , Nod2 Signaling Adaptor Protein/metabolism , Animals , Cell Line , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Mice, Inbred C57BL , Mice, Knockout
15.
Infect Immun ; 84(9): 2439-48, 2016 09.
Article in English | MEDLINE | ID: mdl-27297388

ABSTRACT

Coxiella burnetii is a Gram-negative bacterium that causes Q fever in humans. Q fever is an atypical pneumonia transmitted through inhalation of contaminated aerosols. In mammalian lungs, C. burnetii infects and replicates in several cell types, including alveolar macrophages (AMs). The innate immunity and signaling pathways operating during infection are still poorly understood, in part because of the lack of relevant host cell models for infection in vitro In the study described here, we investigated and characterized the infection of primary murine AMs by C. burnetii phase II in vitro Our data reveal that AMs show a pronounced M2 polarization and are highly permissive to C. burnetii multiplication in vitro Murine AMs present an increased susceptibility to infection in comparison to primary bone marrow-derived macrophages. AMs support more than 2 logs of bacterial replication during 12 days of infection in culture, similar to highly susceptible host cells, such as Vero and THP-1 cells. As a proof of principle that AMs are useful for investigation of C. burnetii replication, we performed experiments with AMs from Nos2(-/-) or Ifng(-/-) mice. In the absence of gamma interferon and nitric oxide synthase 2 (NOS2), AMs were significantly more permissive than wild-type cells. In contrast, AMs from Il4(-/-) mice were more restrictive to C. burnetii replication, supporting the importance of M2 polarization for the permissiveness of AMs to C. burnetii replication. Collectively, our data account for understanding the high susceptibility of alveolar macrophages to bacterial replication and support the use of AMs as a relevant model of C. burnetii growth in primary macrophages.


Subject(s)
Coxiella burnetii/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/microbiology , Animals , Cells, Cultured , Immunity, Innate/immunology , Interferon-gamma/immunology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/immunology , Q Fever/immunology , Q Fever/microbiology , Signal Transduction/immunology
16.
J Leukoc Biol ; 100(2): 423-32, 2016 08.
Article in English | MEDLINE | ID: mdl-26884611

ABSTRACT

In vertebrate hosts, Leishmania braziliensis parasites infect mainly mononuclear phagocytic system cells, which when activated by T helper cell type 1 cytokines produce nitric oxide and kill the pathogens. Chemokine (C-C motif) receptor 2 is a chemokine receptor that binds primarily chemokine (C-C motif) ligand 2 and has an important role in the recruitment of monocytic phagocytes. Although it has been reported that Leishmania braziliensis infection induces CCR2 expression in the lesions, the role of CCR2 during Leishmania braziliensis infection remains unknown. Here, we showed that CCR2 has a role in mediating protection against Leishmania braziliensis infection in mice. The absence of CCR2 resulted in increased susceptibility to infection and was associated with low amounts of Ly6C(+) inflammatory dendritic cells in the lesions, which we found to be the major sources of tumor necrosis factor production and induced nitric oxide synthase expression in C57BL/6 mice lesions. Consequently, CCR2(-/-) mice showed decreased tumor necrosis factor production and induced nitric oxide synthase expression, resulting in impaired parasite elimination. We also demonstrated that CCR2 has a role in directly mediating the differentiation of monocytes into inflammatory dendritic cells at the infection sites, contributing to the accumulation of inflammatory dendritic cells in Leishmania braziliensis lesions and subsequent control of parasite replication. Therefore, these data provide new information on the role of chemokines during the immune response to infections and identify a potential target for therapeutic interventions in cutaneous leishmaniasis.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Inflammation/immunology , Leishmania braziliensis/immunology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/prevention & control , Receptors, CCR2/physiology , Animals , Chemokine CCL2/metabolism , Cytokines/metabolism , Dendritic Cells/parasitology , Female , Inflammation/parasitology , Leishmaniasis, Cutaneous/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/parasitology , Nitric Oxide Synthase Type II/metabolism , Signal Transduction
17.
Infect Immun ; 83(12): 4604-16, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26371124

ABSTRACT

Leishmania infantum is a protozoan parasite that causes visceral leishmaniasis (VL). This infection triggers dendritic cell (DC) activation through the recognition of microbial products by Toll-like receptors (TLRs). Among the TLRs, TLR9 is required for DC activation by different Leishmania species. We demonstrated that TLR9 is upregulated in vitro and in vivo during infection. We show that C57BL/6 mice deficient in TLR9 expression (TLR9(-/-) mice) are more susceptible to infection and display higher parasite numbers in the spleen and liver. The increased susceptibility of TLR9(-/-) mice was due to the impaired recruitment of neutrophils to the infection foci associated with reduced levels of neutrophil chemoattractants released by DCs in the target organs. Moreover, both Th1 and Th17 cells were also committed in TLR9(-/-) mice. TLR9-dependent neutrophil recruitment is mediated via the MyD88 signaling pathway but is TIR domain-containing adapter-inducing interferon beta (TRIF) independent. Furthermore, L. infantum failed to activate both plasmacytoid and myeloid DCs from TLR9(-/-) mice, which presented reduced surface costimulatory molecule expression and chemokine release. Interestingly, neutrophil chemotaxis was affected both in vitro and in vivo when DCs were derived from TLR9(-/-) mice. Our results suggest that TLR9 plays a critical role in neutrophil recruitment during the protective response against L. infantum infection that could be associated with DC activation.


Subject(s)
Leishmania infantum/immunology , Leishmaniasis, Visceral/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Toll-Like Receptor 9/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/parasitology , Dendritic Cells/pathology , Female , Gene Expression Regulation , Host-Pathogen Interactions , Leishmania infantum/pathogenicity , Leishmaniasis, Visceral/genetics , Leishmaniasis, Visceral/parasitology , Leishmaniasis, Visceral/pathology , Liver/immunology , Liver/parasitology , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Neutrophils/parasitology , Neutrophils/pathology , Signal Transduction , Spleen/immunology , Spleen/parasitology , Spleen/pathology , Th1 Cells/immunology , Th1 Cells/parasitology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/parasitology , Th17 Cells/pathology , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics
18.
Immunol Rev ; 265(1): 156-71, 2015 May.
Article in English | MEDLINE | ID: mdl-25879291

ABSTRACT

Inflammasomes are multimeric complexes of proteins that are assembled in the host cell cytoplasm in response to specific stress signals or contamination of the cytoplasm by microbial molecules. The canonical inflammasomes are composed of at least three main components: an inflammatory caspase (caspase-1, caspase-11), an adapter molecule (such as ASC), and a sensor protein (such as NLRP1, NLRP3, NLRP12, NAIP1, NAIP2, NAIP5, or AIM2). The sensor molecule determines the inflammasome specificity by detecting specific microbial products or cell stress signals. Upon activation, these molecular platforms facilitate restriction of microbial replication and trigger an inflammatory form of cell death called pyroptosis, thus accounting for the genesis of inflammatory processes. Inflammasome activation has been widely reported in response to pathogenic bacteria. However, recent reports have highlighted the important role of the inflammasomes in the host response to the pathogenesis of infections caused by intracellular protozoan parasites. Herein, we review the activation and specific roles of inflammasomes in recognition and host responses to intracellular protozoan parasites such as Trypanosoma cruzi, Toxoplasma gondii, Plasmodium spp., and Leishmania spp.


Subject(s)
Inflammasomes/metabolism , Multiprotein Complexes/metabolism , Protozoan Infections/immunology , Animals , Humans , Immunity, Innate , Inflammasomes/immunology , Multiprotein Complexes/immunology , Pyroptosis , Receptors, Pattern Recognition , Signal Transduction
19.
Nat Med ; 19(7): 909-15, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23749230

ABSTRACT

Parasites of the Leishmania genus are the causative agents of leishmaniasis in humans, a disease that affects more than 12 million people worldwide. These parasites replicate intracellularly in macrophages, and the primary mechanisms underlying host resistance involve the production of nitric oxide (NO). In this study we show that the Nlrp3 inflammasome is activated in response to Leishmania infection and is important for the restriction of parasite replication both in macrophages and in vivo as demonstrated through the infection of inflammasome-deficient mice with Leishmania amazonensis, Leishmania braziliensis and Leishmania infantum chagasi. Inflammasome-driven interleukin-1ß (IL-1ß) production facilitated host resistance to infection, as signaling through IL-1 receptor (IL-1R) and MyD88 was necessary and sufficient to trigger inducible nitric oxide synthase (NOS2)-mediated production of NO. In this manuscript we identify a major signaling platform for host resistance to Leishmania spp. infection and describe the molecular mechanisms underlying Leishmania-induced NO production.


Subject(s)
Disease Resistance/drug effects , Inflammasomes/immunology , Interleukin-1beta/metabolism , Leishmania , Nitric Oxide/immunology , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Caspase 1/genetics , Cells, Cultured , Cytoskeletal Proteins/genetics , Disease Resistance/genetics , Disease Resistance/immunology , Female , Leishmaniasis/genetics , Leishmaniasis/immunology , Leishmaniasis/metabolism , Leishmaniasis/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nitric Oxide Synthase Type II/genetics
20.
J Immunol ; 187(8): 4347-59, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21930966

ABSTRACT

Among several pharmacological compounds, Phlebotomine saliva contains substances with anti-inflammatory properties. In this article, we demonstrated the therapeutic activity of salivary gland extract (SGE) of Phlebotomus papatasi in an experimental model of arthritis (collagen-induced arthritis [CIA]) and identified the constituents responsible for such activity. Daily administration of SGE, initiated at disease onset, attenuated the severity of CIA, reducing the joint lesion and proinflammatory cytokine release. In vitro incubation of dendritic cells (DCs) with SGE limited specific CD4(+) Th17 cell response. We identified adenosine (ADO) and 5'AMP as the major salivary molecules responsible for anti-inflammatory activities. Pharmacologic inhibition of ADO A2(A) receptor or enzymatic catabolism of salivary nucleosides reversed the SGE-induced immunosuppressive effect. Importantly, CD73 (ecto-5'-nucleotidase enzyme) is expressed on DC surface during stage of activation, suggesting that ADO is also generated by 5'AMP metabolism. Moreover, both nucleosides mimicked SGE-induced anti-inflammatory activity upon DC function in vitro and attenuated establishment of CIA in vivo. We reveal that ADO and 5'AMP are present in pharmacological amounts in P. papatasi saliva and act preferentially on DC function, consequently reducing Th17 subset activation and suppressing the autoimmune response. Thus, it is plausible that these constituents might be promising therapeutic molecules to target immune inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/immunology , Dendritic Cells/drug effects , Nucleosides/pharmacology , Phlebotomus/chemistry , Salivary Glands/chemistry , Animals , Arthritis, Experimental/pathology , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Dendritic Cells/immunology , Female , Male , Mice , Mice, Inbred DBA , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tissue Extracts/chemistry , Tissue Extracts/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...