Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
J Invest Dermatol ; 144(4): 862-873.e4, 2024 Apr.
Article En | MEDLINE | ID: mdl-37852357

Cutaneous leishmaniasis affects 1 million people worldwide annually. Although conventional treatments primarily target the parasite, there is growing interest in host immune modulation. In this study, we investigated the impact of synthetic ß-carboline harmine (ACB1801), previously shown to be immunoregulatory in cancer, on the pathology caused by a drug-resistant Leishmania major strain causing persistent cutaneous lesions. Exposure to ACB1801 in vitro had a modest impact on parasite burden within host macrophages. Moreover, it significantly increased major histocompatibility complex II and costimulatory molecule expression on infected dendritic cells, suggesting an enhanced immune response. In vivo, ACB1801 monotherapy led to a substantial reduction in lesion development and parasite burden in infected C57BL/6 mice, comparable with efficacy of amphotericin B. Transcriptomics analysis further supported ACB1801 immunomodulatory effects, revealing an enrichment of TNF-α, IFN-γ, and major histocompatibility complex II antigen presentation signatures in the draining lymph nodes of treated mice. Flow cytometry analysis confirmed an increased frequency (1.5×) of protective CD4+IFN-γ+TNF-α+ T cells and a decreased frequency (2×) in suppressive IL-10+FoxP3- T cells at the site of infection and in draining lymph nodes. In addition, ACB1801 downregulated the aryl hydrocarbon receptor signaling, known to enhance immunosuppressive cytokines. Thus, these results suggest a potential use for ACB1801 alone or in combination therapy for cutaneous leishmaniasis.


Leishmania major , Leishmaniasis, Cutaneous , Leishmaniasis , Humans , Animals , Mice , Harmine/pharmacology , Harmine/therapeutic use , Tumor Necrosis Factor-alpha , Mice, Inbred C57BL , Immunity , Mice, Inbred BALB C
2.
Cancers (Basel) ; 14(5)2022 Mar 03.
Article En | MEDLINE | ID: mdl-35267610

Increasing evidence indicates that tumor vasculature normalization could be an appropriate strategy to increase therapies' efficacy in solid tumors by decreasing hypoxia and improving drug delivery. We searched for a novel approach that reduces hypoxia and enhances chemotherapy efficacy in pancreatic adenocarcinoma which is characterized by disrupted blood vasculature associated with poor patient survival. Clinical significance of plasma levels of the angiogenic lipid sphingosine-1-phosphate (S1P) was assessed at baseline in 175 patients. High plasma S1P concentration was found to be a favorable prognostic/predictive marker in advanced/metastatic pancreatic adenocarcinoma patients treated by gemcitabine alone but not in patients receiving a combination gemcitabine and PDGFR-inhibitor. In pancreatic adenocarcinoma PDX models, oral administration of an S1P lyase inhibitor (LX2931) significantly increased plasma S1P levels, decreased tumor expression of the hypoxia marker (CA IX), and enhanced chemotherapy efficacy when combined with gemcitabine treatment. The direct effect of S1P on tumor oxygenation was assessed by administration of S1P onto tumor-grafted CAM model and measuring intra-tumoral pO2 using a tissue oxygen monitor. S1P increased pO2 in a tumor-CAM model. Thus, increasing plasma S1P is a promising strategy to decrease tumor hypoxia and enhance therapy efficacy in solid tumors. S1P may act as a tumor vasculature normalizer.

3.
FASEB J ; 35(5): e21418, 2021 05.
Article En | MEDLINE | ID: mdl-33774873

Constitutively expressed by innate immune cells, the cytokine macrophage migration inhibitory factor (MIF) initiates host immune responses and drives pathogenic responses in infectious, inflammatory, and autoimmune diseases. Dendritic cells (DCs) express high levels of MIF, but the role of MIF in DC function remains poorly characterized. As migration is critical for DC immune surveillance, we investigated whether MIF promoted the migration of DCs. In classical transwell experiments, MIF-/- bone marrow-derived DCs (BMDCs) or MIF+/+ BMDCs treated with ISO-1, an inhibitor of MIF, showed markedly reduced spontaneous migration and chemotaxis. CD74-/- BMDCs that are deficient in the ligand-binding component of the cognate MIF receptor exhibited a migration defect similar to that of MIF-/- BMDCs. Adoptive transfer experiments of LPS-matured MIF+/+ and MIF-/- and of CD74+/+ and CD74-/- BMDCs injected into the hind footpads of homologous or heterologous mice showed that the autocrine and paracrine MIF activity acting via CD74 contributed to the recruitment of DCs to the draining lymph nodes. Mechanistically, MIF activated the Src/PI3K signaling pathway and myosin II complexes, which were required for the migration of BMDCs. Altogether, these data show that the cytokine MIF exerts chemokine-like activity for DC motility and trafficking.


Antigens, Differentiation, B-Lymphocyte/physiology , Chemotaxis , Dendritic Cells/physiology , Histocompatibility Antigens Class II/physiology , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Myosin Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , src-Family Kinases/metabolism , Animals , Cells, Cultured , Chemokines/metabolism , Dendritic Cells/cytology , Immunity , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myosin Type II/genetics , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction , src-Family Kinases/genetics
4.
Vaccine ; 37 Suppl 1: A73-A76, 2019 10 03.
Article En | MEDLINE | ID: mdl-30420115

In 2017, the World Health Organization, the World Organisation for Animal Health, the Food and Agriculture Organization of the United Nations and the Global Alliance for Rabies control developed a strategic plan to end human rabies deaths by 2030. A survey for manufacturing capacity and product characteristics of rabies biologics was conducted to inform this process. Twenty-three of 42 manufacturers, responded, giving a market capacity for 2017 of 90 million vials for human vaccines, 2.5 million vials for rabies immunoglobulins, 2 million vials for monoclonal antibodies and 181 million vials for dog vaccines. Production capacity could be increased by many manufacturers but was limited by country demand, lack of long-term planning and restricted market expansion. Should countries implement national rabies elimination programmes where biologic needs are forecasted and production lead times respected, manufacturers can meet future supply needs towards global elimination of human dog-mediated rabies deaths.


Biological Products/supply & distribution , Immunologic Factors/supply & distribution , Post-Exposure Prophylaxis/methods , Rabies Vaccines/supply & distribution , Rabies/prevention & control , Disease Eradication/organization & administration , Global Health , Humans , Rabies/veterinary
5.
Nat Commun ; 6: 6555, 2015 Mar 24.
Article En | MEDLINE | ID: mdl-25800347

Activation of the NLRP3 inflammasome by microbial ligands or tissue damage requires intracellular generation of reactive oxygen species (ROS). We present evidence that macrophage secretion of IL1ß upon stimulation with ATP, crystals or LPS is mediated by a rapid increase in the activity of xanthine oxidase (XO), the oxidized form of xanthine dehydrogenase, resulting in the formation of uric acid as well as ROS. We show that XO-derived ROS, but not uric acid, is the trigger for IL1ß release and that XO blockade results in impaired IL1ß and caspase1 secretion. XO is localized to both cytoplasmic and mitochondrial compartments and acts upstream to the PI3K-AKT signalling pathway that results in mitochondrial ROS generation. This pathway represents a mechanism for regulating NLRP3 inflammasome activation that may have therapeutic implications in inflammatory diseases.


Carrier Proteins/immunology , Interleukin-1beta/metabolism , Macrophages/metabolism , Reactive Oxygen Species/immunology , Xanthine Dehydrogenase/genetics , Xanthine Oxidase/immunology , Animals , Autophagy , Blotting, Western , Calcium/metabolism , Calcium Phosphates/pharmacology , Carrier Proteins/drug effects , Caspase 1/immunology , Gene Knockdown Techniques , In Vitro Techniques , Lipopolysaccharides/pharmacology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Monocytes , NLR Family, Pyrin Domain-Containing 3 Protein , Peritonitis/immunology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Uric Acid/immunology , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/immunology
6.
PLoS One ; 9(5): e96766, 2014.
Article En | MEDLINE | ID: mdl-24801628

Infections with Leishmania parasites of the Leishmania Viannia subgenus give rise to both localized cutaneous (CL), and metastatic leishmaniasis. Metastasizing disease forms including disseminated (DCL) and mutocutaneous (MCL) leishmaniasis result from parasitic dissemination and lesion formation at sites distal to infection and have increased inflammatory responses. The presence of Leishmania RNA virus (LRV) in L. guyanensis parasites contributes to the exacerbation of disease and impacts inflammatory responses via activation of TLR3 by the viral dsRNA. In this study we investigated other innate immune response adaptor protein modulators and demonstrated that both MyD88 and TLR9 played a crucial role in the development of Th1-dependent healing responses against L. guyanensis parasites regardless of their LRV status. The absence of MyD88- or TLR9-dependent signaling pathways resulted in increased Th2 associated cytokines (IL-4 and IL-13), which was correlated with low transcript levels of IL-12p40. The reliance of IL-12 was further confirmed in IL12AB-/- mice, which were completely susceptible to infection. Protection to L. guyanensis infection driven by MyD88- and TLR9-dependent immune responses arises independently to those induced due to high LRV burden within the parasites.


Immunity, Innate , Leishmania guyanensis/virology , Myeloid Differentiation Factor 88/metabolism , RNA Viruses/physiology , Toll-Like Receptor 9/metabolism , Animals , Disease Susceptibility , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p35/metabolism , Interleukin-12 Subunit p40/deficiency , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Leishmania guyanensis/physiology , Leishmaniasis, Mucocutaneous/immunology , Leishmaniasis, Mucocutaneous/pathology , Leishmaniasis, Mucocutaneous/veterinary , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Signal Transduction , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics
7.
Sci Rep ; 4: 4554, 2014 Apr 01.
Article En | MEDLINE | ID: mdl-24686534

Atherosclerosis is a chronic inflammatory disease due to lipid deposition in the arterial wall. Multiple mechanisms participate in the inflammatory process, including oxidative stress. Xanthine oxidase (XO) is a major source of reactive oxygen species (ROS) and has been linked to the pathogenesis of atherosclerosis, but the underlying mechanisms remain unclear. Here, we show enhanced XO expression in macrophages in the atherosclerotic plaque and in aortic endothelial cells in ApoE(-/-) mice, and that febuxostat, a highly potent XO inhibitor, suppressed plaque formation, reduced arterial ROS levels and improved endothelial dysfunction in ApoE(-/-) mice without affecting plasma cholesterol levels. In vitro, febuxostat inhibited cholesterol crystal-induced ROS formation and inflammatory cytokine release in murine macrophages. These results demonstrate that in the atherosclerotic plaque, XO-mediated ROS formation is pro-inflammatory and XO-inhibition by febuxostat is a potential therapy for atherosclerosis.


Atherosclerosis/drug therapy , Plaque, Atherosclerotic/pathology , Reactive Oxygen Species/metabolism , Thiazoles/pharmacology , Xanthine Oxidase/antagonists & inhibitors , Animals , Aorta/cytology , Aorta/metabolism , Apolipoproteins E/genetics , Atherosclerosis/pathology , Body Weight , Cholesterol/blood , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Febuxostat , Gout Suppressants/pharmacology , Inflammation/drug therapy , L-Lactate Dehydrogenase/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Xanthine Oxidase/biosynthesis
8.
Arthritis Rheumatol ; 66(5): 1327-39, 2014 May.
Article En | MEDLINE | ID: mdl-24470119

OBJECTIVE: Monosodium urate monohydrate (MSU) crystal-induced interleukin-1ß (IL-1ß) secretion is a critical factor in the pathogenesis of gout. However, without costimulation by a proIL-1ß-inducing factor, MSU crystals alone are insufficient to induce IL-1ß secretion. The responsible costimulatory factors that act as a priming endogenous signal in vivo are not yet known. We undertook this study to analyze the costimulatory properties of myeloid-related protein 8 (MRP-8) and MRP-14 (endogenous Toll-like receptor 4 [TLR-4] agonists) in MSU crystal-induced IL-1ß secretion and their relevance in gout. METHODS: MRP-8/MRP-14 was measured in paired serum and synovial fluid samples by enzyme-linked immunosorbent assay (ELISA) and localized in synovial tissue from gout patients by immunohistochemistry. Serum levels were correlated with disease activity, and MSU crystal-induced release of MRPs from human phagocytes was measured. Costimulatory effects of MRP-8 and MRP-14 on MSU crystal-induced IL-1ß secretion from phagocytes were analyzed in vitro by ELISA, Western blotting, and polymerase chain reaction. The impact of MRP was tested in vivo in a murine MSU crystal-induced peritonitis model. RESULTS: MRP-8/MRP-14 levels were elevated in the synovium, tophi, and serum of patients with gout and correlated with disease activity. MRP-8/MRP-14 was released by MSU crystal-activated phagocytes and increased MSU crystal-induced IL-1ß secretion in a TLR-4-dependent manner. Targeted deletion of MRP-14 in mice led to a moderately reduced response of MSU crystal-induced inflammation in vivo. CONCLUSION: MRP-8 and MRP-14, which are highly expressed in gout, are enhancers of MSU crystal-induced IL-1ß secretion in vitro and in vivo. These endogenous TLR-4 ligands released by activated phagocytes contribute to the maintenance of inflammation in gout.


Calgranulin A/metabolism , Calgranulin B/metabolism , Gout/metabolism , Inflammation/metabolism , Uric Acid/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cells, Cultured , Crystallization , Disease Models, Animal , Female , Humans , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phagocytes/metabolism , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
9.
PLoS One ; 8(9): e75527, 2013.
Article En | MEDLINE | ID: mdl-24086554

Excess reactive oxygen species (ROS) formation can trigger various pathological conditions such as inflammation, in which xanthine oxidase (XO) is one major enzymatic source of ROS. Although XO has been reported to play essential roles in inflammatory conditions, the molecular mechanisms underlying the involvement of XO in inflammatory pathways remain unclear. Febuxostat, a selective and potent inhibitor of XO, effectively inhibits not only the generation of uric acid but also the formation of ROS. In this study, therefore, we examined the effects of febuxostat on lipopolysaccharide (LPS)-mediated inflammatory responses. Here we show that febuxostat suppresses LPS-induced MCP-1 production and mRNA expression via activating MAPK phosphatase-1 (MKP-1) which, in turn, leads to dephosphorylation and inactivation of JNK in macrophages. Moreover, these effects of febuxostat are mediated by inhibiting XO-mediated intracellular ROS production. Taken together, our data suggest that XO mediates LPS-induced phosphorylation of JNK through ROS production and MKP-1 inactivation, leading to MCP-1 production in macrophages. These studies may bring new insights into the novel role of XO in regulating inflammatory process through MAPK phosphatase, and demonstrate the potential use of XO inhibitor in modulating the inflammatory processes.


Chemokine CCL2/metabolism , Dual Specificity Phosphatase 1/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 4/metabolism , Thiazoles/pharmacology , Xanthine Oxidase/antagonists & inhibitors , Animals , Cell Line, Tumor , Chemokine CCL2/genetics , Dual Specificity Phosphatase 1/genetics , Febuxostat , Female , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , MAP Kinase Kinase 4/genetics , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Phosphorylation/genetics , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Xanthine Oxidase/metabolism
10.
Nat Rev Rheumatol ; 9(7): 391-9, 2013 Jul.
Article En | MEDLINE | ID: mdl-23670136

Inflammasomes are key inducers of inflammation in response to exogenous and endogenous stimuli, because they regulate the processing and secretion of the proinflammatory cytokines IL-1ß and IL-18. Thus, inflammasomes have a crucial role in host defence against infection, but they can also be involved in inflammatory diseases. Indeed, the NLRP3 (NOD-, LRR- and pyrin domain-containing 3) inflammasome has been shown to play a part in several inflammatory rheumatic disorders, although the mechanisms involved are better elucidated in some of these diseases than in others. In particular, the pathogenesis of cryopyrin-associated periodic syndromes and microcrystal-induced arthritides is thought to be dependent on activation of the NLRP3 inflammasome, and IL-1 inhibition has shown efficacy as a therapeutic strategy in both groups of conditions. In this Review, we describe the current understanding of the mechanisms that trigger the inflammasome, and consider the relevance of the inflammasome to a variety of rheumatic diseases. In addition, we discuss the current therapies targeting this molecular complex, as well as future therapeutic prospects.


Inflammasomes/physiology , Rheumatic Diseases/drug therapy , Rheumatic Diseases/physiopathology , Carrier Proteins/physiology , Cryopyrin-Associated Periodic Syndromes/drug therapy , Cryopyrin-Associated Periodic Syndromes/physiopathology , Humans , Interleukin-1/physiology , Interleukin-18/physiology , NLR Family, Pyrin Domain-Containing 3 Protein
11.
PLoS One ; 8(2): e57352, 2013.
Article En | MEDLINE | ID: mdl-23468973

BACKGROUND: basic calcium phosphate (BCP) crystals are commonly found in osteoarthritis (OA) and are associated with cartilage destruction. BCP crystals induce in vitro catabolic responses with the production of metalloproteases and inflammatory cytokines such as interleukin-1 (IL-1). In vivo, IL-1 production induced by BCP crystals is both dependant and independent of NLRP3 inflammasome. We aimed to clarify 1/ the role of BCP crystals in cartilage destruction and 2/ the role of IL-1 and NLRP3 inflammasome in cartilage degradation related to BCP crystals. METHODOLOGY PRINCIPAL FINDINGS: synovial membranes isolated from OA knees were analysed by alizarin Red and FTIR. Pyrogen free BCP crystals were injected into right knees of WT, NLRP3 -/-, ASC -/-, IL-1α -/- and IL-1ß-/- mice and PBS was injected into left knees. To assess the role of IL-1, WT mice were treated by intra-peritoneal injections of anakinra, the IL-1Ra recombinant protein, or PBS. Articular destruction was studied at d4, d17 and d30 assessing synovial inflammation, proteoglycan loss and chondrocyte apoptosis. BCP crystals were frequently found in OA synovial membranes including low grade OA. BCP crystals injected into murine knee joints provoked synovial inflammation characterized by synovial macrophage infiltration that persisted at day 30, cartilage degradation as evidenced by loss of proteoglycan staining by Safranin-O and concomitant expression of VDIPEN epitopes, and increased chondrocyte apoptosis. BCP crystal-induced synovitis was totally independent of IL-1α and IL-1ß signalling and no alterations of inflammation were observed in mice deficient for components of the NLRP3-inflammasome, IL-1α or IL-1ß. Similarly, treatment with anakinra did not prevent BCP crystal effects. In vitro, BCP crystals elicited enhanced transcription of matrix degrading and pro-inflammatory genes in macrophages. CONCLUSIONS SIGNIFICANCE: intra-articular BCP crystals can elicit synovial inflammation and cartilage degradation suggesting that BCP crystals have a direct pathogenic role in OA. The effects are independent of IL-1 and NLRP3 inflammasome.


Arthritis/etiology , Calcium Phosphates/chemistry , Animals , Apoptosis , Arthritis/pathology , Base Sequence , Crystallization , DNA Primers , Female , Immunohistochemistry , Interleukin-1/genetics , Interleukin-1/physiology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Spectroscopy, Fourier Transform Infrared
12.
J Allergy Clin Immunol ; 131(6): 1635-43, 2013 Jun.
Article En | MEDLINE | ID: mdl-23006543

BACKGROUND: The exact pathogenesis of the pediatric disorder periodic fever, aphthous stomatitis, pharyngitis, cervical adenitis (PFAPA) syndrome is unknown. OBJECTIVES: We hypothesized that PFAPA might be due to dysregulated monocyte IL-1ß production linked to genetic variants in proinflammatory genes. METHODS: Fifteen patients with PFAPA syndrome were studied during and outside a febrile episode. Hematologic profile, inflammatory markers, and cytokine levels were measured in the blood. The capacity of LPS-stimulated PBMCs and monocytes to secrete IL-1ß was assessed by using ELISA, and active IL-1ß secretion was visualized by means of Western blotting. Real-time quantitative PCR was performed to assess cytokine gene expression. DNA was screened for variants of the MEFV, TNFRSF1A, MVK, and NLRP3 genes in a total of 57 patients with PFAPA syndrome. RESULTS: During a febrile attack, patients with PFAPA syndrome revealed significantly increased neutrophil counts, erythrocyte sedimentation rates, and C-reactive protein, serum amyloid A, myeloid-related protein 8/14, and S100A12 levels compared with those seen outside attacks. Stimulated PBMCs secreted significantly more IL-1ß during an attack (during a febrile episode, 575 ± 88 pg/mL; outside a febrile episode, 235 ± 56 pg/mL; P < .001), and this was in the mature active p17 form. IL-1ß secretion was inhibited by ZYVAD, a caspase inhibitor. Similar results were found for stimulated monocytes (during a febrile episode, 743 ± 183 pg/mL; outside a febrile episode, 227 ± 92 pg/mL; P < .05). Genotyping identified variants in 15 of 57 patients, with 12 NLRP3 variants, 1 TNFRSF1A variant, 4 MEFV variants, and 1 MVK variant. CONCLUSION: Our data strongly suggest that IL-1ß monocyte production is dysregulated in patients with PFAPA syndrome. Approximately 20% of them were found to have NLRP3 variants, suggesting that inflammasome-related genes might be involved in this autoinflammatory syndrome.


Fever/metabolism , Interleukin-1beta/biosynthesis , Lymphadenitis/metabolism , Monocytes/metabolism , Pharyngitis/metabolism , Stomatitis, Aphthous/metabolism , Adolescent , Adult , Aged , Child , Female , Fever/genetics , Fever/immunology , Genetic Variation , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/blood , Interleukin 1 Receptor Antagonist Protein/metabolism , Leukocyte Count , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/immunology , Lymphadenitis/genetics , Lymphadenitis/immunology , Male , Middle Aged , Monocytes/immunology , Neutrophils , Pharyngitis/genetics , Pharyngitis/immunology , Stomatitis, Aphthous/genetics , Stomatitis, Aphthous/immunology , Syndrome , Young Adult
13.
PLoS One ; 6(6): e20189, 2011.
Article En | MEDLINE | ID: mdl-21674051

BACKGROUND: DC are activated by pathogen-associated molecular patterns (PAMPs), and this is pivotal for the induction of adaptive immune responses. Thereafter, the clearance of activated DC is crucial to prevent immune pathology. While PAMPs are of major interest for vaccine science due to their adjuvant potential, it is unclear whether and how PAMPs may affect DC viability. We aimed to elucidate the possible apoptotic mechanisms that control activated DC lifespan in response to PAMPs, particularly in vivo. METHODOLOGY/PRINCIPAL FINDINGS: We report that polyinosinic:polycytidylic acid (PolyIC, synthetic analogue of dsRNA) induces dramatic apoptosis of mouse splenic conventional DC (cDC) in vivo, predominantly affecting the CD8α subset, as shown by flow cytometry-based analysis of splenic DC subsets. Importantly, while Bim deficiency conferred only minor protection, cDC depletion was prevented in mice lacking Bim plus one of three other BH3-only proteins, either Puma, Noxa or Bid. Furthermore, we show that Type I Interferon (IFN) is necessary and sufficient for DC death both in vitro and in vivo, and that TLR3 and MAVS co-operate in IFNß production in vivo to induce DC death in response to PolyIC. CONCLUSIONS/SIGNIFICANCE: These results demonstrate for the first time in vivo that apoptosis restricts DC lifespan following activation by PolyIC, particularly affecting the CD8α cDC subset. Such DC apoptosis is mediated by the overlapping action of pro-apoptotic BH3-only proteins, including but not solely involving Bim, and is driven by Type I IFN. While Type I IFNs are important anti-viral factors, CD8α cDC are major cross-presenting cells and critical inducers of CTL. We discuss such paradoxical finding on DC death with PolyIC/Type I IFN. These results could contribute to understand immunosuppression associated with chronic infection, and to the optimization of DC-based therapies and the clinical use of PAMPs and Type I IFNs.


Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Interferon Type I/pharmacology , Poly I-C/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Animals , CD8 Antigens/metabolism , Cell Count , Dendritic Cells/metabolism , Female , Gene Expression Regulation/drug effects , Interferon Type I/biosynthesis , Interferon-beta/biosynthesis , Interferon-beta/pharmacology , Leishmania/physiology , Ligands , Male , Mice , Spleen/immunology , Toll-Like Receptor 3/metabolism
14.
Science ; 331(6018): 775-8, 2011 Feb 11.
Article En | MEDLINE | ID: mdl-21311023

Mucocutaneous leishmaniasis is caused by infections with intracellular parasites of the Leishmania Viannia subgenus, including Leishmania guyanensis. The pathology develops after parasite dissemination to nasopharyngeal tissues, where destructive metastatic lesions form with chronic inflammation. Currently, the mechanisms involved in lesion development are poorly understood. Here we show that metastasizing parasites have a high Leishmania RNA virus-1 (LRV1) burden that is recognized by the host Toll-like receptor 3 (TLR3) to induce proinflammatory cytokines and chemokines. Paradoxically, these TLR3-mediated immune responses rendered mice more susceptible to infection, and the animals developed an increased footpad swelling and parasitemia. Thus, LRV1 in the metastasizing parasites subverted the host immune response to Leishmania and promoted parasite persistence.


Chemokines/metabolism , Cytokines/metabolism , Leishmania guyanensis/pathogenicity , Leishmania guyanensis/virology , Leishmaniasis, Mucocutaneous/immunology , Leishmaniavirus/immunology , Toll-Like Receptor 3/immunology , Animals , Inflammation Mediators/metabolism , Leishmaniasis, Mucocutaneous/parasitology , Leishmaniavirus/physiology , Macrophages/immunology , Macrophages/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Parasitemia , Phagosomes/parasitology , RNA, Double-Stranded/immunology , RNA, Viral/immunology , Toll-Like Receptors/immunology
15.
Eur J Immunol ; 40(6): 1697-707, 2010 Jun.
Article En | MEDLINE | ID: mdl-20333623

Host resistance to Leishmania major is highly dependent on the development of a Th1 immune response. The TLR adaptator myeloid differentiation protein 88 (MyD88) has been implicated in the Th1 immune response associated with the resistant phenotype observed in C57BL/6 mice after infection with L. major. To investigate whether the MyD88 pathway is differentially used by distinct substrains of parasites, MyD88(-/-) C57BL/6 mice were infected with two substrains of L. major, namely L. major LV39 and L. major IR75. MyD88(-/-) mice were susceptible to both substrains of L. major, although with different kinetics of infection. The mechanisms involved during the immune response associated with susceptibility of MyD88(-/-) mice to L. major is however, parasite substrain-dependent. Susceptibility of MyD88(-/-) mice infected with L. major IR75 is a consequence of Th2 immune-deviation, whereas susceptibility of MyD88(-/-) mice to infection with L. major LV39 resulted from an impaired Th1 response. Depletion of regulatory T cells (Treg) partially restored IFN-gamma secretion and the Th1 immune response in MyD88(-/-) mice infected with L. major LV39, demonstrating a role of Treg activity in the development of an impaired Th1 response in these mice.


Leishmaniasis, Cutaneous/immunology , Myeloid Differentiation Factor 88/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Animals , Cytokines/biosynthesis , Disease Susceptibility/immunology , Enzyme-Linked Immunosorbent Assay , Female , Leishmania major/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
16.
J Infect Dis ; 194(8): 1160-7, 2006 Oct 15.
Article En | MEDLINE | ID: mdl-16991092

Mucocutaneous leishmaniasis (MCL) in South and Central America is characterized by the dissemination (metastasis) of Leishmania Viannia subgenus parasites from a cutaneous lesion to nasopharyngeal tissues. Little is known about the pathogenesis of MCL, especially with regard to the virulence of the parasites and the process of metastatic dissemination. We previously examined the functional relationship between cytoplasmic peroxiredoxin and metastatic phenotype using highly, infrequently, and nonmetastatic clones isolated from an L. (V.) guyanensis strain previously shown to be highly metastatic in golden hamsters. Distinct forms of cytoplasmic peroxiredoxin were identified and found to be associated with the metastatic phenotype. We report here that peroxidase activity in the presence of hydrogen peroxide and infectivity differs between metastatic and nonmetastatic L. (V.) guyanensis clones. After hydrogen peroxide treatment or heat shock, peroxiredoxin was detected preferentially as dimers in metastatic L. (V.) guyanensis clones and in L. (V.) panamensis strains from patients with MCL, compared with nonmetastatic parasites. These data provide evidence that resistance to the first microbicidal response of the host cell by Leishmania promastigotes is linked to peroxiredoxin conformation and may be relevant to intracellular survival and persistence, which are prerequisites for the development of metastatic disease.


Drug Resistance/genetics , Leishmania/genetics , Leishmania/pathogenicity , Leishmaniasis, Mucocutaneous/parasitology , Amino Acid Sequence , Animals , Cricetinae , DNA/analysis , Leishmaniasis, Mucocutaneous/pathology , Mesocricetus , Molecular Sequence Data , NADP/pharmacology , Oxidative Stress , Peroxidases/genetics , Peroxiredoxins , Polymerase Chain Reaction , Reactive Oxygen Species/pharmacology , Sequence Alignment , Severity of Illness Index
17.
Clin Cancer Res ; 11(18): 6686-94, 2005 Sep 15.
Article En | MEDLINE | ID: mdl-16166448

PURPOSE: Alpha-fetoprotein (AFP) is a tumor-associated antigen in hepatocellular carcinoma and is a target for the development of cancer vaccine. Four immunodominant AFP-derived HLA-A*0201-restricted peptides have been identified and the administration of these peptides with an adjuvant has stimulated AFP-specific CTL responses in hepatocellular carcinoma patients. However, no AFP-derived CD4 T-cell epitope has yet been reported and the status of AFP-specific CD4(+) T-cell responses in hepatocellular carcinoma patients is not fully understood. The aim of this study was to analyze naturally occurring CD4(+) T-cell responses to AFP. EXPERIMENTAL DESIGN: We analyzed the ability of CD4(+) T cells to recognize an HLA-DR-restricted AFP-derived epitope in 41 hepatocellular carcinoma patients and 24 non-hepatocellular carcinoma control patients using intracellular cytokine assays for IFN-gamma. RESULTS: Here, for the first time, we report the identification of an AFP-derived CD4(+) T-cell epitope that is recognized by circulating lymphocytes from hepatocellular carcinoma patients in association with HLA-DR. The absence of detectable responses in healthy donors and patients with chronic liver disease suggests that AFP-specific CD4(+) T cells in the responder patients had been previously expanded in vivo in response to the tumor. The anti-AFP CD4(+) T-cell response was only detected in hepatocellular carcinoma patients with normal or mildly elevated serum AFP levels who were in the early stage of disease. CONCLUSION: Our data will be instrumental in the development of cancer vaccine using AFP-derived immunogens.


CD4-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/blood , Epitopes/immunology , Liver Neoplasms/blood , alpha-Fetoproteins/immunology , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Cells, Cultured , Epitopes/genetics , Female , Flow Cytometry , HLA-DR Antigens/immunology , Humans , Interferon-gamma/biosynthesis , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Male , Middle Aged , Molecular Sequence Data , Sequence Homology, Amino Acid , alpha-Fetoproteins/genetics
18.
J Immunol ; 173(3): 1772-8, 2004 Aug 01.
Article En | MEDLINE | ID: mdl-15265907

alpha-Fetoprotein (AFP) is a tumor-associated Ag, and its serum level is elevated in patients with hepatocellular carcinoma (HCC). In vitro, AFP induces functional impairment of dendritic cells (DCs). This was demonstrated by the down-regulation of CD40 and CD86 molecules and the impairment of allostimulatory function. Also, AFP was found to induce significant apoptosis of DCs, and AFP-treated DCs produced low levels of IL-12 and TNF-alpha, a cytokine pattern that could hamper an efficient antitumor immune response. Ex vivo, APCs of patients with HCC and high levels of AFP produced lower levels of TNF-alpha than that of healthy individuals. In conclusion, these results illustrate that AFP induces dysfunction and apoptosis of APCs, thereby offering a mechanism by which HCC escapes immunological control.


Antigen Presentation/drug effects , Apoptosis/drug effects , Carcinoma, Hepatocellular/immunology , Dendritic Cells/drug effects , Liver Neoplasms/immunology , Neoplasm Proteins/pharmacology , Tumor Escape/physiology , alpha-Fetoproteins/pharmacology , Antigens, Neoplasm/immunology , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-12/biosynthesis , Lipopolysaccharides/pharmacology , Liver Neoplasms/blood , Liver Neoplasms/pathology , Neoplasm Proteins/physiology , Tumor Escape/drug effects , Tumor Necrosis Factor-alpha/biosynthesis , alpha-Fetoproteins/physiology
...