Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Pathog Dis ; 75(4)2017 06 01.
Article in English | MEDLINE | ID: mdl-28460033

ABSTRACT

Gonorrhea has become resistant to most conventional antimicrobials used in clinical practice. The global spread of multidrug-resistant isolates of Neisseria gonorrhoeae could lead to an era of untreatable gonorrhea. New therapeutic modalities with novel mechanisms of action that do not lend themselves to the development of resistance are urgently needed. Gonococcal lipooligosaccharide (LOS) sialylation is critical for complement resistance and for establishing infection in humans and experimental mouse models. Here we describe two immunotherapeutic approaches that target LOS sialic acid: (i) a fusion protein that comprises the region in the complement inhibitor factor H (FH) that binds to sialylated gonococci and IgG Fc (FH/Fc fusion protein) and (ii) analogs of sialic acid that are incorporated into LOS but fail to protect the bacterium against killing. Both molecules showed efficacy in the mouse vaginal colonization model of gonorrhea and may represent promising immunotherapeutic approaches to target multidrug-resistant isolates. Disabling key gonococcal virulence mechanisms is an effective therapeutic strategy because the reduction of virulence is likely to be accompanied by a loss of fitness, rapid elimination by host immunity and consequently, decreased transmission.


Subject(s)
Gonorrhea/prevention & control , Lipopolysaccharides/metabolism , Neisseria gonorrhoeae/physiology , Sialic Acids/metabolism , Virulence Factors/metabolism , Animals , Complement Factor H/genetics , Complement Factor H/metabolism , Disease Models, Animal , Female , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Mice , Neisseria gonorrhoeae/drug effects , Protein Binding , Recombinant Fusion Proteins/metabolism , Vagina/microbiology
2.
mBio ; 6(6): e01605-15, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26578679

ABSTRACT

UNLABELLED: High levels of circulating immunocomplexes (ICs) are found in patients with either infectious or sterile inflammation. We report that patients with either Plasmodium falciparum or Plasmodium vivax malaria have increased levels of circulating anti-DNA antibodies and ICs containing parasite DNA. Upon stimulation with malaria-induced ICs, monocytes express an NF-κB transcriptional signature. The main source of IC-induced proinflammatory cytokines (i.e., tumor necrosis factor alpha [TNF-α] and interleukin-1ß [IL-1ß])in peripheral blood mononuclear cells from acute malaria patients was found to be a CD14(+) CD16 (FcγRIIIA)(+) CD64 (FcγRI)(high) CD32 (FcγRIIB)(low) monocyte subset. Monocytes from convalescent patients were predominantly of the classical phenotype (CD14(+) CD16(-)) that produces high levels of IL-10 and lower levels of TNF-α and IL-1ß in response to ICs. Finally, we report a novel role for the proinflammatory activity of ICs by demonstrating their ability to induce inflammasome assembly and caspase-1 activation in human monocytes. These findings illuminate our understanding of the pathogenic role of ICs and monocyte subsets and may be relevant for future development of immunity-based interventions with broad applications to systemic inflammatory diseases. IMPORTANCE: Every year, there are approximately 200 million cases of Plasmodium falciparum and P. vivax malaria, resulting in nearly 1 million deaths, most of which are children. Decades of research on malaria pathogenesis have established that the clinical manifestations are often a consequence of the systemic inflammation elicited by the parasite. Recent studies indicate that parasite DNA is a main proinflammatory component during infection with different Plasmodium species. This finding resembles the mechanism of disease in systemic lupus erythematosus, where host DNA plays a central role in stimulating an inflammatory process and self-damaging reactions. In this study, we disclose the mechanism by which ICs containing Plasmodium DNA activate innate immune cells and consequently stimulate systemic inflammation during acute episodes of malaria. Our results further suggest that Toll-like receptors and inflammasomes have a central role in malaria pathogenesis and provide new insights toward developing novel therapeutic interventions for this devastating disease.


Subject(s)
Antigen-Antibody Complex/metabolism , Cytokines/metabolism , DNA, Protozoan/immunology , Inflammasomes/metabolism , Malaria, Falciparum/pathology , Malaria, Vivax/pathology , Monocytes/metabolism , Antigen-Antibody Complex/blood , Antigens, CD/analysis , Humans , Immunophenotyping , Malaria, Falciparum/immunology , Malaria, Vivax/immunology , Monocytes/chemistry , Protein Multimerization
3.
Proc Natl Acad Sci U S A ; 111(39): E4110-8, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25225402

ABSTRACT

The increase of extracellular heme is a hallmark of hemolysis or extensive cell damage. Heme has prooxidant, cytotoxic, and inflammatory effects, playing a central role in the pathogenesis of malaria, sepsis, and sickle cell disease. However, the mechanisms by which heme is sensed by innate immune cells contributing to these diseases are not fully characterized. We found that heme, but not porphyrins without iron, activated LPS-primed macrophages promoting the processing of IL-1ß dependent on nucleotide-binding domain and leucine rich repeat containing family, pyrin domain containing 3 (NLRP3). The activation of NLRP3 by heme required spleen tyrosine kinase, NADPH oxidase-2, mitochondrial reactive oxygen species, and K(+) efflux, whereas it was independent of heme internalization, lysosomal damage, ATP release, the purinergic receptor P2X7, and cell death. Importantly, our results indicated the participation of macrophages, NLRP3 inflammasome components, and IL-1R in the lethality caused by sterile hemolysis. Thus, understanding the molecular pathways affected by heme in innate immune cells might prove useful to identify new therapeutic targets for diseases that have heme release.


Subject(s)
Heme/metabolism , Hemolysis/physiology , Inflammasomes/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 1/deficiency , Caspase 1/genetics , Caspase 1/metabolism , Heme/chemistry , Heme/immunology , Hemolysis/immunology , Humans , Inflammasomes/immunology , Interleukin-1beta/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , NADPH Oxidase 2 , NADPH Oxidases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Potassium/metabolism , Protoporphyrins/chemistry , Protoporphyrins/metabolism , Reactive Oxygen Species/metabolism
4.
mBio ; 5(2): e00949-14, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24643866

ABSTRACT

The pathogenesis of malaria, an insect-borne disease that takes millions of lives every year, is still not fully understood. Complement receptor 1 (CR1) has been described as a receptor for Plasmodium falciparum, which causes cerebral malaria in humans. We investigated the role of CR1 in an experimental model of cerebral malaria. Transgenic mice expressing human CR1 (hCR1(+)) on erythrocytes were infected with Plasmodium berghei ANKA and developed cerebral malaria. No difference in survival was observed in hCR1(+) mice compared to wild-type mice following infection with P. berghei ANKA; however, hCR1 detection was significantly diminished on erythrocytes between days 7 and 10 postinfection. hCR1 levels returned to baseline by day 17 postinfection in surviving animals. Immunoblot assays revealed that total erythrocyte hCR1 levels were diminished, confirming that immune complexes in association with erythrocyte hCR1 were likely removed from erythrocytes in vivo by clearance following immune adherence. Decreases in hCR1 were completely dependent on C3 expression, as mice treated with cobra venom factor (which consumes and depletes C3) retained hCR1 on erythrocytes during C3 depletion through day 7; erythrocyte hCR1 decreases were observed only when C3 levels recovered on day 9. B-cell-deficient mice exhibit a marked increase in survival following infection with P. berghei ANKA, which suggests that immune complexes play a central role in the pathogenesis of experimental cerebral malaria. Together, our findings highlight the importance of complement and immune complexes in experimental cerebral malaria. IMPORTANCE Cerebral malaria is a deadly complication of infection with Plasmodium falciparum. Despite its high prevalence, relatively little is understood about its pathogenesis. We have determined that immune complexes are generated and deposited on erythrocytes specifically expressing human complement receptor 1 in a mouse model of cerebral malaria. We also provide evidence demonstrating the importance of immunoglobulins in the pathogenesis of cerebral malaria in mice. These findings may have important implications in human cerebral malaria.


Subject(s)
Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/toxicity , B-Lymphocytes/immunology , Malaria, Cerebral/immunology , Malaria, Cerebral/pathology , Plasmodium berghei/immunology , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Complement 3b/genetics , Receptors, Complement 3b/immunology , Survival Analysis
5.
J Biol Chem ; 288(15): 10715-21, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23443666

ABSTRACT

Natural killer (NK) cells are important for innate immunity in particular through the production of IFN-γ and GM-CSF. Both cytokines are important in restoration of immune function of tolerized leukocytes under inflammatory events. The expression of TLRs in NK cells has been widely studied by analyzing the mRNA of these receptors, rarely seeking their protein expression. We previously showed that murine spleen NK cells express TLR9 intracellularly and respond to CpG oligodeoxynucleotide (CpG-ODN) by producing IFN-γ and GM-CSF. However, to get such production the presence of accessory cytokines (such as IL-15 and IL-18) was required, whereas CpG-ODN or accessory cytokines alone did not induce IFN-γ or GM-CSF. We show here that TLR9 overlaps with the Golgi apparatus in NK cells. Furthermore, CpG-ODN stimulation in the presence of accessory cytokines induces the phosphorylation of c-Jun, STAT3, and IκBα. IFN-γ and GM-CSF production requires NF-κB and STAT3 activation as well as Erk-dependent mechanisms for IFN-γ and p38 signaling for GM-CSF. Using knock-out-mice, we show that UNC93b1 and IL-12 (produced by NK cells themselves) are also necessary for IFN-γ and GM-CSF production. IFN-γ production was found to be MyD88- and TLR9-dependent, whereas GM-CSF was TLR9-independent but dependent on STING (stimulator of interferon genes), a cytosolic adaptor recently described for DNA sensing. Our study thereby allows us to gain insight into the mechanisms of synergy between accessory cytokines and CpG-ODN in NK cells. It also identifies a new and alternative signaling pathway for CpG-ODN in murine NK cells.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Interferon-gamma/biosynthesis , Killer Cells, Natural/metabolism , MAP Kinase Signaling System/physiology , Membrane Proteins/metabolism , Spleen/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , I-kappa B Kinase/metabolism , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-15/biosynthesis , Interleukin-15/genetics , Interleukin-15/immunology , Interleukin-18/biosynthesis , Interleukin-18/genetics , Interleukin-18/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , MAP Kinase Signaling System/drug effects , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Oligodeoxyribonucleotides/pharmacology , Phosphorylation/drug effects , Phosphorylation/immunology , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/immunology , Proto-Oncogene Proteins c-jun/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Spleen/cytology , Spleen/immunology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
6.
Proc Natl Acad Sci U S A ; 108(9): 3689-94, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21303985

ABSTRACT

Excessive release of proinflammatory cytokines by innate immune cells is an important component of the pathogenic basis of malaria. Proinflammatory cytokines are a direct output of Toll-like receptor (TLR) activation during microbial infection. Thus, interference with TLR function is likely to render a better clinical outcome by preventing their aberrant activation and the excessive release of inflammatory mediators. Herein, we describe the protective effect and mechanism of action of E6446, a synthetic antagonist of nucleic acid-sensing TLRs, on experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA. We show that in vitro, low doses of E6446 specifically inhibited the activation of human and mouse TLR9. Tenfold higher concentrations of this compound also inhibited the human TLR8 response to single-stranded RNA. In vivo, therapy with E6446 diminished the activation of TLR9 and prevented the exacerbated cytokine response observed during acute Plasmodium infection. Furthermore, severe signs of ECM, such as limb paralysis, brain vascular leak, and death, were all prevented by oral treatment with E6446. Hence, we provide evidence that supports the involvement of nucleic acid-sensing TLRs in malaria pathogenesis and that interference with the activation of these receptors is a promising strategy to prevent deleterious inflammatory responses that mediate pathogenesis and severity of malaria.


Subject(s)
Hydrocarbons, Aromatic/pharmacology , Malaria, Cerebral/prevention & control , Malaria, Cerebral/therapy , Nucleic Acids/metabolism , Toll-Like Receptors/antagonists & inhibitors , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Humans , Hydrocarbons, Aromatic/chemistry , Inflammation/complications , Inflammation/pathology , Lipopolysaccharides/pharmacology , Malaria, Cerebral/chemically induced , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Plasmodium chabaudi/drug effects , Plasmodium chabaudi/physiology , Shock, Septic/chemically induced , Shock, Septic/complications , Toll-Like Receptors/metabolism
7.
Proc Natl Acad Sci U S A ; 106(14): 5789-94, 2009 Apr 07.
Article in English | MEDLINE | ID: mdl-19297619

ABSTRACT

Malaria-induced sepsis is associated with an intense proinflammatory cytokinemia for which the underlying mechanisms are poorly understood. It has been demonstrated that experimental infection of humans with Plasmodium falciparum primes Toll-like receptor (TLR)-mediated proinflammatory responses. Nevertheless, the relevance of this phenomenon during natural infection and, more importantly, the mechanisms by which malaria mediates TLR hyperresponsiveness are unclear. Here we show that TLR responses are boosted in febrile patients during natural infection with P. falciparum. Microarray analyses demonstrated that an extraordinary percentage of the up-regulated genes, including genes involving TLR signaling, had sites for IFN-inducible transcription factors. To further define the mechanism involved in malaria-mediated "priming," we infected mice with Plasmodium chabaudi. The human data were remarkably predictive of what we observed in the rodent malaria model. Malaria-induced priming of TLR responses correlated with increased expression of TLR mRNA in a TLR9-, MyD88-, and IFNgamma-dependent manner. Acutely infected WT mice were highly susceptible to LPS-induced lethality while TLR9(-/-), IL12(-/-) and to a greater extent, IFNgamma(-/-) mice were protected. Our data provide unprecedented evidence that TLR9 and MyD88 are essential to initiate IL12 and IFNgamma responses and favor host hyperresponsiveness to TLR agonists resulting in overproduction of proinflammatory cytokines and the sepsis-like symptoms of acute malaria.


Subject(s)
Immunity, Innate , Interferon-gamma/immunology , Interleukin-12/immunology , Malaria/immunology , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptors/immunology , Animals , Cytokines , Fever , Gene Expression Profiling , Humans , Inflammation , Mice , Plasmodium chabaudi , Plasmodium falciparum , Sepsis/parasitology , Sepsis/pathology , Toll-Like Receptor 9/immunology , Toll-Like Receptors/genetics , Transcription Factors , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...