Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Front Immunol ; 14: 1143012, 2023.
Article in English | MEDLINE | ID: mdl-37143657

ABSTRACT

Introduction: Plasmodium sporozoites (SPZ) inoculated by Anopheles mosquitoes into the skin of the mammalian host migrate to the liver before infecting hepatocytes. Previous work demonstrated that early production of IL-6 in the liver is detrimental for the parasite growth, contributing to the acquisition of a long-lasting immune protection after immunization with live attenuated parasites. Methods: Considering that IL-6 as a critical pro-inflammatory signal, we explored a novel approach whereby the parasite itself encodes for the murine IL-6 gene. We generated transgenic P. berghei parasites that express murine IL-6 during liver stage development. Results and Discussion: Though IL-6 transgenic SPZ developed into exo-erythrocytic forms in hepatocytes in vitro and in vivo, these parasites were not capable of inducing a blood stage infection in mice. Furthermore, immunization of mice with transgenic IL-6-expressing P. berghei SPZ elicited a long-lasting CD8+ T cell-mediated protective immunity against a subsequent infectious SPZ challenge. Collectively, this study demonstrates that parasite-encoded IL-6 attenuates parasite virulence with abortive liver stage of Plasmodium infection, forming the basis of a novel suicide vaccine strategy to elicit protective antimalarial immunity.


Subject(s)
Liver Diseases , Malaria Vaccines , Animals , Mice , CD8-Positive T-Lymphocytes , Interleukin-6 , Mammals , Plasmodium berghei
2.
J Med Chem ; 64(14): 10403-10417, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34185525

ABSTRACT

Epigenetic post-translational modifications are essential for human malaria parasite survival and progression through its life cycle. Here, we present new functionalized suberoylanilide hydroxamic acid (SAHA) derivatives that chemically combine the pan-histone deacetylase inhibitor SAHA with the DNA methyltransferase inhibitor procainamide. A three- or four-step chemical synthesis was designed starting from cheap raw materials. Compared to the single drugs, the combined molecules showed a superior activity in Plasmodium and a potent inhibition against human HDAC6, exerting no cytotoxicity in human cell lines. These new compounds are fully active in multidrug-resistant Plasmodium falciparum Cambodian isolates. They target transmission of the parasite by inducing irreversible morphological changes in gametocytes and inhibiting exflagellation. The compounds are slow-acting and have an additive antimalarial effect in combination with fast-acting epidrugs and dihydroartemisinin. The lead compound decreases parasitemia in mice in a severe malaria model. Taken together, this novel fused molecule offers an affordable alternative to current failing antimalarial therapy.


Subject(s)
Antimalarials/pharmacology , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Procainamide/pharmacology , Antimalarials/chemical synthesis , Antimalarials/chemistry , Dose-Response Relationship, Drug , Drug Resistance, Multiple/drug effects , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Hydroxamic Acids/chemistry , Molecular Structure , Procainamide/chemistry , Structure-Activity Relationship
3.
Front Immunol ; 11: 585502, 2020.
Article in English | MEDLINE | ID: mdl-33329563

ABSTRACT

Deciphering the mechanisms by which Plasmodium parasites develop inside hepatocytes is an important step toward the understanding of malaria pathogenesis. We propose that the nature and the magnitude of the inflammatory response in the liver are key for the establishment of the infection. Here, we used mice deficient in the multidrug resistance-2 gene (Mdr2-/-)-encoded phospholipid flippase leading to the development of liver inflammation. Infection of Mdr2-/- mice with Plasmodium berghei ANKA (PbANKA) sporozoites (SPZ) resulted in the blockade of hepatic exo-erythrocytic forms (EEFs) with no further development into blood stage parasites. Interestingly, cultured primary hepatocytes from mutant and wild-type mice are equally effective in supporting EEF development. The abortive infection resulted in a long-lasting immunity in Mdr2-/- mice against infectious SPZ where neutrophils and IL-6 appear as key effector components along with CD8+ and CD4+ effector and central memory T cells. Inflammation-induced breakdown of liver tolerance promotes anti-parasite immunity and provides new approaches for the design of effective vaccines against malaria disease.


Subject(s)
Hepatitis/immunology , Hepatocytes/parasitology , Malaria , ATP Binding Cassette Transporter, Subfamily B/deficiency , Animals , Female , Hepatocytes/immunology , Inflammation/immunology , Liver/immunology , Liver/parasitology , Malaria/immunology , Malaria/parasitology , Mice , Plasmodium berghei , Sporozoites , ATP-Binding Cassette Sub-Family B Member 4
4.
PLoS One ; 15(5): e0232183, 2020.
Article in English | MEDLINE | ID: mdl-32379764

ABSTRACT

Due to the lack of efficiency to control malaria elicited by sub-unit vaccine preparations, vaccination with live-attenuated Plasmodium parasite as reported 70 years ago with irradiated sporozoites regained recently a significant interest. The complex life cycle of the parasite and the different stages of development between mammal host and anopheles do not help to propose an easy vaccine strategy. In order to achieve a complete long-lasting protection against Plasmodium infection and disease, we considered a genetically attenuated blood stage parasite in the hmgb2 gene coding for the high-mobility-group-box 2 (HMGB2). This Plasmodium protein belongs to the HMGB family and hold as the mammal proteins, a double life since it acts first as a nuclear factor involved in chromatin remodelling and transcription regulation and second, when secreted as an active pro-inflammatory alarmin protein. Even though the number of reports on whole living attenuated blood stage parasites is limited when compared to attenuated sporozoites, the results reported with Plasmodium KO parasites are very encouraging. In this report, we present a novel strategy based on pre-immunization with Δhmgb2PbNK65 parasitized red blood cells that confer long-lasting protection in a murine experimental cerebral malaria model against two highly pathogenic homologous and heterologous parasites.


Subject(s)
HMGB2 Protein/genetics , Malaria, Cerebral/prevention & control , Plasmodium berghei/genetics , Animals , Anopheles/immunology , CD8-Positive T-Lymphocytes/immunology , Cross Protection/immunology , Disease Models, Animal , Erythrocytes/parasitology , Female , HMGB2 Protein/metabolism , Immunization/methods , Malaria Vaccines/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Plasmodium berghei/pathogenicity , Sporozoites/genetics , Vaccination/methods , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
5.
ACS Cent Sci ; 6(1): 16-21, 2020 Jan 22.
Article in English | MEDLINE | ID: mdl-31989022

ABSTRACT

Malaria is the deadliest parasitic disease affecting over 200 million people worldwide. The increasing number of treatment failures due to multi-drug-resistant parasites in South-East Asia hinders the efforts for elimination. It is thus urgent to develop new antimalarials to contain these resistant parasites. Based on a previous report showing the presence of DNA methylation in Plasmodium, we generated new types of DNA methylation inhibitors against malaria parasites. The quinoline-quinazoline-based inhibitors kill parasites, including artemisinin-resistant field isolates adapted to culture, in the low nanomolar range. The compounds target all stages of the asexual cycle, including early rings, during a 6 h treatment period; they reduce DNA methylation in the parasite and show in vivo activity at 10 mg/kg. These potent inhibitors are a new starting point to develop fast-acting antimalarials that could be used in combination with artemisinins.

6.
Cell Microbiol ; 21(7): e13021, 2019 07.
Article in English | MEDLINE | ID: mdl-30835870

ABSTRACT

Protozoan pathogens secrete nanosized particles called extracellular vesicles (EVs) to facilitate their survival and chronic infection. Here, we show the inhibition by Plasmodium berghei NK65 blood stage-derived EVs of the proliferative response of CD4+ T cells in response to antigen presentation. Importantly, these results were confirmed in vivo by the capacity of EVs to diminish the ovalbumin-specific delayed type hypersensitivity response. We identified two proteins associated with EVs, the histamine releasing factor (HRF) and the elongation factor 1α (EF-1α) that were found to have immunosuppressive activities. Interestingly, in contrast to WT parasites, EVs from genetically HRF- and EF-1α-deficient parasites failed to inhibit T cell responses in vitro and in vivo. At the level of T cells, we demonstrated that EVs from WT parasites dephosphorylate key molecules (PLCγ1, Akt, and ERK) of the T cell receptor signalling cascade. Remarkably, immunisation with EF-1α alone or in combination with HRF conferred a long-lasting antiparasite protection and immune memory. In conclusion, we identified a new mechanism by which P. berghei-derived EVs exert their immunosuppressive functions by altering T cell responses. The identification of two highly conserved immune suppressive factors offers new conceptual strategies to overcome EV-mediated immune suppression in malaria-infected individuals.


Subject(s)
Biomarkers, Tumor/genetics , Extracellular Vesicles/immunology , Malaria/genetics , Peptide Elongation Factor 1/genetics , Animals , Antigen Presentation/immunology , Antigens/genetics , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , Cell Proliferation/genetics , Extracellular Vesicles/genetics , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Malaria/parasitology , Malaria/pathology , Plasmodium berghei/genetics , Plasmodium berghei/pathogenicity , T-Lymphocytes/immunology , T-Lymphocytes/parasitology , Tumor Protein, Translationally-Controlled 1
7.
Sci Rep ; 7(1): 9129, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28831137

ABSTRACT

While most subunit malaria vaccines provide only limited efficacy, pre-erythrocytic and erythrocytic genetically attenuated parasites (GAP) have been shown to confer complete sterilizing immunity. We recently generated a Plasmodium berghei (PbNK65) parasite that lacks a secreted factor, the histamine releasing factor (HRF) (PbNK65 hrfΔ), and induces in infected mice a self-resolving blood stage infection accompanied by a long lasting immunity. Here, we explore the immunological mechanisms underlying the anti-parasite protective properties of the mutant PbNK65 hrfΔ and demonstrate that in addition to an up-regulation of IL-6 production, CD4+ but not CD8+ T effector lymphocytes are indispensable for the clearance of malaria infection. Maintenance of T cell-associated protection is associated with the reduction in CD4+PD-1+ and CD8+PD-1+ T cell numbers. A higher number of central and effector memory B cells in mutant-infected mice also plays a pivotal role in protection. Importantly, we also demonstrate that prior infection with WT parasites followed by a drug cure does not prevent the induction of PbNK65 hrfΔ-induced protection, suggesting that such protection in humans may be efficient even in individuals that have been infected and who repeatedly received antimalarial drugs.


Subject(s)
Biomarkers, Tumor/genetics , Host-Parasite Interactions , Immunologic Memory , Malaria/immunology , Malaria/parasitology , Plasmodium/genetics , Plasmodium/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cytokines , Disease Models, Animal , Erythrocytes/immunology , Erythrocytes/parasitology , Female , Gene Expression , Life Cycle Stages , Mice , Plasmodium/growth & development , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Sequence Deletion , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Protein, Translationally-Controlled 1
8.
Sci Rep ; 7: 42243, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28181563

ABSTRACT

According to the WHO, and despite reduction in mortality rates, there were an estimated 438 000 malaria deaths in 2015. Therefore new antimalarials capable of limiting organ damage are still required. We show that systemic and lung adenovirus (Ad)-mediated over-expression of trappin-2 (T-2) an antibacterial molecule with anti-inflammatory activity, increased mice survival following infection with the cerebral malaria-inducing Plasmodium berghei ANKA (PbANKA) strain. Systemically, T-2 reduced PbANKA sequestration in spleen, lung, liver and brain, associated with a decrease in pro-inflammatory cytokines (eg TNF-α in spleen and lung) and an increase in IL-10 production in the lung. Similarly, local lung instillation of Ad-T-2 resulted in a reduced organ parasite sequestration and a shift towards an anti-inflammatory/repair response, potentially implicating monocytes in the protective phenotype. Relatedly, we demonstrated in vitro that human monocytes incubated with Plasmodium falciparum-infected red blood cells (Pf-iRBCs) and IgGs from hyper-immune African human sera produced T-2 and that the latter colocalized with merozoites and inhibited Pf multiplication. This array of data argues for the first time for the potential therapeutic usefulness of this host defense peptide in human malaria patients, with the aim to limit acute lung injury and respiratory distress syndrom often observed during malaria episodes.


Subject(s)
Anti-Infective Agents/therapeutic use , Antiparasitic Agents/therapeutic use , Elafin/therapeutic use , Malaria, Cerebral/drug therapy , Malaria, Cerebral/parasitology , Plasmodium berghei/drug effects , Administration, Intranasal , Animals , Anti-Infective Agents/pharmacology , Antiparasitic Agents/pharmacology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Elafin/pharmacology , Erythrocytes/parasitology , Female , Humans , Malaria, Cerebral/blood , Merozoites/metabolism , Mice, Inbred C57BL , Monocytes/metabolism , Parasitemia/drug therapy , Parasitemia/parasitology , Parasitemia/pathology , Plasmodium falciparum/growth & development , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism
9.
J Exp Med ; 213(8): 1419-28, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27432939

ABSTRACT

Although most vaccines against blood stage malaria in development today use subunit preparations, live attenuated parasites confer significantly broader and more lasting protection. In recent years, Plasmodium genetically attenuated parasites (GAPs) have been generated in rodent models that cause self-resolving blood stage infections and induce strong protection. All such GAPs generated so far bear mutations in housekeeping genes important for parasite development in red blood cells. In this study, using a Plasmodium berghei model compatible with tracking anti-blood stage immune responses over time, we report a novel blood stage GAP that lacks a secreted factor related to histamine-releasing factor (HRF). Lack of HRF causes an IL-6 increase, which boosts T and B cell responses to resolve infection and leave a cross-stage, cross-species, and lasting immunity. Mutant-induced protection involves a combination of antiparasite IgG2c antibodies and FcγR(+) CD11b(+) cell phagocytes, especially neutrophils, which are sufficient to confer protection. This immune-boosting GAP highlights an important role of opsonized parasite-mediated phagocytosis, which may be central to protection induced by all self-resolving blood stage GAP infections.


Subject(s)
Biomarkers, Tumor/genetics , Malaria , Plasmodium berghei , Protozoan Proteins , T-Lymphocytes/immunology , Animals , Antibodies, Protozoan/immunology , B-Lymphocytes/immunology , Disease Models, Animal , Female , Immunoglobulin G/immunology , Interleukin-6/immunology , Malaria/genetics , Malaria/immunology , Mice , Neutrophils/immunology , Phagocytosis/immunology , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Tumor Protein, Translationally-Controlled 1
10.
Infect Immun ; 83(7): 2771-84, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25916985

ABSTRACT

Eukaryotic high-mobility-group-box (HMGB) proteins are nuclear factors involved in chromatin remodeling and transcription regulation. When released into the extracellular milieu, HMGB1 acts as a proinflammatory cytokine that plays a central role in the pathogenesis of several immune-mediated inflammatory diseases. We found that the Plasmodium genome encodes two genuine HMGB factors, Plasmodium HMGB1 and HMGB2, that encompass, like their human counterparts, a proinflammatory domain. Given that these proteins are released from parasitized red blood cells, we then hypothesized that Plasmodium HMGB might contribute to the pathogenesis of experimental cerebral malaria (ECM), a lethal neuroinflammatory syndrome that develops in C57BL/6 (susceptible) mice infected with Plasmodium berghei ANKA and that in many aspects resembles human cerebral malaria elicited by P. falciparum infection. The pathogenesis of experimental cerebral malaria was suppressed in C57BL/6 mice infected with P. berghei ANKA lacking the hmgb2 gene (Δhmgb2 ANKA), an effect associated with a reduction of histological brain lesions and with lower expression levels of several proinflammatory genes. The incidence of ECM in pbhmgb2-deficient mice was restored by the administration of recombinant PbHMGB2. Protection from experimental cerebral malaria in Δhmgb2 ANKA-infected mice was associated with reduced sequestration in the brain of CD4(+) and CD8(+) T cells, including CD8(+) granzyme B(+) and CD8(+) IFN-γ(+) cells, and, to some extent, neutrophils. This was consistent with a reduced parasite sequestration in the brain, lungs, and spleen, though to a lesser extent than in wild-type P. berghei ANKA-infected mice. In summary, Plasmodium HMGB2 acts as an alarmin that contributes to the pathogenesis of cerebral malaria.


Subject(s)
HMGB2 Protein/metabolism , Malaria, Cerebral/pathology , Malaria, Cerebral/parasitology , Plasmodium berghei/pathogenicity , Virulence Factors/metabolism , Animals , Brain/pathology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Disease Models, Animal , Gene Deletion , Gene Knockout Techniques , HMGB2 Protein/genetics , Histocytochemistry , Mice, Inbred C57BL , Neutrophils/immunology , Plasmodium berghei/genetics , Virulence , Virulence Factors/genetics
11.
Cell Microbiol ; 17(4): 542-58, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25329441

ABSTRACT

Plasmodium spp., which causes malaria, produces a histamine-releasing factor (HRF), an orthologue of mammalian HRF. Histamine-releasing factor produced by erythrocytic stages of the parasite is thought to play a role in the pathogenesis of severe malaria. Here, we show in a rodent model that HRF is not important during the erythrocytic but pre-erythrocytic phase of infection, which mainly consists in the transformation in the liver of the mosquito-injected parasite form into the erythrocyte-infecting form. Development of P. berghei ANKA cl15cy1 liver stages lacking HRF is impaired and associated with an early rise in systemic IL-6, a cytokine that strongly suppresses development of Plasmodium liver stages. The defect is rescued by injection of anti-IL-6 antibodies or infection in IL-6-deficient mice and parasite HRF is sufficient to decrease IL-6 synthesis, indicating a direct role of parasite HRF in reducing host IL-6. The target cells modulated by HRF for IL-6 production at early time points during liver infection are neutrophils. Parasite HRF is thus used to down-regulate a cytokine with anti-parasite activity. Our data also highlight the link between a prolonged transition from liver to blood-stage infection and reduced incidence of experimental cerebral malaria.


Subject(s)
Biomarkers, Tumor/metabolism , Host-Pathogen Interactions , Interleukin-6/antagonists & inhibitors , Liver/parasitology , Malaria/pathology , Plasmodium berghei/physiology , Animals , Disease Models, Animal , Liver/pathology , Mice , Mice, Knockout , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Treatment Outcome , Tumor Protein, Translationally-Controlled 1
12.
J Exp Med ; 208(11): 2225-36, 2011 Oct 24.
Article in English | MEDLINE | ID: mdl-21967768

ABSTRACT

The role of the IgE-FcεRI complex in malaria severity in Plasmodium falciparum-hosting patients is unknown. We demonstrate that mice genetically deficient for the high-affinity receptor for IgE (FcεRIα-KO) or for IgE (IgE-KO) are less susceptible to experimental cerebral malaria (ECM) after infection with Plasmodium berghei (PbANKA). Mast cells and basophils, which are the classical IgE-expressing effector cells, are not involved in disease as mast cell-deficient and basophil-depleted mice developed a disease similar to wild-type mice. However, we show the emergence of an FcεRI(+) neutrophil population, which is not observed in mice hosting a non-ECM-inducing PbNK65 parasite strain. Depletion of this FcεRI(+) neutrophil population prevents ECM, whereas transfer of this population into FcεRIα-KO mice restores ECM susceptibility. FcεRI(+) neutrophils preferentially home to the brain and induce elevated levels of proinflammatory cytokines. These data define a new pathogenic mechanism of ECM and implicate an FcεRI-expressing neutrophil subpopulation in malaria disease severity.


Subject(s)
Immunoglobulin E/immunology , Malaria, Cerebral/immunology , Malaria, Cerebral/pathology , Neutrophils/immunology , Receptors, IgE/immunology , Adoptive Transfer , Animals , Basophils/cytology , Basophils/immunology , Cytokines/immunology , Eosinophils/cytology , Eosinophils/immunology , Female , Immunoglobulin E/genetics , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Plasmodium berghei/immunology , Plasmodium berghei/pathogenicity , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, IgE/genetics
13.
Vector Borne Zoonotic Dis ; 11(4): 423-32, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21395422

ABSTRACT

Malaria accounts for the greatest morbidity and mortality of any arthropod-borne disease globally. Recently, it was determined that the protective antisporozoite CD8+ T-cell response originates predominantly from cutaneous lymph nodes draining the site of parasite inoculation by an Anopheles mosquito. The female mosquito inoculates sporozoites along with an assortment of salivary proteins into the skin of its mammalian host. Mosquito saliva has demonstrable antihemostatic as well as various immunomodulatory activities, and studies with mosquito-borne viruses support a role for mosquito saliva in enhancement of transmission and exacerbation of disease. Early differences in immune response can be detected, which discriminate between mice that are resistant and susceptible to neurological pathology. This supports the idea that early divergence in the immune response may influence the likelihood of progression to the more severe forms of malaria. To evaluate the effect of mosquito feeding on the pathogenesis and immune response to malaria, we injected washed Plasmodium berghei sporozoites intradermally in the presence or absence of mosquito feeding. We observed that mice exposed to mosquito feeding in tandem with the inoculation of sporozoites had higher parasitemias and an elevated progression to cerebral malaria. This was associated with, in particular, elevated levels of interleukin-4 and interleukin-10, suppression of overall transcription in response to infection, and decreased extravasation of dendritic cells and monocytes. This study enhances to our understanding of the complexity of the interactions between the malaria parasite, its host, and the mosquito vector.


Subject(s)
Anopheles/parasitology , Malaria, Cerebral/parasitology , Mice, Inbred BALB C/immunology , Mice, Inbred C57BL/immunology , Plasmodium berghei/immunology , Animals , CD8-Positive T-Lymphocytes , Disease Models, Animal , Female , Host-Parasite Interactions , Insect Vectors/parasitology , Interleukin-10 , Interleukin-4 , Malaria, Cerebral/transmission , Mice , Mice, Inbred BALB C/parasitology , Mice, Inbred C57BL/parasitology , Plasmodium berghei/pathogenicity , Polymerase Chain Reaction , RNA, Ribosomal, 18S/analysis , Saliva/parasitology
14.
PLoS One ; 4(6): e6004, 2009 Jun 23.
Article in English | MEDLINE | ID: mdl-19547708

ABSTRACT

BACKGROUND: Histamine is a biogenic amine that has been shown to contribute to several pathological conditions, such as allergic conditions, experimental encephalomyelitis, and malaria. In humans, as well as in murine models of malaria, increased plasma levels of histamine are associated with severity of infection. We reported recently that histamine plays a critical role in the pathogenesis of experimental cerebral malaria (CM) in mice infected with Plasmodium berghei ANKA. Histamine exerts its biological effects through four different receptors designated H1R, H2R, H3R, and H4R. PRINCIPAL FINDINGS: In the present work, we explored the role of histamine signaling via the histamine H3 receptor (H3R) in the pathogenesis of murine CM. We observed that the lack of H3R expression (H3R(-/-) mice) accelerates the onset of CM and this was correlated with enhanced brain pathology and earlier and more pronounced loss of blood brain barrier integrity than in wild type mice. Additionally tele-methylhistamine, the major histamine metabolite in the brain, that was initially present at a higher level in the brain of H3R(-/-) mice was depleted more quickly post-infection in H3R(-/-) mice as compared to wild-type counterparts. CONCLUSIONS: Our data suggest that histamine regulation through the H3R in the brain suppresses the development of CM. Thus modulating histamine signaling in the central nervous system, in combination with standard therapies, may represent a novel strategy to reduce the risk of progression to cerebral malaria.


Subject(s)
Brain/parasitology , Malaria, Cerebral/metabolism , Receptors, Histamine H3/metabolism , Signal Transduction , Animals , Brain/immunology , Brain/metabolism , Disease Progression , Female , Histamine/metabolism , Immune System , Inflammation , Malaria, Cerebral/parasitology , Malaria, Cerebral/prevention & control , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Plasmodium berghei/metabolism
15.
Med Sci (Paris) ; 25(4): 377-81, 2009 Apr.
Article in French | MEDLINE | ID: mdl-19409190

ABSTRACT

A hallmark of the host response to Plasmodium parasite is an inflammatory reaction characterized by elevated histaminemia levels. Since histamine, which acts through four different receptors and which synthesis is under the control of the histidine decarboxylase (HDC), is endowed with pro-inflammatory and immunosuppressive activities, we hypothesized that this vaso-active amine may participe to malaria pathogenesis. Combining genetic and pharmacologic approaches by using H1R(-/-), H2R(-/-), H3R(-/-), HDC(-/-) mice and H1R, H2R-, and H3R-antagonists, respectively, we found that cerebral malaria-associated pathogenetic processes such as blood brain barrier disruption, and T lymphocyte sequestration to cerebral vascular endothelium in mice were associated with histamine production. The identification of this novel inflammatory pathway and its implication in Plasmodium infection may lead to novel strategies to manipulate the anti-Plasmodium immune response and may provide new therapeutic tools to alleviate malaria disease.


Subject(s)
Histamine/physiology , Malaria/physiopathology , Receptors, Histamine/physiology , Adult , Animals , Basophils/physiology , Blood-Brain Barrier/physiology , Child, Preschool , Histamine/blood , Histamine/deficiency , Histidine Ammonia-Lyase/deficiency , Histidine Ammonia-Lyase/physiology , Host-Parasite Interactions , Humans , Infant , Malaria, Cerebral/physiopathology , Malaria, Falciparum/physiopathology , Mast Cells/physiology , Mice , Mice, Knockout , Models, Biological , Parasitemia/physiopathology , Plasmodium berghei , Receptors, Histamine/deficiency , Receptors, Histamine/genetics
16.
J Exp Med ; 205(2): 395-408, 2008 Feb 18.
Article in English | MEDLINE | ID: mdl-18227221

ABSTRACT

From the inoculation of Plasmodium sporozoites via Anopheles mosquito bites to the development of blood-stage parasites, a hallmark of the host response is an inflammatory reaction characterized by elevated histamine levels in the serum and tissues. Given the proinflammatory and immunosuppressive activities associated with histamine, we postulated that this vasoactive amine participates in malaria pathogenesis. Combined genetic and pharmacologic approaches demonstrated that histamine binding to H1R and H2R but not H3R and H4R increases the susceptibility of mice to infection with Plasmodium. To further understand the role of histamine in malaria pathogenesis, we used histidine decarboxylase-deficient (HDC(-/-)) mice, which are free of histamine. HDC(-/-) mice were highly resistant to severe malaria whether infected by mosquito bites or via injection of infected erythrocytes. HDC(-/-) mice displayed resistance to two lethal strains: Plasmodium berghei (Pb) ANKA, which triggers cerebral malaria (CM), and Pb NK65, which causes death without neurological symptoms. The resistance of HDC(-/-) mice to CM was associated with preserved blood-brain barrier integrity, the absence of infected erythrocyte aggregation in the brain vessels, and a lack of sequestration of CD4 and CD8 T cells. We demonstrate that histamine-mediated signaling contributes to malaria pathogenesis. Understanding the basis for these biological effects of histamine during infection may lead to novel therapeutic strategies to alleviate the severity of malaria.


Subject(s)
Histamine/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/pathogenicity , Animals , Blood-Brain Barrier/metabolism , Brain/immunology , Brain/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/blood , Cytokines/genetics , Cytokines/immunology , Female , Histamine/blood , Histamine H1 Antagonists/pharmacology , Histidine Decarboxylase/deficiency , Histidine Decarboxylase/genetics , Histidine Decarboxylase/immunology , Malaria, Cerebral/blood , Mice , Mice, Knockout , Mice, Transgenic , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Histamine/deficiency , Receptors, Histamine/genetics , Receptors, Histamine/immunology
17.
J Immunol ; 176(7): 4141-6, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16547250

ABSTRACT

While probing host skin to search for blood vessels, the female Anopheles mosquito delivers Plasmodium parasites in the presence of saliva. Saliva from various blood-feeding vectors which contains several pharmacologically active components is believed to facilitate blood feeding as well as parasite transmission to the host. Recently, we found that mosquito saliva has the capacity to activate dermal mast cells and to induce local inflammatory cell influx. Our main objective in the present work is to investigate whether saliva, through mosquito bites, controls the magnitude of Ag-specific immune responses and whether this control is dependent on the mast cell-mediated inflammatory response. Using a mast cell knockin mouse model, we found that mosquito bites consistently induced MIP-2 in the skin and IL-10 in draining lymph nodes, and down-regulate Ag-specific T cell responses by a mechanism dependent on mast cells and mediated by IL-10. Our results provide evidence for new mechanisms which may operate during Plasmodium parasite transmission by mosquito bites.


Subject(s)
Anopheles/immunology , Antigens/immunology , Bites and Stings/immunology , Down-Regulation , Mast Cells/immunology , Animals , Antigens/metabolism , Cells, Cultured , Chemokines, CXC/metabolism , Cytokines/metabolism , Female , Hypersensitivity/immunology , Hypersensitivity/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mast Cells/metabolism , Mice , Saliva/immunology , Skin/immunology , Time Factors
18.
J Immunol ; 174(7): 3932-40, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15778349

ABSTRACT

When Anopheles mosquitoes probe the skin for blood feeding, they inject saliva in dermal tissue. Mosquito saliva is known to exert various biological activities, but its perception by the immune system and its role in parasite transmission remain poorly understood. In the present study, we report on the cellular changes occurring in the mouse skin and draining lymph nodes after a Anopheles stephensi mosquito bite. We show that mosquito bites induce dermal mast cell degranulation, leading to fluid extravasation and neutrophil influx. This inflammatory response does not occur in mast cell-deficient W/W(v) mice, unless these are reconstituted specifically with mast cells. Mast cell activation caused by A. stephensi mosquito bites is followed by hyperplasia of the draining lymph node due to the accumulation of CD3(+), B220(+), CD11b(+), and CD11c(+) leukocytes. The T cell enrichment of the draining lymph nodes results from their sequestration from the circulation rather than local proliferation. These data demonstrate that mosquito bites and very likely saliva rapidly trigger the immune system, emphasizing the critical contribution of peripheral mast cells in inducing T cell and dendritic cell recruitment within draining lymph nodes, a prerequisite for the elicitation of T and B lymphocyte priming.


Subject(s)
Anopheles/pathogenicity , Hyperplasia/etiology , Inflammation/etiology , Insect Bites and Stings/pathology , Lymph Nodes/pathology , Mast Cells/pathology , Animals , Cell Degranulation , Chemotaxis, Leukocyte/immunology , Female , Inflammation/pathology , Insect Bites and Stings/immunology , Mice , Mice, Inbred Strains , Neutrophils/physiology , Saliva/immunology , Skin/pathology , T-Lymphocytes/physiology
19.
J Immunol ; 170(6): 3037-45, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12626558

ABSTRACT

Mast cells (MCs) are considered major players in IgE-mediated allergic responses, but have also recently been recognized as active participants in innate as well as specific immune responses. Recent work provided evidence that MCs are able to activate B and T lymphocytes through the release of vesicles called exosomes. Here we demonstrate that exosomes, which are located in the endocytic pathway, harbor exogenous Ags that associate with other molecules endowed with immunomodulatory functions, including 60- and 70-kDa heat shock proteins. Administration to naive mice of Ag-containing exosomes in the absence of conventional adjuvants elicits specific Ab responses across the MHC II haplotype barrier. We demonstrate that MC-exosomes induce immature dendritic cells (DCs) to up-regulate MHC class II, CD80, CD86, and CD40 molecules and to acquire potent Ag-presenting capacity to T cells. Uptake and processing of Ag-associated exosomes by endogenous DCs were also demonstrated. Finally, exosome-associated heat shock proteins are critical for the acquisition by DCs of the Ag-presenting function. This work demonstrates a heretofore unrecognized collaborative interaction between MCs and DCs leading to the elicitation of specific immune responses.


Subject(s)
Cytoplasmic Vesicles/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Mast Cells/cytology , Mast Cells/immunology , Animals , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cattle , Cell Differentiation/immunology , Chaperonin 60/immunology , Chaperonin 60/metabolism , Cytoplasmic Vesicles/metabolism , Endocytosis/immunology , HSC70 Heat-Shock Proteins , HSP70 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/metabolism , Immunophenotyping , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred DBA , Transferrin/immunology , Transferrin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL