Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1143012, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37143657

RESUMO

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.


Assuntos
Hepatopatias , Vacinas Antimaláricas , Animais , Camundongos , Linfócitos T CD8-Positivos , Interleucina-6 , Mamíferos , Plasmodium berghei
2.
Front Immunol ; 11: 585502, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329563

RESUMO

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.


Assuntos
Hepatite/imunologia , Hepatócitos/parasitologia , Malária , Subfamília B de Transportador de Cassetes de Ligação de ATP/deficiência , Animais , Feminino , Hepatócitos/imunologia , Inflamação/imunologia , Fígado/imunologia , Fígado/parasitologia , Malária/imunologia , Malária/parasitologia , Camundongos , Plasmodium berghei , Esporozoítos , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
3.
PLoS One ; 15(5): e0232183, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32379764

RESUMO

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.


Assuntos
Proteína HMGB2/genética , Malária Cerebral/prevenção & controle , Plasmodium berghei/genética , Animais , Anopheles/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteção Cruzada/imunologia , Modelos Animais de Doenças , Eritrócitos/parasitologia , Feminino , Proteína HMGB2/metabolismo , Imunização/métodos , Vacinas Antimaláricas/imunologia , Malária Cerebral/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei/patogenicidade , Esporozoítos/genética , Vacinação/métodos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
4.
Cell Microbiol ; 21(7): e13021, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30835870

RESUMO

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.


Assuntos
Biomarcadores Tumorais/genética , Vesículas Extracelulares/imunologia , Malária/genética , Fator 1 de Elongação de Peptídeos/genética , Animais , Apresentação de Antígeno/imunologia , Antígenos/genética , Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/parasitologia , Proliferação de Células/genética , Vesículas Extracelulares/genética , Humanos , Evasão da Resposta Imune/genética , Evasão da Resposta Imune/imunologia , Malária/parasitologia , Malária/patologia , Plasmodium berghei/genética , Plasmodium berghei/patogenicidade , Linfócitos T/imunologia , Linfócitos T/parasitologia , Proteína Tumoral 1 Controlada por Tradução
5.
Sci Rep ; 7(1): 9129, 2017 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-28831137

RESUMO

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.


Assuntos
Biomarcadores Tumorais/genética , Interações Hospedeiro-Parasita , Memória Imunológica , Malária/imunologia , Malária/parasitologia , Plasmodium/genética , Plasmodium/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Citocinas , Modelos Animais de Doenças , Eritrócitos/imunologia , Eritrócitos/parasitologia , Feminino , Expressão Gênica , Estágios do Ciclo de Vida , Camundongos , Plasmodium/crescimento & desenvolvimento , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Deleção de Sequência , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Proteína Tumoral 1 Controlada por Tradução
6.
Sci Rep ; 7: 42243, 2017 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-28181563

RESUMO

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.


Assuntos
Anti-Infecciosos/uso terapêutico , Antiparasitários/uso terapêutico , Elafina/uso terapêutico , Malária Cerebral/tratamento farmacológico , Malária Cerebral/parasitologia , Plasmodium berghei/efeitos dos fármacos , Administração Intranasal , Animais , Anti-Infecciosos/farmacologia , Antiparasitários/farmacologia , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Elafina/farmacologia , Eritrócitos/parasitologia , Feminino , Humanos , Malária Cerebral/sangue , Merozoítos/metabolismo , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Parasitemia/tratamento farmacológico , Parasitemia/parasitologia , Parasitemia/patologia , Plasmodium falciparum/crescimento & desenvolvimento , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/metabolismo
7.
J Exp Med ; 213(8): 1419-28, 2016 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-27432939

RESUMO

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.


Assuntos
Biomarcadores Tumorais/genética , Malária , Plasmodium berghei , Proteínas de Protozoários , Linfócitos T/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Linfócitos B/imunologia , Modelos Animais de Doenças , Feminino , Imunoglobulina G/imunologia , Interleucina-6/imunologia , Malária/genética , Malária/imunologia , Camundongos , Neutrófilos/imunologia , Fagocitose/imunologia , Plasmodium berghei/genética , Plasmodium berghei/imunologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteína Tumoral 1 Controlada por Tradução
8.
Artigo em Inglês | MEDLINE | ID: mdl-26306096

RESUMO

BACKGROUND: The World Allergy Organization estimates that 40 % of the world's population is affected by allergic diseases. The International Study of Asthma and Allergies in Childhood has completed Phase III and it has now become clear that these diseases have increased in developing countries, especially Africa, where prevalence rates were formerly low. Despite an increase in studies in Africa, few sub-Saharan West African countries are represented; the focus has remained on urban populations and little attention has been paid to rural sub-Saharan Africa. METHODS: We performed an allergy survey in a birth cohort of children aged less than 15 years in rural Senegal and implemented an ISAAC questionnaire. We carried out a complete blood count and serological analyses for IgE levels against common allergens and mosquito saliva. RESULTS: The prevalence rates of asthma, rhinoconjunctivitis (RC) and atopic dermatitis (AD) were 12.8, 12.5 and 12.2 % respectively. Specific IgE (sIgE) levels against mosquito spp. salivary gland antigens were significantly associated with AD; sIgE levels against selected true grasses (Poaceae) were significantly associated with RC. sIgE levels against house dust mite spp. were not associated with asthma, but were significantly correlated with mosquito IgE levels. Such cross-reactivity may blur the association between HDM sIgE and asthma. Consumption of seafood, storing whey cream, using plant fibre bedding and presence of carpet were significantly associated with increased risk of RC. The association of seafood may be the result of histamine intoxication from molluscs prepared by putrefaction. Cat presence and dog contact were associated with increased risk of asthma. Cow contact was associated with increased risk of AD. CONCLUSIONS: Our allergy study in rural West Africa revealed lower prevalence rates than the majority of African urban settings. Although several associated known risk factors were identified, there were associations specific to the region. The identification of probable artefactual dietary phenomena is a challenge for robust diagnosis of allergic disease. The association AD with mosquito saliva, a common allergen in rural settings, warrants specific attention. Further studies in rural Africa are needed to address the aetiology of allergy in a non-urban environment.

9.
Cell Microbiol ; 17(4): 542-58, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25329441

RESUMO

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.


Assuntos
Biomarcadores Tumorais/metabolismo , Interações Hospedeiro-Patógeno , Interleucina-6/antagonistas & inibidores , Fígado/parasitologia , Malária/patologia , Plasmodium berghei/fisiologia , Animais , Modelos Animais de Doenças , Fígado/patologia , Camundongos , Camundongos Knockout , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/metabolismo , Resultado do Tratamento , Proteína Tumoral 1 Controlada por Tradução
10.
BMJ Open ; 3(7)2013.
Artigo em Inglês | MEDLINE | ID: mdl-23883878

RESUMO

OBJECTIVES: To assess the impact of atopy and allergy on the risk of clinical malaria. DESIGN: A clinical and immunological allergy cross-sectional survey in a birth cohort of 175 children from 1 month to 14 years of age followed for up to 15 years in a longitudinal open cohort study of malaria in Senegal. Malaria incidence data were available for 143 of these children (aged 4 months to 14 years of age) for up to 15 years. Mixed-model regression analysis was used to determine the impact of allergy status on malaria incidence, adjusting for age, gender, sickle-cell trait and force of infection. MAIN OUTCOME MEASURES: Asthma, allergic rhinoconjunctivitis and atopic dermatitis status, the number of clinical Plasmodium falciparum malaria episodes since birth and associated parasite density. RESULTS: 12% of the children were classified as asthmatic and 10% as having atopic dermatitis. These groups had respectively a twofold (OR 2.12 95%; CI 1.46 to 3.08; p=8×10(-5)) and threefold (OR 3.15; 1.56 to 6.33; p=1.3×10(-3)) increase in the risk of clinical P falciparum malaria once older than the age of peak incidence of clinical malaria (3-4 years of age). They also presented with higher P falciparum parasite densities (asthma: mean 105.3 parasites/µL±SE 41.0 vs 51.3±9.7; p=6.2×10(-3). Atopic dermatitis: 135.4±70.7 vs 52.3±11.0; p=0.014). There was no effect of allergy on the number of non-malaria clinical presentations. Individuals with allergic rhinoconjunctivitis did not have an increased risk of clinical malaria nor any difference in parasite densities. CONCLUSIONS: These results demonstrate that asthma and atopic dermatitis delay the development of clinical immunity to P falciparum. Despite the encouraging decrease in malaria incidence rates in Africa, a significant concern is the extent to which the increase in allergy will exacerbate the burden of malaria. Given the demonstrated antiparasitic effect of antihistamines, administration to atopic children will likely reduce the burden of clinical malaria in these children, increase the efficacy of first-line treatment antimalarials and alleviate the non-infectious consequences of atopy.

11.
Cancer Res ; 72(11): 2757-67, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22427351

RESUMO

During cancer development, a number of regulatory cell subsets and immunosuppressive cytokines subvert adaptive immune responses. Although it has been shown that tumor-derived interleukin (IL)-18 participates in the PD-1-dependent tumor progression in NK cell-controlled cancers, the mechanistic cues underlying this immunosuppression remain unknown. Here, we show that IL-18 converts a subset of Kit(-) (CD11b(-)) into Kit(+) natural killer (NK) cells, which accumulate in all lymphoid organs of tumor bearers and mediate immunoablative functions. Kit(+) NK cells overexpressed B7-H1/PD-L1, a ligand for PD-1. The adoptive transfer of Kit(+) NK cells promoted tumor growth in two pulmonary metastases tumor models and significantly reduced the dendritic and NK cell pools residing in lymphoid organs in a B7-H1-dependent manner. Neutralization of IL-18 by RNA interference in tumors or systemically by IL-18-binding protein dramatically reduced the accumulation of Kit(+)CD11b(-) NK cells in tumor bearers. Together, our findings show that IL-18 produced by tumor cells elicits Kit(+)CD11b(-) NK cells endowed with B7-H1-dependent immunoablative functions in mice.


Assuntos
Interleucina-18/imunologia , Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Animais , Antígeno B7-H1/análise , Antígeno B7-H1/fisiologia , Antígeno CD11b/análise , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-kit/análise
12.
Biochim Biophys Acta ; 1822(1): 49-56, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21354307

RESUMO

Plasmodium falciparum, the aetiological agent of human lethal malaria, is responsible for over 2 million deaths per year and malaria episodes may vary considerably in their severity and clinical manifestations. Dysregulated balance of the inflammatory response and a defect in the anti-Plasmodium parasite immune response represent the hallmarks of malaria disease. Among the many possible mechanisms, it is now widely recognized that the production of pro-inflammatory mediators and cytokines and upregulation of endothelial cell adhesion molecules play important roles in malaria pathogenesis. We and others provided evidence that some components of allergic inflammatory response to malaria parasites or elicited by by-products of parasite infection may contribute to malaria pathogenesis. This review provides some clue regarding these mechanisms where mast cells and histamine, an inflammatory mediator generated following IgE-independent or IgE-mediated immune response, were found to play a major role in parasite transmission and malaria pathogenesis, respectively. This article is part of a Special Issue entitled: Mast cells in inflammation.


Assuntos
Hipersensibilidade/parasitologia , Malária Falciparum/patologia , Mastócitos/patologia , Plasmodium falciparum/imunologia , Animais , Anopheles/metabolismo , Anopheles/parasitologia , Histamina/metabolismo , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Mordeduras e Picadas de Insetos/imunologia , Mordeduras e Picadas de Insetos/patologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Mastócitos/imunologia , Mastócitos/metabolismo , Saliva/imunologia , Transdução de Sinais , Pele/imunologia , Pele/patologia
13.
J Exp Med ; 208(11): 2225-36, 2011 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-21967768

RESUMO

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.


Assuntos
Imunoglobulina E/imunologia , Malária Cerebral/imunologia , Malária Cerebral/patologia , Neutrófilos/imunologia , Receptores de IgE/imunologia , Transferência Adotiva , Animais , Basófilos/citologia , Basófilos/imunologia , Citocinas/imunologia , Eosinófilos/citologia , Eosinófilos/imunologia , Feminino , Imunoglobulina E/genética , Malária Cerebral/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/citologia , Plasmodium berghei/imunologia , Plasmodium berghei/patogenicidade , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Receptores de IgE/genética
14.
Vector Borne Zoonotic Dis ; 11(4): 423-32, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21395422

RESUMO

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.


Assuntos
Anopheles/parasitologia , Malária Cerebral/parasitologia , Camundongos Endogâmicos BALB C/imunologia , Camundongos Endogâmicos C57BL/imunologia , Plasmodium berghei/imunologia , Animais , Linfócitos T CD8-Positivos , Modelos Animais de Doenças , Feminino , Interações Hospedeiro-Parasita , Insetos Vetores/parasitologia , Interleucina-10 , Interleucina-4 , Malária Cerebral/transmissão , Camundongos , Camundongos Endogâmicos BALB C/parasitologia , Camundongos Endogâmicos C57BL/parasitologia , Plasmodium berghei/patogenicidade , Reação em Cadeia da Polimerase , RNA Ribossômico 18S/análise , Saliva/parasitologia
15.
Front Immunol ; 2: 78, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22566867

RESUMO

One of the effector arms of the pathogenesis of severe forms of malaria disease is the development of uncontrolled or excessive inflammatory responses. A characteristic inflammatory response may arise from the propensity of some individuals to produce IgE antibodies against environmental antigens or against parasite components. We believe that an allergic inflammatory response which develops concomitantly with a malaria episode may drive the disease course toward severe forms. The role of the IgE-FcεRI complex in malaria severity in Plasmodium falciparum-hosting patients is unknown. Subsequently, except a very limited number of reports, study of effector cells that express this complex such as mast cells and basophils and that may contribute to malaria pathogenesis have been particularly neglected. A better understanding of this type of inflammatory response and its implication in malaria disease and how it impacts Plasmodium parasite development may provide additional tools to alleviate or to cure this deadly disease.

16.
J Invest Dermatol ; 131(4): 916-24, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21191401

RESUMO

CD160 is expressed by human and mouse natural killer (NK) cells and other cytotoxic lymphocyte subpopulations. CD160 is mostly expressed as a trimeric 83 kDa glycosylphosphatidylinositol (GPI)-anchored activating NK receptor, cleaved upon IL-15 stimulation in a secreted trimeric soluble form (sCD160) that binds to major histocompatibility complex (MHC) class I molecules, while a transmembrane isoform appears. sCD160 exhibits immunoregulatory function as it inhibits CD8(+) T-lymphocyte cytotoxic activity. We show that human mast cells (MCs) express CD160. In human and mouse skin, resident MCs expressed CD160, whereas in C57BL/6-Kit(W-sh/W-sh) mice, CD160(+) cells were only identified at the site of reconstitution with syngeneic cultured MCs. In the human mast cell line, HMC-1, we only identified the transcripts of the GPI-anchored CD160 isoform. Furthermore, CD160 was identified in HMC-1 and mouse MC supernatants, suggesting that MCs release sCD160. Supporting this hypothesis, HMC-1 express the GPI-specific phospholipase D variant 2 involved in the NK lymphocyte membrane cleavage of CD160, and morphological studies highlighted a relative loss of CD160 expression in inflammatory skin sites, where MC degranulation is expected to occur. We also demonstrated an inhibition of T-cell cytotoxicity by HMC-1 supernatant that was partially reversed by anti-CD160 mAb. In conclusion, sCD160, produced by MCs, may have a role in T-cell-MC interactions in vivo.


Assuntos
Dermatite/fisiopatologia , Proteínas Ligadas por GPI/metabolismo , Mastócitos/metabolismo , Mastócitos/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Antígenos CD/genética , Antígenos CD/imunologia , Células COS , Comunicação Celular/imunologia , Chlorocebus aethiops , Meios de Cultivo Condicionados/farmacologia , Dermatite/metabolismo , Dermatite/patologia , Derme/citologia , Derme/imunologia , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Expressão Gênica/imunologia , Humanos , Células Jurkat , Células Matadoras Naturais/citologia , Mastócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , RNA Mensageiro/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Solubilidade
17.
PLoS One ; 5(9): e12558, 2010 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-20838433

RESUMO

BACKGROUND: Pregnancy-associated malaria (PAM) is a serious consequence of the adhesion to the placental receptor chondroitin sulfate A (CSA) of Plasmodium falciparum-infected erythrocytes (PE) expressing the large cysteine-rich multi-domain protein var2CSA. Women become resistant to PAM, and develop strain-transcending immunity against CSA-binding parasites. The identification of var2CSA regions that could elicit broadly neutralizing and adhesion-blocking antibodies is a key step for the design of prophylactic vaccine strategies. METHODOLOGY: Escherichia coli expressed var2CSA DBL domains were refolded and purified prior to immunization of mice and a goat. Protein-G-purified antibodies were tested for their ability to block FCR3(CSA)-infected erythrocytes binding to placental (BeWo) and monkey brain endothelial (ScC2) cell lines using a flow cytoadhesion inhibition assay mimicking closely the physiological conditions present in the placenta at shear stress of 0.05 Pa. DBL5-ε, DBL6-ε and DBL5-6-ε induced cross-reactive antibodies using Alum and Freund as adjuvants, which blocked cytoadhesion at values ranging between 40 to 96% at 0.5 mg IgG per ml. Importantly, antibodies raised against recombinant DBL5-ε from 3 distinct parasites genotypes (HB3, Dd2 and 7G8) showed strain-transcending inhibition ranging from 38 to 64% for the heterologuous FCR3(CSA). CONCLUSIONS: Using single and double DBL domains from var2CSA and Alum as adjuvant, we identified recombinant subunits inducing an immune response in experimental animals which is able to block efficiently parasite adhesion in a flow cytoadhesion assay that mimics closely the erythrocyte flow in the placenta. These subunits show promising features for inclusion into a vaccine aiming to protect against PAM.


Assuntos
Antígenos de Protozoários/imunologia , Malária/imunologia , Placenta/parasitologia , Plasmodium falciparum/fisiologia , Complicações Parasitárias na Gravidez/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Adesão Celular , Linhagem Celular , Sulfatos de Condroitina/imunologia , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Humanos , Malária/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Placenta/citologia , Placenta/imunologia , Plasmodium falciparum/química , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Gravidez , Complicações Parasitárias na Gravidez/parasitologia , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Saimiri , Especificidade da Espécie
18.
PLoS One ; 4(6): e6004, 2009 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-19547708

RESUMO

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.


Assuntos
Encéfalo/parasitologia , Malária Cerebral/metabolismo , Receptores Histamínicos H3/metabolismo , Transdução de Sinais , Animais , Encéfalo/imunologia , Encéfalo/metabolismo , Progressão da Doença , Feminino , Histamina/metabolismo , Sistema Imunitário , Inflamação , Malária Cerebral/parasitologia , Malária Cerebral/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Plasmodium berghei/metabolismo
19.
J Exp Med ; 205(2): 395-408, 2008 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-18227221

RESUMO

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.


Assuntos
Histamina/imunologia , Malária Cerebral/imunologia , Plasmodium berghei/patogenicidade , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/imunologia , Encéfalo/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/sangue , Citocinas/genética , Citocinas/imunologia , Feminino , Histamina/sangue , Antagonistas dos Receptores Histamínicos H1/farmacologia , Histidina Descarboxilase/deficiência , Histidina Descarboxilase/genética , Histidina Descarboxilase/imunologia , Malária Cerebral/sangue , Camundongos , Camundongos Knockout , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores Histamínicos/deficiência , Receptores Histamínicos/genética , Receptores Histamínicos/imunologia
20.
J Immunol ; 176(7): 4141-6, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16547250

RESUMO

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.


Assuntos
Anopheles/imunologia , Antígenos/imunologia , Mordeduras e Picadas/imunologia , Regulação para Baixo , Mastócitos/imunologia , Animais , Antígenos/metabolismo , Células Cultivadas , Quimiocinas CXC/metabolismo , Citocinas/metabolismo , Feminino , Hipersensibilidade/imunologia , Hipersensibilidade/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Mastócitos/metabolismo , Camundongos , Saliva/imunologia , Pele/imunologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...