Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 378
Filtrer
1.
Immunobiology ; 229(5): 152837, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39089130

RÉSUMÉ

It is known that conventional antigen presentation involves phagocytosis of antigens followed by its internalization in endocytic compartments and presentation of epitopes through MHC class II molecules for CD4 T cells. However, since 1976 a cross-presentation pathway has been studied, in which CD8 T cells are activated via MHC class I with antigens acquired through phagocytosis or endocytosis by dendritic cells (DCs). Among some important molecules involved in the cross-presentation, the C-type lectin receptor of the Dectin-1 cluster (CLECs), particularly the CLEC9A receptor, not only is expressed in dendritic cells but also presents a pivotal role in this context. In special, CLEC12A has been highlighted as a malaria pigment hemozoin (HZ) receptor. During Plasmodium infection, hemozoin crystals defend the parasite against heme toxicity within erythrocytes, as well as the released native HZ elicits pro-inflammatory responses and can induce cross-presentation. Particularly, this crystal can be synthesized from hematin anhydride and mimics the native form, and the gaps generated between the nanocrystal domains during its synthesis allow for substance coupling followed by its coating. Therefore, this study aimed to assess whether synthetic hemozoin (sHz) or hematin anhydride could be a nanocarrier and promote cross-presentation in dendritic cells. Firstly, it was verified that sHz can carry coated and coupled antigens, the compounds can associate to LAMP1-positive vesicles and decrease overall intracellular pH, which can potentially enhance the cross-presentation of ovalbumin and Leishmania infantum antigens. Thus, this study adds important data in the molecular intricacies of antigen presentation by showing not only the sHz immunomodulatory properties but also its potential applications as an antigen carrier.


Sujet(s)
Présentation d'antigène , Cross-priming , Cellules dendritiques , Hémoprotéines , Hémoprotéines/immunologie , Cross-priming/immunologie , Animaux , Cellules dendritiques/immunologie , Souris , Nanoparticules/composition chimique , Humains , Paludisme/immunologie , Lectines de type C/métabolisme , Lectines de type C/immunologie , Ovalbumine/immunologie
2.
Immunobiology ; 229(4): 152823, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38861873

RÉSUMÉ

Acute lung injury caused by severe malaria (SM) is triggered by a dysregulated immune response towards the infection with Plasmodium parasites. Postmortem analysis of human lungs shows diffuse alveolar damage (DAD), the presence of CD8 lymphocytes, neutrophils, and increased expression of Intercellular Adhesion Molecule 1 (ICAM-1). P. berghei ANKA (PbA) infection in C57BL/6 mice reproduces many SM features, including acute lung injury characterized by DAD, CD8+ T lymphocytes and neutrophils in the lung parenchyma, and tissular expression of proinflammatory cytokines and adhesion molecules, such as IFNγ, TNFα, ICAM, and VCAM. Since this is related to a dysregulated immune response, immunomodulatory agents are proposed to reduce the complications of SM. The monocyte locomotion inhibitory factor (MLIF) is an immunomodulatory pentapeptide isolated from axenic cultures of Entamoeba hystolitica. Thus, we evaluated if the MLIF intraperitoneal (i.p.) treatment prevented SM-induced acute lung injury. The peptide prevented SM without a parasiticidal effect, indicating that its protective effect was related to modifications in the immune response. Furthermore, peripheral CD8+ leukocytes and neutrophil proportions were higher in infected treated mice. However, the treatment prevented DAD, CD8+ cell infiltration into the pulmonary tissue and downregulated IFNγ. Moreover, VCAM-1 expression was abrogated. These results indicate that the MLIF treatment downregulated adhesion molecule expression, impeding cell migration and proinflammatory cytokine tissular production, preventing acute lung injury induced by SM. Our findings represent a potential novel strategy to avoid this complication in various events where a dysregulated immune response triggers lung injury.


Sujet(s)
Lésion pulmonaire aigüe , Modèles animaux de maladie humaine , Paludisme , Plasmodium berghei , Animaux , Lésion pulmonaire aigüe/immunologie , Lésion pulmonaire aigüe/étiologie , Souris , Paludisme/immunologie , Plasmodium berghei/immunologie , Souris de lignée C57BL , Granulocytes neutrophiles/immunologie , Lymphocytes T CD8+/immunologie , Cytokines/métabolisme , Poumon/immunologie , Poumon/anatomopathologie , Humains , Femelle , Oligopeptides
3.
Front Immunol ; 14: 1140426, 2023.
Article de Anglais | MEDLINE | ID: mdl-36993971

RÉSUMÉ

Introduction: This study provides evidence of how Th1 cell metabolism is modulated by the purinergic receptor P2X7 (P2RX7), a cation cannel activated by high extracellular concentrations of adenosine triphosphate (ATP). Methods: In vivo analysis was performed in the Plasmodium chabaudi model of malaria in view of the great relevance of this infectious disease for human health, as well as the availability of data concerning Th1/Tfh differentiation. Results: We show that P2RX7 induces T-bet expression and aerobic glycolysis in splenic CD4+ T cells that respond to malaria, at a time prior to Th1/Tfh polarization. Cell-intrinsic P2RX7 signaling sustains the glycolytic pathway and causes bioenergetic mitochondrial stress in activated CD4+ T cells. We also show in vitro the phenotypic similarities of Th1-conditioned CD4+ T cells that do not express P2RX7 and those in which the glycolytic pathway is pharmacologically inhibited. In addition, in vitro ATP synthase blockade and the consequent inhibition of oxidative phosphorylation, which drives cellular metabolism for aerobic glycolysis, is sufficient to promote rapid CD4+ T cell proliferation and polarization to the Th1 profile in the absence of P2RX7. Conclusion: These data demonstrate that P2RX7-mediated metabolic reprograming for aerobic glycolysis is a key event for Th1 differentiation and suggest that ATP synthase inhibition is a downstream effect of P2RX7 signaling that potentiates the Th1 response.


Sujet(s)
Glycolyse , Paludisme , Récepteurs purinergiques P2X7 , Lymphocytes auxiliaires Th1 , Animaux , Souris , Souris de lignée C57BL , Récepteurs purinergiques P2X7/métabolisme , Lymphocytes auxiliaires Th1/cytologie , Lymphocytes auxiliaires Th1/métabolisme , Différenciation cellulaire , Plasmodium chabaudi , Paludisme/immunologie , Adénosine triphosphate , Adenosine triphosphatases , Mitochondries/métabolisme , Protéines à domaine boîte-T/métabolisme , Phosphorylation oxydative , Transduction du signal , Cellules cultivées
4.
Front Immunol ; 12: 584660, 2021.
Article de Anglais | MEDLINE | ID: mdl-34248924

RÉSUMÉ

The immune response of Anopheles mosquitoes to Plasmodium invasion has been extensively studied and shown to be mediated mainly by the nitric oxide synthase (NOS), dual oxidase (DUOX), phenoloxidase (PO), and antimicrobial peptides activity. Here, we studied the correlation between a heat shock insult, transcription of immune response genes, and subsequent susceptibility to Plasmodium berghei infection in Anopheles albimanus. We found that transcript levels of many immune genes were drastically affected by the thermal stress, either positively or negatively. Furthermore, the transcription of genes associated with modifications of nucleic acid methylation was affected, suggesting an increment in both DNA and RNA methylation. The heat shock increased PO and NOS activity in the hemolymph, as well as the transcription of several immune genes. As consequence, we observed that heat shock increased the resistance of mosquitoes to Plasmodium invasion. The data provided here could help the understanding of infection transmission under the ever more common heat waves.


Sujet(s)
Anopheles/immunologie , Anopheles/parasitologie , Réaction de choc thermique/immunologie , Hémolymphe/parasitologie , Paludisme/immunologie , Plasmodium berghei/immunologie , Animaux , Anopheles/génétique , Femelle , Réaction de choc thermique/génétique , Immunité/génétique , Paludisme/parasitologie
5.
Malar J ; 20(1): 296, 2021 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-34210332

RÉSUMÉ

BACKGROUND: Ascariasis and malaria are highly prevalent parasitic diseases in tropical regions and often have overlapping endemic areas, contributing to high morbidity and mortality rates in areas with poor sanitary conditions. Several studies have previously aimed to correlate the effects of Ascaris-Plasmodium coinfections but have obtained contradictory and inconclusive results. Therefore, the present study aimed to investigate parasitological and immunopathological aspects of the lung during murine experimental concomitant coinfection by Plasmodium berghei and Ascaris suum during larvae ascariasis. METHODS: C57BL/6J mice were inoculated with 1 × 104 P. berghei strain NK65-NY-infected red blood cells (iRBCs) intraperitoneally and/or 2500 embryonated eggs of A. suum by oral gavage. P. berghei parasitaemia, morbidity and the survival rate were assessed. On the seventh day postinfection (dpi), A. suum lung burden analysis; bronchoalveolar lavage (BAL); histopathology; NAG, MPO and EPO activity measurements; haematological analysis; and respiratory mechanics analysis were performed. The concentrations of interleukin (IL)-1ß, IL-12/IL-23p40, IL-6, IL-4, IL-33, IL-13, IL-5, IL-10, IL-17A, IFN-γ, TNF and TGF-ß were assayed by sandwich ELISA. RESULTS: Animals coinfected with P. berghei and A. suum show decreased production of type 1, 2, and 17 and regulatory cytokines; low leukocyte recruitment in the tissue; increased cellularity in the circulation; and low levels of NAG, MPO and EPO activity that lead to an increase in larvae migration, as shown by the decrease in larvae recovered in the lung parenchyma and increase in larvae recovered in the airway. This situation leads to severe airway haemorrhage and, consequently, an impairment respiratory function that leads to high morbidity and early mortality. CONCLUSIONS: This study demonstrates that the Ascaris-Plasmodium interaction is harmful to the host and suggests that this coinfection may potentiate Ascaris-associated pathology by dampening the Ascaris-specific immune response, resulting in the early death of affected animals.


Sujet(s)
Ascaridiose , Co-infection , Régulation négative/immunologie , Immunité innée/génétique , Paludisme , Animaux , Ascaridiose/immunologie , Ascaridiose/parasitologie , Ascaridiose/anatomopathologie , Ascaris suum/génétique , Ascaris suum/physiologie , Co-infection/immunologie , Co-infection/parasitologie , Co-infection/anatomopathologie , Régulation de l'expression des gènes , Poumon/anatomopathologie , Paludisme/immunologie , Paludisme/parasitologie , Paludisme/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Plasmodium berghei/physiologie
6.
Crit Care ; 25(1): 236, 2021 07 06.
Article de Anglais | MEDLINE | ID: mdl-34229735

RÉSUMÉ

Infectious diseases may affect brain function and cause encephalopathy even when the pathogen does not directly infect the central nervous system, known as infectious disease-associated encephalopathy. The systemic inflammatory process may result in neuroinflammation, with glial cell activation and increased levels of cytokines, reduced neurotrophic factors, blood-brain barrier dysfunction, neurotransmitter metabolism imbalances, and neurotoxicity, and behavioral and cognitive impairments often occur in the late course. Even though infectious disease-associated encephalopathies may cause devastating neurologic and cognitive deficits, the concept of infectious disease-associated encephalopathies is still under-investigated; knowledge of the underlying mechanisms, which may be distinct from those of encephalopathies of non-infectious cause, is still limited. In this review, we focus on the pathophysiology of encephalopathies associated with peripheral (sepsis, malaria, influenza, and COVID-19), emerging therapeutic strategies, and the role of neuroinflammation.


Sujet(s)
Encéphalopathies/immunologie , COVID-19/complications , Cytokines/immunologie , Grippe humaine/complications , Paludisme/complications , Sepsie/complications , Barrière hémato-encéphalique/immunologie , Encéphalopathies/prévention et contrôle , COVID-19/immunologie , Humains , Grippe humaine/immunologie , Paludisme/immunologie , Sepsie/immunologie
7.
Infect Immun ; 89(8): e0080920, 2021 07 15.
Article de Anglais | MEDLINE | ID: mdl-34061587

RÉSUMÉ

Pregnancy-associated malaria is often associated with adverse pregnancy outcomes. Placental circulatory impairments are an intriguing and unsolved component of malaria pathophysiology. Here, we uncovered a Toll-like receptor 4 (TLR4)-TRIF-endothelin axis that controls trophoblast motility and is linked to fetal protection during Plasmodium infection. In a cohort of 401 pregnancies from northern Brazil, we found that infection during pregnancy reduced expression of endothelin receptor B in syncytiotrophoblasts, while endothelin expression was only affected during acute infection. We further show that quantitative expression of placental endothelin and endothelin receptor B proteins are differentially controlled by maternal and fetal TLR4 alleles. Using murine malaria models, we identified placental autonomous responses to malaria infection mediated by fetally encoded TLR4 that not only controlled placental endothelin gene expression but also correlated with fetal viability protection. In vitro assays showed that control of endothelin expression in fetal syncytiotrophoblasts exposed to Plasmodium-infected erythrocytes was dependent on TLR4 via the TRIF pathway but not MyD88 signaling. Time-lapse microscopy in syncytiotrophoblast primary cultures and cell invasion assays demonstrated that ablation of TLR4 or endothelin receptor blockade abrogates trophoblast collective motility and cell migration responses to infected erythrocytes. These results cohesively substantiate the hypothesis that fetal innate immune sensing, namely, the TRL4-TRIF pathway, exerts a fetal protective role during malaria infection by mediating syncytiotrophoblast vasoregulatory responses that counteract placental insufficiency.


Sujet(s)
Endothélines/métabolisme , Placenta/métabolisme , Placenta/parasitologie , Transduction du signal , Récepteur de type Toll-4/métabolisme , Trophoblastes/métabolisme , Marqueurs biologiques , Brésil , Femelle , Interactions hôte-pathogène/immunologie , Humains , Paludisme/immunologie , Paludisme/métabolisme , Paludisme/parasitologie , Placenta/immunologie , Grossesse , Complications parasitaires de la grossesse , Issue de la grossesse
8.
Am J Trop Med Hyg ; 104(6): 2139-2145, 2021 04 05.
Article de Anglais | MEDLINE | ID: mdl-33819177

RÉSUMÉ

Haiti is targeting malaria elimination by 2025. The Grand'Anse department in southwestern Haiti experiences one-third to half of all nationally reported Plasmodium falciparum cases. Although there are historical reports of Plasmodium vivax and Plasmodium malariae, today, non-falciparum infections would remain undetected because of extensive use of falciparum-specific histidine-rich protein 2 (HRP2) rapid diagnostic tests (RDT) at health facilities. A recent case-control study was conducted in Grand'Anse to identify risk factors for P. falciparum infection using HRP2-based RDTs (n = 1,107). Post hoc multiplex Plasmodium antigenemia and antibody (IgG) detection by multiplex bead assay revealed one blood sample positive for pan-Plasmodium aldolase, negative for P. falciparum HRP2, and positive for IgG antibodies to P. malariae. Based on this finding, we selected 52 samples with possible P. malariae infection using IgG and antigenemia data and confirmed infection status by species-specific PCR. We confirmed one P. malariae infection in a 6-month-old infant without travel history. Congenital P. malariae could not be excluded. However, our finding-in combination with historical reports of P. malariae-warrants further investigation into the presence and possible extent of non-falciparum malaria in Haiti. Furthermore, we showed the use of multiplex Plasmodium antigen and IgG detection in selecting samples of interest for subsequent PCR analysis, thereby reducing costs as opposed to testing all available samples by PCR. This is of specific use in low-transmission or eliminating settings where infections are rare.


Sujet(s)
Anticorps antiprotozoaires/sang , Antigènes de protozoaire/sang , Éradication de maladie/méthodes , Paludisme/diagnostic , Paludisme/prévention et contrôle , Dépistage de masse/méthodes , Plasmodium malariae/immunologie , Protéines de protozoaire/sang , Adolescent , Antigènes de protozoaire/immunologie , Études cas-témoins , Enfant , Enfant d'âge préscolaire , Éradication de maladie/normes , Haïti/épidémiologie , Humains , Immunoglobuline G/sang , Nourrisson , Paludisme/épidémiologie , Paludisme/immunologie , Dépistage de masse/statistiques et données numériques , Plasmodium malariae/composition chimique , Plasmodium malariae/génétique , Protéines de protozoaire/immunologie
9.
Cells ; 10(2)2021 02 23.
Article de Anglais | MEDLINE | ID: mdl-33672278

RÉSUMÉ

Malaria is a parasitic disease (caused by different Plasmodium species) that affects millions of people worldwide. The lack of effective malaria drugs and a vaccine contributes to this disease, continuing to cause major public health and socioeconomic problems, especially in low-income countries. Cell death is implicated in malaria immune responses by eliminating infected cells, but it can also provoke an intense inflammatory response and lead to severe malaria outcomes. The study of the pathophysiological role of cell death in malaria in mammalians is key to understanding the parasite-host interactions and design prophylactic and therapeutic strategies for malaria. In this work, we review malaria-triggered cell death pathways (apoptosis, autophagy, necrosis, pyroptosis, NETosis, and ferroptosis) and we discuss their potential role in the development of new approaches for human malaria therapies.


Sujet(s)
Paludisme/anatomopathologie , Transduction du signal , Animaux , Mort cellulaire , Humains , Immunité , Paludisme/immunologie , Modèles biologiques , Pyroptose
10.
Arq. ciências saúde UNIPAR ; 25(1): 53-59, jan-abr. 2021.
Article de Portugais | LILACS | ID: biblio-1151416

RÉSUMÉ

O referido trabalho tem como objetivo analisar e avaliar a atual conjuntura das pesquisas científicas na busca da imunização eficaz contra a malária, destacando os principais mecanismos imunológicos e moleculares subjacentes à referida proteção, bem como, as perspectivas a curto e médio prazo. O presente estudo de revisão selecionou pesquisas nas bases de dados da Medical Literature Analysis and Retrieval System Online (Medline), National Library of Medicine (Pubmed), Scientific Electronic Library Online (SciELO), Web of Science e Scopus. Foram combinados os termos Malaria, Immunization, Vaccine and Epidemiology, com seus sinônimos remissivos e outros descritores associados, no período compreendido entre janeiro e julho de 2019. Como fator preponderante dos critérios de inclusão, foram selecionadas revisões sistemáticas com ou sem metanálise, publicadas nos últimos 5 anos, que discorressem detalhadamente sobre o tema, ou que apresentassem informações estatísticas ou históricas relevantes, relacionada ao tema. Como critérios de exclusão foram considerados: materiais literários e científicos, anteriores ao período de 2014 e que não apresentassem informações estatísticas ou histórica relevantes ao tema, ou que, não se adequassem à temática da pesquisa. Após a aplicação dos critérios de inclusão e exclusão, foi realizada a análise e seleção dos artigos. Dos 451 artigos identificados, 44 foram selecionados. As informações extraídas dos referidos trabalhos convergem no sentido de que a erradicação da malária é uma tarefa demasiadamente complexa, a qual não será alcançada com as vacinas atuais, havendo necessidade do desenvolvimento de ferramentas imunizadoras de maior eficácia. Apesar dos esforços, atualmente ainda não existe uma vacina eficaz na prevenção da infecção, mas vários estudos se encontram em andamento nessa vertente, tornando promissor o surgimento de uma vacina eficaz contra o parasita.


This study aims at analyzing and evaluating the current status of scientific research in the search for effective immunization against malaria, highlighting the key immunological and molecular mechanisms of such protection and the short- and medium-term perspectives. The search and selection of studies took place in the databases of the Medical Literature Analysis and Retrieval System Online (Medline); National Library of Medicine (Pubmed); Scientific Electronic Library Online (SciELO); Web of Science; and Scopus. The terms Malaria, Immunization, Vaccine, and Epidemiology were used, with their corresponding cross-referenced synonyms and other associated descriptors, including the period from January to July 2019. As a main factor in the inclusion criteria, systematic reviews with or without meta-analysis published in the last 5 years, presenting a detailed discourse about the topic, or relevant statistical or historical information related to the topic were selected. The following exclusion criteria were considered: literary and scientific materials, prior to 2014, and without statistical or historical information relevant to the theme, or which did not fit the research theme. After applying the inclusion and exclusion criteria, the articles were analyzed and selected. From a total of 451 identified articles, 44 were selected. The information extracted from the referred studies converge in the sense that malaria eradication is an overly complex task, which will not be achieved with the current vaccines, requiring the development of more effective immunizing tools. Despite all the efforts, there is no effective vaccine for preventing infection yet, but several studies are being developed in this area, making the emergence of an effective vaccine against the disease promising.


Sujet(s)
Immunisation , Paludisme/immunologie , Parasites , Protozooses/prévention et contrôle , Vaccins/immunologie , Épidémiologie/statistiques et données numériques , Sporozoïtes/immunologie , Infections/épidémiologie
11.
J. Venom. Anim. Toxins incl. Trop. Dis. ; 27: e20200073, 2021. tab, graf, ilus
Article de Anglais | VETINDEX | ID: vti-31982

RÉSUMÉ

he resistance against antimalarial drugs represents a global challenge in the fight and control of malaria. The Brazilian biodiversity can be an important tool for research and development of new medicinal products. In this context, toxinology is a multidisciplinary approach on the development of new drugs, including the isolation, purification, and evaluation of the pharmacological activities of natural toxins. The present study aimed to evaluate the cytotoxicity, as well as the antimalarial activity in silico and in vitro of four compounds isolated from Rhinella marina venom as potential oral drug prototypes. Methods: Four compounds were challenged against 35 target proteins from P. falciparum and screened to evaluate their physicochemical properties using docking assay in Brazilian Malaria Molecular Targets (BraMMT) software and in silico assay in OCTOPUS® software. The in vitro antimalarial activity of the compounds against the 3D7 Plasmodium falciparum clones were assessed using the SYBR Green I based assay (IC50). For the cytotoxic tests, the LD50 was determined in human pulmonary fibroblast cell line using the [3(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] (MTT) assay. Results: All compounds presented a ligand-receptor interaction with ten Plasmodium falciparum-related protein targets, as well as antimalarial activity against chloroquine resistant strain (IC50 = 3.44 µM to 19.11 µM). Three of them (dehydrobufotenine, marinobufagin, and bufalin) showed adequate conditions for oral drug prototypes, with satisfactory prediction of absorption, permeability, and absence of toxicity. In the cell viability assay, only dehydrobufotenin was selective for the parasite. Conclusions: Dehydrobufotenin revealed to be a potential oral drug prototype presenting adequate antimalarial activity and absence of cytotoxicity, therefore should be subjected to further studies.(AU)


Sujet(s)
Bufanolide/administration et posologie , Bufonidae , Biodiversité , Paludisme/immunologie , Antipaludiques , Techniques in vitro , Simulation numérique
12.
J. venom. anim. toxins incl. trop. dis ; J. venom. anim. toxins incl. trop. dis;27: e20200073, 2021. tab, graf, ilus
Article de Anglais | LILACS, VETINDEX | ID: biblio-1154769

RÉSUMÉ

he resistance against antimalarial drugs represents a global challenge in the fight and control of malaria. The Brazilian biodiversity can be an important tool for research and development of new medicinal products. In this context, toxinology is a multidisciplinary approach on the development of new drugs, including the isolation, purification, and evaluation of the pharmacological activities of natural toxins. The present study aimed to evaluate the cytotoxicity, as well as the antimalarial activity in silico and in vitro of four compounds isolated from Rhinella marina venom as potential oral drug prototypes. Methods: Four compounds were challenged against 35 target proteins from P. falciparum and screened to evaluate their physicochemical properties using docking assay in Brazilian Malaria Molecular Targets (BraMMT) software and in silico assay in OCTOPUS® software. The in vitro antimalarial activity of the compounds against the 3D7 Plasmodium falciparum clones were assessed using the SYBR Green I based assay (IC50). For the cytotoxic tests, the LD50 was determined in human pulmonary fibroblast cell line using the [3(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] (MTT) assay. Results: All compounds presented a ligand-receptor interaction with ten Plasmodium falciparum-related protein targets, as well as antimalarial activity against chloroquine resistant strain (IC50 = 3.44 µM to 19.11 µM). Three of them (dehydrobufotenine, marinobufagin, and bufalin) showed adequate conditions for oral drug prototypes, with satisfactory prediction of absorption, permeability, and absence of toxicity. In the cell viability assay, only dehydrobufotenin was selective for the parasite. Conclusions: Dehydrobufotenin revealed to be a potential oral drug prototype presenting adequate antimalarial activity and absence of cytotoxicity, therefore should be subjected to further studies.(AU)


Sujet(s)
Bufanolide/administration et posologie , Bufonidae , Biodiversité , Paludisme/immunologie , Antipaludiques , Techniques in vitro , Simulation numérique
13.
PLoS Pathog ; 16(9): e1008840, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32913355

RÉSUMÉ

P. vivax-infected Retics (iRetics) express human leukocyte antigen class I (HLA-I), are recognized by CD8+ T cells and killed by granulysin (GNLY) and granzymes. However, how Plasmodium infection induces MHC-I expression on Retics is unknown. In addition, whether GNLY helps control Plasmodium infection in vivo has not been studied. Here, we examine these questions using rodent infection with the P. yoelii 17XNL strain, which has tropism for Retics. Infection with P. yoelii caused extramedullary erythropoiesis, reticulocytosis and expansion of CD8+CD44+CD62L- IFN-γ-producing T cells that form immune synapses with iRetics. We now provide evidence that MHC-I expression by iRetic is dependent on IFN-γ-induced transcription of IRF-1, MHC-I and ß2-microglobulin (ß2-m) in erythroblasts. Consistently, CTLs from infected wild type (WT) mice formed immune synapses with iRetics in an IFN-γ- and MHC-I-dependent manner. When challenged with P. yoelii 17XNL, WT mice cleared parasitemia and survived, while IFN-γ KO mice remained parasitemic and all died. ß2-m KO mice that do not express MHC-I and have virtually no CD8+ T cells had prolonged parasitemia, and 80% survived. Because mice do not express GNLY, GNLY-transgenic mice can be used to assess the in vivo importance of GNLY. Parasite clearance was accelerated in GNLY-transgenic mice and depletion of CD8+ T cells ablated the GNLY-mediated resistance to P. yoelii. Altogether, our results indicate that in addition to previously described mechanisms, IFN-γ promotes host resistance to the Retic-tropic P. yoelii 17XNL strain by promoting MHC-I expression on iRetics that become targets for CD8+ cytotoxic T lymphocytes and GNLY.


Sujet(s)
Antigènes de différenciation des lymphocytes T/immunologie , Lymphocytes T CD8+/immunologie , Interféron gamma/immunologie , Paludisme/immunologie , Plasmodium yoelii/immunologie , Animaux , Antigènes de différenciation des lymphocytes T/génétique , Lymphocytes T CD8+/anatomopathologie , Interféron gamma/génétique , Paludisme/génétique , Paludisme/anatomopathologie , Souris , Souris de lignée BALB C , Souris knockout
14.
PLoS Pathog ; 16(8): e1008230, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32797076

RÉSUMÉ

Neutrophil extracellular traps (NETs) evolved as a unique effector mechanism contributing to resistance against infection that can also promote tissue damage in inflammatory conditions. Malaria infection can trigger NET release, but the mechanisms and consequences of NET formation in this context remain poorly characterized. Here we show that patients suffering from severe malaria had increased amounts of circulating DNA and increased neutrophil elastase (NE) levels in plasma. We used cultured erythrocytes and isolated human neutrophils to show that Plasmodium-infected red blood cells release macrophage migration inhibitory factor (MIF), which in turn caused NET formation by neutrophils in a mechanism dependent on the C-X-C chemokine receptor type 4 (CXCR4). NET production was dependent on histone citrullination by peptidyl arginine deiminase-4 (PAD4) and independent of reactive oxygen species (ROS), myeloperoxidase (MPO) or NE. In vitro, NETs functioned to restrain parasite dissemination in a mechanism dependent on MPO and NE activities. Finally, C57/B6 mice infected with P. berghei ANKA, a well-established model of cerebral malaria, presented high amounts of circulating DNA, while treatment with DNAse increased parasitemia and accelerated mortality, indicating a role for NETs in resistance against Plasmodium infection.


Sujet(s)
Érythrocytes/immunologie , Pièges extracellulaires/immunologie , Facteurs inhibiteurs de la migration des macrophages/métabolisme , Paludisme/immunologie , Granulocytes neutrophiles/immunologie , Plasmodium/immunologie , Récepteurs CXCR4/métabolisme , Animaux , Érythrocytes/métabolisme , Érythrocytes/parasitologie , Pièges extracellulaires/métabolisme , Pièges extracellulaires/parasitologie , Humains , Paludisme/métabolisme , Paludisme/parasitologie , Paludisme/anatomopathologie , Souris , Souris de lignée C57BL , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/parasitologie , Parasitémie/immunologie , Parasitémie/métabolisme , Parasitémie/parasitologie , Parasitémie/anatomopathologie
15.
Cytometry A ; 97(9): 872-881, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32686260

RÉSUMÉ

Malaria is a threat to human mankind and kills about half a million people every year. On the other hand, COVID-19 resulted in several hundred thousand deaths since December 2019 and remains without an efficient and safe treatment. The antimalarials chloroquine (CQ) and its analog, hydroxychloroquine (HCQ), have been tested for COVID-19 treatment, and several conflicting evidence has been obtained. Therefore, the aim of this review was to summarize the evidence regarding action mechanisms of these compounds against Plasmodium and SARS-CoV-2 infection, together with cytometry applications. CQ and HCQ act on the renin angiotensin system, with possible implications on the cardiorespiratory system. In this context, flow and image cytometry emerge as powerful technologies to investigate the mechanism of therapeutic candidates, as well as for the identification of the immune response and prognostics of disease severity. Data from the large randomized trials support the conclusion that CQ and HCQ do not provide any clinical improvements in disease severity and progression of SARS-CoV-2 patients, as well as they do not present any solid evidence of increased serious side effects. These drugs are safe and effective antimalarials agents, but in SARS-CoV-2 patients, they need further studies in the context of clinical trials. © 2020 International Society for Advancement of Cytometry.


Sujet(s)
Antipaludiques/usage thérapeutique , Antiviraux/usage thérapeutique , Betacoronavirus/effets des médicaments et des substances chimiques , Chloroquine/usage thérapeutique , Infections à coronavirus/traitement médicamenteux , Paludisme/traitement médicamenteux , Plasmodium/effets des médicaments et des substances chimiques , Pneumopathie virale/traitement médicamenteux , Animaux , Antipaludiques/effets indésirables , Antiviraux/effets indésirables , Betacoronavirus/immunologie , Betacoronavirus/pathogénicité , COVID-19 , Chloroquine/effets indésirables , Infections à coronavirus/diagnostic , Infections à coronavirus/immunologie , Infections à coronavirus/virologie , Cytométrie en flux , Interactions hôte-microbes , Interactions hôte-parasite , Humains , Paludisme/diagnostic , Paludisme/immunologie , Paludisme/parasitologie , Pandémies , Plasmodium/immunologie , Plasmodium/pathogénicité , Pneumopathie virale/diagnostic , Pneumopathie virale/immunologie , Pneumopathie virale/virologie , SARS-CoV-2 , Résultat thérapeutique , Traitements médicamenteux de la COVID-19
16.
Front Immunol ; 11: 1010, 2020.
Article de Anglais | MEDLINE | ID: mdl-32655546

RÉSUMÉ

Trypanosoma cruzi P21 protein (P21) is a putative secreted and immunomodulatory molecule with potent bioactive properties such as induction of phagocytosis and actin cytoskeleton polymerization. Despite the bioactive properties described so far, the action of P21 on parasite replication in muscle cell lineage or T. cruzi parasitism during acute experimental infection is unclear. We observed that recombinant P21 (rP21) decreased the multiplication of T. cruzi in C2C12 myoblasts, phenomenon associated with greater actin polymerization and IFN-γ and IL-4 higher expression. During experimental infection, lower cardiac nests, inflammatory infiltrate and fibrosis were observed in mice infected and treated with rP21. These results were correlated with large expression of IFN-γ counterbalanced by high levels of IL-10, which was consistent with the lower cardiac tissue injury found in these mice. We have also observed that upon stress, such as that induced by the presence of the IFN-γ cytokine, T. cruzi produced more P21. The effect of P21 in controlling the replication of T. cruzi, may indicate an evolutionary mechanism of survival developed by the parasite. Thus, when subjected to different stress conditions, the protozoan produces more P21, which induces T. cruzi latency in the host organism, enabling the protozoan to evade the host's immune system.


Sujet(s)
Protéines et peptides de signalisation intercellulaire/métabolisme , Paludisme/parasitologie , Myoblastes/parasitologie , Myocarde/anatomopathologie , Protéines de protozoaire/métabolisme , Trypanosoma cruzi/physiologie , Maladie aigüe , Animaux , Lignée cellulaire , Interactions hôte-parasite , Humains , Échappement immunitaire , Protéines et peptides de signalisation intercellulaire/génétique , Interféron gamma/métabolisme , Paludisme/immunologie , Souris , Souris de lignée C57BL , Modèles animaux , Charge parasitaire , Protéines de protozoaire/génétique
17.
Trends Parasitol ; 36(5): 459-472, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-32298633

RÉSUMÉ

Inflammasomes are cytosolic complexes that assemble in response to cellular stress or upon sensing microbial molecules, culminating in cytokine processing and an inflammatory form of cell death called pyroptosis. Inflammasomes are usually composed of a sensor molecule, an adaptor protein, and an inflammatory caspase, such as Caspase-1, which cleaves and activates multiple substrates, including Gasdermin-D, pro-IL-1ß, and pro-IL-18. Ultimately, inflammasome activation promotes inflammation and restriction of the microbial infection. In recent years, many studies have addressed the role of inflammasomes during fungal, bacterial, viral, and parasitic diseases, revealing sophisticated aspects of the host-pathogen interaction. In this review, we summarize recent advances on inflammasome activation in response to intracellular parasites, including Leishmania spp., Plasmodium spp., Trypanosoma cruzi, and Toxoplasma gondii.


Sujet(s)
Interactions hôte-pathogène/immunologie , Inflammasomes/immunologie , Protozooses/immunologie , Animaux , Eucaryotes/immunologie , Humains , Leishmaniose/immunologie , Leishmaniose/parasitologie , Paludisme/immunologie , Paludisme/parasitologie , Protozooses/parasitologie , Recherche/tendances , Toxoplasmose/immunologie , Toxoplasmose/parasitologie , Trypanosomiase/immunologie , Trypanosomiase/parasitologie
18.
Am J Hypertens ; 33(8): 695-702, 2020 08 04.
Article de Anglais | MEDLINE | ID: mdl-32211753

RÉSUMÉ

Malaria etiologies with pathophysiological similarities to hypertension currently constitute a major subject of research. The malaria-high blood pressure hypothesis is strongly supported by observations of the increasing incidence of hypertension in malaria-endemic, low- and middle-income countries with poor socioeconomic conditions, particularly in sub-Saharan African countries. Malnutrition and low birth weight with persistent symptomatic malaria presentations in pregnancy correlate strongly with the development of preeclampsia, gestational hypertension and subsequent hypertension in adult life. Evidence suggest that the link between malaria infection and high blood pressure involves interactions between malaria parasites and erythrocytes, the inflammatory process, effects of the infection during pregnancy; effects on renal and vascular functions as well as effects in sickle cell disease. Possible mechanisms which provide justification for the malaria-high blood pressure hypothesis include the following: endothelial dysfunction (reduced nitric oxide (NO) levels), impaired release of local neurotransmitters and cytokines, decrease in vascular smooth muscle cell viability and/or alterations in cellular calcium signaling leading to enhanced vascular reactivity, remodeling, and cardiomyopathies, deranged homeostasis through dehydration, elevated intracellular mediators and proinflammatory cytokine responses, possible genetic regulations, activation of the renin-angiotensin-aldosterone system mechanisms and renal derangements, severe anemia and hemolysis, renal failure, and end organ damage. Two key mediators of the malaria-high blood pressure association are: endothelial dysfunction (reduced NO) and increased angiotensin-converting enzyme activity/angiotensin II levels. Sickle cell disease is associated with protection against malaria infection and reduced blood pressure. In this review, we present the state of knowledge about the malaria-blood pressure hypothesis and suggest insights for future studies.


Sujet(s)
Endothélium/physiopathologie , Hypertension artérielle/physiopathologie , Paludisme/physiopathologie , Système rénine-angiotensine/physiologie , Angiotensine-II/métabolisme , Signalisation calcique , Survie cellulaire , Cytokines/métabolisme , Pays en voie de développement , Endothélium/métabolisme , Femelle , Humains , Hypertension artérielle/épidémiologie , Hypertension artérielle/immunologie , Hypertension artérielle/métabolisme , Hypertension artérielle gravidique/épidémiologie , Inflammation/immunologie , Inflammation/métabolisme , Paludisme/épidémiologie , Paludisme/immunologie , Paludisme/métabolisme , Muscles lisses vasculaires , Myocytes du muscle lisse/métabolisme , Agents neuromédiateurs/métabolisme , Monoxyde d'azote/métabolisme , Peptidyl-Dipeptidase A/métabolisme , Pré-éclampsie/épidémiologie , Grossesse
19.
Sci Adv ; 6(10): eaax6346, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-32181339

RÉSUMÉ

Placental malaria (PM) is associated with severe inflammation leading to abortion, preterm delivery, and intrauterine growth restriction. Innate immunity responses play critical roles, but the mechanisms underlying placental immunopathology are still unclear. Here, we investigated the role of inflammasome activation in PM by scrutinizing human placenta samples from an endemic area and ablating inflammasome components in a PM mouse model. The reduction in birth weight in babies from infected mothers is paralleled by increased placental expression of AIM2 and NLRP3 inflammasomes. Using genetic dissection, we reveal that inflammasome activation pathways are involved in the production and detrimental action of interleukin-1ß (IL-1ß) in the infected placenta. The IL-1R pharmacological antagonist Anakinra improved pregnancy outcomes by restoring fetal growth and reducing resorption in an experimental model. These findings unveil that IL-1ß-mediated signaling is a determinant of PM pathogenesis, suggesting that IL-1R antagonists can improve clinical outcomes of malaria infection in pregnancy.


Sujet(s)
Inflammasomes/effets des médicaments et des substances chimiques , Interleukine-1 bêta/immunologie , Paludisme à Plasmodium falciparum/immunologie , Paludisme/immunologie , Plasmodium falciparum/pathogénicité , Complications parasitaires de la grossesse/immunologie , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Caspase-1/génétique , Caspase-1/immunologie , Lignée cellulaire , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/immunologie , Femelle , Régulation de l'expression des gènes , Humains , Immunité innée , Facteurs immunologiques/pharmacologie , Inflammasomes/génétique , Inflammasomes/immunologie , Interféron gamma/génétique , Interféron gamma/immunologie , Antagoniste du récepteur à l'interleukine-1/pharmacologie , Interleukine-1 bêta/antagonistes et inhibiteurs , Interleukine-1 bêta/génétique , Paludisme/traitement médicamenteux , Paludisme/génétique , Paludisme/parasitologie , Paludisme à Plasmodium falciparum/génétique , Paludisme à Plasmodium falciparum/parasitologie , Paludisme à Plasmodium falciparum/anatomopathologie , Souris , Souris knockout , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Protéine-3 de la famille des NLR contenant un domaine pyrine/immunologie , Plasmodium berghei/immunologie , Plasmodium berghei/pathogénicité , Plasmodium falciparum/immunologie , Grossesse , Complications parasitaires de la grossesse/génétique , Complications parasitaires de la grossesse/parasitologie , Complications parasitaires de la grossesse/prévention et contrôle , Récepteurs à l'interleukine-1/génétique , Récepteurs à l'interleukine-1/immunologie , Transduction du signal/immunologie , Cellules THP-1 , Trophoblastes/effets des médicaments et des substances chimiques , Trophoblastes/immunologie , Trophoblastes/parasitologie , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/immunologie
20.
Int J Mol Sci ; 21(3)2020 Jan 21.
Article de Anglais | MEDLINE | ID: mdl-31973044

RÉSUMÉ

Insect saliva induces significant antibody responses associated with the intensity of exposure to bites and the risk of disease in humans. Several salivary biomarkers have been characterized to determine exposure intensity to Old World Anopheles mosquito species. However, new tools are needed to quantify the intensity of human exposure to Anopheles bites and understand the risk of malaria in low-transmission areas in the Americas. To address this need, we conducted proteomic and bioinformatic analyses of immunogenic candidate proteins present in the saliva of uninfected Anopheles albimanus from two separate colonies-one originating from Central America (STECLA strain) and one originating from South America (Cartagena strain). A ~65 kDa band was identified by IgG antibodies in serum samples from healthy volunteers living in a malaria endemic area in Colombia, and a total of five peptides were designed from the sequences of two immunogenic candidate proteins that were shared by both strains. ELISA-based testing of human IgG antibody levels against the peptides revealed that the transferrin-derived peptides, TRANS-P1, TRANS-P2 and a salivary peroxidase peptide (PEROX-P3) were able to distinguish between malaria-infected and uninfected groups. Interestingly, IgG antibody levels against PEROX-P3 were significantly lower in people that have never experienced malaria, suggesting that it may be a good marker for mosquito bite exposure in naïve populations such as travelers and deployed military personnel. In addition, the strength of the differences in the IgG levels against the peptides varied according to location, suggesting that the peptides may able to detect differences in intensities of bite exposure according to the mosquito population density. Thus, the An. albimanus salivary peptides TRANS-P1, TRANS-P2, and PEROX-P3 are promising biomarkers that could be exploited in a quantitative immunoassay for determination of human-vector contact and calculation of disease risk.


Sujet(s)
Anopheles/métabolisme , Paludisme/immunologie , Protéines et peptides salivaires/immunologie , Protéines et peptides salivaires/isolement et purification , Animaux , Production d'anticorps , Antigènes , Marqueurs biologiques/sang , Colombie , Humains , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Morsures et piqûres d'insectes , Protéines d'insecte/immunologie , Vecteurs moustiques , Projets pilotes , Protéomique , Salive/composition chimique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE