Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.299
Filtrar
1.
Immunohorizons ; 8(5): 371-383, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38780542

RESUMO

Our previous work demonstrated that basophils regulate a suite of malaria phenotypes, including intestinal mastocytosis and permeability, the immune response to infection, gametocytemia, and parasite transmission to the malaria mosquito Anopheles stephensi. Given that activated basophils are primary sources of the regulatory cytokines IL-4 and IL-13, we sought to examine the contributions of these mediators to basophil-dependent phenotypes in malaria. We generated mice with basophils depleted for IL-4 and IL-13 (baso IL-4/IL-13 (-)) and genotype controls (baso IL-4/IL-13 (+)) by crossing mcpt8-Cre and Il4/Il13fl/fl mice and infected them with Plasmodium yoelii yoelii 17XNL. Conditional deletion was associated with ileal mastocytosis and mast cell (MC) activation, increased intestinal permeability, and increased bacterial 16S levels in blood, but it had no effect on neutrophil activation, parasitemia, or transmission to A. stephensi. Increased intestinal permeability in baso IL-4/IL-13 (-) mice was correlated with elevated plasma eotaxin (CCL11), a potent eosinophil chemoattractant, and increased ileal MCs, proinflammatory IL-17A, and the chemokines MIP-1α (CCL3) and MIP-1ß (CCL4). Blood bacterial 16S copies were positively but weakly correlated with plasma proinflammatory cytokines IFN-γ and IL-12p40, suggesting that baso IL-4/IL-13 (-) mice failed to control bacterial translocation into the blood during malaria infection. These observations suggest that basophil-derived IL-4 and IL-13 do not contribute to basophil-dependent regulation of parasite transmission, but these cytokines do orchestrate protection of intestinal barrier integrity after P. yoelii infection. Specifically, basophil-dependent IL-4/IL-13 control MC activation and prevent infection-induced intestinal barrier damage and bacteremia, perhaps via regulation of eosinophils, macrophages, and Th17-mediated inflammation.


Assuntos
Translocação Bacteriana , Basófilos , Interleucina-13 , Interleucina-4 , Malária , Plasmodium yoelii , Animais , Interleucina-13/metabolismo , Basófilos/imunologia , Basófilos/metabolismo , Malária/imunologia , Camundongos , Plasmodium yoelii/imunologia , Interleucina-4/metabolismo , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos Endogâmicos C57BL , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/parasitologia , Camundongos Knockout , Feminino , Anopheles/parasitologia , Anopheles/imunologia , Anopheles/microbiologia
2.
Infect Immun ; 92(5): e0011324, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38624215

RESUMO

Malaria, one of the major infectious diseases in the world, is caused by the Plasmodium parasite. Plasmodium antigens could modulate the inflammatory response by binding to macrophage membrane receptors. As an export protein on the infected erythrocyte membrane, Plasmodium surface-related antigen (SRA) participates in the erythrocyte invasion and regulates the immune response of the host. This study found that the F2 segment of P. yoelii SRA activated downstream MAPK and NF-κB signaling pathways by binding to CD68 on the surface of the macrophage membrane and regulating the inflammatory response. The anti-PySRA-F2 antibody can protect mice against P. yoelii, and the pro-inflammatory responses such as IL-1ß, TNF-α, and IL-6 after infection with P. yoelii are attenuated. These findings will be helpful for understanding the involvement of the pathogenic mechanism of malaria with the exported protein SRA.


Assuntos
Antígenos CD , Antígenos de Diferenciação Mielomonocítica , Macrófagos , Malária , Plasmodium yoelii , Plasmodium yoelii/imunologia , Animais , Camundongos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/parasitologia , Malária/imunologia , Malária/parasitologia , Antígenos CD/metabolismo , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/metabolismo , Antígenos de Diferenciação Mielomonocítica/imunologia , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/metabolismo , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Humanos , Feminino , Antígenos de Superfície/imunologia , Antígenos de Superfície/metabolismo , Ligação Proteica , Transdução de Sinais , NF-kappa B/metabolismo , NF-kappa B/imunologia , Membrana Celular/metabolismo , Membrana Celular/imunologia , Inflamação/imunologia , Inflamação/metabolismo
3.
Front Immunol ; 14: 1140426, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36993971

RESUMO

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.


Assuntos
Glicólise , Malária , Receptores Purinérgicos P2X7 , Células Th1 , Animais , Camundongos , Camundongos Endogâmicos C57BL , Receptores Purinérgicos P2X7/metabolismo , Células Th1/citologia , Células Th1/metabolismo , Diferenciação Celular , Plasmodium chabaudi , Malária/imunologia , Trifosfato de Adenosina , Adenosina Trifosfatases , Mitocôndrias/metabolismo , Proteínas com Domínio T/metabolismo , Fosforilação Oxidativa , Transdução de Sinais , Células Cultivadas
4.
Adv Sci (Weinh) ; 9(22): e2103701, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35635376

RESUMO

Cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) play critical roles in the innate immunity against infectious diseases and are required to link pathogen DNA sensing to immune responses. However, the mechanisms by which cGAS-STING-induced cytokines suppress the adaptive immune response against malaria infections remain poorly understood. Here, cGAS-STING signaling is identified to play a detrimental role in regulating anti-malaria immunity. cGAS or STING deficiency in mice markedly prolongs mouse survival during lethal malaria Plasmodium yoelii nigeriensis N67C infections by reducing late interleukin (IL)-6 production. Mechanistically, cGAS/STING recruits myeloid differentiation factor 88 (MyD88) and specifically induces the p38-dependent signaling pathway for late IL-6 production, which, in turn, expands CD11b+ Ly6Chi proinflammatory monocytes to inhibit immunity. Moreover, the blockage or ablation of the cGAS-STING-MyD88-p38-IL-6 signaling axis or the depletion of CD11b+ Ly6Chi proinflammatory monocytes provides mice a significant survival benefit during N67C and other lethal malaria-strain infections. Taken together, these findings identify a previously unrecognized detrimental role of cGAS-STING-MyD88-p38 axis in infectious diseases through triggering the late IL-6 production and proinflammatory monocyte expansion and provide insight into how targeting the DNA sensing pathway, dysregulated cytokines, and proinflammatory monocytes enhances immunity against infection.


Assuntos
Malária , Monócitos , Animais , DNA , Interleucina-6/metabolismo , Malária/imunologia , Malária/mortalidade , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Monócitos/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo
5.
Front Immunol ; 13: 801120, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154114

RESUMO

An increase in mast cells (MCs) and MCs mediators has been observed in malaria-associated bacteremia, however, the role of these granulocytes in malarial immunity is poorly understood. Herein, we studied the role of mouse MC protease (Mcpt) 4, an ortholog of human MC chymase, in malaria-induced bacteremia using Mcpt4 knockout (Mcpt4-/-) mice and Mcpt4+/+ C57BL/6J controls, and the non-lethal mouse parasite Plasmodium yoelii yoelii 17XNL. Significantly lower parasitemia was observed in Mcpt4-/- mice compared with Mcpt4+/+ controls by day 10 post infection (PI). Although bacterial 16S DNA levels in blood were not different between groups, increased intestinal permeability to FITC-dextran and altered ileal adherens junction E-cadherin were observed in Mcpt4-/- mice. Relative to infected Mcpt4+/+ mice, ileal MC accumulation in Mcpt4-/- mice occurred two days earlier and IgE levels were higher by days 8-10 PI. Increased levels of circulating myeloperoxidase were observed at 6 and 10 days PI in Mcpt4+/+ but not Mcpt4-/- mice, affirming a role for neutrophil activation that was not predictive of parasitemia or bacterial 16S copies in blood. In contrast, early increased plasma levels of TNF-α, IL-12p40 and IL-3 were observed in Mcpt4-/- mice, while levels of IL-2, IL-10 and MIP1ß (CCL4) were increased over the same period in Mcpt4+/+ mice, suggesting that the host response to infection was skewed toward a type-1 immune response in Mcpt4-/- mice and type-2 response in Mcpt4+/+ mice. Spearman analysis revealed an early (day 4 PI) correlation of Mcpt4-/- parasitemia with TNF-α and IFN-γ, inflammatory cytokines known for their roles in pathogen clearance, a pattern that was observed in Mcpt4+/+ mice much later (day 10 PI). Transmission success of P. y. yoelii 17XNL to Anopheles stephensi was significantly higher from infected Mcpt4-/- mice compared with infected Mcpt4+/+ mice, suggesting that Mcpt4 also impacts transmissibility of sexual stage parasites. Together, these results suggest that early MCs activation and release of Mcpt4 suppresses the host immune response to P. y. yoelii 17XNL, perhaps via degradation of TNF-α and promotion of a type-2 immune response that concordantly protects epithelial barrier integrity, while limiting the systemic response to bacteremia and parasite transmissibility.


Assuntos
Anopheles/parasitologia , Permeabilidade da Membrana Celular/imunologia , Quimases/genética , Quimases/imunologia , Interações Hospedeiro-Parasita/imunologia , Malária/imunologia , Mastócitos/enzimologia , Plasmodium yoelii/imunologia , Animais , Feminino , Íleo/citologia , Íleo/patologia , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo
6.
Eur J Immunol ; 52(2): 270-284, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34773640

RESUMO

Recognition of pathogen-associated molecular patterns (PAMPs) through Toll-like receptors (TLRs) plays a pivotal role in first-line pathogen defense. TLRs are also likely triggered during a Plasmodium infection in vivo by parasite-derived components. However, the contribution of innate responses to liver infection and to the subsequent clinical outcome of a blood infection is not well understood. To assess the potential effects of enhanced TLR-signalling on Plasmodium infection, we systematically examined the effect of agonist-primed immune responses to sporozoite inoculation in the P. berghei/C57Bl/6 murine malaria model. We could identify distinct stage-specific effects on the course of infection after stimulation with two out of four TLR-ligands tested. Priming with a TLR9 agonist induced killing of pre-erythrocytic stages in the liver that depended on macrophages and the expression of inducible nitric oxide synthase (iNOS). These factors have previously not been recognized as antigen-independent effector mechanisms against Plasmodium liver stages. Priming with TLR4 and -9 agonists also translated into blood stage-specific protection against experimental cerebral malaria (ECM). These insights are relevant to the activation of TLR signalling pathways by adjuvant systems of antimalaria vaccine strategies. The protective role of TLR4-activation against ECM might also explain some unexpected clinical effects observed with pre-erythrocytic vaccine approaches.


Assuntos
Hepatopatias , Fígado , Ativação de Macrófagos , Macrófagos/imunologia , Malária , Plasmodium berghei/imunologia , Transdução de Sinais , Receptor Toll-Like 9/imunologia , Animais , Feminino , Fígado/imunologia , Fígado/parasitologia , Hepatopatias/genética , Hepatopatias/imunologia , Hepatopatias/parasitologia , Malária/genética , Malária/imunologia , Camundongos , Camundongos Transgênicos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor Toll-Like 9/genética
7.
Immunity ; 54(12): 2859-2876.e7, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34788599

RESUMO

Repeat antigens, such as the Plasmodium falciparum circumsporozoite protein (PfCSP), use both sequence degeneracy and structural diversity to evade the immune response. A few PfCSP-directed antibodies have been identified that are effective at preventing malaria infection, including CIS43, but how these repeat-targeting antibodies might be improved has been unclear. Here, we engineered a humanized mouse model in which B cells expressed inferred human germline CIS43 (iGL-CIS43) B cell receptors and used both vaccination and bioinformatic analysis to obtain variant CIS43 antibodies with improved protective capacity. One such antibody, iGL-CIS43.D3, was significantly more potent than the current best-in-class PfCSP-directed antibody. We found that vaccination with a junctional epitope peptide was more effective than full-length PfCSP at recruiting iGL-CIS43 B cells to germinal centers. Structure-function analysis revealed multiple somatic hypermutations that combinatorically improved protection. This mouse model can thus be used to understand vaccine immunogens and to develop highly potent anti-malarial antibodies.


Assuntos
Subpopulações de Linfócitos B/imunologia , Epitopos/imunologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/imunologia , Vacinas de DNA/imunologia , Transferência Adotiva , Animais , Anticorpos Antiprotozoários/metabolismo , Modelos Animais de Doenças , Epitopos/genética , Engenharia Genética , Humanos , Evasão da Resposta Imune , Imunogenicidade da Vacina , Camundongos , Camundongos SCID , Proteínas de Protozoários/genética , Relação Estrutura-Atividade , Vacinação
8.
Int Immunopharmacol ; 101(Pt A): 108320, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34741871

RESUMO

Plant-derived antimalarials are indispensable for malaria treatment and a platform for new drugs. The present study explores sinigrin, for malaria using in vitro, in silico and in vivo strategies and the immune response generated after administration. The compound exhibited promising activity against chloroquine (CQ)-resistant (RKL-9) IC50 5.14 µg/mL and CQ-sensitive (3D7) IC50 5.47 µg/mL strains of P. falciparum and was safe in both in vitro (CC50 > 640 µg/mL) and in vivo (LD50 > 2 g/kg) toxicity studies. In addition, virtual screening showed hydrogen bonding, hydrophobic and van der Waals interactions with amino acid residues of 3BPM (falcipain-3). In vivo studies revealed promising antimalarial activity of sinigrin (200 mg/kg) with 87.44% chemo-suppression on day 5 and significantly (p < 0.0001) enhanced the mean survival time (21 ± 4.74 days) in contrast to the infected control (5.4 ± 1.14 days). In combination therapy, sinigrin (100 mg/kg and 200 mg/kg) augmented the efficacy of artesunate (AS 50 mg/kg) with 100% survival and no recrudescence. These observations are further corresponded and supported by DLC, NO production, cytokine analysis, biochemical and histopathological studies. Treatment with the combination resulted in a regulated interplay of immune cells and cytokines aiding in parasite clearance in addition to its specific inhibitory activity. We report the antimalarial activity of sinigrin first time with best D-score against falcipain-3. These findings highlight sinigrin as a HIT molecule, which may potentially be used in drug and vaccine development approaches.


Assuntos
Antimaláricos/uso terapêutico , Artesunato/uso terapêutico , Cisteína Endopeptidases/metabolismo , Glucosinolatos/uso terapêutico , Malária/tratamento farmacológico , Animais , Antimaláricos/administração & dosagem , Artesunato/administração & dosagem , Cisteína Endopeptidases/efeitos dos fármacos , Citocinas/metabolismo , Quimioterapia Combinada , Feminino , Glucosinolatos/administração & dosagem , Contagem de Leucócitos , Malária/imunologia , Malária Falciparum/tratamento farmacológico , Malária Falciparum/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Simulação de Acoplamento Molecular , Óxido Nítrico/metabolismo , Plasmodium berghei/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Células RAW 264.7/efeitos dos fármacos , Células RAW 264.7/metabolismo
9.
J Immunol ; 207(11): 2631-2635, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34716185

RESUMO

Radiation-attenuated sporozoite (RAS) vaccination offers hope for global malaria control through induction of protective liver-stage-specific memory CD8 T cells. Effective RAS vaccination regimens exist; however, widespread implementation remains unfeasible. A key difficulty resides in the need to administer three or more doses i.v. to achieve sufficient immunity. Strategies to reduce the number of RAS doses are therefore desirable. Here we used mice to model human immune responses to a single, suboptimal weight-normalized RAS dose administered i.v. followed by subunit vaccination to amplify liver-stage-specific memory CD8 T cells. RAS+subunit prime-boost regimens increased the numbers of liver-stage-specific memory CD8 T cells to a level greater than is present after one RAS vaccination. Both i.v. and i.m. subunit vaccine delivery induced immunity in mice, and many vaccinated mice completely cleared liver infection. These findings are particularly relevant to human vaccine development because RAS+subunit prime-boost vaccination would reduce the logistical challenges of multiple RAS-only immunizations.


Assuntos
Hepatopatias/imunologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Esporozoítos/imunologia , Vacinas Atenuadas/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Animais , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vacinação
10.
J Parasitol ; 107(4): 639-647, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34358312

RESUMO

Some antimalarial drugs are immune-modulators that impact multiple pathways of innate immunity in malarial treatment. However, information on the immunomodulatory effects of artequine and rutin in the treatment of malaria remains elusive. Twenty-five Swiss mice (18 ± 2 g) were used for this study. Twenty were infected with Plasmodium berghei (NK65). Parasitemia was confirmed, and the animals were grouped (n = 5) as follows: Group A was not infected but treated orally with vehicle. Groups B to E were infected and treated (B) orally with vehicle (10 ml/kg), (C) with 10 mg/kg artequine, (D) with 10 mg/kg of artequine supplemented with 100 mg rutin/kg, and (D) with 10 mg/kg of artequine supplemented with 200 mg rutin/kg, for 7 days. Blood was collected for hematological, inflammatory cytokines, and immunoglobulins G and M assays. Post mitochondrial supernatant fraction was used for antioxidant assays. Rutin co-administration (200 mg/kg) significantly (P < 0.001) increased platelet and neutrophil counts (P < 0.01) but significantly (P < 0.01) decreased white blood cell count and lymphocyte relative to parasitized control. Also, it significantly (P < 0.05) decreased lipid peroxidation, xanthine oxidase, and superoxide dismutase activities but significantly (P < 0.05) increased reduced glutathione and glutathione S-transferase activity. Rutin co-administration also caused a significant (P < 0.001) increase in tumor necrosis factor-alpha, interleukin-6, and immunoglobulin M levels, while interleukin-1ß and immunoglobulin G decreased significantly (P < 0.001) compared with parasitized control. These results showed that rutin co-administration with artequine improved host antioxidant status and modulated the immune and inflammatory responses.


Assuntos
Antimaláricos/uso terapêutico , Artesunato/uso terapêutico , Malária/tratamento farmacológico , Plasmodium berghei/efeitos dos fármacos , Rutina/uso terapêutico , Animais , Antioxidantes/análise , Citocinas/efeitos dos fármacos , Quimioterapia Combinada , Imunoglobulinas/efeitos dos fármacos , Contagem de Leucócitos , Leucócitos/efeitos dos fármacos , Malária/imunologia , Masculino , Camundongos
11.
Infect Immun ; 89(11): e0024921, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34424755

RESUMO

Malaria remains a grave concern for humans, as effective medical countermeasures for Plasmodium infection have yet to be developed. Phagocytic clearance of parasitized red blood cells (pRBCs) by macrophages is an important front-line innate host defense against Plasmodium infection. We previously showed that repeated injections of low-dose lipopolysaccharide (LPS) prior to bacterial infection, called LPS preconditioning, strongly augmented phagocytic/bactericidal activity in murine macrophages. However, whether LPS preconditioning prevents murine Plasmodium infection is unclear. We investigated the protective effects of LPS preconditioning against lethal murine Plasmodium infection, focusing on CD11bhigh F4/80low liver macrophages, which are increased by LPS preconditioning. Mice were subjected to LPS preconditioning by intraperitoneal injections of low-dose LPS for 3 consecutive days, and 24 h later, they were intravenously infected with pRBCs of Plasmodium yoelii 17XL. LPS preconditioning markedly increased the murine survival and reduced parasitemia, while it did not reduce tumor necrosis factor (TNF) secretions, only delaying the peak of plasma gamma interferon (IFN-γ) after Plasmodium infection in mice. An in vitro phagocytic clearance assay of pRBCs showed that the CD11bhigh F4/80low liver macrophages, but not spleen macrophages, in the LPS-preconditioned mice had significantly augmented phagocytic activity against pRBCs. The adoptive transfer of CD11bhigh F4/80low liver macrophages from LPS-preconditioned mice to control mice significantly improved survival after Plasmodium infection. We conclude that LPS preconditioning stimulated CD11bhigh F4/80low liver macrophages to augment the phagocytic clearance of pRBCs, which may play a central role in resistance against Plasmodium infection.


Assuntos
Eritrócitos/parasitologia , Lipopolissacarídeos/farmacologia , Fígado/imunologia , Macrófagos/imunologia , Malária/imunologia , Fagocitose/efeitos dos fármacos , Plasmodium yoelii , Transferência Adotiva , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium yoelii/crescimento & desenvolvimento , Fator de Necrose Tumoral alfa/sangue
12.
Cell Commun Signal ; 19(1): 74, 2021 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-34243757

RESUMO

Our murine cancer model studies have demonstrated that Plasmodium infection activates the immune system that has been inhibited by cancer cells, counteracts tumor immunosuppressive microenvironment, inhibits tumor angiogenesis, inhibits tumor growth and metastasis, and prolongs the survival time of tumor-bearing mice. Based on these studies, three clinical trials of Plasmodium immunotherapy for advanced cancers have been approved and are ongoing in China. After comparing the mechanisms of action of Plasmodium immunotherapy with those of immune checkpoint blockade therapy, we propose the notion that cancer is an ecological disease and that Plasmodium immunotherapy is a systemic ecological counterattack therapy for this ecological disease, with limited side effects and without danger to public health based on the use of artesunate and other measures. Recent reports of tolerance to treatment and limitations in majority of patients associated with the use of checkpoint blockers further support this notion. We advocate further studies on the mechanisms of action of Plasmodium infection against cancer and investigations on Plasmodium-based combination therapy in the coming future. Video Abstract.


Assuntos
Imunoterapia , Malária/imunologia , Neoplasias/imunologia , Microambiente Tumoral/imunologia , Animais , Artesunato/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Malária/complicações , Malária/parasitologia , Malária/terapia , Camundongos , Neoplasias/complicações , Neoplasias/parasitologia , Neoplasias/terapia , Microambiente Tumoral/efeitos dos fármacos
13.
Am J Trop Med Hyg ; 105(2): 477-479, 2021 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-34125702

RESUMO

The protective effect of semi-immunity to alleviate clinical complications of malaria remains incompletely understood. This ecological study quantified the proportion of unfavorable clinical outcomes among patient populations with imported malaria as a function of the reported proportion of absent semi-immunity in a patient population. Group-level proportions were extracted from published studies on imported malaria. Linear regression analyses demonstrate a consistent positive trend between the average proportion of absent semi-immunity in patient populations of imported malaria and the proportion of unfavorable clinical outcomes therein. Regression equations provide a group-level estimate of attributable fractions of clinical complications resulting from absent semi-immunity to malaria.


Assuntos
Malária , Plasmodium/imunologia , Antimaláricos/uso terapêutico , Quimioprevenção , Doenças Transmissíveis Importadas/epidemiologia , Doenças Transmissíveis Importadas/imunologia , Humanos , Imunidade , Malária/tratamento farmacológico , Malária/epidemiologia , Malária/imunologia , Adesão à Medicação , Mortalidade , Vigilância da População , Prevalência , Viagem , Resultado do Tratamento
14.
Comput Biol Chem ; 92: 107493, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33962170

RESUMO

Vaccine based strategies offer a promising future in malaria control by generating protective immunity against natural infection. However, vaccine development is hindered by the Plasmodium sp. genetic diversity. Previously, we have shown P41 protein from 6-Cysteine shared by Plasmodium sp. and could be used for cross-species anti-malaria vaccines. Two different approaches, ancestral, and consensus sequence, could produce a single target for all human-infecting Plasmodium. In this study, we investigated the efficacy of ancestral and consensus of P41 protein. Phylogenetic and time tree reconstruction was conducted by RAXML and BEAST2 package to determine the relationship of known P41 sequences. Ancestral and consensus sequences were reconstructed by the GRASP server and Unipro Ugene software, respectively. The structural prediction was made using the Psipred and Rosetta program. The protein characteristic was analyzed by assessing hydrophobicity and Post-Translational Modification sites. Meanwhile, the immunogenicity score for B-cell, T-cell, and MHC was determined using an immunoinformatic approach. The result suggests that ancestral and consensus have a distinct protein characteristic with high immunogenicity scores for all immune cells. We found one shared conserved epitope with phosphorylation modification from the ancestral sequence to target the cross-species vaccine. Thus, this study provides detailed insight into P41 efficacy for the cross-species anti-malaria blood-stage vaccine.


Assuntos
Antígenos de Protozoários/imunologia , Antígeno CD48/imunologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Antígeno CD48/química , Antígeno CD48/genética , Vacinas Antimaláricas/química , Vacinas Antimaláricas/genética
15.
Cell Rep ; 35(2): 108996, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33852850

RESUMO

Antibodies targeting the NANP/NVDP repeat domain of the Plasmodium falciparum circumsporozoite protein (CSPRepeat) can protect against malaria. However, it has also been suggested that the CSPRepeat is a decoy that prevents the immune system from mounting responses against other domains of CSP. Here, we show that, following parasite immunization, B cell responses to the CSPRepeat are immunodominant over responses to other CSP domains despite the presence of similar numbers of naive B cells able to bind these regions. We find that this immunodominance is driven by avid binding of the CSPRepeat to cognate B cells that are able to expand at the expense of B cells with other specificities. We further show that mice immunized with repeat-truncated CSP molecules develop responses to subdominant epitopes and are protected against malaria. These data demonstrate that the CSPRepeat functions as a decoy, but truncated CSP molecules may be an approach for malaria vaccination.


Assuntos
Anticorpos Antiprotozoários/biossíntese , Imunização/métodos , Vacinas Antimaláricas/administração & dosagem , Malária/prevenção & controle , Peptídeos/administração & dosagem , Plasmodium berghei/efeitos dos fármacos , Proteínas de Protozoários/genética , Animais , Anopheles/parasitologia , Anticorpos Neutralizantes/biossíntese , Linfócitos B/imunologia , Linfócitos B/parasitologia , Feminino , Expressão Gênica , Malária/imunologia , Malária/parasitologia , Vacinas Antimaláricas/biossíntese , Vacinas Antimaláricas/genética , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/genética , Peptídeos/imunologia , Plasmodium berghei/imunologia , Plasmodium berghei/patogenicidade , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Ligação Proteica , Proteínas de Protozoários/imunologia , Esporozoítos/imunologia , Esporozoítos/efeitos da radiação , Transgenes , Vacinas Atenuadas
16.
Front Endocrinol (Lausanne) ; 12: 643851, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33841336

RESUMO

Malaria is the leading cause of parasitic infection-related death globally. Additionally, malaria-associated mortality is higher in men than in women, and this sexual dimorphism reflects differences in innate and adaptive immune responses that are influenced by sex hormones. Normally, females develop more robust immune responses against parasites than males. However, most clinical and laboratory studies related to the immune response to malaria do not consider sex as a variable, and relatively few studies have compared the sex-dependent role of 17ß-estradiol in this process. In this study, we decreased in vivo the levels of 17ß-estradiol by gonadectomy or administered 17ß-estradiol to intact or gonadectomized male and female CBA/Ca mice infected with Plasmodium berghei ANKA. Subsequently, we assessed the effects of 17ß-estradiol on parasite load; the percentages of different immune cells in the spleen; the plasma levels of antibodies and pro- and anti-inflammatory cytokines; and the mRNA expression levels of cytokine-encoding genes in the brain. The results showed that the administration of 17ß-estradiol increased parasitemia and decreased body weight in intact female mice. Moreover, intact females exhibited higher levels of CD8+ T cells and lower levels of NK1.1+ cells than their male counterparts under the same condition. Gonadectomy increased IFN-γ and decreased TNF-α concentrations only in intact female mice. Additionally, IL-10 levels were higher in intact females than in their male counterparts. Finally, the mRNA expression levels of cytokines coding genes in the brain showed a dimorphic pattern, i.e., gonadectomy upregulated Tnf, Il1b, and Il10 expression in males but not in females. Our findings explain the sexual dimorphism in the immune response to malaria, at least in part, and suggest potential sex-dependent implications for the efficacy of vaccines or drugs targeting malaria.


Assuntos
Estradiol/metabolismo , Sistema Imunitário/imunologia , Malária/imunologia , Malária/metabolismo , Parasitemia/imunologia , Fatores Sexuais , Animais , Temperatura Corporal , Linfócitos T CD8-Positivos/citologia , Citocinas/metabolismo , Feminino , Hemoglobinas/análise , Humanos , Interferon gama/metabolismo , Interleucina-10/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos CBA , Orquiectomia , Ovariectomia , Parasitemia/parasitologia , Plasmodium berghei , Baço/imunologia , Baço/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
17.
Int Immunol ; 33(8): 409-422, 2021 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-33914894

RESUMO

Upon activation, specific CD4+ T cells up-regulate the expression of CD11a and CD49d, surrogate markers of pathogen-specific CD4+ T cells. However, using T-cell receptor transgenic mice specific for a Plasmodium antigen, termed PbT-II, we found that activated CD4+ T cells develop not only to CD11ahiCD49dhi cells, but also to CD11ahiCD49dlo cells during acute Plasmodium infection. CD49dhi PbT-II cells, localized in the red pulp of spleens, expressed transcription factor T-bet and produced IFN-γ, indicating that they were type 1 helper T (Th1)-type cells. In contrast, CD49dlo PbT-II cells resided in the white pulp/marginal zones and were a heterogeneous population, with approximately half of them expressing CXCR5 and a third expressing Bcl-6, a master regulator of follicular helper T (Tfh) cells. In adoptive transfer experiments, both CD49dhi and CD49dlo PbT-II cells differentiated into CD49dhi Th1-type cells after stimulation with antigen-pulsed dendritic cells, while CD49dhi and CD49dlo phenotypes were generally maintained in mice infected with Plasmodium chabaudi. These results suggest that CD49d is expressed on Th1-type Plasmodium-specific CD4+ T cells, which are localized in the red pulp of the spleen, and can be used as a marker of antigen-specific Th1 CD4+ T cells, rather than that of all pathogen-specific CD4+ T cells.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Integrina alfa4/imunologia , Malária/imunologia , Plasmodium chabaudi/imunologia , Células T Auxiliares Foliculares/imunologia , Células Th1/imunologia , Transferência Adotiva/métodos , Animais , Células Cultivadas , Células Dendríticas/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-bcl-6/imunologia , Baço/imunologia , Linfócitos T Auxiliares-Indutores/imunologia
18.
Mol Med Rep ; 23(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33846776

RESUMO

Postoperative recurrence causes a high mortality rate among patients with hepatocellular carcinoma (HCC). The current study aimed to determine the effects of Plasmodium infection on HCC metastasis and recurrence. The antitumor effects of Plasmodium infection were determined using two murine orthotopic HCC models: The non­resection model and the resection model. Tumour tissues derived from tumour­bearing mice treated with or without Plasmodium infection were harvested 15 days post­tumour inoculation. The expression levels of biomarkers related to epithelial­mesenchymal transition (EMT) and molecules associated with CC­chemokine receptor 10 (CCR10)­mediated PI3K/Akt/GSK­3ß/Snail signalling were identified using reverse transcription­quantitative PCR and western blotting. The results demonstrated that Plasmodium infection significantly suppressed the progression, recurrence and metastasis of HCC in the two mouse models. The expression levels of E­cadherin were significantly higher in the Plasmodium­treated group compared with that in the control group, whereas the expression levels of Vimentin and Snail were significantly lower in the Plasmodium­treated group. Furthermore, Plasmodium infection inhibited the activation of Akt and GSK­3ß in the tumour tissues by downregulating the expression levels of CCR10 and subsequently suppressing the accumulation of Snail, which may contribute to the suppression of EMT and the prevention of tumour recurrence and metastasis. In conclusion, the results of the present study demonstrated that Plasmodium infection inhibited the recurrence and metastasis and improved the prognosis of HCC by suppressing CCR10­mediated PI3K/Akt/GSK­3ß/Snail signalling and preventing the EMT. These results may be important for the development of novel therapies for HCC recurrence and metastasis, especially for patients in the perioperative period.


Assuntos
Carcinoma Hepatocelular/prevenção & controle , Transição Epitelial-Mesenquimal , Neoplasias Hepáticas/prevenção & controle , Malária , Animais , Biomarcadores Tumorais/metabolismo , Caderinas/metabolismo , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Neoplasias Hepáticas/genética , Malária/imunologia , Malária/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Recidiva Local de Neoplasia/genética , Fosfatidilinositol 3-Quinases/metabolismo , Receptores CCR10 , Transdução de Sinais , Vimentina/metabolismo
19.
Nutrients ; 13(3)2021 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-33799736

RESUMO

Plasmodium falciparum is a protozoan parasite which causes malarial disease in humans. Infections commonly occur in sub-Saharan Africa, a region with high rates of inadequate nutrient consumption resulting in malnutrition. The complex relationship between malaria and malnutrition and their effects on gut immunity and physiology are poorly understood. Here, we investigated the effect of malaria infection in the guts of moderately malnourished mice. We utilized a well-established low protein diet that is deficient in zinc and iron to induce moderate malnutrition and investigated mucosal tissue phenotype, permeability, and innate immune response in the gut. We observed that the infected moderately malnourished mice had lower parasite burden at the peak of infection, but damaged mucosal epithelial cells and high levels of FITC-Dextran concentration in the blood serum, indicating increased intestinal permeability. The small intestine in the moderately malnourished mice were also shorter after infection with malaria. This was accompanied with lower numbers of CD11b+ macrophages, CD11b+CD11c+ myeloid cells, and CD11c+ dendritic cells in large intestine. Despite the lower number of innate immune cells, macrophages in the moderately malnourished mice were highly activated as determined by MHCII expression and increased IFNγ production in the small intestine. Thus, our data suggest that malaria infection may exacerbate some of the abnormalities in the gut induced by moderate malnutrition.


Assuntos
Imunidade Inata , Imunidade nas Mucosas , Mucosa Intestinal/patologia , Malária/complicações , Desnutrição/complicações , Plasmodium chabaudi , Animais , Citocinas/biossíntese , Mucosa Intestinal/imunologia , Intestino Grosso/imunologia , Intestino Grosso/patologia , Intestino Delgado/imunologia , Intestino Delgado/patologia , Macrófagos/imunologia , Malária/imunologia , Malária/patologia , Masculino , Desnutrição/imunologia , Desnutrição/patologia , Camundongos , Camundongos Endogâmicos C57BL
20.
Trop Biomed ; 38(1): 187-204, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33797545

RESUMO

Malaria infection still remains as one of the most prominent parasitic diseases afflicting mankind in tropical and subtropical regions. The severity of malaria infection has often been associated to exuberant host immune inflammatory responses that could possibly lead to severe immunopathological conditions and subsequent death of host tissues. Activin A is a protein belonging to the transforming growth factor-beta (TGF-ß) family that regulates multiple physiological processes and pathological-associated diseases. The biological roles of activin A have been associated with manipulation of inflammation-related processes and modulation of host immune responses. This implies that activin A protein could play a role in malaria pathogenesis since malaria infection has been closely linked to severe immune responses leading to death, However, the actual in vivo role of activin A in malaria infection remains elusive. Hence, this study was undertaken to investigate the involvement of activin A in malaria infection as well as to assess the modulating effects of activin A on the cytokine releases (TNF-α, IFN-γ and IL-10) and histopathological changes in major affected organs (kidney, liver, lung, brain and spleen) in malarial mice infected with Plasmodium berghei ANKA. Our results showed that the concentrations of plasma activin A were significantly increased in malarial mice throughout the study periods. Also. the systemic activin A level was positively correlated with malaria parasitemia. This indicates that activin A could play a role in malaria pathogenesis and malaria parasitemia development. Plasma TNF-α, IFN-γ and IL-10 cytokine levels were significantly increased in malarial mice at day-5 post infection, suggesting that these cytokines attributed to severe malaria pathogenesis. Histopathological features such as sequestration of parasitized red blood cells (pRBCs) and hemozoin formation were amongst the most common pathological conditions observed in tissues of major affected organs (kidney, liver, lung, brain and spleen) in malarial mice. Neutralization of activin A production via recombinant mouse activin RIIA Fc chimera (rmActivin RIIA Fc chimera) had significantly reduced the parasitemia levels in malarial mice. The release of TNF-α cytokine was significantly reduced as well as the sequestration of parasitized pRBCs and hemozoin formation in major affected organs in malarial mice were also alleviated following inhibition of activin A production. Overall, this preliminary study suggests that activin A could play an immune modulation role in malaria pathogenesis through modulation of TNF-α release that benefits host from severe pathological destructions provoked by intensified inflammatory responses. Further studies are warranted to elucidate the precise mechanism of immune modulation mediated by activin A and its associated immune-modulation mediators in regulating the inflammatory responses elicited during the course of malaria infection.


Assuntos
Ativinas/antagonistas & inibidores , Malária/tratamento farmacológico , Fator de Necrose Tumoral alfa/sangue , Ativinas/imunologia , Animais , Citocinas/sangue , Interferon gama/sangue , Interleucina-10/sangue , Malária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Plasmodium berghei
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA