Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 469
Filter
1.
J Neuroinflammation ; 21(1): 119, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715061

ABSTRACT

BACKGROUND: Cerebral malaria (CM) is the most lethal complication of malaria, and survivors usually endure neurological sequelae. Notably, the cytotoxic effect of infiltrating Plasmodium-activated CD8+ T cells on cerebral microvasculature endothelial cells is a prominent feature of the experimental CM (ECM) model with blood-brain barrier disruption. However, the damage effect of CD8+ T cells infiltrating the brain parenchyma on neurons remains unclear. Based on the immunosuppressive effect of the PD-1/PD-L1 pathway on T cells, our previous study demonstrated that the systemic upregulation of PD-L1 to inhibit CD8+ T cell function could effectively alleviate the symptoms of ECM mice. However, it has not been reported whether neurons can suppress the pathogenic effect of CD8+ T cells through the PD-1/PD-L1 negative immunomodulatory pathway. As the important inflammatory factor of CM, interferons can induce the expression of PD-L1 via different molecular mechanisms according to the neuro-immune microenvironment. Therefore, this study aimed to investigate the direct interaction between CD8+ T cells and neurons, as well as the mechanism of neurons to alleviate the pathogenic effect of CD8+ T cells through up-regulating PD-L1 induced by IFNs. METHODS: Using the ECM model of C57BL/6J mice infected with Plasmodium berghei ANKA (PbA), morphological observations were conducted in vivo by electron microscope and IF staining. The interaction between the ECM CD8+ T cells (immune magnetic bead sorting from spleen of ECM mice) and primary cultured cortical neurons in vitro was observed by IF staining and time-lapse photography. RNA-seq was performed to analyze the signaling pathway of PD-L1 upregulation in neurons induced by IFNß or IFNγ, and verified through q-PCR, WB, IF staining, and flow cytometry both in vitro and in vivo using IFNAR or IFNGR gene knockout mice. The protective effect of adenovirus-mediated PD-L1 IgGFc fusion protein expression was verified in ECM mice with brain stereotaxic injection in vivo and in primary cultured neurons via viral infection in vitro. RESULTS: In vivo, ECM mice showed infiltration of activated CD8+ T cells and neuronal injury in the brain parenchyma. In vitro, ECM CD8+ T cells were in direct contact with neurons and induced axonal damage, as an active behavior. The PD-L1 protein level was elevated in neurons of ECM mice and in primary cultured neurons induced by IFNß, IFNγ, or ECM CD8+ T cells in vitro. Furthermore, the IFNß or IFNγ induced neuronal expression of PD-L1 was mediated by increasing STAT1/IRF1 pathway via IFN receptors. The increase of PD-L1 expression in neurons during PbA infection was weakened after deleting the IFNAR or IFNGR. Increased PD-L1 expression by adenovirus partially protected neurons from CD8+ T cell-mediated damage both in vitro and in vivo. CONCLUSION: Our study demonstrates that both type I and type II IFNs can induce neurons to upregulate PD-L1 via the STAT1/IRF1 pathway mediated by IFN receptors to protect against activated CD8+ T cell-mediated damage, providing a targeted pathway to alleviate neuroinflammation during ECM.


Subject(s)
B7-H1 Antigen , CD8-Positive T-Lymphocytes , Malaria, Cerebral , Mice, Inbred C57BL , Neurons , STAT1 Transcription Factor , Up-Regulation , Animals , Mice , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/metabolism , Malaria, Cerebral/immunology , Malaria, Cerebral/metabolism , Malaria, Cerebral/pathology , Mice, Knockout , Neurons/metabolism , Plasmodium berghei , Signal Transduction/physiology , STAT1 Transcription Factor/metabolism , Up-Regulation/drug effects
2.
Int Immunopharmacol ; 132: 111982, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38569430

ABSTRACT

RTS,S is the first malaria vaccine recommended for implementation among young children at risk. However, vaccine efficacy is modest and short-lived. To mitigate the risk of cerebral malaria (CM) among children under the age of 5, it is imperative to develop new vaccines. EVs are potential vaccine candidates as they obtain the ability of brain-targeted delivery and transfer plasmodium antigens and immunomodulators during infections. This study extracted EVs from BALB/c mice infected with Plasmodium yoelii 17XNL (P.y17XNL). C57BL/6J mice were intravenously immunized with EVs (EV-I.V. + CM group) or subcutaneously vaccinated with the combination of EVs and CpG ODN-1826 (EV + CPG ODN-S.C. + CM group) on days 0 and 20, followed by infection with Plasmodium berghei ANKA (P.bANKA) on day 20 post-second immunization. We monitored Parasitemia and survival rate. The integrity of the Blood-brain barrier (BBB) was examined using Evans blue staining.The levels of cytokines and adhesion molecules were evaluated using Luminex, RT-qPCR, and WB. Brain pathology was evaluated by hematoxylin and eosin and immunohistochemical staining. The serum levels of IgG, IgG1, and IgG2a were analyzed by enzyme-linked immunosorbent assay. Compared with those in the P.bANKA-infected group, parasitemia increased slowly, death was delayed (day 10 post-infection), and the survival rate reached 75 %-83.3 % in the EV-I.V. + ECM and EV + CPG ODN-S.C. + ECM groups. Meanwhile, compared with the EV + CPG ODN-S.C. + ECM group, although parasitemia was almost the same, the survival rate increased in the EV-I.V. + ECM group.Additionally, EVs immunization markedly downregulated inflammatory responses in the spleen and brain and ameliorated brain pathological changes, including BBB disruption and infected red blood cell (iRBC) sequestration. Furthermore, the EVs immunization group exhibited enhanced antibody responses (upregulation of IgG1 and IgG2a production) compared to the normal control group. EV immunization exerted protective effects, improving the integrity of the BBB, downregulating inflammation response of brain tissue, result in reduces the incidence of CM. The protective effects were determined by immunological pathways and brain targets elicited by EVs. Intravenous immunization exhibited better performance than subcutaneous immunization, which perhaps correlated with EVs, which can naturally cross BBB to play a better role in brain protection.


Subject(s)
Blood-Brain Barrier , Erythrocytes , Extracellular Vesicles , Malaria, Cerebral , Mice, Inbred BALB C , Mice, Inbred C57BL , Oligodeoxyribonucleotides , Plasmodium berghei , Animals , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Malaria, Cerebral/prevention & control , Plasmodium berghei/immunology , Extracellular Vesicles/immunology , Erythrocytes/parasitology , Erythrocytes/immunology , Blood-Brain Barrier/immunology , Mice , Oligodeoxyribonucleotides/administration & dosage , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Female , Brain/parasitology , Brain/immunology , Brain/pathology , Cytokines/metabolism , Cytokines/blood , Plasmodium yoelii/immunology , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Parasitemia/immunology , Disease Models, Animal , Immunoglobulin G/blood , Immunoglobulin G/immunology
3.
Mol Immunol ; 152: 240-254, 2022 12.
Article in English | MEDLINE | ID: mdl-36395532

ABSTRACT

Th9, a new subgroup of CD4+T cells is characterized by its specific cytokine IL-9, is a critical factor in allergic diseases, cancers and parasitic infections. This study aimed to explore the potential roles of Th9 cells in the immunopathogenesis of ECM. In splenocytes sourced from uninfected, PbA and Py infected mice, Th9 cells were characterised by flow cytometry, cell sorting and qPCR. Enhancement of CD4+IL-9+ (Th9) cells were observed in both the infections, which corroborated with increased expression of the differentiating transcription factors. Moreover, crucial cytokine receptors (IL-4R, TGF-ßR, IL-6R) as well as chemokine receptors (CCR3, CCR6 and CCR7) and activation marker (CD96), demonstrated elevation upon PbA infection in splenic Th9 cells. Furthermore, Neutralization of IL-9 along with IL-6 enhanced host survivability, reduced mean neurological score of ECM. However, anti- IL-9 treatment also down regulated frequency of Th17 cells, and its transcription factors pSTAT3, RORγT along with depleted Il-1ß and Il-6 expression. In sum, understanding how IL-9 producing CD4+ T-cells can alter Th17/Treg ratio and by that modulate host's immune response, could pave the way for developing immunomodulatory interventions against cerebral malaria.


Subject(s)
Interleukin-9 , Malaria, Cerebral , Th17 Cells , Animals , Mice , Interleukin-6/immunology , Interleukin-9/immunology , Malaria, Cerebral/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Transcription Factors/immunology
4.
Proc Natl Acad Sci U S A ; 119(36): e2206327119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037380

ABSTRACT

Cerebral malaria (CM) is a life-threatening form of Plasmodium falciparum infection caused by brain inflammation. Brain endothelium dysfunction is a hallmark of CM pathology, which is also associated with the activation of the type I interferon (IFN) inflammatory pathway. The molecular triggers and sensors eliciting brain type I IFN cellular responses during CM remain largely unknown. We herein identified the stimulator of interferon response cGAMP interactor 1 (STING1) as the key innate immune sensor that induces Ifnß1 transcription in the brain of mice infected with Plasmodium berghei ANKA (Pba). This STING1/IFNß-mediated response increases brain CXCL10 governing the extent of brain leukocyte infiltration and blood-brain barrier (BBB) breakdown, and determining CM lethality. The critical role of brain endothelial cells (BECs) in fueling type I IFN-driven brain inflammation was demonstrated in brain endothelial-specific IFNß-reporter and STING1-deficient Pba-infected mice, which were significantly protected from CM lethality. Moreover, extracellular particles (EPs) released from Pba-infected erythrocytes activated the STING1-dependent type I IFN response in BECs, a response requiring intracellular acidification. Fractionation of the EPs enabled us to identify a defined fraction carrying hemoglobin degradation remnants that activates STING1/IFNß in the brain endothelium, a process correlated with heme content. Notably, stimulation of STING1-deficient BECs with heme, docking experiments, and in vitro binding assays unveiled that heme is a putative STING1 ligand. This work shows that heme resultant from the parasite heterotrophic activity operates as an alarmin, triggering brain endothelial inflammatory responses via the STING1/IFNß/CXCL10 axis crucial to CM pathogenesis and lethality.


Subject(s)
Brain , Heme , Interferon-beta , Malaria, Cerebral , Membrane Proteins , Animals , Brain/parasitology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/parasitology , Endothelium/immunology , Endothelium/parasitology , Heme/metabolism , Interferon-beta/immunology , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Plasmodium berghei/metabolism , Transcriptional Activation/immunology
5.
Cytokine ; 155: 155910, 2022 07.
Article in English | MEDLINE | ID: mdl-35594680

ABSTRACT

Myeloid derived suppressor cells (MDSCs) are a group of heterogeneous cell populations that can suppress T cell responses. Various aspects of MDSCs in regulating immune responses in several cancer and infectious diseases have been reported till date. But the role and regulation of MDSCs have not been systematically studied in the context of malaria. This study depicts the phenotypic and functional characteristics of splenic MDSCs and how they regulate Th-17 mediated immune response during Experimental Cerebral Malaria (ECM). Flow cytometric analysis reveals that MDSCs in the spleen and bone marrow expand at 8 dpi during ECM. Among subtypes of MDSCs, PMN-MDSCs show significant expansion in the spleen but M-MDSCs remain unaltered. Functional analysis of sorted MDSCs from spleens of Plasmodium berghei ANKA (PbA) infected mice shows suppressive nature of these cells and high production of Nitric oxide (NO). Besides, MDSCs were also found to express various inflammatory markers during ECM suggesting the M1 type phenotype of these cells. In-vivo depletion of MDSCs by the use of Anti Gr-1 increases mice survival but doesn't significantly alter the parasitemia. Previously, it has been reported that Treg/Th-17 balance in the spleen is skewed towards Th-17 during ECM. Depletion of MDSCs was found to regulate Th-17 percentages to homeostatic levels and subvert various inflammatory changes in the spleen. Among different factors, IL-6 was found to play an important role in the expansion of MDSCs and expression of inflammatory markers on MDSCs in a STAT3-dependent manner. These findings provide a unique insight into the role of IL-6 in the expansion of the MDSC population which causes inflammatory changes and increased Th-17 responses during ECM.


Subject(s)
Interleukin-6 , Malaria, Cerebral , Myeloid-Derived Suppressor Cells , Th17 Cells , Animals , Interleukin-6/immunology , Malaria, Cerebral/immunology , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , Spleen , Th17 Cells/immunology
6.
CNS Neurosci Ther ; 28(1): 46-63, 2022 01.
Article in English | MEDLINE | ID: mdl-34766463

ABSTRACT

INTRODUCTION: The experimental cerebral malaria (ECM) model in C57BL/6 mice infected with Plasmodium berghei ANKA (PbA) has revealed microglia are involved in the ECM immune microenvironment. However, the regulation of microglia in the ECM immune response is not clear, and there is no safe and efficient treatment clinically for the protection of the nerve cells. AIMS: To elucidate the negative regulation mechanism in the ECM brain mediated by microglia. Furthermore, to investigate protective effect of the appropriate enhancement of the PD-1/PD-L1 pathway in the brain against ECM through the intrathecal injection of the adenovirus expressing PDL1-IgG1Fc fusion protein. RESULTS: The PD-1/PD-L1 pathway was induced in the ECM brain and showed an upregulation in the microglia. Deep single-cell analysis of immune niches in the ECM brainstem indicated that the microglia showed obvious heterogeneity and activation characteristics. Intrathecal injection of recombinant adenovirus expressing PD-L1 repressed the neuroinflammation and alleviated ECM symptoms. In addition, the synergistic effect of artemisinin and intracranial immunosuppression mediated by PD-L1 was more efficacious than either treatment alone. CONCLUSION: The appropriate enhancement of the PD-1/PD-L1 pathway in the early stage of ECM has an obvious protective effect on the maintenance of immune microenvironment homeostasis in the brain. Regulating microglia and the PD-1/PD-L1 pathway could be considered as a promising approach for protection against human cerebral malaria in the future.


Subject(s)
Inflammation , Malaria, Cerebral/immunology , Microglia/immunology , Plasmodium berghei/immunology , Programmed Cell Death 1 Receptor , Signal Transduction , Animals , B7-H1 Antigen , Brain/immunology , Brain/metabolism , Disease Models, Animal , Injections, Spinal , Mice , Mice, Inbred C57BL , Neuroinflammatory Diseases/immunology
7.
Front Immunol ; 12: 711876, 2021.
Article in English | MEDLINE | ID: mdl-34659202

ABSTRACT

Cerebral malaria is a potentially lethal disease, which is caused by excessive inflammatory responses to Plasmodium parasites. Here we use a newly developed transgenic Plasmodium berghei ANKA (PbAAma1OVA) parasite that can be used to study parasite-specific T cell responses. Our present study demonstrates that Ifnar1-/- mice, which lack type I interferon receptor-dependent signaling, are protected from experimental cerebral malaria (ECM) when infected with this novel parasite. Although CD8+ T cell responses generated in the spleen are essential for the development of ECM, we measured comparable parasite-specific cytotoxic T cell responses in ECM-protected Ifnar1-/- mice and wild type mice suffering from ECM. Importantly, CD8+ T cells were increased in the spleens of ECM-protected Ifnar1-/- mice and the blood-brain-barrier remained intact. This was associated with elevated splenic levels of CCL5, a T cell and eosinophil chemotactic chemokine, which was mainly produced by eosinophils, and an increase in eosinophil numbers. Depletion of eosinophils enhanced CD8+ T cell infiltration into the brain and increased ECM induction in PbAAma1OVA-infected Ifnar1-/- mice. However, eosinophil-depletion did not reduce the CD8+ T cell population in the spleen or reduce splenic CCL5 concentrations. Our study demonstrates that eosinophils impact CD8+ T cell migration and proliferation during PbAAma1OVA-infection in Ifnar1-/- mice and thereby are contributing to the protection from ECM.


Subject(s)
Brain/immunology , Eosinophils/physiology , Malaria, Cerebral/immunology , Parasitemia/immunology , Plasmodium berghei , T-Lymphocytes/immunology , Animals , Animals, Outbred Strains , Anopheles/parasitology , Antigens, Protozoan/immunology , Cell Movement , Chemokine CCL5/analysis , Chemokine CCL5/physiology , Cytotoxicity, Immunologic , Female , Leukocyte Count , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mosquito Vectors/parasitology , Organisms, Genetically Modified , Ovalbumin , Parasitemia/parasitology , Peptide Fragments , Plasmodium berghei/genetics , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptors, CCR5/physiology , Spleen/chemistry , Spleen/immunology
8.
J Immunol ; 207(6): 1578-1590, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34400523

ABSTRACT

In the Plasmodium berghei ANKA mouse model of malaria, accumulation of CD8+ T cells and infected RBCs in the brain promotes the development of experimental cerebral malaria (ECM). In this study, we used malaria-specific transgenic CD4+ and CD8+ T cells to track evolution of T cell immunity during the acute and memory phases of P. berghei ANKA infection. Using a combination of techniques, including intravital multiphoton and confocal microscopy and flow cytometric analysis, we showed that, shortly before onset of ECM, both CD4+ and CD8+ T cell populations exit the spleen and begin infiltrating the brain blood vessels. Although dominated by CD8+ T cells, a proportion of both T cell subsets enter the brain parenchyma, where they are largely associated with blood vessels. Intravital imaging shows these cells moving freely within the brain parenchyma. Near the onset of ECM, leakage of RBCs into areas of the brain can be seen, implicating severe damage. If mice are cured before ECM onset, brain infiltration by T cells still occurs, but ECM is prevented, allowing development of long-term resident memory T cell populations within the brain. This study shows that infiltration of malaria-specific T cells into the brain parenchyma is associated with cerebral immunopathology and the formation of brain-resident memory T cells. The consequences of these resident memory populations is unclear but raises concerns about pathology upon secondary infection.


Subject(s)
Blood-Brain Barrier/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Adoptive Transfer/methods , Animals , Disease Models, Animal , Female , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parasitemia/immunology , Spleen/immunology
9.
Immunology ; 164(4): 737-753, 2021 12.
Article in English | MEDLINE | ID: mdl-34407221

ABSTRACT

Experimental cerebral malaria (ECM) is a severe complication of Plasmodium berghei ANKA (PbA) infection in mice, characterized by CD8+ T-cell accumulation within the brain. Whilst the dynamics of CD8+ T-cell activation and migration during extant primary PbA infection have been extensively researched, the fate of the parasite-specific CD8+ T cells upon resolution of ECM is not understood. In this study, we show that memory OT-I cells persist systemically within the spleen, lung and brain following recovery from ECM after primary PbA-OVA infection. Whereas memory OT-I cells within the spleen and lung exhibited canonical central memory (Tcm) and effector memory (Tem) phenotypes, respectively, memory OT-I cells within the brain post-PbA-OVA infection displayed an enriched CD69+ CD103- profile and expressed low levels of T-bet. OT-I cells within the brain were excluded from short-term intravascular antibody labelling but were targeted effectively by longer-term systemically administered antibodies. Thus, the memory OT-I cells were extravascular within the brain post-ECM but were potentially not resident memory cells. Importantly, whilst memory OT-I cells exhibited strong reactivation during secondary PbA-OVA infection, preventing activation of new primary effector T cells, they had dampened reactivation during a fourth PbA-OVA infection. Overall, our results demonstrate that memory CD8+ T cells are systemically distributed but exhibit a unique phenotype within the brain post-ECM, and that their reactivation characteristics are shaped by infection history. Our results raise important questions regarding the role of distinct memory CD8+ T-cell populations within the brain and other tissues during repeat Plasmodium infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Host-Parasite Interactions/immunology , Malaria/immunology , Malaria/parasitology , Plasmodium berghei/physiology , Animals , Biomarkers , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Chemotaxis, Leukocyte/immunology , Disease Susceptibility , Epitopes, T-Lymphocyte/immunology , Erythrocytes/immunology , Erythrocytes/parasitology , Extracellular Matrix , Immunologic Memory , Immunophenotyping , Life Cycle Stages , Lymphocyte Activation/immunology , Malaria/metabolism , Malaria/pathology , Malaria, Cerebral/immunology , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Mice , Mice, Transgenic , Organ Specificity/immunology
10.
Int Immunol ; 33(11): 587-594, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34455438

ABSTRACT

Cerebral malaria (CM) is a life-threatening complication of the malaria disease caused by Plasmodium falciparum infection and is responsible for the death of half a million people annually. The molecular pathogenesis underlying CM in humans is not completely understood, although sequestration of infected erythrocytes in cerebral microvessels is thought to play a major role. In contrast, experimental cerebral malaria (ECM) models in mice have been thought to be distinct from human CM, and are mainly caused by inflammatory mediators. Here, to understand the spatial distribution and the potential sequestration of parasites in the whole-brain microvessels during a mouse model of ECM, we utilized the new tissue-clearing method CUBIC (Clear, Unobstructed, Brain/Body Imaging Cocktails and Computational analysis) with light-sheet fluorescent microscopy (LSFM), and reconstructed images in three dimensions (3D). We demonstrated significantly greater accumulation of Plasmodium berghei ANKA (PbANKA) parasites in the olfactory bulb (OB) of mice, compared with the other parts of the brain, including the cerebral cortex, cerebellum and brainstem. Furthermore, we show that PbANKA parasites preferentially accumulate in the brainstem when the OB is surgically removed. This study therefore not only highlights a successful application of CUBIC tissue-clearing technology to visualize the whole brain and its microvessels during ECM, but it also shows CUBIC's future potential for visualizing pathological events in the whole ECM brain at the cellular level, an achievement that would greatly advance our understanding of human cerebral malaria.


Subject(s)
Brain/pathology , Malaria, Cerebral/pathology , Animals , Brain/immunology , Brain/parasitology , Disease Models, Animal , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Plasmodium berghei/immunology
11.
Front Immunol ; 12: 642585, 2021.
Article in English | MEDLINE | ID: mdl-34025654

ABSTRACT

Reticulon and the REEP family of proteins stabilize the high curvature of endoplasmic reticulum tubules. The REEP5 homolog in Plasmodium, Plasmodium berghei YOP1 (PbYOP1), plays an important role in the erythrocytic cycle of the P. berghei ANKA and the pathogenesis of experimental cerebral malaria (ECM), but the mechanisms are largely unknown. Here, we show that protection from ECM in Pbyop1Δ-infected mice is associated with reduced intracerebral Th1 accumulation, decreased expression of pro-inflammatory cytokines and chemokines, and attenuated pathologies in the brainstem, though the total number of CD4+ and CD8+ T cells sequestered in the brain are not reduced. Expression of adhesive molecules on brain endothelial cells, including ICAM-1, VCAM-1, and CD36, are decreased, particularly in the brainstem, where fatal pathology is always induced during ECM. Subsequently, CD8+ T cell-mediated cell apoptosis in the brain is compromised. These findings suggest that Pbyop1Δ parasites can be a useful tool for mechanistic investigation of cerebral malaria pathogenesis.


Subject(s)
Malaria, Cerebral/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Plasmodium berghei/immunology , Plasmodium berghei/metabolism , Protozoan Proteins/immunology , Protozoan Proteins/metabolism
12.
Int Immunopharmacol ; 97: 107674, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34044183

ABSTRACT

Cerebral malaria (CM) is a neurological complication derived from the Plasmodium falciparum infection in humans. The mechanisms involved in the disease progression are still not fully understood, but both the sequestration of infected red blood cells (iRBC) and leukocytes and an exacerbated host inflammatory immune response are significant factors. In this study, we investigated the effect of Monocyte Locomotion Inhibitory Factor (MLIF), an anti-inflammatory peptide, in a well-characterized murine model of CM. Our data showed that the administration of MLIF increased the survival and avoided the neurological signs of CM in Plasmodium berghei ANKA (PbA) infected C57BL/6 mice. MLIF administration down-regulated systemic inflammatory mediators such as IFN-γ, TNF-α, IL-6, CXCL2, and CCL2, as well as the in situ expression of TNF-α in the brain. In the same way, MLIF reduced the expression of CD31, CD36, CD54, and CD106 in the cerebral endothelium of infected animals and prevented the sequestration of iRBC and leucocytes in the brain microvasculature. Furthermore, MLIF inhibited the activation of astrocytes and microglia and preserved the integrity of the blood-brain barrier (BBB). In conclusion, our results demonstrated that the administration of MLIF increased survival and conferred neuroprotection by decreasing neuroinflammation in murine CM.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Malaria, Cerebral/prevention & control , Neuroprotective Agents/administration & dosage , Oligopeptides/administration & dosage , Animals , Astrocytes/drug effects , Astrocytes/immunology , Brain/drug effects , Brain/immunology , Brain/pathology , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/immunology , Female , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mice , Microglia/drug effects , Microglia/immunology , Plasmodium berghei/immunology
13.
J Biol Chem ; 296: 100615, 2021.
Article in English | MEDLINE | ID: mdl-33798555

ABSTRACT

Persistent high levels of proinflammatory and Th1 responses contribute to cerebral malaria (CM). Suppression of inflammatory responses and promotion of Th2 responses prevent pathogenesis. IL-4 commonly promotes Th2 responses and inhibits inflammatory and Th1 responses. Therefore, IL-4 is widely considered as a beneficial cytokine via its Th2-promoting role that is predicted to provide protection against severe malaria by inhibiting inflammatory responses. However, IL-4 may also induce inflammatory responses, as the result of IL-4 action depends on the timing and levels of its production and the tissue environment in which it is produced. Recently, we showed that dendritic cells (DCs) produce IL-4 early during malaria infection in response to a parasite protein and that this IL-4 response may contribute to severe malaria. However, the mechanism by which IL-4 produced by DCs contributing to lethal malaria is unknown. Using Plasmodium berghei ANKA-infected C57BL/6 mice, a CM model, we show here that mice lacking IL-4Rα only in CD8α+ DCs are protected against CM pathogenesis and survive, whereas WT mice develop CM and die. Compared with WT mice, mice lacking IL-4Rα in CD11c+ or CD8α+ DCs showed reduced inflammatory responses leading to decreased Th1 and cytotoxic CD8+ T cell responses, lower infiltration of CD8+ T cells to the brain, and negligible brain pathology. The novel results presented here reveal a paradoxical role of IL-4Rα signaling in CM pathogenesis that promotes CD8α+ DC-mediated inflammatory responses that generate damaging Th1 and cytotoxic CD8+ T cell responses.


Subject(s)
CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Receptors, Cell Surface/immunology , Signal Transduction/immunology , Th1 Cells/immunology , Animals , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Interleukin-4/genetics , Interleukin-4/immunology , Malaria, Cerebral/genetics , Malaria, Cerebral/pathology , Mice , Mice, Knockout , Plasmodium berghei/genetics , Receptors, Cell Surface/genetics , Signal Transduction/genetics , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology
14.
Sci Rep ; 11(1): 3680, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33574457

ABSTRACT

The Plasmodium falciparum erythrocyte-membrane-protein-1 (PF3D7_1150400/PF11_0521) contains both domain cassette DC13 and DBLß3 domain binding to EPCR and ICAM-1 receptors, respectively. This type of PfEMP1 proteins with dual binding specificity mediate specific interactions with brain micro-vessels endothelium leading to the development of cerebral malaria (CM). Using plasma collected from children at time of hospital admission and after 30 days, we study an acquisition of IgG response to PF3D7_1150400/PF11_0521 DC13 and DBLß3_D4 recombinant constructs, and five peptides located within these constructs, specifically in DBLα1.7_D2 and DBLß3_D4 domains. We found significant IgG responses against the entire DC13, PF11_0521_DBLß3_D4 domain, and peptides. The responses varied against different peptides and depended on the clinical status of children. The response was stronger at day 30, and mostly did not differ between CM and uncomplicated malaria (UM) groups. Specifically, the DBLß3 B3-34 peptide that contains essential residues involved in the interaction between PF11_0521 DBLß3_D4 domain and ICAM-1 receptor demonstrated significant increase in reactivity to IgG1 and IgG3 antibodies at convalescence. Further, IgG reactivity in CM group at time of admission against functionally active (ICAM-1-binding) PF11_0521 DBLß3_D4 domain was associated with protection against severe anemia. These results support development of vaccine based on the PF3D7_1150400/PF11_0521 structures to prevent CM.


Subject(s)
Immunoglobulin G/blood , Malaria, Cerebral/immunology , Malaria, Falciparum/immunology , Peptides/immunology , Protozoan Proteins/immunology , Anemia/complications , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antigens, Protozoan/blood , Antigens, Protozoan/immunology , Brain/immunology , Brain/metabolism , Brain/parasitology , Brain/pathology , Child, Preschool , Endothelial Protein C Receptor/genetics , Endothelial Protein C Receptor/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/parasitology , Erythrocytes/parasitology , Female , Humans , Immunoglobulin G/immunology , Infant , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Malaria, Cerebral/blood , Malaria, Cerebral/genetics , Malaria, Cerebral/parasitology , Malaria, Falciparum/blood , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Male , Peptides/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Protein Binding/genetics , Protein Binding/immunology , Protozoan Proteins/genetics
15.
J Immunol ; 206(1): 118-131, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33239419

ABSTRACT

Cytokine responses to malaria play important roles in both protective immunity development and pathogenesis. Although the roles of cytokines such as TNF-α, IL-12, IFN-γ, and IL-10 in immunity and pathogenesis to the blood stage malaria are largely known, the role of IL-4 remains less understood. IL-4 targets many cell types and induces multiple effects, including cell proliferation, gene expression, protection from apoptosis, and immune regulation. Accordingly, IL-4 has been exploited as a therapeutic for several inflammatory diseases. Malaria caused by Plasmodium falciparum manifests in many organ-specific fatal pathologies, including cerebral malaria (CM), driven by a high parasite load, leading to parasite sequestration in organs and consequent excessive inflammatory responses and endothelial damage. We investigated the therapeutic potential of IL-4 against fatal malaria in Plasmodium berghei ANKA-infected C57BL/6J mice, an experimental CM model. IL-4 treatment significantly reduced parasitemia, CM pathology, and mortality. The therapeutic effect of IL-4 is mediated through multiple mechanisms, including enhanced parasite clearance mediated by upregulation of phagocytic receptors and increased IgM production, and decreased brain inflammatory responses, including reduced chemokine (CXCL10) production, reduced chemokine receptor (CXCR3) and adhesion molecule (LFA-1) expression by T cells, and downregulation of cytotoxic T cell lytic potential. IL-4 treatment markedly reduced the infiltration of CD8+ T cells and brain pathology. STAT6, PI3K-Akt-NF-κB, and Src signaling mediated the cellular and molecular events that contributed to the IL-4-dependent decrease in parasitemia. Overall, our results provide mechanistic insights into how IL-4 treatment mitigates experimental CM and have implications in developing treatment strategies for organ-specific fatal malaria.


Subject(s)
Antimalarials/therapeutic use , Brain/pathology , Interleukin-4/therapeutic use , Malaria, Cerebral/immunology , Plasmodium falciparum/physiology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cytotoxicity, Immunologic , Humans , Inflammation , Malaria, Cerebral/therapy , Mice , Mice, Inbred C57BL , Models, Animal , Parasitemia , Signal Transduction
16.
Med Sci (Paris) ; 36(8-9): 817-821, 2020.
Article in French | MEDLINE | ID: mdl-32821058

ABSTRACT

TITLE: Brèves. ABSTRACT: L'unité d'enseignement « Immunopathologie ¼ qui propose les brèves de ce numéro est suivie par des étudiants des sept parcours recherche du Master Biologie Santé de l'Université de Montpellier. On y étudie les bases physiopathologiques des maladies immunologiques, les cibles thérapeutiques et les mécanismes d'échappement des microorganismes et des tumeurs. Ce Master rassemble des étudiants issus du domaine des sciences et technologies et de celui de la santé. Les articles présentés ont été choisis par les étudiants selon leur domaine de prédilection.


Subject(s)
Allergy and Immunology/trends , Plasmodium falciparum/immunology , Animals , Antibodies, Monoclonal/therapeutic use , B-Lymphocytes/physiology , Dysbiosis/etiology , Dysbiosis/metabolism , Fatty Acids, Volatile/physiology , Genes, cdc/drug effects , Genes, cdc/immunology , HIV/immunology , HIV Infections/immunology , HIV Infections/therapy , Humans , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/trends , Lymphocyte Subsets/physiology , Malaria, Cerebral/immunology , Malaria, Cerebral/therapy , Mice , Orthomyxoviridae/physiology , Protein Kinase Inhibitors/therapeutic use , Streptococcus pneumoniae/physiology , Superinfection/etiology , Superinfection/metabolism
17.
J Leukoc Biol ; 108(4): 1183-1197, 2020 10.
Article in English | MEDLINE | ID: mdl-32362022

ABSTRACT

Heterogeneity and high plasticity are common features of cells from the mononuclear phagocyte system: monocytes (MOs), macrophages, and dendritic cells (DCs). Upon activation by microbial agents, MO can differentiate into MO-derived DCs (MODCs). In previous work, we have shown that during acute infection with Plasmodium berghei ANKA (PbA), MODCs become, transiently, the main CD11b+ myeloid population in the spleen (SP) and once recruited to the brain play an important role in the development of experimental cerebral malaria (ECM). Here, we isolated 4 cell populations: bone marrow (BM) MOs (BM-MOs) and SP-MOs from uninfected mice; BM inflammatory MOs (BM-iMOs) and SP-MODCs from PbA-infected mice and used a system biology approach to a holistic transcriptomic comparison and provide an interactome analysis by integrating differentially expressed miRNAs (DEMs) and their differentially expressed gene targets (DEGs) data. The Jaccard index (JI) was used for gauging the similarity and diversity among these cell populations. Whereas BM-MOs, BM-iMOs, and SP-MOs presented high similarity of DEGs, SP-MODCs distinguished by showing a greater number of DEGs. Moreover, functional analysis identified an enrichment in canonical pathways, such as DC maturation, neuroinflammation, and IFN signaling. Upstream regulator analysis identified IFNγ as the potential upstream molecule that can explain the observed DEMs-Target DEGs intersections in SP-MODCs. Finally, directed target analysis and in vivo/ex vivo assays indicate that SP-MODCs differentiate in the SP and IFNγ is a main driver of this process.


Subject(s)
Dendritic Cells/immunology , Gene Expression Regulation/immunology , Malaria, Cerebral/immunology , MicroRNAs/immunology , Monocytes/immunology , Plasmodium berghei/immunology , RNA, Messenger/immunology , Animals , Dendritic Cells/pathology , Malaria, Cerebral/genetics , Malaria, Cerebral/pathology , Mice , Mice, Knockout , MicroRNAs/genetics , Monocytes/pathology , RNA, Messenger/genetics , Transcriptome/immunology
18.
mBio ; 11(2)2020 04 07.
Article in English | MEDLINE | ID: mdl-32265335

ABSTRACT

Coinfections shape immunity and influence the development of inflammatory diseases, resulting in detrimental or beneficial outcome. Coinfections with concurrent Plasmodium species can alter malaria clinical evolution, and malaria infection itself can modulate autoimmune reactions. Yet, the underlying mechanisms remain ill defined. Here, we demonstrate that the protective effects of some rodent malaria strains on T cell-mediated inflammatory pathologies are due to an RNA virus cohosted in malaria-parasitized blood. We show that live and extracts of blood parasitized by Plasmodium berghei K173 or Plasmodium yoelii 17X YM, protect against P. berghei ANKA-induced experimental cerebral malaria (ECM) and myelin oligodendrocyte glycoprotein (MOG)/complete Freund's adjuvant (CFA)-induced experimental autoimmune encephalomyelitis (EAE), and that protection is associated with a strong type I interferon (IFN-I) signature. We detected the presence of the RNA virus lactate dehydrogenase-elevating virus (LDV) in the protective Plasmodium stabilates and we established that LDV infection alone was necessary and sufficient to recapitulate the protective effects on ECM and EAE. In ECM, protection resulted from an IFN-I-mediated reduction in the abundance of splenic conventional dendritic cell and impairment of their ability to produce interleukin (IL)-12p70, leading to a decrease in pathogenic CD4+ Th1 responses. In EAE, LDV infection induced IFN-I-mediated abrogation of IL-23, thereby preventing the differentiation of granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing encephalitogenic CD4+ T cells. Our work identifies a virus cohosted in several Plasmodium stabilates across the community and deciphers its major consequences on the host immune system. More generally, our data emphasize the importance of considering contemporaneous infections for the understanding of malaria-associated and autoimmune diseases.IMPORTANCE Any infection modifies the host immune status, potentially ameliorating or aggravating the pathophysiology of a simultaneous inflammatory condition. In the course of investigating how malaria infection modulates the severity of contemporaneous inflammatory diseases, we identified a nonpathogenic mouse virus in stabilates of two widely used rodent parasite lines: Plasmodium berghei K173 and Plasmodium yoelii 17X YM. We established that the protective effects of these Plasmodium lines on cerebral malaria and multiple sclerosis are exclusively due to this virus. The virus induces a massive type I interferon (IFN-I) response and causes quantitative and qualitative defects in the ability of dendritic cells to promote pathogenic T cell responses. Beyond revealing a possible confounding factor in rodent malaria models, our work uncovers some bases by which a seemingly innocuous viral (co)infection profoundly changes the immunopathophysiology of inflammatory diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Inflammation/immunology , Interferon Type I/immunology , Lactate dehydrogenase-elevating virus/immunology , Malaria, Cerebral/immunology , Animals , Coinfection/immunology , Coinfection/parasitology , Coinfection/virology , Cytokines/immunology , Dendritic Cells/immunology , Inflammation/physiopathology , Interferon-gamma/immunology , Malaria, Cerebral/blood , Malaria, Cerebral/parasitology , Male , Mice , Mice, Inbred C57BL , Plasmodium berghei , Plasmodium yoelii , Spleen/cytology , Spleen/immunology
19.
J Immunol ; 204(2): 327-334, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31907275

ABSTRACT

Malaria is a deadly infectious disease caused by parasites of the Plasmodium spp. that takes an estimated 435,000 lives each year, primarily among young African children. For most children, malaria is a febrile illness that resolves with time, but in ∼1% of cases, for reasons we do not understand, malaria becomes severe and life threatening. Cerebral malaria (CM) is the most common form of severe malaria, accounting for the vast majority of childhood deaths from malaria despite highly effective antiparasite chemotherapy. Thus, CM is one of the most prevalent lethal brain diseases, and one for which we have no effective therapy. CM is, in part, an immune-mediated disease, and to fully understand CM, it is essential to appreciate the complex relationship between the malarial parasite and the human immune system. In this study, we provide a primer on malaria for immunologists and, in this context, review progress identifying targets for therapeutic intervention.


Subject(s)
Malaria, Cerebral/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Child, Preschool , Female , Humans , Infant , Male
20.
Immunol Rev ; 293(1): 230-252, 2020 01.
Article in English | MEDLINE | ID: mdl-31562653

ABSTRACT

Malaria, a mosquito-borne infectious disease caused by parasites of the genus Plasmodium continues to be a major health problem worldwide. The unicellular Plasmodium-parasites have the unique capacity to infect and replicate within host erythrocytes. By expressing variant surface antigens Plasmodium falciparum has evolved to avoid protective immune responses; as a result in endemic areas anti-malaria immunity develops gradually over many years of multiple and repeated infections. We are studying the role of Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) expressed by asexual stages of P. falciparum responsible for the pathogenicity of severe malaria. The immunopathology of falciparum malaria has been linked to cyto-adhesion of infected erythrocytes to specific host receptors. A greater appreciation of the PfEMP1 molecules important for the development of protective immunity and immunopathology is a prerequisite for the rational discovery and development of a safe and protective anti-disease malaria vaccine. Here we review the role of ICAM-1 and EPCR receptor adhering falciparum-parasites in the development of severe malaria; we discuss our current research to understand the factors involved in the pathogenesis of cerebral malaria and the feasibility of developing a vaccine targeted specifically to prevent this disease.


Subject(s)
Host-Parasite Interactions/immunology , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium falciparum/immunology , Antigenic Variation , Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Humans , Immunity , Malaria Vaccines/immunology , Malaria, Cerebral/prevention & control , Malaria, Falciparum/prevention & control , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...