Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Int J Mol Sci ; 25(7)2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38612941

ABSTRACT

The WHO's global strategy for malaria targets a reduction of at least 90% of both incidence and mortality rates for 2030 [...].


Subject(s)
Malaria , Humans , Malaria/epidemiology
2.
Brain Behav Immun ; 117: 20-35, 2024 03.
Article in English | MEDLINE | ID: mdl-38157948

ABSTRACT

BACKGROUND: Cerebral malaria (CM) is a fatal neuroinflammatory syndrome caused (in humans) by the protozoa Plasmodium (P.) falciparum. Glial cell activation is one of the mechanisms that contributes to neuroinflammation in CM. RESULT: By studying a mouse model of CM (caused by P. berghei ANKA), we describe that the induction of autophagy promoted p21-dependent senescence in astrocytes and that CXCL-10 was part of the senescence-associated secretory phenotype. Furthermore, p21 expression was observed in post-mortem brain and peripheral blood samples from patients with CM. Lastly, we found that the depletion of senescent astrocytes with senolytic drugs abrogated inflammation and protected mice from CM. CONCLUSION: Our data provide evidence for a novel mechanism through which astrocytes could be involved in the neuropathophysiology of CM. p21 gene expression in blood cell and an elevated plasma CXCL-10 concentration could be valuable biomarkers of CM in humans. In the end, we believe senolytic drugs shall open up new avenues to develop newer treatment options.


Subject(s)
Malaria, Cerebral , Humans , Animals , Mice , Neuroinflammatory Diseases , Astrocytes , Senotherapeutics , Autophagy
3.
Front Immunol ; 14: 1134020, 2023.
Article in English | MEDLINE | ID: mdl-37575260

ABSTRACT

Cutaneous leishmaniasis (CL) caused by infection with the parasite Leishmania exhibits a large spectrum of clinical manifestations ranging from single healing to severe chronic lesions with the manifestation of resistance or not to treatment. Depending on the specie and multiple environmental parameters, the evolution of lesions is determined by a complex interaction between parasite factors and the early immune responses triggered, including innate and adaptive mechanisms. Moreover, lesion resolution requires parasite control as well as modulation of the pathologic local inflammation responses and the initiation of wound healing responses. Here, we have summarized recent advances in understanding the in situ immune response to cutaneous leishmaniasis: i) in North Africa caused by Leishmania (L.) major, L. tropica, and L. infantum, which caused in most cases localized autoresolutives forms, and ii) in French Guiana resulting from L. guyanensis and L. braziliensis, two of the most prevalent strains that may induce potentially mucosal forms of the disease. This review will allow a better understanding of local immune parameters, including cellular and cytokines release in the lesion, that controls infection and/or protect against the pathogenesis in new world compared to old world CL.


Subject(s)
Leishmania , Leishmaniasis, Cutaneous , Humans , French Guiana/epidemiology , Africa, Northern , Cytokines
4.
Front Immunol ; 13: 1024998, 2022.
Article in English | MEDLINE | ID: mdl-36569929

ABSTRACT

Many parasitic diseases (including cerebral malaria, human African trypanosomiasis, cerebral toxoplasmosis, neurocysticercosis and neuroschistosomiasis) feature acute or chronic brain inflammation processes, which are often associated with deregulation of glial cell activity and disruption of the brain blood barrier's intactness. The inflammatory responses of astrocytes and microglia during parasite infection are strongly influenced by a variety of environmental factors. Although it has recently been shown that the gut microbiota influences the physiology and immunomodulation of the central nervous system in neurodegenerative diseases like Alzheimer's disease and Parkinson's, the putative link in parasite-induced neuroinflammatory diseases has not been well characterized. Likewise, the central nervous system can influence the gut microbiota. In parasite infections, the gut microbiota is strongly perturbed and might influence the severity of the central nervous system inflammation response through changes in the production of bacterial metabolites. Here, we review the roles of astrocytes and microglial cells in the neuropathophysiological processes induced by parasite infections and their possible regulation by the gut microbiota.


Subject(s)
Gastrointestinal Microbiome , Malaria, Cerebral , Humans , Gastrointestinal Microbiome/physiology , Neuroinflammatory Diseases , Central Nervous System/metabolism , Inflammation/metabolism
5.
Int J Mol Sci ; 23(21)2022 Nov 03.
Article in English | MEDLINE | ID: mdl-36362246

ABSTRACT

Interleukin-33 (IL-33) is an immunomodulatory cytokine which plays critical roles in tissue function and immune-mediated diseases. IL-33 is abundant within the brain and spinal cord tissues where it acts as a key cytokine to coordinate the exchange between the immune and central nervous system (CNS). In this review, we report the recent advances to our knowledge regarding the role of IL-33 and of its receptor ST2 in cerebral malaria, and in particular, we highlight the pivotal role that IL-33/ST2 signaling pathway could play in brain and cerebrospinal barriers permeability. IL-33 serum levels are significantly higher in children with severe Plasmodium falciparum malaria than children without complications or noninfected children. IL-33 levels are correlated with parasite load and strongly decrease with parasite clearance. We postulate that sequestration of infected erythrocytes or merozoites liberation from schizonts could amplify IL-33 production in endothelial cells, contributing either to malaria pathogenesis or recovery.


Subject(s)
Malaria, Cerebral , Malaria, Falciparum , Child , Humans , Interleukin-33/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Endothelial Cells/metabolism , Malaria, Falciparum/parasitology , Cytokines , Plasmodium falciparum/metabolism
6.
Life (Basel) ; 12(3)2022 Mar 12.
Article in English | MEDLINE | ID: mdl-35330166

ABSTRACT

Malaria, caused by Plasmodium species (spp.), is a deadly parasitic disease that results in approximately 400,000 deaths per year globally. Autophagy pathways play a fundamental role in the developmental stages of the parasite within the mammalian host. They are also involved in the production of Plasmodium-derived extracellular vesicles (EVs), which play an important role in the infection process, either by providing nutrients for parasite growth or by contributing to the immunopathophysiology of the disease. For example, during the hepatic stage, Plasmodium-derived EVs contribute to parasite virulence by modulating the host immune response. EVs help in evading the different autophagy mechanisms deployed by the host for parasite clearance. During cerebral malaria, on the other hand, parasite-derived EVs promote an astrocyte-mediated inflammatory response, through the induction of a non-conventional host autophagy pathway. In this review, we will discuss the cross-talk between Plasmodium-derived microvesicles and autophagy, and how it influences the outcome of infection.

7.
Autophagy ; 18(7): 1583-1598, 2022 07.
Article in English | MEDLINE | ID: mdl-34747313

ABSTRACT

Cerebral malaria is a neuroinflammatory disease induced by P. falciparum infection. In animal models, the neuro-pathophysiology of cerebral malaria results from the sequestration of infected red blood cells (iRBCs) in microvessels that promotes the activation of glial cells in the brain. This activation provokes an exacerbated inflammatory response characterized by the secretion of proinflammatory cytokines and chemokines, leading to brain infiltration by pathogenic CD8+ T lymphocytes. Astrocytes are a major subtype of brain glial cells that play an important role in maintaining the homeostasis of the central nervous system, the integrity of the brain-blood barrier and in mounting local innate immune responses. We have previously shown that parasitic microvesicles (PbA-MVs) are transferred from iRBCs to astrocytes. The present study shows that an unconventional LC3-mediated autophagy pathway independent of ULK1 is involved in the transfer and degradation of PbA-MVs inside the astrocytes. We further demonstrate that inhibition of the autophagy process by treatment with 3-methyladenine blocks the transfer of PbA-MVs, which remain localized in the astrocytic cell membrane and are not internalized. Moreover, bafilomycin A1, another drug against autophagy promotes the accumulation of PbA-MVs inside the astrocytes by inhibiting the fusion with lysosomes, and prevents ECM in mice infected with PbA. Finally, we establish that RUBCN/rubicon or ATG5 silencing impede astrocyte production in CCL2 and CXCL10 chemokines induced by PbA stimulation. Altogether, our data suggest that a non-canonical autophagy-lysosomal pathway may play a key role in cerebral malaria through regulation of brain neuro-inflammation by astrocytes.


Subject(s)
Malaria, Cerebral , Plasmodium , Animals , Astrocytes/metabolism , Autophagy , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Plasmodium berghei
8.
J Immunol ; 205(11): 3071-3082, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33148715

ABSTRACT

Malaria is associated with complicated immunopathogenesis. In this study, we provide evidence for an unexpected role of TLR3 in promoting the establishment of Plasmodium yoelii infection through delayed clearance of parasitemia in wild type C57BL/6jRj (B6) compared with TLR3 knockout mice. In this study, we confirmed an increased expression of Tlr3, Trif, Tbk1, and Irf7/Irf3 in the liver 42 h postinfection and the initiation of an early burst of proinflammatory response such as Ifng, NF-kB, and Tnfa in B6 mice that may promote parasite fitness. Interestingly, in the absence of TLR3, we showed the involvement of high IFN-γ and lower type I IFN response in the early clearance of parasitemia. In parallel, we observed an increase in splenic NK and NKT cells expressing TLR3 in infected B6 mice, suggesting a role for TLR sensing in the innate immune response. Finally, we find evidence that the increase in the frequency of CD19+TLR3+ B cells along with reduced levels of total IgG in B6 mice possibly suggests the initiation of TLR3-dependent pathway early during P. yoelii infection. Our results thus reveal a new mechanism in which a parasite-activated TLR3 pathway promotes blood stage infection along with quantitative and qualitative differences in Ab responses.


Subject(s)
Malaria/immunology , Mammals/immunology , Mammals/parasitology , Plasmodium yoelii/immunology , Toll-Like Receptor 3/immunology , Animals , B-Lymphocytes/immunology , Immunity, Innate/immunology , Immunoglobulin G/immunology , Inflammation/immunology , Inflammation/parasitology , Interferon Type I/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/parasitology , Malaria/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/immunology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/parasitology , Parasitemia/immunology , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/immunology
9.
Genes Immun ; 21(1): 45-62, 2020 01.
Article in English | MEDLINE | ID: mdl-31501529

ABSTRACT

Genetic mapping and genome-wide studies provide evidence for the association of several genetic polymorphisms with malaria, a complex pathological disease with multiple severity degrees. We have previously described Berr1and Berr2 as candidate genes identified in the WLA/Pas inbreed mouse strain predisposing to resistance to cerebral malaria (CM) induced by P. berghei ANKA. We report in this study the phenotypic and functional characteristics of a congenic strain we have derived for Berr2WLA allele on the C57BL/6JR (B6) background. B6.WLA-Berr2 was found highly resistant to CM compared to C57BL/6JR susceptible mice. The mechanisms associated with CM resistance were analyzed by combining genotype, transcriptomic and immune response studies. We found that B6.WLA-Berr2 mice showed a reduced parasite sequestration and blood-brain barrier disruption with low CXCR3+ T cell infiltration in the brain along with altered glial cell response upon P. berghei ANKA infection compared to B6. In addition, we have identified the CD300f, belonging to a family of Ig-like encoding genes, as a potential candidate associated with CM resistance. Microglia cells isolated from the brain of infected B6.WLA-Berr2 mice significantly expressed higher level of CD300f compared to CMS mice and were associated with inhibition of inflammatory response.


Subject(s)
Malaria, Cerebral/genetics , Microglia/metabolism , Receptors, Immunologic/metabolism , Alleles , Animals , Brain/immunology , Brain/metabolism , Brain/pathology , Chromosome Mapping , Disease Models, Animal , Disease Resistance/genetics , Disease Resistance/immunology , Female , Genotype , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Microglia/physiology , Receptors, Immunologic/genetics
10.
Parasite Immunol ; 40(10): e12580, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30102786

ABSTRACT

B cell-mediated humoral responses are essential for controlling malarial infection. Studies have addressed the effects of Plasmodium falciparum infection on peripheral B-cell subsets but not much is known for P. vivax infection. Furthermore, majority of the studies investigate changes during acute infection, but not after parasite clearance. In this prospective study, we analysed peripheral B-cell profiles and antibody responses during acute P. vivax infection and upon recovery (30 days post-treatment) in a low-transmission area in India. Dengue patients were included as febrile-condition controls. Both dengue and malaria patients showed a transient increase in atypical memory B cells during acute infection. However, transient B cell-activating factor (BAFF)-independent increase in the percentage of total and activated immature B cells was observed in malaria patients. Naïve B cells from malaria patients also showed increased TLR4 expression. Total IgM levels remained unchanged during acute infection but increased significantly at recovery. Serum antibody profiling showed a parasite-specific IgM response that persisted at recovery. A persistent IgM autoantibody response was also observed in malaria but not dengue patients. Our data suggest that in hypoendemic regions acute P. vivax infection skews peripheral B-cell subsets and results in a persistent parasite-specific and autoreactive IgM response.


Subject(s)
Antibodies, Protozoan/blood , B-Lymphocyte Subsets/immunology , Immunoglobulin M/blood , Malaria, Vivax/immunology , Plasmodium vivax/immunology , Adult , Antibodies, Protozoan/immunology , Antibody Formation , B-Cell Activating Factor/metabolism , Female , Humans , Immunoglobulin M/immunology , India , Malaria, Vivax/parasitology , Male , Middle Aged , Prospective Studies , Toll-Like Receptor 4/biosynthesis
11.
Glia ; 65(1): 75-92, 2017 01.
Article in English | MEDLINE | ID: mdl-27696532

ABSTRACT

Astrocytes and microglia are activated during cerebral malaria (CM) and contribute to the production and release of several mediators during neuroinflammatory processes. Whether these changes are the consequence of a direct crosstalk between glial cells and the malarial parasite and how these cells participate in the pathogenesis of CM is not yet clear. We therefore examined the interaction of astrocytes and microglia with Plasmodium berghei ANKA-infected red blood cells using primary cell cultures derived from newborn C57BL/6 mice. We observed a dynamic transfer of vesicles from the parasite to astrocytes within minutes of contact, and the phagocytosis of infected red blood cells by microglia. Differential gene expression studies using the Affymetrix GeneChip® microarray, and quantitative PCR analyses showed the increase in expression of the set of genes belonging to the immune response network in parasite activated astrocytes and microglia. Interestingly, expression of these genes was also significantly upregulated in brains of mice dying from CM compared with uninfected mice or infected mice that did not develop the neuropathology. Accumulation of parasite-derived vesicles within astrocytes, and the phagocytosis of infected red blood cells by microglia induced a subsequent increase in interferon gamma inducible protein 10 (IP10) in both the brain and plasma of infected mice at the onset of CM, confirming a role for this molecule in CM pathogenesis. Altogether, these observations point to a possible role for glial cells in the neuropathological processes leading to CM. GLIA 2016 GLIA 2017;65:75-92.


Subject(s)
Astrocytes/parasitology , Erythrocytes/parasitology , Malaria, Cerebral/parasitology , Microglia/parasitology , Phagocytosis/physiology , Animals , Astrocytes/metabolism , Brain/parasitology , Brain/pathology , Cells, Cultured , Cytokines/metabolism , Female , Malaria, Cerebral/pathology , Mice, Inbred C57BL , Microglia/metabolism
12.
Brain Behav Immun ; 58: 280-290, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27477919

ABSTRACT

Cerebral malaria is the deadliest complication of Plasmodium falciparum infection. Its pathophysiology is associated with a strong pro-inflammatory reaction and the activation of glial cells. Among modulators released during the infection, heme seems to play a controversial role in the pathophysiology of malaria. Herein, we first investigated the phenotype of glial cells during cerebral malaria in C57BL/6 mice infected with P. berghei ANKA. Given the fact that high levels of heme were associated with cerebral malaria, we then investigated its impact on microglial, astrocyte, and T cell responses to further clarify its contribution in the neuropathophysiology. Surprisingly, we found that administration of heme twice a day from day three of infection induced the expression of the Heme oxygenase-1 (Hmox1) gene and prevented brain damages. More specifically, heme inhibited the M1 phenotype of microglia, hampered the activation of astrocytes, and decreased the cerebral expression of Ifng, Tnfa and Ip10. Heme might that way alter the migration of pathogenic CD4 and CD8 T lymphocytes within the brain observed during cerebral malaria. Taking into account that cerebral malaria results from a complex interplay between host- and parasite-derived factors, it is possible that genetic polymorphisms of Hmox1, which could be associated with the control of systemic levels of heme during P. falciparum infection, might explain its dual role and its contribution to the resistance to cerebral malaria.


Subject(s)
Astrocytes/immunology , Brain/immunology , Brain/parasitology , Heme/metabolism , Malaria, Cerebral/immunology , Microglia/immunology , T-Lymphocytes/metabolism , Animals , Female , Heme/administration & dosage , Heme Oxygenase-1/metabolism , Infectious Encephalitis/complications , Malaria, Cerebral/complications , Malaria, Cerebral/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , Plasmodium berghei/pathogenicity , Spleen
13.
Mol Biosyst ; 12(11): 3324-3332, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27546486

ABSTRACT

The human malarial parasite Plasmodium falciparum causes the most severe forms of malarial infections, which include cerebral malaria and various organ dysfunctions amongst adults in India. So far no dependable clinical descriptor is available that can distinguish cerebral malaria from other symptomatically similar diseases such as sepsis and encephalitis. This study aims at evaluating the differential metabolic features of plasma samples from P. falciparum patients with varying severities, and patients suffering from symptomatically similar diseases. 1H Nuclear Magnetic Resonance (NMR) based metabolic profiling of the plasma of the infected individuals and the control population was performed. The differences in the plasma profiles were evaluated through multivariate statistical analyses. The results suggest malaria-specific elevation of plasma lipoproteins. Such an increase was absent in control populations. In addition, cerebral malaria patients exhibited a decrease in plasma glycoproteins; such a reduction was not observed in malarial patients without cerebral symptoms. The data presented here indicates that the metabolism and/or transport of the plasma lipids is specifically perturbed by malarial infections. The differential perturbation of the plasma glycoprotein levels in cerebral malaria patients may have important implications in the diagnosis of cerebral malaria.


Subject(s)
Energy Metabolism , Host-Parasite Interactions , Malaria, Falciparum/metabolism , Metabolome , Metabolomics , Plasmodium falciparum , Proton Magnetic Resonance Spectroscopy , Adolescent , Adult , Case-Control Studies , Encephalitis/blood , Encephalitis/metabolism , Female , Humans , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Male , Metabolomics/methods , Middle Aged , Sepsis/blood , Sepsis/metabolism , Young Adult
14.
PLoS One ; 11(7): e0158420, 2016.
Article in English | MEDLINE | ID: mdl-27441662

ABSTRACT

Cerebral malaria (CM) caused by Plasmodium falciparum parasites often leads to the death of infected patients or to persisting neurological sequelae despite anti-parasitic treatments. Erythropoietin (EPO) was recently suggested as a potential adjunctive treatment for CM. However diverging results were obtained in patients from Sub-Saharan countries infected with P. falciparum. In this study, we measured EPO levels in the plasma of well-defined groups of P. falciparum-infected patients, from the state of Odisha in India, with mild malaria (MM), CM, or severe non-CM (NCM). EPO levels were then correlated with biological parameters, including parasite biomass, heme, tumor necrosis factor (TNF)-α, interleukin (IL)-10, interferon gamma-induced protein (IP)-10, and monocyte chemoattractant protein (MCP)-1 plasma concentrations by Spearman's rank and multiple correlation analyses. We found a significant increase in EPO levels with malaria severity degree, and more specifically during fatal CM. In addition, EPO levels were also found correlated positively with heme, TNF-α, IL-10, IP-10 and MCP-1 during CM. We also found a significant multivariate correlation between EPO, TNF-α, IL-10, IP-10 MCP-1 and heme, suggesting an association of EPO with a network of immune factors in CM patients. The contradictory levels of circulating EPO reported in CM patients in India when compared to Africa highlights the need for the optimization of adjunctive treatments according to the targeted population.


Subject(s)
Erythropoietin/blood , Heme/metabolism , Interleukin-10/blood , Malaria, Cerebral/blood , Tumor Necrosis Factor-alpha/blood , Adult , Antigens, Protozoan/metabolism , Chemokine CCL2/blood , Female , Hemopexin/metabolism , Humans , India , Malaria, Cerebral/parasitology , Male , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Severity of Illness Index , Young Adult
15.
Infect Genet Evol ; 43: 146-50, 2016 09.
Article in English | MEDLINE | ID: mdl-27166096

ABSTRACT

Toll-interleukin-1 receptor domain containing adapter protein (TIRAP) plays a crucial role in TLR2 and TLR4 signaling pathways. Glycosylphospatidylinositol (GPI), considered a toxin molecule of Plasmodium falciparum, interacts with TLR2 and 4 to induce an immune inflammatory response. A single nucleotide polymorphism at coding region of TIRAP (S180L) has been reported to influence TLRs signaling. In the present study, we investigated the association of TIRAP (S180L) polymorphism with susceptibility/resistance to severe P. falciparum malaria in a cohort of adult patients from India. TIRAP S180L polymorphism was typed in 347 cases of severe malaria (SM), 232 uncomplicated malaria and 150 healthy controls. Plasma levels of TNF-α was quantified by ELISA. Heterozygous mutation (S/L) conferred significant protection against MOD (multi organ dysfunction), NCSM (non-cerebral severe malaria) as well as mortality. Interestingly, homozygous mutants (L/L) had 16 fold higher susceptibility to death. TIRAP mutants (S/L and L/L) were associated with significantly higher plasma TNF-α levels compared to wild type (S/S). The results of the present study demonstrate that TIRAP S180L heterozygous mutation may protect patients against severe malaria and mortality.


Subject(s)
Malaria, Falciparum/mortality , Malaria, Falciparum/prevention & control , Membrane Glycoproteins/genetics , Polymorphism, Single Nucleotide , Receptors, Interleukin-1/genetics , Case-Control Studies , Female , Gene Frequency , Heterozygote , Humans , Malaria, Falciparum/genetics , Male , Signal Transduction , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/blood
16.
PLoS One ; 11(2): e0147871, 2016.
Article in English | MEDLINE | ID: mdl-26844551

ABSTRACT

Cerebral Malaria (CM) is associated with a pathogenic T cell response. Mice infected by P. berghei ANKA clone 1.49 (PbA) developing CM (CM+) present an altered PBL TCR repertoire, partly due to recurrently expanded T cell clones, as compared to non-infected and CM- infected mice. To analyse the relationship between repertoire alteration and CM, we performed a kinetic analysis of the TRBV repertoire during the course of the infection until CM-related death in PbA-infected mice. The repertoires of PBL, splenocytes and brain lymphocytes were compared between infected and non-infected mice using a high-throughput CDR3 spectratyping method. We observed a modification of the whole TCR repertoire in the spleen and blood of infected mice, from the fifth and the sixth day post-infection, respectively, while only three TRBV were significantly perturbed in the brain of infected mice. Using multivariate analysis and statistical modelling, we identified a unique TCRß signature discriminating CM+ from CTR mice, enriched during the course of the infection in the spleen and the blood and predicting CM onset. These results highlight a dynamic modification and compartmentalization of the TCR diversity during the course of PbA infection, and provide a novel method to identify disease-associated TCRß signature as diagnostic and prognostic biomarkers.


Subject(s)
Genetic Variation , Malaria, Cerebral/genetics , Malaria, Cerebral/parasitology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Brain/immunology , Brain/parasitology , Complementarity Determining Regions/genetics , Disease Models, Animal , Malaria, Cerebral/diagnosis , Malaria, Cerebral/immunology , Mice , Plasmodium berghei , Prognosis , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
Infect Immun ; 84(1): 329-38, 2016 01.
Article in English | MEDLINE | ID: mdl-26553468

ABSTRACT

The role of naturally occurring CD4(+) CD25(+) Foxp3(+) regulatory T cells (nTreg) in the pathogenesis of cerebral malaria (CM), which involves both pathogenic T cell responses and parasite sequestration in the brain, is still unclear. To assess the contribution and dynamics of nTreg during the neuropathogenesis, we unbalanced the ratio between nTreg and naive CD4(+) T cells in an attenuated model of Plasmodium berghei ANKA-induced experimental CM (ECM) by using a selective cell enrichment strategy. We found that nTreg adoptive transfer accelerated the onset and increased the severity of CM in syngeneic C57BL/6 (B6) P. berghei ANKA-infected mice without affecting the level of parasitemia. In contrast, naive CD4(+) T cell enrichment prevented CM and promoted parasite clearance. Furthermore, early during the infection nTreg expanded in the spleen but did not efficiently migrate to the site of neuroinflammation, suggesting that nTreg exert their pathogenic action early in the spleen by suppressing the protective naive CD4(+) T cell response to P. berghei ANKA infection in vivo in both CM-susceptible (B6) and CM-resistant (B6-CD4(-/-)) mice. However, their sole transfer was not sufficient to restore CM susceptibility in two CM-resistant congenic strains tested. Altogether, these results demonstrate that nTreg are activated and functional during P. berghei ANKA infection and that they contribute to the pathogenesis of CM. They further suggest that nTreg may represent an early target for the modulation of the immune response to malaria.


Subject(s)
Brain/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Brain/cytology , Brain/parasitology , CD4 Antigens/genetics , CD4 Antigens/metabolism , Cell Movement/immunology , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/transplantation
18.
J Transl Med ; 13: 369, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26602091

ABSTRACT

BACKGROUND: Plasmodium falciparum malaria in India is characterized by high rates of severe disease, with multiple organ dysfunction (MOD)-mainly associated with acute renal failure (ARF)-and increased mortality. The objective of this study is to identify cytokine signatures differentiating severe malaria patients with MOD, cerebral malaria (CM), and cerebral malaria with MOD (CM-MOD) in India. We have previously shown that two cytokines clusters differentiated CM from mild malaria in Maharashtra. Hence, we also aimed to determine if these cytokines could discriminate malaria subphenotypes in Odisha. METHODS: P. falciparum malaria patients from the SCB Medical College Cuttack in the Odisha state in India were enrolled along with three sets of controls: healthy individuals, patients with sepsis and encephalitis (n = 222). We determined plasma concentrations of pro- and anti-inflammatory cytokines and chemokines for all individuals using a multiplex assay. We then used an ensemble of statistical analytical methods to ascertain whether particular sets of cytokines/chemokines were predictors of severity or signatures of a disease category. RESULTS: Of the 26 cytokines/chemokines tested, 19 increased significantly during malaria and clearly distinguished malaria patients from controls, as well as sepsis and encephalitis patients. High amounts of IL-17, IP-10, and IL-10 predicted MOD, decreased IL-17 and MIP-1α segregated CM-MOD from MOD, and increased IL-12p40 differentiated CM from CM-MOD. Most severe malaria patients with ARF exhibited high levels of IL-17. CONCLUSION: We report distinct differences in cytokine production correlating with malarial disease severity in Odisha and Maharashtra populations in India. We show that CM, CM-MOD and MOD are clearly distinct malaria-associated pathologies. High amounts of IL-17, IP-10, and IL-10 were predictors of MOD; decreased IL-17 and MIP-1α separated CM-MOD from MOD; and increased IL-12p40 differentiated CM from CM-MOD. Data also suggest that the IL-17 pathway may contribute to malaria pathogenesis via different regulatory mechanisms and may represent an interesting target to mitigate the pathological processes in malaria-associated ARF.


Subject(s)
Acute Kidney Injury/physiopathology , Chemokine CXCL10/physiology , Interleukin-10/physiology , Interleukin-17/physiology , Malaria, Falciparum/physiopathology , Multiple Organ Failure/physiopathology , Acute Kidney Injury/pathology , Chemokine CXCL10/blood , Humans , Interleukin-10/blood , Interleukin-17/blood , Malaria, Falciparum/pathology , Multiple Organ Failure/pathology
19.
Infect Immun ; 83(10): 3793-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26169278

ABSTRACT

Several immunomodulatory factors are involved in malaria pathogenesis. Among them, heme has been shown to play a role in the pathophysiology of severe malaria in rodents, but its role in human severe malaria remains unclear. Circulating levels of total heme and its main scavenger, hemopexin, along with cytokine/chemokine levels and biological parameters, including hemoglobin and creatinine levels, as well as transaminase activities, were measured in the plasma of 237 Plasmodium falciparum-infected patients living in the state of Odisha, India, where malaria is endemic. All patients were categorized into well-defined groups of mild malaria, cerebral malaria (CM), or severe noncerebral malaria, which included acute renal failure (ARF) and hepatopathy. Our results show a significant increase in total plasma heme levels with malaria severity, especially for CM and malarial ARF. Spearman rank correlation and canonical correlation analyses have shown a correlation between total heme, hemopexin, interleukin-10, tumor necrosis factor alpha, gamma interferon-induced protein 10 (IP-10), and monocyte chemotactic protein 1 (MCP-1) levels. In addition, canonical correlations revealed that heme, along with IP-10, was associated with the CM pathophysiology, whereas both IP-10 and MCP-1 together with heme discriminated ARF. Altogether, our data indicate that heme, in association with cytokines and chemokines, is involved in the pathophysiology of both CM and ARF but through different mechanisms.


Subject(s)
Heme/metabolism , Malaria, Falciparum/blood , Plasmodium falciparum/physiology , Adult , Chemokine CCL2/blood , Disease Progression , Female , Hemopexin/metabolism , Humans , India , Interleukin-10/blood , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Male , Middle Aged , Tumor Necrosis Factor-alpha/blood , Young Adult
20.
Malar J ; 14: 162, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25889717

ABSTRACT

BACKGROUND: Mechanisms of acquired protection to malaria in asymptomatic Plasmodium falciparum carriers are only partially understood. Among them, the role plays by the self-reactive antibodies has not been clarified yet. In this study, the relationship between repertoires of circulating self-reactive and parasite-specific immunoglobulin G (IgG), their correlation with cytokine levels, and their association with protection against malaria was investigated in asymptomatic Plasmodium falciparum-infected Gabonese children. METHODS: The diversity of P. falciparum-specific antibody repertoire was analysed using a protein micro-array immunoassay, the total auto-antibody repertoire by quantitative immunoblotting and circulating cytokine levels were measured by ELISA in endemic controls (EC) and P. falciparum-infected children from Gabon with asymptomatic (AM) or mild malaria (MM). The association of self- and parasite-specific antibody repertoires with circulating cytokines was evaluated using single linkage hierarchical clustering, Kruskal-Wallis tests and Spearman's rank correlation. RESULTS: Children with AM exhibited an IgG response to merozoite surface protein 3 (MSP3) but not to MSP1-19, although their levels of total P. falciparum-specific IgG were similar to those in the MM group. Moreover, the asymptomatic children had increased levels of autoantibodies recognising brain antigens. In addition, a correlation between IL-10 levels and parasite load was found in AM and MM children. These two groups also exhibited significant correlations between plasma levels of IL-10 and IFN-γ with age and with total plasma IgG levels. IL-10 and IFN-γ levels were also associated with auto-antibody responses in AM. CONCLUSIONS: Altogether, these results indicate that a self-reactive polyclonal response associated with increased IgG to MSP3 and high plasma levels of IL-10 and IFN-γ may contribute to protective immune mechanisms triggered in asymptomatic P. falciparum infection in Gabonese children.


Subject(s)
Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Autoantibodies/blood , Interleukin-10/blood , Malaria, Falciparum/immunology , Plasmodium falciparum/physiology , Protozoan Proteins/immunology , Asymptomatic Infections , Autoantibodies/biosynthesis , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Gabon , Humans , Infant , Malaria, Falciparum/parasitology , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...