Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Exp Parasitol ; 257: 108686, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38158008

ABSTRACT

BACKGROUND: Based on understanding of placental pathological features and safe medication in pregnancy-associated malaria (PAM), establishment of a stable pregnant mouse infection model with Plasmodium was urgently needed. METHODS: ICR mice with vaginal plugs detected were randomly divided into post-pregnancy infection (Malaria+) and uninfected pregnancy (Malaria-) cohorts. Age-matched mice that had not been mated were infected as pre-pregnancy infection group (Virgin control), which were subsequently mated with ICR males. All mice were inoculated with 1 × 106Plasmodium berghei ANKA-infected RBCs by intraperitoneal injection, and the same amount of saline was given to Malaria- group. We recorded the incidence of adverse pregnancy outcomes and the amounts of offspring in each group. RESULTS: The Virgin group mice were unable to conceive normally, and vaginal bleeding, abortion, or stillbirth appeared in the Malaria+ group. The incidence of adverse pregnancy outcomes was extremely high and statistically significant compared with the control (Malaria-) group (P < 0.05), of which placenta exhibited pathological features associated with human gestational malaria. CONCLUSIONS: The intraperitoneal injection of 1 × 106Plasmodium berghei ANKA-infected RBCs could establish a model of pregnancy-associated malaria in ICR mouse.


Subject(s)
Malaria , Pregnancy Outcome , Male , Pregnancy , Female , Mice , Animals , Humans , Mice, Inbred ICR , Placenta/pathology , Malaria/drug therapy , Plasmodium berghei
2.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Article in English | MEDLINE | ID: mdl-33836556

ABSTRACT

CD47 is an antiphagocytic "don't eat me" signal that inhibits programmed cell removal of self. As red blood cells (RBCs) age they lose CD47 expression and become susceptible to programmed cell removal by macrophages. CD47-/- mice infected with Plasmodium yoelii, which exhibits an age-based preference for young RBCs, were previously demonstrated to be highly resistant to malaria infection. Our study sought to test the therapeutic benefit of CD47 blockade on ameliorating the clinical syndromes of experimental cerebral malaria (ECM), using the Plasmodium berghei ANKA (Pb-A) murine model. In vitro we tested the effect of anti-CD47 mAb on Plasmodium-infected RBC phagocytosis and found that anti-CD47 treatment significantly increased clearance of Plasmodium-infected RBCs. Infection of C57BL/6 mice with Pb-A is lethal and mice succumb to the clinical syndromes of CM between days 6 and 10 postinfection. Strikingly, treatment with anti-CD47 resulted in increased survival during the cerebral phase of Pb-A infection. Anti-CD47-treated mice had increased lymphocyte counts in the peripheral blood and increased circulating levels of IFN-γ, TNF-α, and IL-22. Despite increased circulating levels of inflammatory cytokines, anti-CD47-treated mice had reduced pathological features in the brain. Survival of ECM in anti-CD47-treated mice was correlated with reduced cellular accumulation in the cerebral vasculature, improved blood-brain barrier integrity, and reduced cytotoxic activity of infiltrating CD8+ T cells. These results demonstrate the therapeutic benefit of anti-CD47 to reduce morbidity in a lethal model of ECM, which may have implications for preventing mortality in young African children who are the highest casualties of CM.


Subject(s)
CD47 Antigen/antagonists & inhibitors , Host-Parasite Interactions , Malaria, Cerebral/pathology , Animals , Antibodies, Monoclonal/immunology , CD47 Antigen/immunology , Erythrocytes/parasitology , Humans , Malaria, Cerebral/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phagocytosis
3.
Int J Mol Sci ; 24(14)2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37511092

ABSTRACT

Newly emerging data suggest that several neutrophil defense mechanisms may play a role in both aggravating and protecting against malaria. These exciting findings suggest that the balance of these cells in the host body may have an impact on the pathogenesis of malaria. To fully understand the role of neutrophils in severe forms of malaria, such as cerebral malaria (CM), it is critical to gain a comprehensive understanding of their behavior and functions. This study investigated the dynamics of neutrophil and T cell responses in C57BL/6 and BALB/c mice infected with Plasmodium berghei ANKA, murine models of experimental cerebral malaria (ECM) and non-cerebral experimental malaria, respectively. The results demonstrated an increase in neutrophil percentage and neutrophil-T cell ratios in the spleen and blood before the development of clinical signs of ECM, which is a phenomenon not observed in the non-susceptible model of cerebral malaria. Furthermore, despite the development of distinct forms of malaria in the two strains of infected animals, parasitemia levels showed equivalent increases throughout the infection period evaluated. These findings suggest that the neutrophil percentage and neutrophil-T cell ratios may be valuable predictive tools for assessing the dynamics and composition of immune responses involved in the determinism of ECM development, thus contributing to the advancing of our understanding of its pathogenesis.


Subject(s)
Malaria, Cerebral , Animals , Mice , Neutrophils/pathology , Mice, Inbred C57BL , Plasmodium berghei , CD8-Positive T-Lymphocytes , Disease Models, Animal
4.
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
5.
J Cell Mol Med ; 24(18): 10636-10647, 2020 09.
Article in English | MEDLINE | ID: mdl-32779889

ABSTRACT

Malaria in pregnancy (MiP) induces intrauterine growth restriction (IUGR) and preterm labour (PTL). However, its effects on yolk sac morphology and function are largely unexplored. We hypothesized that MiP modifies yolk sac morphology and efflux transport potential by modulating ABC efflux transporters. C57BL/6 mice injected with Plasmodium berghei ANKA (5 × 105 infected erythrocytes) at gestational day (GD) 13.5 were subjected to yolk sac membrane harvesting at GD 18.5 for histology, qPCR and immunohistochemistry. MiP did not alter the volumetric proportion of the yolk sac's histological components. However, it increased levels of Abcb1a mRNA (encoding P-glycoprotein) and macrophage migration inhibitory factor (Mif chemokine), while decreasing Abcg1 (P < 0.05); without altering Abca1, Abcb1b, Abcg2, Snat1, Snat2, interleukin (Il)-1ß and C-C Motif chemokine ligand 2 (Ccl2). Transcripts of Il-6, chemokine (C-X-C motif) ligand 1 (Cxcl1), Glut1 and Snat4 were not detectible. ABCA1, ABCG1, breast cancer resistance protein (BCRP) and P-gp were primarily immunolocalized to the cell membranes and cytoplasm of endodermic epithelium but also in the mesothelium and in the endothelium of mesodermic blood vessels. Intensity of P-gp labelling was stronger in both endodermic epithelium and mesothelium, whereas ABCA1 labelling increased in the endothelium of the mesodermic blood vessels. The presence of ABC transporters in the yolk sac wall suggests that this fetal membrane acts as an important protective gestational barrier. Changes in ABCA1 and P-gp in MiP may alter the biodistribution of toxic substances, xenobiotics, nutrients and immunological factors within the fetal compartment and participate in the pathogenesis of malaria-induced IUGR and PTL.


Subject(s)
ATP Binding Cassette Transporter 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B/biosynthesis , Gene Expression Regulation , Malaria/metabolism , Pregnancy Complications, Infectious/metabolism , Yolk Sac/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Biological Transport , Cytokines/biosynthesis , Cytokines/genetics , Female , Fetal Growth Retardation/etiology , Inflammation , Malaria/complications , Malaria/genetics , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Organ Size , Plasmodium berghei , Pregnancy , Pregnancy Complications, Infectious/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Yolk Sac/ultrastructure
6.
Proc Natl Acad Sci U S A ; 114(29): 7701-7706, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28673996

ABSTRACT

Severe malaria and associated high parasite burdens occur more frequently in humans lacking robust adaptive immunity to Plasmodium falciparum Nevertheless, the host may partly control blood-stage parasite numbers while adaptive immunity is gradually established. Parasite control has typically been attributed to enhanced removal of parasites by the host, although in vivo quantification of this phenomenon remains challenging. We used a unique in vivo approach to determine the fate of a single cohort of semisynchronous, Plasmodium berghei ANKA- or Plasmodium yoelii 17XNL-parasitized red blood cells (pRBCs) after transfusion into naive or acutely infected mice. As previously shown, acutely infected mice, with ongoing splenic and systemic inflammatory responses, controlled parasite population growth more effectively than naive controls. Surprisingly, however, this was not associated with accelerated removal of pRBCs from circulation. Instead, transfused pRBCs remained in circulation longer in acutely infected mice. Flow cytometric assessment and mathematical modeling of intraerythrocytic parasite development revealed an unexpected and substantial slowing of parasite maturation in acutely infected mice, extending the life cycle from 24 h to 40 h. Importantly, impaired parasite maturation was the major contributor to control of parasite growth in acutely infected mice. Moreover, by performing the same experiments in rag1-/- mice, which lack T and B cells and mount weak inflammatory responses, we revealed that impaired parasite maturation is largely dependent upon the host response to infection. Thus, impairment of parasite maturation represents a host-mediated, immune system-dependent mechanism for limiting parasite population growth during the early stages of an acute blood-stage Plasmodium infection.


Subject(s)
Host-Parasite Interactions , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium berghei/physiology , Plasmodium falciparum/physiology , Adaptive Immunity , Animals , Cytokines/metabolism , Erythrocytes/parasitology , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Immune System , Inflammation , Malaria , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Theoretical , Plasmodium yoelii/physiology
7.
Parasitol Res ; 119(4): 1301-1315, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32179986

ABSTRACT

Malaria and lymphatic filariasis (LF) are two leading and common mosquito-borne parasitic diseases worldwide. These two diseases are co-endemic in many tropical and sub-tropical regions and are known to share vectors. The interactions between malaria and filarial parasites are poorly understood. Thus, this study aimed at establishing the interactions that occur between Brugia pahangi and Plasmodium berghei ANKA (PbA) co-infection in gerbils. Briefly, the gerbils were matched according to age, sex, and weight and grouped into filarial-only infection, PbA-only infection, co-infection, and control group. The parasitemia, survival and clinical assessment of the gerbils were monitored for a period of 30 days post Plasmodium infection. The immune responses of gerbils to both mono and co-infection were monitored. Findings show that co-infected gerbils have higher survival rate than PbA-infected gerbils. Food and water consumption were significantly reduced in both PbA-infected and co-infected gerbils, although loss of body weight, hypothermia, and anemia were less severe in co-infected gerbils. Plasmodium-infected gerbils also suffered hypoglycemia, which was not observed in co-infected gerbils. Furthermore, gerbil cytokine responses to co-infection were significantly higher than PbA-only-infected gerbils, which is being suggested as a factor for their increased longevity. Co-infected gerbils had significantly elicited interleukin-4, interferon-gamma, and tumor necrotic factor at early stage of infection than PbA-infected gerbils. Findings from this study suggest that B. pahangi infection protect against severe anemia and hypoglycemia, which are manifestations of PbA infection.


Subject(s)
Brugia pahangi/immunology , Filariasis/veterinary , Gerbillinae/parasitology , Malaria/veterinary , Plasmodium berghei/immunology , Animals , Coinfection/immunology , Coinfection/parasitology , Cytokines/blood , Female , Filariasis/parasitology , Host-Parasite Interactions/immunology , Hypoglycemia/parasitology , Malaria/parasitology , Male , Mosquito Vectors/parasitology , Parasitemia/parasitology , Parasitemia/veterinary , Survival Rate
8.
Mol Biol Rep ; 46(4): 3801-3808, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31012028

ABSTRACT

A direct role for IgA either for elimination of malaria parasite or for improvement in tissue pathology has not been investigated in case of Malaria infection while IgG, IgE and IgM were all implicated in the adverse pathology. In this communication, we delineate further that Malaria specific IgA appears to be significant among individuals who had multiple episodes of infection. Interestingly, the IgA elicited by immunization of the homologous peptides derived from Plasmodium berghei ANKA have also resulted in protection of host from adverse lung pathology, while the parasite load is unaffected. The PfrVI immunized mice and mice infected with repeated cycles of 'infection and recovery', simulating an endemic like situation, have resulted in development of B cell population that secretes the IgA specific to this region VI. Summarily, our results suggest that the IgA specific to the malarial antigen can confer significant advantage to hosts in protecting the overall tissue pathology.


Subject(s)
Immunoglobulin A/immunology , Malaria/immunology , Malaria/metabolism , Plasmodium berghei/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Erythrocytes/metabolism , Immunization , Ligands , Malaria/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmodium berghei/pathogenicity , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
9.
Parasitology ; 146(12): 1571-1577, 2019 10.
Article in English | MEDLINE | ID: mdl-31244453

ABSTRACT

Cerebral malaria (CM) is the severe neurological complication causing acute non-traumatic encephalopathy in tropical countries. The mechanisms underlying the fatal cerebral complications are still not fully understood. Glutamate, a major excitatory neurotransmitter in the central nervous system of the mammalian brain, plays a key role in the development of neuronal cells, motor function, synaptic plasticity, learning and memory processes under normal physiological conditions. The subtypes of ionotropic glutamate receptor are N-methyl-D-aspartate receptors (NMDARs) which are involved in cellular mechanisms of learning and memory, synaptic plasticity and also mediate excitotoxic neuronal injury. In the present study, we found that glutamate level in synaptosomes, as well as expression of NMDAR, was elevated during the extreme condition of CM in C57BL6 mice. Arteether at 50 mg kg-1 × 1, 25 mg kg-1 × 2, days decreased the NMDAR expression and increased the overall survival of the experimental CM mice.


Subject(s)
Antimalarials/pharmacology , Artemisinins/pharmacology , Gene Expression/drug effects , Malaria, Cerebral/drug therapy , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Female , Longevity/drug effects , Mice , Mice, Inbred C57BL
10.
Parasitol Res ; 118(10): 2969-2977, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31482465

ABSTRACT

C57BL/6 mice infected with Plasmodium berghei ANKA (PbA) develop neurological symptoms and die 6--7-day post-inoculation in the absence of high parasitemia. The effects of chronic intake of a high-fat diet on this process are largely unknown. In this study, we assessed the effect of a high-fat diet on the host-parasite response to malarial infection. Mice were fed ad libitum with either standard or a high-fat diet for 8 weeks and afterwards were infected with PbA. PbA-infected mice feeding a standard diet presented blood parasitemia, hepatic and cerebral histopathological alterations, and hepatic injury with increased hemozoin deposition in the liver. By contrast, these changes were not observed in the malaria high-fat diet group. In addition, mice fed a high-fat diet did not develop the expected neurological symptoms of cerebral malaria and were resistant to death. Taken together, our results indicate that chronic ingestion of high-fat diet prevents the development of experimental malaria induced by PbA injection, suggesting a relationship between a high-fat diet and malaria, which is an interesting subject for further study in humans.


Subject(s)
Diet, High-Fat , Malaria/prevention & control , Plasmodium berghei/physiology , Animals , Brain/pathology , Disease Models, Animal , Hemeproteins/metabolism , Liver/metabolism , Liver/pathology , Malaria/parasitology , Malaria/pathology , Mice, Inbred C57BL , Parasitemia/parasitology , Parasitemia/prevention & control , Plasmodium berghei/growth & development
11.
Infect Immun ; 86(7)2018 07.
Article in English | MEDLINE | ID: mdl-29685989

ABSTRACT

Recent studies have demonstrated that a subpopulation of neutrophils express the TCRαß combinatorial immunoreceptor in humans and mice. Here, we report that a Plasmodium berghei ANKA murine malaria infection induces expansion of TCRß expressing CD11b+ Ly6G+ neutrophils in the spleen during the early phase of infection. Measurement of TCRß transcript and protein levels of neutrophils in wild-type versus nude and Rag1 knockout mice establishes that the observed expression is not a consequence of nonspecific antibody staining or passive receptor expression due to phagocytosis or trogocytosis of peripheral T cells. Remarkably, on day 3 postinfection, we observed a highly significant correlation between the proportion of neutrophils that express TCRß and peripheral blood parasite burden. In addition, TCRß+ neutrophils phagocytose parasitized erythrocytes with 4-fold greater efficiency than TCRß- neutrophils. Together these results signify that TCR expression by the neutrophil plays an important role in the regulation of parasite burden by enhancing the phagocytic capacity of the neutrophil.


Subject(s)
Malaria/immunology , Neutrophils/immunology , Parasitemia/immunology , Phagocytosis , Plasmodium berghei , Receptors, Antigen, T-Cell, alpha-beta/analysis , Animals , Brain/immunology , Female , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/genetics , Spleen/immunology
12.
Infect Immun ; 86(8)2018 08.
Article in English | MEDLINE | ID: mdl-29784863

ABSTRACT

In animal models of experimental cerebral malaria (ECM), the glycosylphosphatidylinositols (GPIs) and GPI anchors are the major factors that induce nuclear factor kappa B (NF-κB) activation and proinflammatory responses, which contribute to malaria pathogenesis. GPIs and GPI anchors are transported to the cell surface via a process called GPI transamidation, which involves the GPI transamidase (GPI-T) complex. In this study, we showed that GPI16, one of the GPI-T subunits, is highly conserved among Plasmodium species. Genetic knockout of pbgpi16 (Δpbgpi16) in the rodent malaria parasite Plasmodium berghei strain ANKA led to a significant reduction of the amounts of GPIs in the membranes of merozoites, as well as surface display of several GPI-anchored merozoite surface proteins. Compared with the wild-type parasites, Δpbgpi16 parasites in C57BL/6 mice caused much less NF-κB activation and elicited a substantially attenuated T helper type 1 response. As a result, Δpbgpi16 mutant-infected mice displayed much less severe brain pathology, and considerably fewer Δpbgpi16 mutant-infected mice died from ECM. This study corroborated the GPI toxin as a significant inducer of ECM and further suggested that vaccines against parasite GPIs may be a promising strategy to limit the severity of malaria.


Subject(s)
Aminoacyltransferases/metabolism , Glycosylphosphatidylinositols/metabolism , Malaria, Cerebral/pathology , Malaria, Cerebral/parasitology , Plasmodium berghei/enzymology , Protozoan Proteins/metabolism , Virulence Factors/metabolism , Aminoacyltransferases/genetics , Animals , Brain/pathology , Cell Membrane/chemistry , Disease Models, Animal , Female , Gene Knockout Techniques , Membrane Proteins/analysis , Merozoites/chemistry , Mice, Inbred C57BL , NF-kappa B/metabolism , Plasmodium berghei/genetics , Protozoan Proteins/analysis , Survival Analysis , Th1 Cells/immunology , Virulence Factors/genetics
13.
Cell Physiol Biochem ; 50(4): 1414-1428, 2018.
Article in English | MEDLINE | ID: mdl-30355912

ABSTRACT

BACKGROUND/AIMS: Malaria is the most deadly parasitic infection in the world, resulting in damage to various organs, including the liver, of the infected organism; however, the mechanism causing this damage in the liver remains unclear. Liver fibrosis, a major characteristic of liver diseases, occurs in response to liver injury and is regulated by a complex network of signaling pathways. Hedgehog (Hh) signaling orchestrates a number of hepatic responses including hepatic fibrogenesis. Therefore, we investigated whether Hh signaling influenced the liver's response to malarial infection. METHODS: Eight-week-old male C57BL/6 mice inoculated with blood containing Plasmodium berghei ANKA (PbA)-infected erythrocytes were sacrificed when the level of parasitemia in the blood reached 10% or 30%, and the livers were collected for biochemical analysis. Liver responses to PbA infection were examined by hematoxylin and eosin staining, real-time polymerase chain reaction, immunohistochemistry and western blot. RESULTS: Severe hepatic injury, such as ballooned hepatocytes, sinusoidal dilatation, and infiltrated leukocytes, was evident in the livers of the malaria-infected mice. Hypoxia was also induced in 30% parasitemia group. With the accumulation of Kupffer cells, inflammation markers, TNF-α, interleukin-1ß, and chemokine (C-X-C motif) ligand 1, were significantly upregulated in the infected group compared with the control group. Expression of fibrotic markers, including transforming growth factor-ß, α-smooth muscle actin (α-SMA), collagen 1a1, thymosin ß4, and vimentin, were significantly higher in the infected groups than in the control group. With increased collagen deposition, hepatic stellate cells expressing α-SMA accumulated in the liver of the PbA-infected mice, whereas those cells were rarely detected in the livers of the control mice. The levels of Hh signaling and Yes-associated protein (YAP), two key regulators for hepatic fibrogenesis, were significantly elevated in the infected groups compared with the control group. Treatment of mice with Hh inhibitor, GDC-0449, reduced hepatic inflammation and fibrogenesis with Hh suppression in PbA-infected mice. CONCLUSION: Our results demonstrate that HSCs are activated in and Hh and YAP signaling are associated with this process, contributing to increased hepatic fibrosis in malaria-infected livers.


Subject(s)
Hedgehog Proteins/metabolism , Liver/metabolism , Plasmodium berghei/pathogenicity , Signal Transduction/physiology , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Anilides/therapeutic use , Animals , Cell Cycle Proteins , Chemokines, C/metabolism , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Hedgehog Proteins/antagonists & inhibitors , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Liver/parasitology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Malaria/parasitology , Malaria/pathology , Malaria/veterinary , Male , Mice , Mice, Inbred C57BL , Phosphoproteins/metabolism , Plasmodium berghei/growth & development , Pyridines/therapeutic use , Thymosin/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation , Vimentin/metabolism , YAP-Signaling Proteins
14.
Malar J ; 17(1): 169, 2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29665817

ABSTRACT

BACKGROUND: Alterations in inflammatory cytokines and genetic background of the host contribute to the outcome of malaria infection. Despite the promising protective role of IL-17 in infections, little attention is given to further understand its importance in the pathogenesis of severe malaria anaemia in chronic/endemic situations. The objective of this study, therefore, was to evaluate IL-17 levels in anaemic condition and its association with host genetic factors. METHODS: Two mice strains (Balb/c and CBA) were crossed to get the F1 progeny, and were (F1, Balb/c, CBA) taken through 6 cycles of Plasmodium berghei (ANKA strain) infection and chloroquine/pyrimethamine treatment to generate semi-immune status. Cytokine levels and kinetics of antibody production, CD4+CD25+T regulatory cells were evaluated by bead-based multiplex assay kit, ELISA and FACs, respectively. RESULTS: High survival with high Hb loss at significantly low parasitaemia was observed in Balb/c and F1. Furthermore, IgG levels were two times higher in Balb/c, F1 than CBA. While CD4+CD25+ Treg cells were lower in CBA; IL-4, IFN-γ, IL-12α and IL-17 were significantly higher (p < 0.05) in Balb/c, F1. CONCLUSIONS: In conclusion, elevated IL-17 levels together with high IL-4, IL-12α and IFN-γ levels may be a marker of protection, and the mechanism may be controlled by host factor (s). Further studies of F2 between the F1 and Balb/c will be informative in evaluating if these genes are segregated or further apart.


Subject(s)
Adaptive Immunity/immunology , Anemia/immunology , Interleukin-17/genetics , Malaria/immunology , Plasmodium berghei/physiology , Adaptive Immunity/genetics , Anemia/genetics , Anemia/parasitology , Animals , Female , Interleukin-17/metabolism , Malaria/complications , Malaria/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred CBA
15.
Malar J ; 17(1): 68, 2018 Feb 05.
Article in English | MEDLINE | ID: mdl-29402267

ABSTRACT

BACKGROUND: Plasmodium falciparum malaria is still one of the most deadly pathology worldwide. Efficient treatment is jeopardized by parasite resistance to artemisinin and its derivatives, and by poor access to treatment in endemic regions. Anti-malarial traditional remedies still offer new tracks for identifying promising antiplasmodial molecules, and a way to ensure that all people have access to care. The present study aims to validate the traditional use of Terminalia macroptera, a Malian plant used in traditional medicine. METHODS: Terminalia macroptera was collected in Mali. Leaves (TML) and roots ethanolic extracts (TMR) were prepared and tested at 2000 mg/kg for in vivo acute toxicity in Albino Swiss mice. Antiplasmodial activity of the extracts was assessed against a chloroquine resistant strain P. falciparum (FcB1) in vitro. In vivo, anti-malarial efficacy was assessed by a 4-day suppressive test at 100 mg/kg in two malaria murine models of uncomplicated malaria (Plasmodium chabaudi chabaudi infection) and cerebral malaria (Plasmodium berghei strain ANKA infection). Constituents of TMR were characterized by ultra-high-performance liquid chromatography coupled to high resolution mass spectrometry. Top ranked compounds were putatively identified using plant databases and in silico fragmentation pattern. RESULTS: Lethal dose of TML and TMR were greater than 2000 mg/kg in Albino Swiss mice. According to the OECD's Globally Harmonized System of Classification, both extracts are non-toxic orally. Antiplasmodial activity of T. macroptera extracts was confirmed in vitro against P. falciparum FcB1 strain with IC50 values of 1.2 and 1.6 µg/mL for TML and TMR, respectively. In vivo, oral administration of TML and TMR induced significant reduction of parasitaemia (37.2 and 46.4% respectively) in P. chabaudi chabaudi infected mice at the 7th day of infection compared to untreated mice. In the cerebral malaria experimental model, mice treated with TMR and TML presented respectively 50 and 66.7% survival rates at day 9 post-infection when all untreated mice died. Eleven major compounds were found in TMR. Among them, several molecules already known could be responsible for the antiplasmodial activity of the roots extract of T. macroptera. CONCLUSIONS: This study confirms both safety and anti-malarial activity of T. macroptera, thus validating its traditional use.


Subject(s)
Antimalarials/pharmacology , Plasmodium berghei/drug effects , Plasmodium chabaudi/drug effects , Terminalia/chemistry , Animals , Female , Mali , Medicine, Traditional , Mice , Plant Extracts/pharmacology , Plant Leaves/chemistry , Plant Roots/chemistry , Plants, Medicinal , Toxicity Tests, Acute
16.
Infect Immun ; 85(9)2017 09.
Article in English | MEDLINE | ID: mdl-28674030

ABSTRACT

Artesunate remains the mainstay of treatment for cerebral malaria, but it is less effective in later stages of disease when the host inflammatory response and blood-brain barrier integrity dictate clinical outcomes. Nitric oxide (NO) is an important regulator of inflammation and microvascular integrity, and impaired NO bioactivity is associated with fatal outcomes in malaria. Endogenous NO bioactivity in mammals is largely mediated by S-nitrosothiols (SNOs). Based on these observations, we hypothesized that animals deficient in the SNO-metabolizing enzyme, S-nitrosoglutathione reductase (GSNOR), which exhibit enhanced S-nitrosylation, would have improved outcomes in a preclinical model of cerebral malaria. GSNOR knockout (KO) mice infected with Plasmodium berghei ANKA had significantly delayed mortality compared to WT animals (P < 0.0001), despite higher parasite burdens (P < 0.01), and displayed markedly enhanced survival versus the wild type (WT) when treated with the antimalarial drug artesunate (77% versus 38%; P < 0.001). Improved survival was associated with higher levels of protein-bound NO, decreased levels of CD4+ and CD8+ T cells in the brain, improved blood-brain barrier integrity, and improved coma scores, as well as higher levels of gamma interferon. GSNOR KO animals receiving WT bone marrow had significantly reduced survival following P. berghei ANKA infection compared to those receiving KO bone barrow (P < 0.001). Reciprocal transplants established that survival benefits of GSNOR deletion were attributable primarily to the T cell compartment. These data indicate a role for GSNOR in the host response to malaria infection and suggest that strategies to disrupt its activity will improve clinical outcomes by enhancing microvascular integrity and modulating T cell tissue tropism.


Subject(s)
Alcohol Dehydrogenase/deficiency , Malaria, Cerebral/pathology , Plasmodium berghei/pathogenicity , Animals , Antimalarials/administration & dosage , Artemisinins/administration & dosage , Artesunate , Disease Models, Animal , Female , Malaria, Cerebral/drug therapy , Male , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Survival Analysis , T-Lymphocytes/immunology , Treatment Outcome
17.
Cytokine ; 99: 249-259, 2017 11.
Article in English | MEDLINE | ID: mdl-28803696

ABSTRACT

Modulation of pro-inflammatory and anti-inflammatory axis and orientation of glial cell function towards neuroinflammation, were hallmark signs of cerebral malaria (CM). CM pathogenesis was concerned with the circulating levels of Interleukin 6 (IL 6) and Transforming growth factor ß (TGF ß). Definite roles of these two cytokines in brain related pathology remained largely unexplored. To study the effect of these two cytokines, we have examined changes in morphology and in activation profile of the glial cells after TGF ß and IL 6 neutralization during CM in cortex and cerebellum of the Plasmodium berghei ANKA (PbA) infected male swiss albino mice. PbA infection caused severe inflammation by inducing changes in morphological features as well as in activation profile of the astrocytes and microglia. Similar inflammatory signs were evident in Anti TGF ß treated set. Interestingly in the Anti IL 6 treated set, reduced level of activation of these glial cells corresponds to the reduced level of inflammatory profile. Microglial activation was found to be synchronous with TLR4 engagement. Neuronal death was triggered by neuroinflammatory milieu seen in PbA and PbA+Anti TGF ß treated set. In conclusion, it can be said that IL 6 and TGF ß perform essential role in CM pathogenesis by modulating the level of glial cell induced neuroinflammation.


Subject(s)
Brain/pathology , Inflammation/pathology , Interleukin-6/metabolism , Malaria, Cerebral/pathology , Neuroglia/metabolism , Plasmodium berghei/physiology , Transforming Growth Factor beta/metabolism , Animals , Apoptosis , Astrocytes/metabolism , Biomarkers/metabolism , CD11b Antigen/metabolism , Cell Aggregation , DNA-Binding Proteins , Glial Fibrillary Acidic Protein/metabolism , Inflammation Mediators/metabolism , Malaria, Cerebral/metabolism , Male , Mice , Nerve Tissue Proteins/metabolism , Neuroglia/pathology , Nuclear Proteins/metabolism , Silver Staining , Toll-Like Receptor 4/metabolism
18.
Malar J ; 16(1): 25, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28086782

ABSTRACT

BACKGROUND: Malaria is one of the most life-threatening health problems worldwide and treatment has been compromised by drug resistance. Identifying lead molecules from natural products might help to find better anti-malarial drugs, since those obtained from natural sources are still effective against malarial parasites. This study aimed at investigating the in vivo antiplasmodial activity of crude extract of the leaves of Ajuga remota together with its safety in mice models. METHODS: In vivo parasite growth inhibitory effect of crude extract was assessed in mice inoculated with Plasmodium berghei (ANKA strain). The in vivo antiplasmodial activity of the test extract was performed against early infection (4-day suppressive test), curative effect against established infection and prophylactic effect against residual infection. Acute and sub-acute toxicity were carried out according to OECD guidelines. RESULTS: In vivo parasite growth inhibition effect of hydroethanolic crude extract of A. remota was evaluated at 30, 50 and 100 mg/kg dose levels. It suppressed parasitaemia by 77.34% at 100 mg/kg dose level in the 4-day test. In curative and prophylactic potential tests, it suppressed parasitaemia by 66.67 and 59.66% at 100 mg/kg dose level, respectively. In vivo toxicity tests revealed no toxicity. All parasitaemia suppressions were statistically significant at P < 0.05 as compared to the vehicle-treated group. The crude extract also prolonged survival time in a dose dependent manner. CONCLUSIONS: The investigation results suggest that the leave extract of Ajuga remota possesses antimalarial activity.


Subject(s)
Ajuga/chemistry , Antiprotozoal Agents/administration & dosage , Complex Mixtures/administration & dosage , Malaria/drug therapy , Plant Extracts/administration & dosage , Plasmodium berghei/drug effects , Animals , Antiprotozoal Agents/isolation & purification , Antiprotozoal Agents/pharmacology , Complex Mixtures/isolation & purification , Complex Mixtures/pharmacology , Disease Models, Animal , Female , Malaria/parasitology , Malaria/prevention & control , Mice , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Survival Analysis , Treatment Outcome
19.
J Infect Dis ; 214(10): 1547-1556, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27638944

ABSTRACT

Cell-mediated immune responses play important roles in immune protection against Plasmodium infection. However, impaired immunity, such as lymphocyte exhaustion, is a common phenomenon in malaria. T-cell immunoglobulin- and mucin-domain-containing molecule 3 (Tim-3) is an important regulatory molecule in cell-mediated immunity and has been implicated in malaria. In this study, it was found that Tim-3 expression on key populations of lymphocytes was significantly increased in both Plasmodium falciparum-infected patients and Plasmodium berghei ANKA (PbANKA)-infected C57BL/6 mice. Upregulation of Tim-3 led to lymphocyte exhaustion, while blocking Tim-3 signaling with an anti-Tim-3 antibody restored lymphocyte activity in Plasmodium infections. Further, anti-Tim-3 treatment accelerated the parasite clearance and relieved the symptoms of cerebral malaria in PbANKA-infected mice. In conclusion, Tim-3 on immune cells negatively regulates cell-mediated immunity against Plasmodium infection, and blocking Tim-3 signaling enhances sterile immunity and may play a protective role during malarial parasite infections.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/metabolism , Immunity, Cellular , Malaria/immunology , Plasmodium falciparum/immunology , Animals , Healthy Volunteers , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Humans , Male , Mice, Inbred C57BL , Plasmodium berghei/immunology
20.
Eur J Immunol ; 45(5): 1354-65, 2015 May.
Article in English | MEDLINE | ID: mdl-25682948

ABSTRACT

Cerebral malaria, a severe complication of Plasmodium falciparum infection, can be modeled in murine Plasmodium berghei ANKA (PbA) infection. PbA-induced experimental cerebral malaria (ECM) is CD8(+) T-cell mediated, and influenced by TH 1/TH 2 balance. Here, we show that IL-33 expression is increased in brain undergoing ECM and we address the role of the IL-33/ST2 pathway in ECM development. ST2-deficient mice were resistant to PbA-induced neuropathology. They survived >20 days with no ECM neurological sign and a preserved cerebral microcirculation, while WT mice succumbed within 10 days with ECM, brain vascular leakage, distinct microvascular pathology obstruction, and hemorrhages. Parasitemia and brain parasite load were similar in ST2-deficient and WT mice. Protection was accompanied by reduced brain sequestration of activated CD4(+) T cells and perforin(+) CD8(+) T cells. While IFN-γ and T-cell-attracting chemokines CXCL9 and CXCL10 were not affected in the absence of functional ST2 pathway, the local expression of ICAM-1, CXCR3, and LT-α, crucial for ECM development, was strongly reduced, and this may explain the diminished pathogenic T-cell recruitment and resistance to ECM. Therefore, IL-33 is induced in PbA sporozoite infection, and the pathogenic T-cell responses with local microvascular pathology are dependent on IL-33/ST2 signaling, identifying IL-33 as a new actor in ECM development.


Subject(s)
Malaria, Cerebral/etiology , Plasmodium berghei , Receptors, Interleukin/metabolism , Animals , Brain/immunology , Brain/parasitology , Brain/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Female , Inflammation/etiology , Inflammation/immunology , Inflammation/pathology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/metabolism , Lymphocyte Activation , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasmodium berghei/immunology , Plasmodium berghei/pathogenicity , Receptors, Interleukin/deficiency , Receptors, Interleukin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL