Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Life Sci Alliance ; 5(6)2022 06.
Article in English | MEDLINE | ID: mdl-35260473

ABSTRACT

Brain swelling occurs in cerebral malaria (CM) and may either reverse or result in fatal outcome. It is currently unknown how brain swelling in CM reverses, as brain swelling at the acute stage is difficult to study in humans and animal models with reliable induction of reversible edema are not known. In this study, we show that reversible brain swelling in experimental murine CM can be induced reliably after single vaccination with radiation-attenuated sporozoites as proven by in vivo high-field magnetic resonance imaging. Our results provide evidence that brain swelling results from transcellular blood-brain barrier disruption (BBBD), as revealed by electron microscopy. This mechanism enables reversal of brain swelling but does not prevent persistent focal brain damage, evidenced by microhemorrhages, in areas of most severe BBBD. In adult CM patients magnetic resonance imaging demonstrate microhemorrhages in more than one third of patients with reversible edema, emphasizing similarities of the experimental model and human disease. Our data suggest that targeting transcellular BBBD may represent a promising adjunct therapeutic approach to reduce edema and may improve neurological outcome.


Subject(s)
Brain Edema , Malaria, Cerebral , Animals , Blood-Brain Barrier/diagnostic imaging , Brain/diagnostic imaging , Brain/pathology , Brain Edema/diagnostic imaging , Brain Edema/etiology , Brain Edema/pathology , Edema/pathology , Humans , Malaria, Cerebral/pathology , Mice
2.
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
3.
Vaccines (Basel) ; 8(4)2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33316929

ABSTRACT

Clinical and experimental evidence suggests that the tuberculosis vaccine BCG offers protection against unrelated pathogens including the malaria parasite. Cerebral malaria (CM) is the most severe complication associated with Plasmodium falciparum infection in humans and is responsible for most of the fatalities attributed to malaria. We investigated whether BCG protected C57BL/6 mice from P. berghei ANKA (PbA)-induced experimental CM (ECM). The majority of PbA-infected mice that were immunized with BCG showed prolonged survival without developing clinical symptoms of ECM. However, this protective effect waned over time and was associated with the recovery of viable BCG from liver and spleen. Intriguingly, BCG-mediated protection from ECM was not associated with a reduction in parasite burden, indicating that BCG immunization did not improve anti-parasite effector mechanisms. Instead, we found a significant reduction in pro-inflammatory mediators and CD8+ T cells in brains of BCG-vaccinated mice. Together these data suggest that brain recruitment of immune cells involved in the pathogenesis of ECM decreased after BCG vaccination. Understanding the mechanisms underlying the protective effects of BCG on PbA-induced ECM can provide a rationale for developing effective adjunctive therapies to reduce the risk of death and brain damage in CM.

4.
Amino Acids ; 52(5): 693-710, 2020 May.
Article in English | MEDLINE | ID: mdl-32367435

ABSTRACT

In this study, a determination of Troponin I and creatine kinase activity in whole-blood samples in a cohort of 100 small infants in the age of 2-5 years from Uganda with complicated Plasmodium falciparum malaria suggests the prevalence of cardiac symptoms in comparison to non-infected, healthy patients. Troponin I and creatine kinase activity increased during infection. Different reports showed that complicated malaria coincides with hypoxia in children. The obtained clinical data prompted us to further elucidate the underlying regulatory mechanisms of cardiac involvement in human cardiac ventricular myocytes. Complicated malaria is the most common clinical presentation and might induce cardiac impairment by hypoxia. Eukaryotic initiation factor 5A (eIF-5A) is involved in hypoxia induced factor (HIF-1α) expression. EIF-5A is a protein posttranslationally modified by hypusination involving catalysis of the two enzymes deoxyhypusine synthase (DHS) and deoxyhypusine hydroxylase. Treatment of human cardiomyocytes with GC7, an inhibitor of DHS, catalyzing the first step in hypusine biosynthesis led to a decrease in proinflammatory and proapoptotic myocardial caspase-1 activity in comparison to untreated cardiomyocytes. This effect was even more pronounced after co-administration of GC7 and GPI from P. falciparum simulating the pathology of severe malaria. Moreover, in comparison to untreated and GC7-treated cardiomyocytes, co-administration of GC7 and GPI significantly decreased the release of cytochrome C and lactate from damaged mitochondria. In sum, coadministration of GC7 prevented cardiac damage driven by hypoxia in vitro. Our approach demonstrates the potential of the pharmacological inhibitor GC7 to ameliorate apoptosis in cardiomyocytes in an in vitro model simulating severe malaria. This regulatory mechanism is based on blocking EIF-5A hypusination.


Subject(s)
Apoptosis , Malaria/pathology , Myocytes, Cardiac/pathology , Parasitemia/pathology , Peptide Initiation Factors/metabolism , Plasmodium berghei/isolation & purification , RNA-Binding Proteins/metabolism , Animals , Child, Preschool , Female , Humans , Infant , Malaria/metabolism , Malaria/parasitology , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/parasitology , Parasitemia/metabolism , Parasitemia/parasitology , Peptide Initiation Factors/genetics , RNA-Binding Proteins/genetics , Eukaryotic Translation Initiation Factor 5A
5.
Nucleic Acids Res ; 48(1): e2, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31680162

ABSTRACT

The lack of endogenous RNAi machinery in the malaria parasite Plasmodium hampers gene annotation and hence antimalarial drug and vaccine development. Here, we engineered rodent Plasmodium berghei to express a minimal, non-canonical RNAi machinery that solely requires Argonaute 2 (Ago2) and a modified short hairpin RNA, so-called AgoshRNA. Using this strategy, we achieved robust and specific gene knockdown throughout the entire parasite life cycle. We also successfully silenced the endogenous gene perforin-like protein 2, phenocopying a full gene knockout. Transcriptionally restricting Ago2 expression to the liver stage further enabled us to perform a stage-specific gene knockout. The RNAi-competent Plasmodium lines reported here will be a valuable resource for loss-of-function phenotyping of the many uncharacterized genes of Plasmodium in low or high throughput, without the need to engineer the target gene locus. Thereby, our new strategy and transgenic Plasmodium lines will ultimately benefit the discovery of urgently needed antimalarial drug and vaccine candidates. Generally, the ability to render RNAi-negative organisms RNAi-competent by mere introduction of two components, Ago2 and AgoshRNA, is a unique paradigm that should find broad applicability in other species.


Subject(s)
Argonaute Proteins/genetics , Genetic Engineering/methods , Plasmodium berghei/genetics , Protozoan Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Animals , Anopheles/parasitology , Argonaute Proteins/metabolism , Female , Genes, Reporter , Green Fluorescent Proteins/antagonists & inhibitors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Life Cycle Stages/genetics , Mice , Mice, Inbred C57BL , Mosquito Vectors/parasitology , Organisms, Genetically Modified , Perforin/genetics , Perforin/metabolism , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/metabolism , RNA, Small Interfering/metabolism , Transgenes
6.
Immunology ; 159(2): 193-204, 2020 02.
Article in English | MEDLINE | ID: mdl-31631339

ABSTRACT

Excessive inflammatory immune responses during infections with Plasmodium parasites are responsible for severe complications such as cerebral malaria (CM) that can be studied experimentally in mice. Dendritic cells (DCs) activate cytotoxic CD8+ T-cells and initiate immune responses against the parasites. Batf3-/- mice lack a DC subset, which efficiently induces strong CD8 T-cell responses by cross-presentation of exogenous antigens. Here we show that Batf3-/- mice infected with Plasmodium berghei ANKA (PbA) were protected from experimental CM (ECM), characterized by a stable blood-brain barrier (BBB) and significantly less infiltrated peripheral immune cells in the brain. Importantly, the absence of ECM in Batf3-/- mice correlated with attenuated responses of cytotoxic T-cells, as their parasite-specific lytic activity as well as the production of interferon gamma and granzyme B were significantly decreased. Remarkably, spleens of ECM-protected Batf3-/- mice had elevated levels of regulatory immune cells and interleukin 10. Thus, protection from ECM in PbA-infected Batf3-/- mice was associated with the absence of strong CD8+ T-cell activity and induction of immunoregulatory mediators and cells.


Subject(s)
Basic-Leucine Zipper Transcription Factors/deficiency , Brain/immunology , Dendritic Cells/immunology , Malaria, Cerebral/prevention & control , Plasmodium berghei/pathogenicity , Repressor Proteins/deficiency , T-Lymphocytes, Cytotoxic/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Blood-Brain Barrier/immunology , Blood-Brain Barrier/parasitology , Brain/metabolism , Brain/parasitology , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/parasitology , Disease Models, Animal , Female , Granzymes/immunology , Granzymes/metabolism , Host-Parasite Interactions , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Malaria, Cerebral/immunology , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Mice, Inbred C57BL , Mice, Knockout , Plasmodium berghei/immunology , Repressor Proteins/genetics , Spleen/immunology , Spleen/metabolism , Spleen/parasitology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/parasitology
7.
Cell Microbiol ; 21(10): e13088, 2019 10.
Article in English | MEDLINE | ID: mdl-31364224

ABSTRACT

Intracellular Plasmodium parasites develop inside a parasitophorous vacuole (PV), a specialised compartment enclosed by a membrane (PVM) that contains proteins of both host and parasite origin. Although exported protein 1 (EXP1) is one of the earliest described parasitic PVM proteins, its function throughout the Plasmodium life cycle remains insufficiently understood. Here, we show that whereas the N-terminus of Plasmodium berghei EXP1 (PbEXP1) is essential for parasite survival in the blood, parasites lacking PbEXP1's entire C-terminal (CT) domain replicate normally in the blood but cause less severe pathology than their wild-type counterparts. Moreover, truncation of PbEXP1's CT domain not only impairs parasite development in the mosquito but also abrogates PbEXP1 localization to the PVM of intrahepatic parasites, severely limiting their replication and preventing their egress into the blood. Our findings highlight the importance of EXP1 during the Plasmodium life cycle and identify this protein as a promising target for antiplasmodial intervention.


Subject(s)
Culicidae/parasitology , Liver/parasitology , Plasmodium berghei/genetics , Protein Domains/genetics , Protozoan Proteins/genetics , Animals , Cell Line, Tumor , Erythrocytes/parasitology , Female , Humans , Intracellular Membranes/metabolism , Intracellular Membranes/parasitology , Life Cycle Stages/genetics , Liver/metabolism , Mice , Mice, Inbred C57BL , Plasmodium berghei/growth & development , Plasmodium berghei/pathogenicity , Protozoan Proteins/metabolism , Vacuoles/metabolism , Vacuoles/parasitology
8.
Radiology ; 290(2): 359-367, 2019 02.
Article in English | MEDLINE | ID: mdl-30615566

ABSTRACT

Purpose To investigate the association of inflammation and brain edema in a cerebral malaria (CM) mouse model with a combination of bis-5-hydroxy-tryptamide-diethylenetriaminepentaacetate gadolinium, referred to as MPO-Gd, and cross-linked iron oxide nanoparticle (CLIO-NP) imaging. Materials and Methods Female wild-type (n = 23) and myeloperoxidase (MPO) knock-out (n = 5) mice were infected with the Plasmodium berghei ANKA strain from May 2016 to July 2018. Seven healthy mice served as control animals. At a Rapid Murine Coma and Behavioral Scale (RMCBS) score of less than 15, mice underwent MRI at 9.4 T and received gadodiamide, MPO-Gd, or CLIO-NPs. T1-weighted MRI was used to assess MPO activity, and T2*-weighted MRI was used to track CLIO-NPs. Immunofluorescent staining and flow cytometric analyses characterized CLIO-NPs, MPO, endothelial cells, and leukocytes. An unpaired, two-tailed Student t test was used to compare groups; Spearman correlation analysis was used to determine the relationship of imaging parameters to clinical severity. Results MPO-Gd enhancement occurred in inflammatory CM hotspots (olfactory bulb > rostral migratory stream > brainstem > cortex, P < .05 for all regions compared with control mice; mean olfactory bulb signal intensity ratio: 1.40 ± 0.07 vs 0.96 ± 0.01, P < .01). The enhancement was reduced in MPO knockout mice (mean signal intensity ratio at 60 minutes: 1.13 ± 0.04 vs 1.40 ± 0.07 in CM, P < .05). Blood-brain barrier compromise was suggested by parenchymal gadolinium enhancement, leukocyte recruitment, and endothelial activation. CLIO-NPs accumulated mainly intravascularly and at the vascular endothelium. CLIO-NPs were also found in the choroid plexus, indicating inflammation of the ventricular system. Blood-cerebrospinal fluid barrier breakdown showed correlation with brain swelling (r2: 0.55, P < .01) and RMCBS score (r2: 0.75, P < .001). Conclusion Iron oxide nanoparticle imaging showed strong inflammatory involvement of the microvasculature in a murine model of cerebral malaria. Furthermore, bis-5-hydroxy-tryptamide-diethylenetriaminepentaacetate gadolinium imaging depicted parenchymal and intraventricular inflammation. This combined molecular imaging approach links vascular inflammation to breakdown of the blood-brain barrier and blood-cerebrospinal fluid barrier that correlate with global brain edema and disease severity. © RSNA, 2018 Online supplemental material is available for this article. See also the editorial by Kiessling in this issue.


Subject(s)
Brain Edema , Encephalitis , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/chemistry , Malaria, Cerebral , Peroxidase/metabolism , Animals , Brain/diagnostic imaging , Brain/enzymology , Brain/pathology , Brain Edema/diagnostic imaging , Brain Edema/enzymology , Brain Edema/parasitology , Brain Edema/pathology , Disease Models, Animal , Encephalitis/diagnostic imaging , Encephalitis/enzymology , Encephalitis/parasitology , Encephalitis/pathology , Female , Malaria, Cerebral/complications , Malaria, Cerebral/diagnostic imaging , Malaria, Cerebral/enzymology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
Cell Microbiol ; 21(5): e12999, 2019 05.
Article in English | MEDLINE | ID: mdl-30597708

ABSTRACT

Ferlins mediate calcium-dependent vesicular fusion. Although conserved throughout eukaryotic evolution, their function in unicellular organisms including apicomplexan parasites is largely unknown. Here, we define a crucial role for a ferlin-like protein (FLP) in host-to-vector transmission of the rodent malaria parasite Plasmodium berghei. Infection of the mosquito vectors requires the formation of free gametes and their fertilisation in the mosquito midgut. Mature gametes will only emerge upon secretion of factors that stimulate the disruption of the red blood cell membrane and the parasitophorous vacuole membrane. Genetic depletion of FLP in sexual stages leads to a complete life cycle arrest in the mosquito. Although mature gametes form normally, mutants lacking FLP remain trapped in the red blood cell. The egress defect is rescued by detergent-mediated membrane lysis. In agreement with ferlin vesicular localisation, HA-tagged FLP labels intracellular speckles, which relocalise to the cell periphery during gamete maturation. Our data define FLP as a novel critical factor for Plasmodium fertilisation and transmission and suggest an evolutionarily conserved example of ferlin-mediated exocytosis.


Subject(s)
Erythrocyte Membrane/metabolism , Erythrocytes/metabolism , Germ Cells/metabolism , Malaria/transmission , Plasmodium berghei/growth & development , Protozoan Proteins/metabolism , Animals , Culicidae/parasitology , Detergents/pharmacology , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/genetics , Erythrocyte Membrane/parasitology , Erythrocytes/drug effects , Erythrocytes/parasitology , Exocytosis/genetics , Female , Germ Cells/cytology , Germ Cells/growth & development , Germ Cells/ultrastructure , Host-Pathogen Interactions , Life Cycle Stages/genetics , Malaria/genetics , Malaria/metabolism , Malaria/parasitology , Mice , Mice, Inbred C57BL , Mosquito Vectors/genetics , Mosquito Vectors/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/pathogenicity , Protein Domains/genetics , Protozoan Proteins/genetics
10.
Front Immunol ; 9: 1875, 2018.
Article in English | MEDLINE | ID: mdl-30154793

ABSTRACT

Cerebral malaria is a complex neurological syndrome caused by an infection with Plasmodium falciparum parasites and is exclusively attributed to a series of host-parasite interactions at the pathological blood-stage of infection. In contrast, the preceding intra-hepatic phase of replication is generally considered clinically silent and thereby excluded from playing any role in the development of neurological symptoms. In this study, however, we present an antigen PbmaLS_05 that is presented to the host immune system by both pre-erythrocytic and intra-erythrocytic stages and contributes to the development of cerebral malaria in mice. Although deletion of the endogenous PbmaLS_05 prevented the development of experimental cerebral malaria (ECM) in susceptible mice after both sporozoite and infected red blood cell (iRBC) infections, we observed significant differences in contribution of the host immune response between both modes of inoculation. Moreover, PbmaLS_05-specific CD8+ T cells contributed to the development of ECM after sporozoite but not iRBC-infection, suggesting that pre-erythrocytic antigens like PbmaLS_05 can also contribute to the development of cerebral symptoms. Our data thus highlight the importance of the natural route of infection in the study of ECM, with potential implications for vaccine and therapeutic strategies against malaria.


Subject(s)
Antigens, Protozoan/immunology , Disease Susceptibility , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Plasmodium berghei/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cross-Priming/immunology , Disease Models, Animal , Gene Expression , Genes, Protozoan , Genes, Reporter , Life Cycle Stages , Magnetic Resonance Imaging , Malaria, Cerebral/diagnosis , Malaria, Cerebral/pathology , Mice , Plasmodium berghei/genetics , Plasmodium berghei/growth & development
11.
Front Immunol ; 9: 1137, 2018.
Article in English | MEDLINE | ID: mdl-29892289

ABSTRACT

Whole sporozoite vaccines represent one of the most promising strategies to induce protection against malaria. However, the development of efficient vaccination protocols still remains a major challenge. To understand how the generation of immunity is affected by variations in vaccination dosage and frequency, we systematically analyzed intrasplenic and intrahepatic CD8+ T cell responses following varied immunizations of mice with radiation-attenuated sporozoites. By combining experimental data and mathematical modeling, our analysis indicates a reversing role of spleen and liver in the generation of protective liver-resident CD8+ T cells during priming and booster injections: While the spleen acts as a critical source compartment during priming, the increase in vaccine-induced hepatic T cell levels is likely due to local reactivation in the liver in response to subsequent booster injections. Higher dosing accelerates the efficient generation of liver-resident CD8+ T cells by especially affecting their local reactivation. In addition, we determine the differentiation and migration pathway from splenic precursors toward hepatic memory cells thereby presenting a mechanistic framework for the impact of various vaccination protocols on these dynamics. Thus, our work provides important insights into organ-specific CD8+ T cell dynamics and their role and interplay in the formation of protective immunity against malaria.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Malaria/immunology , Malaria/parasitology , Plasmodium/immunology , Plasmodium/radiation effects , Sporozoites/immunology , Sporozoites/radiation effects , Algorithms , Animals , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/metabolism , Female , Host-Pathogen Interactions/immunology , Immunization , Immunologic Memory , Immunophenotyping , Liver/immunology , Liver/parasitology , Lymphocyte Count , Malaria/prevention & control , Mice , Models, Biological , Models, Theoretical , Organ Specificity/immunology , Plasmodium berghei/immunology , Spleen/immunology , Spleen/parasitology , Vaccination
12.
Front Microbiol ; 9: 166, 2018.
Article in English | MEDLINE | ID: mdl-29515528

ABSTRACT

The blood-stage of the Plasmodium parasite is one of the key phases within its life cycle that influences disease progression during a malaria infection. The efficiency of the parasite in infecting red blood cells (RBC) determines parasite load and parasite-induced hemolysis that is responsible for the development of anemia and potentially drives severe disease progression. However, the molecular factors defining the infectivity of Plasmodium parasites have not been completely identified so far. Using the Plasmodium berghei mouse model for malaria, we characterized and compared the blood-stage infection dynamics of PbANKA WT and a mutant parasite strain lacking a novel Plasmodium antigen, PbmaLS_05, that is well conserved in both human and animal Plasmodium parasite strains. Infection of mice with parasites lacking PbmaLS_05 leads to lower parasitemia levels and less severe disease progression in contrast to mice infected with the wildtype PbANKA strain. To specifically determine the effect of deleting PbmaLS_05 on parasite infectivity we developed a mathematical model describing erythropoiesis and malarial infection of RBC. By applying our model to experimental data studying infection dynamics under normal and drug-induced altered erythropoietic conditions, we found that both PbANKA and PbmaLS_05 (-) parasite strains differed in their infectivity potential during the early intra-erythrocytic stage of infection. Parasites lacking PbmaLS_05 showed a decreased ability to infect RBC, and immature reticulocytes in particular that are usually a preferential target of the parasite. These altered infectivity characteristics limit parasite burden and affect disease progression. Our integrative analysis combining mathematical models and experimental data suggests that deletion of PbmaLS_05 affects productive infection of reticulocytes, which makes this antigen a useful target to analyze the actual processes relating RBC preferences to the development of severe disease outcomes in malaria.

13.
Sci Rep ; 8(1): 3085, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29449638

ABSTRACT

Cerebral malaria is a life-threatening complication of Plasmodia infection and a major cause of child mortality in Sub-Saharan Africa. We report that protection from experimental cerebral malaria in the rodent model is obtained by a single intravenous or subcutaneous whole-parasite immunization. Whole-parasite immunization with radiation-attenuated sporozoites was equally protective as immunization with non-attenuated sporozoites under chemoprophylaxis. Both immunization regimens delayed the development of blood-stage parasites, but differences in cellular and humoral immune mechanisms were observed. Single-dose whole-parasite vaccination might serve as a relatively simple and feasible immunization approach to prevent life-threatening cerebral malaria.


Subject(s)
Malaria Vaccines/administration & dosage , Malaria, Cerebral/prevention & control , Malaria, Cerebral/parasitology , Plasmodium berghei/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Injections, Intravenous , Injections, Subcutaneous , Malaria Vaccines/immunology , Malaria, Cerebral/immunology , Mice , Mice, Inbred C57BL , Sporozoites/immunology
14.
Int J Med Microbiol ; 308(1): 107-117, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28964681

ABSTRACT

The intracellular development and differentiation of the Plasmodium parasite in the host liver is a prerequisite for the actual onset of malaria disease pathology. Since liver-stage infection is clinically silent and can be completely eliminated by sterilizing immune responses, it is a promising target for urgently needed innovative antimalarial drugs and/or vaccines. Discovered more than 65 years ago, these stages remain poorly understood regarding their molecular repertoire and interaction with their host cells in comparison to the pathogenic erythrocytic stages. The differentiating and replicative intrahepatic parasite resides in a membranous compartment called the parasitophorous vacuole, separating it from the host-cell cytoplasm. Here we outline seminal work that contributed to our present understanding of the fundamental dynamic cellular processes of the intrahepatic malarial parasite with both specific host-cell factors and compartments.


Subject(s)
Host-Parasite Interactions , Liver/parasitology , Plasmodium/growth & development , Vacuoles/parasitology , Animals , Hepatocytes/metabolism , Hepatocytes/parasitology , Humans , Intracellular Membranes/metabolism , Malaria/parasitology , Plasmodium/metabolism , Protozoan Proteins/metabolism , Vacuoles/metabolism
15.
J Vis Exp ; (124)2017 06 08.
Article in English | MEDLINE | ID: mdl-28654030

ABSTRACT

Cerebral malaria is a sign of severe malarial disease and is often a harbinger of death. While aggressive management can be life-saving, the detection of cerebral malaria can be difficult. We present an experimental mouse model of cerebral malaria that shares multiple features of the human disease, including edema and microvascular pathology. Using magnetic resonance imaging (MRI), we can detect and track the blood-brain barrier disruption, edema development, and subsequent brain swelling. We describe multiple MRI techniques that can visualize these pertinent pathological changes. Thus, we show that MRI represents a valuable tool to visualize and track pathological changes, such as edema, brain swelling, and microvascular pathology, in vivo.


Subject(s)
Edema/diagnostic imaging , Magnetic Resonance Imaging/methods , Malaria, Cerebral/diagnostic imaging , Animals , Brain/blood supply , Disease Models, Animal , Edema/pathology , Humans , Malaria, Cerebral/pathology , Mice
16.
Proc Natl Acad Sci U S A ; 114(7): E1138-E1147, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28137845

ABSTRACT

The first, obligatory replication phase of malaria parasite infections is characterized by rapid expansion and differentiation of single parasites in liver cells, resulting in the formation and release of thousands of invasive merozoites into the bloodstream. Hepatic Plasmodium development occurs inside a specialized membranous compartment termed the parasitophorous vacuole (PV). Here, we show that, during the parasite's hepatic replication, the C-terminal region of the parasitic PV membrane protein exported protein 1 (EXP-1) binds to host Apolipoprotein H (ApoH) and that this molecular interaction plays a pivotal role for successful Plasmodium liver-stage development. Expression of a truncated EXP-1 protein, missing the specific ApoH interaction site, or down-regulation of ApoH expression in either hepatic cells or mouse livers by RNA interference resulted in impaired intrahepatic development. Furthermore, infection of mice with sporozoites expressing a truncated version of EXP-1 resulted in both a significant reduction of liver burden and delayed blood-stage patency, leading to a disease outcome different from that generally induced by infection with wild-type parasites. This study identifies a host-parasite protein interaction during the hepatic stage of infection by Plasmodium parasites. The identification of such vital interactions may hold potential toward the development of novel malaria prevention strategies.


Subject(s)
Liver/parasitology , Malaria/parasitology , Membrane Proteins/metabolism , Plasmodium berghei/physiology , Protozoan Proteins/metabolism , beta 2-Glycoprotein I/metabolism , Animals , Animals, Genetically Modified , Binding Sites , Down-Regulation , Genes, Protozoan , HEK293 Cells , Hepatocytes/parasitology , Humans , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Protein Binding , RNA Interference , RNA, Small Interfering/genetics , Sequence Deletion , Sporozoites/physiology , Vacuoles/parasitology , beta 2-Glycoprotein I/antagonists & inhibitors , beta 2-Glycoprotein I/genetics
17.
Nat Commun ; 7: 13401, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27824335

ABSTRACT

Plasmodium falciparum infections can cause severe malaria, but not every infected person develops life-threatening complications. In particular, carriers of the structural haemoglobinopathies S and C and infants are protected from severe disease. Protection is associated with impaired parasite-induced host actin reorganization, required for vesicular trafficking of parasite-encoded adhesins, and reduced cytoadherence of parasitized erythrocytes in the microvasculature. Here we show that aberrant host actin remodelling and the ensuing reduced cytoadherence result from a redox imbalance inherent to haemoglobinopathic and fetal erythrocytes. We further show that a transient oxidative insult to wild-type erythrocytes before infection with P. falciparum induces the phenotypic features associated with the protective trait of haemoglobinopathic and fetal erythrocytes. Moreover, pretreatment of mice with the pro-oxidative nutritional supplement menadione mitigate the development of experimental cerebral malaria. Our results identify redox imbalance as a causative principle of protection from severe malaria, which might inspire host-directed intervention strategies.


Subject(s)
Anemia, Sickle Cell/blood , Erythrocytes/parasitology , Fetus/pathology , Malaria, Falciparum/pathology , Malaria, Falciparum/parasitology , Oxidative Stress , Actins/metabolism , Animals , Cytoplasm/metabolism , Erythrocytes/ultrastructure , Female , Hemoglobins/metabolism , Mice, Inbred C57BL , Models, Biological , Oxidation-Reduction , Phenotype , Plasmodium berghei/drug effects , Plasmodium berghei/physiology , Plasmodium falciparum/metabolism , Plasmodium falciparum/ultrastructure , Vitamin K 3/pharmacology
18.
FEBS Open Bio ; 6(8): 860-72, 2016 08.
Article in English | MEDLINE | ID: mdl-27516964

ABSTRACT

The biological function of the post-translational modification hypusine in the eukaryotic initiation factor 5A (EIF-5A) in eukaryotes is still not understood. Hypusine is formed by two sequential enzymatic steps at a specific lysine residue in the precursor protein EIF-5A. One important biological function of EIF-5A which was recently identified is the translation of polyproline-rich mRNA, suggesting its biological relevance in a variety of biological processes. Hypusinated eIF-5A controls the proliferation of cancer cells and inflammatory processes in malaria. It was shown that pharmacological inhibition of the enzymes involved in this pathway, deoxyhypusine synthase (DHS) and the deoxyhypusine hydroxylase (DOHH), arrested the growth of malaria parasites. Down-regulation of both the malarial eIF-5A and dhs genes by in vitro and in vivo silencing led to decreased transcript levels and protein expression and, in turn, to low parasitemia, confirming a critical role of hypusination in eIF-5A for proliferation in Plasmodium. To further investigate whether eIF-5A and the activating enzyme DHS are essential for Plasmodium erythrocytic stages, targeted gene disruption was performed in the rodent malaria parasite Plasmodium berghei. Full disruption of both genes was not successful; instead parasites harboring the episome for eIF-5A and dhs genes were obtained, suggesting that these genes may perform vital functions during the pathogenic blood cell stage. Next, a knock-in strategy was pursued for both endogenous genes eIF-5A and dhs from P. berghei. The latter resulted in viable recombinant parasites, strengthening the observation that they might be essential for proliferation during asexual development of the malaria parasite.

19.
PLoS Pathog ; 12(7): e1005710, 2016 07.
Article in English | MEDLINE | ID: mdl-27409081

ABSTRACT

Parasites causing malaria need to migrate in order to penetrate tissue barriers and enter host cells. Here we show that the actin filament-binding protein coronin regulates gliding motility in Plasmodium berghei sporozoites, the highly motile forms of a rodent malaria-causing parasite transmitted by mosquitoes. Parasites lacking coronin show motility defects that impair colonization of the mosquito salivary glands but not migration in the skin, yet result in decreased transmission efficiency. In non-motile sporozoites low calcium concentrations mediate actin-independent coronin localization to the periphery. Engagement of extracellular ligands triggers an intracellular calcium release followed by the actin-dependent relocalization of coronin to the rear and initiation of motility. Mutational analysis and imaging suggest that coronin organizes actin filaments for productive motility. Using coronin-mCherry as a marker for the presence of actin filaments we found that protein kinase A contributes to actin filament disassembly. We finally speculate that calcium and cAMP-mediated signaling regulate a switch from rapid parasite motility to host cell invasion by differentially influencing actin dynamics.


Subject(s)
Actin Cytoskeleton/metabolism , Malaria/parasitology , Microfilament Proteins/metabolism , Plasmodium berghei/metabolism , Sporozoites/metabolism , Animals , Blotting, Western , Culicidae/microbiology , DNA Mutational Analysis , Disease Models, Animal , Hep G2 Cells , Humans , Insect Vectors/microbiology , Mice , Mice, Inbred C57BL , Plasmodium berghei/pathogenicity , Protozoan Proteins/metabolism , Transfection
20.
Sci Rep ; 6: 26824, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27241521

ABSTRACT

During the clinically silent liver stage of a Plasmodium infection the parasite replicates from a single sporozoite into thousands of merozoites. Infection of humans and rodents with large numbers of sporozoites that arrest their development within the liver can cause sterile protection from subsequent infections. Disruption of genes essential for liver stage development of rodent malaria parasites has yielded a number of attenuated parasite strains. A key question to this end is how increased attenuation relates to vaccine efficacy. Here, we generated rodent malaria parasite lines that arrest during liver stage development and probed the impact of multiple gene deletions on attenuation and protective efficacy. In contrast to P. berghei strain ANKA LISP2(-) or uis3(-) single knockout parasites, which occasionally caused breakthrough infections, the double mutant lacking both genes was completely attenuated even when high numbers of sporozoites were administered. However, different vaccination protocols showed that LISP2(-) parasites protected better than uis3(-) and double mutants. Hence, deletion of several genes can yield increased safety but might come at the cost of protective efficacy.


Subject(s)
Liver/parasitology , Malaria Vaccines , Malaria/genetics , Plasmodium berghei/genetics , Animals , Female , Gene Deletion , Malaria/immunology , Malaria/prevention & control , Mice, Inbred C57BL , Plasmodium berghei/immunology , Sporozoites/genetics , Sporozoites/immunology , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...