Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 801111, 2022.
Article in English | MEDLINE | ID: mdl-35734173

ABSTRACT

The skin is the site of host invasion by the mosquito-borne Plasmodium parasite, which caused an estimated 229 million infections and 409,000 deaths in 2019 according to WHO World Malaria report 2020. In our previous studies, we have shown that skin scarification (SS) with a P. falciparum circumsporozoite (CS) peptide in the oil-in-water adjuvant AddaVax containing a combination of TLR 7/8 and TLR 9 agonists can elicit sporozoite neutralizing antibodies. SS with AddaVax + TLR agonists, but not AddaVax alone, elicited CD4+ Th1 cells and IgG2a/c anti-repeat antibody. To explore the innate immune responses that may contribute to development of adaptive immunity following SS, we examined the skin at 4h and 24h post priming with CS peptide in AddaVax with or without TLR agonists. H&E stained and IHC-labeled dorsal skin sections obtained 24h post SS demonstrated a marked difference in the pattern of infiltration with F4/80+, CD11b+ and Ly6G+ cells at the immunization site, with the lowest intensity noted following SS with AddaVax + TLR agonists. Serum collected at 4h post SS, had reproducible increases in IL-6, MIP-3α, IL-22 and IP-10 (CXCL10) following SS with AddaVax + TLR agonists, but not with AddaVax alone. To begin to decipher the complex roles of these pro-inflammatory cytokines/chemokines, we utilized IP-10 deficient (IP-10 -/-) mice to examine the role of this chemokine in the development of anti-repeat antibody response following SS. In the absence of IP-10, the levels of Th1-type IgG2a/c antibody and kinetics of the primary anti-repeat antibody response were reduced following prime and boost. The IP-10 chemokine, present as early as 4h post prime, may provide an early serological marker for rapid screening of adjuvant formulations and delivery platforms to optimize SS-induced humoral immunity to CS repeats as well as other pathogens.


Subject(s)
Antibodies, Protozoan , Immunity, Innate , Malaria, Falciparum , Plasmodium falciparum , Vaccination , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Neutralizing , Chemokine CXCL10 , Immunoglobulin G , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Mice , Protozoan Proteins
3.
Sci Rep ; 6: 32575, 2016 09 14.
Article in English | MEDLINE | ID: mdl-27624667

ABSTRACT

Malaria eradication will require a combination of vector control, chemotherapy and an easily administered vaccine. Sterile immunity can be elicited in humans by immunization with sporozoites, the infective stage injected by bite of the mosquito vector, however, whole parasite vaccines present formidable logistical challenges for production, storage and administration. The "gold standard" for infectious disease eradiation, the Smallpox Eradication Programme, utilized mass immunization using the skin scarification (SS) route. SS may more closely mimic the natural route of malaria infection initiated by sporozoites injected by mosquito bite which elicits both neutralizing antibodies and protective cell mediated immunity. We investigated the potential of SS immunization using a malaria repeat peptide containing a protective B cell epitope of Plasmodium falciparum, the most lethal human species, and delivery vehicles containing TLR agonists as adjuvants. In a murine model, SS immunization with peptide in combination with TLR-7/8 and -9 agonists elicited high levels of systemic sporozoite neutralizing antibody, Th1- type CD4+ T cells and resistance to challenge by bites of infected mosquitoes. SS provides the potential to elicit humoral immunity to target Plasmodium at multiple stages of its complex life cycle.


Subject(s)
Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Plasmodium falciparum/drug effects , Sporozoites/drug effects , Adjuvants, Immunologic/administration & dosage , Animals , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Humans , Immunization , Life Cycle Stages/drug effects , Life Cycle Stages/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mice , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/immunology , Sporozoites/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
5.
Front Microbiol ; 6: 482, 2015.
Article in English | MEDLINE | ID: mdl-26074888

ABSTRACT

Plasmodium falciparum malaria remains one of the most serious health problems globally. Immunization with attenuated parasites elicits multiple cellular effector mechanisms capable of eliminating Plasmodium liver stages. However, malaria liver stage (LS) immunity is complex and the mechanisms effector T cells use to locate the few infected hepatocytes in the large liver in order to kill the intracellular LS parasites remain a mystery to date. Here, we review our current knowledge on the behavior of CD8 effector T cells in the hepatic microvasculature, in malaria and other hepatic infections. Taking into account the unique immunological and lymphogenic properties of the liver, we discuss whether classical granule-mediated cytotoxicity might eliminate infected hepatocytes via direct cell contact or whether cytokines might operate without cell-cell contact and kill Plasmodium LSs at a distance. A thorough understanding of the cellular effector mechanisms that lead to parasite death hence sterile protection is a prerequisite for the development of a successful malaria vaccine to protect the 40% of the world's population currently at risk of Plasmodium infection.

6.
PLoS Pathog ; 10(12): e1004528, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25474413

ABSTRACT

Cerebral malaria claims the lives of over 600,000 African children every year. To better understand the pathogenesis of this devastating disease, we compared the cellular dynamics in the cortical microvasculature between two infection models, Plasmodium berghei ANKA (PbA) infected CBA/CaJ mice, which develop experimental cerebral malaria (ECM), and P. yoelii 17XL (PyXL) infected mice, which succumb to malarial hyperparasitemia without neurological impairment. Using a combination of intravital imaging and flow cytometry, we show that significantly more CD8(+) T cells, neutrophils, and macrophages are recruited to postcapillary venules during ECM compared to hyperparasitemia. ECM correlated with ICAM-1 upregulation on macrophages, while vascular endothelia upregulated ICAM-1 during ECM and hyperparasitemia. The arrest of large numbers of leukocytes in postcapillary and larger venules caused microrheological alterations that significantly restricted the venous blood flow. Treatment with FTY720, which inhibits vascular leakage, neurological signs, and death from ECM, prevented the recruitment of a subpopulation of CD45(hi) CD8(+) T cells, ICAM-1(+) macrophages, and neutrophils to postcapillary venules. FTY720 had no effect on the ECM-associated expression of the pattern recognition receptor CD14 in postcapillary venules suggesting that endothelial activation is insufficient to cause vascular pathology. Expression of the endothelial tight junction proteins claudin-5, occludin, and ZO-1 in the cerebral cortex and cerebellum of PbA-infected mice with ECM was unaltered compared to FTY720-treated PbA-infected mice or PyXL-infected mice with hyperparasitemia. Thus, blood brain barrier opening does not involve endothelial injury and is likely reversible, consistent with the rapid recovery of many patients with CM. We conclude that the ECM-associated recruitment of large numbers of activated leukocytes, in particular CD8(+) T cells and ICAM(+) macrophages, causes a severe restriction in the venous blood efflux from the brain, which exacerbates the vasogenic edema and increases the intracranial pressure. Thus, death from ECM could potentially occur as a consequence of intracranial hypertension.


Subject(s)
Blood-Brain Barrier/immunology , Cerebral Cortex/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Plasmodium yoelii/immunology , Animals , Blood-Brain Barrier/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cerebral Cortex/parasitology , Cerebral Cortex/pathology , Claudin-5/immunology , Disease Models, Animal , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/pharmacology , Intercellular Adhesion Molecule-1/immunology , Macrophages/immunology , Macrophages/pathology , Malaria, Cerebral/drug therapy , Malaria, Cerebral/pathology , Mice , Neutrophils/immunology , Neutrophils/pathology , Occludin/immunology , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Zonula Occludens-1 Protein/immunology
7.
Article in English | MEDLINE | ID: mdl-25414834

ABSTRACT

Most Plasmodium falciparum-infected children with cerebral malaria (CM) die from respiratory arrest, but the underlying pathology is unclear. Here we present a model in which the ultimate cause of death from CM is severe intracranial hypertension. Dynamic imaging of mice infected with P. berghei ANKA, an accepted model for experimental CM, revealed that leukocyte adhesion impairs the venous blood flow by reducing the functional lumen of postcapillary venules (PCV). The resulting increase in intracranial pressure (ICP) exacerbates cerebral edema formation, a hallmark of both murine and pediatric CM. We propose that two entirely different pathogenetic mechanisms-cytoadherence of P. falciparum-infected erythrocytes in pediatric CM and leukocyte arrest in murine CM-result in the same pathological outcome: a severe increase in ICP leading to brainstem herniation and death from respiratory arrest. The intracranial hypertension (IH) model unifies previous hypotheses, applies to human and experimental CM alike, eliminates the need to explain any selective recognition mechanism Plasmodium might use to target multiple sensitive sites in the brain, and explains how an intravascular parasite can cause so much neuronal dysfunction.


Subject(s)
Intracranial Hypertension/etiology , Intracranial Hypertension/mortality , Malaria, Cerebral/complications , Plasmodium falciparum , Animals , Cause of Death , Disease Models, Animal , Humans , Intracranial Hypertension/diagnosis , Mice
8.
PLoS Pathog ; 10(6): e1004080, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24967715

ABSTRACT

Alternatively activated macrophages (AAM) that accumulate during chronic T helper 2 inflammatory conditions may arise through proliferation of resident macrophages or recruitment of monocyte-derived cells. Liver granulomas that form around eggs of the helminth parasite Schistosoma mansoni require AAM to limit tissue damage. Here, we characterized monocyte and macrophage dynamics in the livers of infected CX3CR1(GFP/+) mice. CX3CR1-GFP⁺ monocytes and macrophages accumulated around eggs and in granulomas during infection and upregulated PD-L2 expression, indicating differentiation into AAM. Intravital imaging of CX3CR1-GFP⁺ Ly6C(low) monocytes revealed alterations in patrolling behavior including arrest around eggs that were not encased in granulomas. Differential labeling of CX3CR1-GFP⁺ cells in the blood and the tissue showed CD4⁺ T cell dependent accumulation of PD-L2⁺ CX3CR1-GFP⁺ AAM in the tissues as granulomas form. By adoptive transfer of Ly6C(high) and Ly6C(low) monocytes into infected mice, we found that AAM originate primarily from transferred Ly6C(high) monocytes, but that these cells may transition through a Ly6C(low) state and adopt patrolling behavior in the vasculature. Thus, during chronic helminth infection AAM can arise from recruited Ly6C(high) monocytes via help from CD4⁺ T cells.


Subject(s)
Antigens, Ly/blood , CD4-Positive T-Lymphocytes/immunology , Granuloma/immunology , Liver/immunology , Macrophages/immunology , Monocytes/immunology , Schistosomiasis mansoni/immunology , Animals , Antigens, Ly/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/parasitology , Cell Communication , Cell Transdifferentiation , Crosses, Genetic , Female , Granuloma/parasitology , Granuloma/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunologic Surveillance , Liver/metabolism , Liver/parasitology , Liver/pathology , Macrophage Activation , Macrophages/metabolism , Macrophages/parasitology , Male , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/metabolism , Monocytes/parasitology , Ovum/growth & development , Ovum/immunology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Recombinant Proteins/metabolism , Schistosoma mansoni/growth & development , Schistosoma mansoni/immunology , Schistosomiasis mansoni/metabolism , Schistosomiasis mansoni/parasitology , Schistosomiasis mansoni/physiopathology , Up-Regulation
9.
Parasitol Int ; 63(1): 171-86, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24076429

ABSTRACT

Plasmodium falciparum malaria is responsible for the deaths of over half a million African children annually. Until a decade ago, dynamic analysis of the malaria parasite was limited to in vitro systems with the typical limitations associated with 2D monocultures or entirely artificial surfaces. Due to extremely low parasite densities, the liver was considered a black box in terms of Plasmodium sporozoite invasion, liver stage development, and merozoite release into the blood. Further, nothing was known about the behavior of blood stage parasites in organs such as the brain where clinical signs manifest and the ensuing immune response of the host that may ultimately result in a fatal outcome. The advent of fluorescent parasites, advances in imaging technology, and availability of an ever-increasing number of cellular and molecular probes have helped illuminate many steps along the pathogenetic cascade of this deadly tropical parasite.


Subject(s)
Brain/parasitology , Liver/parasitology , Lung/parasitology , Microscopy/methods , Plasmodium/cytology , Animals , Brain/immunology , Liver/immunology , Lung/immunology , Plasmodium/physiology
10.
Infect Immun ; 81(12): 4350-62, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24042110

ABSTRACT

Immunization with Plasmodium sporozoites can elicit high levels of sterile immunity, and neutralizing antibodies from protected hosts are known to target the repeat region of the circumsporozoite (CS) protein on the parasite surface. CS-based subunit vaccines have been hampered by suboptimal immunogenicity and the requirement for strong adjuvants to elicit effective humoral immunity. Pathogen-associated molecular patterns (PAMPs) that signal through Toll-like receptors (TLRs) can function as potent adjuvants for innate and adaptive immunity. We examined the immunogenicity of recombinant proteins containing a TLR5 agonist, flagellin, and either full-length or selected epitopes of the Plasmodium falciparum CS protein. Mice immunized with either of the flagellin-modified CS constructs, administered intranasally (i.n.) or subcutaneously (s.c.), developed similar levels of malaria-specific IgG1 antibody and interleukin-5 (IL-5)-producing T cells. Importantly, immunization via the i.n. but not the s.c. route elicited sporozoite neutralizing antibodies capable of inhibiting >90% of sporozoite invasion in vitro and in vivo, as measured using a transgenic rodent parasite expressing P. falciparum CS repeats. These findings demonstrate that functional sporozoite neutralizing antibody can be elicited by i.n. immunization with a flagellin-modified P. falciparum CS protein and raise the potential of a scalable, safe, needle-free vaccine for the 40% of the world's population at risk of malaria.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Antibodies, Protozoan/immunology , Cells, Cultured , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , Flagellin/immunology , Humans , Immunity, Humoral/immunology , Immunization , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Interleukin-5/biosynthesis , Malaria Vaccines/administration & dosage , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protozoan Proteins/administration & dosage , Recombinant Proteins/immunology , Sporozoites/immunology , Toll-Like Receptor 5/agonists , Vaccines, Subunit/immunology
11.
PLoS One ; 8(8): e70842, 2013.
Article in English | MEDLINE | ID: mdl-23967119

ABSTRACT

Plasmodium falciparum malaria remains one of the most serious health problems globally and a protective malaria vaccine is desperately needed. Vaccination with attenuated parasites elicits multiple cellular effector mechanisms that lead to Plasmodium liver stage elimination. While granule-mediated cytotoxicity requires contact between CD8+ effector T cells and infected hepatocytes, cytokine secretion should allow parasite killing over longer distances. To better understand the mechanism of parasite elimination in vivo, we monitored the dynamics of CD8+ T cells in the livers of naïve, immunized and sporozoite-infected mice by intravital microscopy. We found that immunization of BALB/c mice with attenuated P. yoelii 17XNL sporozoites significantly increases the velocity of CD8+ T cells patrolling the hepatic microvasculature from 2.69±0.34 µm/min in naïve mice to 5.74±0.66 µm/min, 9.26±0.92 µm/min, and 7.11±0.73 µm/min in mice immunized with irradiated, early genetically attenuated (Pyuis4-deficient), and late genetically attenuated (Pyfabb/f-deficient) parasites, respectively. Sporozoite infection of immunized mice revealed a 97% and 63% reduction in liver stage density and volume, respectively, compared to naïve controls. To examine cellular mechanisms of immunity in situ, naïve mice were passively immunized with hepatic or splenic CD8+ T cells. Unexpectedly, adoptive transfer rendered the motile CD8+ T cells from immunized mice immotile in the liver of P. yoelii infected mice. Similarly, when mice were simultaneously inoculated with viable sporozoites and CD8+ T cells, velocities 18 h later were also significantly reduced to 0.68±0.10 µm/min, 1.53±0.22 µm/min, and 1.06±0.26 µm/min for CD8+ T cells from mice immunized with irradiated wild type sporozoites, Pyfabb/f-deficient parasites, and P. yoelii CS280-288 peptide, respectively. Because immobilized CD8+ T cells are unable to make contact with infected hepatocytes, soluble mediators could potentially play a key role in parasite elimination under these experimental conditions.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Immunization , Liver/immunology , Liver/parasitology , Malaria/immunology , Plasmodium yoelii/physiology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , Hepatocytes/immunology , Hepatocytes/parasitology , Malaria/prevention & control , Mice , Mice, Inbred BALB C , Sporozoites/physiology
12.
PLoS Pathog ; 8(10): e1002982, 2012.
Article in English | MEDLINE | ID: mdl-23133375

ABSTRACT

Plasmodium falciparum malaria is responsible for nearly one million annual deaths worldwide. Because of the difficulty in monitoring the pathogenesis of cerebral malaria in humans, we conducted a study in various mouse models to better understand disease progression in experimental cerebral malaria (ECM). We compared the effect on the integrity of the blood brain barrier (BBB) and the histopathology of the brain of P. berghei ANKA, a known ECM model, P. berghei NK65, generally thought not to induce ECM, P. yoelii 17XL, originally reported to induce human cerebral malaria-like histopathology, and P. yoelii YM. As expected, P. berghei ANKA infection caused neurological signs, cerebral hemorrhages, and BBB dysfunction in CBA/CaJ and Swiss Webster mice, while Balb/c and A/J mice were resistant. Surprisingly, PbNK induced ECM in CBA/CaJ mice, while all other mice were resistant. P. yoelii 17XL and P. yoelii YM caused lethal hyperparasitemia in all mouse strains; histopathological alterations, BBB dysfunction, or neurological signs were not observed. Intravital imaging revealed that infected erythrocytes containing mature parasites passed slowly through capillaries making intimate contact with the endothelium, but did not arrest. Except for relatively rare microhemorrhages, mice with ECM presented no obvious histopathological alterations that would explain the widespread disruption of the BBB. Intravital imaging did reveal, however, that postcapillary venules, but not capillaries or arterioles, from mice with ECM, but not hyperparasitemia, exhibit platelet marginalization, extravascular fibrin deposition, CD14 expression, and extensive vascular leakage. Blockage of LFA-1 mediated cellular interactions prevented leukocyte adhesion, vascular leakage, neurological signs, and death from ECM. The endothelial barrier-stabilizing mediators imatinib and FTY720 inhibited vascular leakage and neurological signs and prolonged survival to ECM. Thus, it appears that neurological signs and coma in ECM are due to regulated opening of paracellular-junctional and transcellular-vesicular fluid transport pathways at the neuroimmunological BBB.


Subject(s)
Blood-Brain Barrier/pathology , Malaria, Cerebral/pathology , Plasmodium berghei/pathogenicity , Plasmodium yoelii/pathogenicity , Animals , Benzamides , Blood-Brain Barrier/immunology , Blood-Brain Barrier/physiopathology , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/etiology , Disease Models, Animal , Fingolimod Hydrochloride , Imatinib Mesylate , Lipopolysaccharide Receptors/biosynthesis , Lymphocyte Function-Associated Antigen-1/metabolism , Malaria, Cerebral/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Neuroimmunomodulation , Parasitemia , Piperazines/pharmacology , Plasmodium falciparum/pathogenicity , Propylene Glycols/pharmacology , Pyrimidines/pharmacology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
13.
PLoS One ; 7(8): e43913, 2012.
Article in English | MEDLINE | ID: mdl-22952808

ABSTRACT

Human African trypanosomiasis or sleeping sickness is a vector-borne parasitic disease that has a major impact on human health and welfare in sub-Saharan countries. Based mostly on data from animal models, it is currently thought that trypanosome entry into the brain occurs by initial infection of the choroid plexus and the circumventricular organs followed days to weeks later by entry into the brain parenchyma. However, Trypanosoma brucei bloodstream forms rapidly cross human brain microvascular endothelial cells in vitro and appear to be able to enter the murine brain without inflicting cerebral injury. Using a murine model and intravital brain imaging, we show that bloodstream forms of T. b. brucei and T. b. rhodesiense enter the brain parenchyma within hours, before a significant level of microvascular inflammation is detectable. Extravascular bloodstream forms were viable as indicated by motility and cell division, and remained detectable for at least 3 days post infection suggesting the potential for parasite survival in the brain parenchyma. Vascular inflammation, as reflected by leukocyte recruitment and emigration from cortical microvessels, became apparent only with increasing parasitemia at later stages of the infection, but was not associated with neurological signs. Extravascular trypanosomes were predominantly associated with postcapillary venules suggesting that early brain infection occurs by parasite passage across the neuroimmunological blood brain barrier. Thus, trypanosomes can invade the murine brain parenchyma during the early stages of the disease before meningoencephalitis is fully established. Whether individual trypanosomes can act alone or require the interaction from a quorum of parasites remains to be shown. The significance of these findings for disease development is now testable.


Subject(s)
Brain/parasitology , Trypanosoma brucei brucei/physiology , Animals , Blood/parasitology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/parasitology , Brain/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Species Specificity , Time Factors , Trypanosoma brucei brucei/metabolism
14.
Int J Parasitol ; 40(6): 751-60, 2010 May.
Article in English | MEDLINE | ID: mdl-20018192

ABSTRACT

The infective stage of Entamoeba parasites is an encysted form. This stage can be readily generated in vitro, which has allowed identification of stimuli that trigger the differentiation of the parasite trophozoite stage into the cyst stage. Studies of the second differentiation event, emergence of the parasite from the cyst upon infection of a host, have been hampered by the lack of an efficient means to excyst the parasite and complete the life cycle in vitro. We have determined that a combination of exposures to water, bicarbonate and bile induces rapid excystment of Entamoeba invadens cysts. The high efficiency of this method has allowed the visualization of the dynamics of the process by electron and confocal microscopy, and should permit the analysis of stage-specific gene expression and high-throughput screening of inhibitory compounds.


Subject(s)
Entamoeba/growth & development , Entamoebiasis/parasitology , Upper Gastrointestinal Tract/chemistry , Upper Gastrointestinal Tract/parasitology , Animals , Bicarbonates/metabolism , Bile/metabolism , Entamoeba/cytology , Microscopy, Confocal , Microscopy, Electron
15.
Int J Parasitol ; 39(1): 119-32, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18723023

ABSTRACT

Malaria vaccines, comprised of irradiated Plasmodium falciparum sporozoites or a synthetic peptide containing T and B cell epitopes of the circumsporozoite protein (CSP), elicit multifunctional cytotoxic and non-cytotoxic CD4(+) T cells in immunised volunteers. Both lytic and non-lytic CD4(+)T cell clones recognised a series of overlapping epitopes within a 'universal' T cell epitope EYLNKIQNSLSTEWSPCSVT of CSP (NF54 isolate) that was presented in the context of multiple DR molecules. Lytic activity directly correlated with T cell receptor (TCR) functional avidity as measured by stimulation indices and recognition of naturally occurring variant peptides. CD4(+) T cell-mediated cytotoxicity was contact-dependent and did not require de novo synthesis of cytotoxic mediators, suggesting a granule-mediated mechanism. Live cell imaging of the interaction of effector and target cells demonstrated that CD4(+) cytotoxic T cells recognise target cells with their leading edge, reorient their cytotoxic granules towards the zone of contact, and form a stable immunological synapse. CTL attacks induced chromatin condensation, nuclear fragmentation and formation of apoptotic bodies in target cells. Together, these findings suggest that CD4(+) CTLs trigger target cell apoptosis via classical perforin/granzyme-mediated cytotoxicity, similar to CD8(+) CTLs, and these multifunctional sporozoite- and peptide-induced CD4(+) T cells have the potential to play a direct role as effector cells in targeting the exoerythrocytic forms within the liver.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Protozoan/immunology , Clone Cells , Cytokines/biosynthesis , Cytotoxicity, Immunologic/immunology , Epitopes, T-Lymphocyte/immunology , Humans , Immunity, Cellular , Immunological Synapses , Malaria, Falciparum/immunology , Receptors, Antigen, T-Cell/immunology
16.
Cell Microbiol ; 10(10): 1956-67, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18647171

ABSTRACT

Advances in our understanding of the molecular and cell biology of the malaria parasite have led to new vaccine development efforts resulting in a pipeline of over 40 candidates undergoing clinical phase I-III trials. Vaccine-induced CD4+ and CD8+ T cells specific for pre-erythrocytic stage antigens have been found to express cytolytic and multi-cytokine effector functions that support a key role for these T cells within the hepatic environment. However, little is known of the cellular interactions that occur during the effector phase in which the intracellular hepatic stage of the parasite is targeted and destroyed. This review focuses on cell biological aspects of the interaction between malaria-specific effector cells and the various antigen-presenting cells that are known to exist within the liver, including hepatocytes, dendritic cells, Kupffer cells, stellate cells and sinusoidal endothelia. Considering the unique immune properties of the liver, it is conceivable that these different hepatic antigen-presenting cells fulfil distinct but complementary roles during the effector phase against Plasmodium liver stages.


Subject(s)
Antigen-Presenting Cells/immunology , Liver/immunology , Liver/parasitology , Plasmodium/immunology , Plasmodium/physiology , T-Lymphocytes/immunology , Animals , Humans
17.
Int J Parasitol ; 38(14): 1639-50, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18656478

ABSTRACT

Plasmodium sporozoites traverse Kupffer cells on their way into the liver. Sporozoite contact does not elicit a respiratory burst in these hepatic macrophages and blocks the formation of reactive oxygen species in response to secondary stimuli via elevation of the intracellular cAMP concentration. Here we show that increasing the cAMP level with dibutyryl cyclic adenosine monophosphate (db-cAMP) or isobutylmethylxanthine (IBMX) also modulates cytokine secretion in murine Kupffer cells towards an overall anti-inflammatory profile. Stimulation of Plasmodium yoelii sporozoite-exposed Kupffer cells with lipopolysaccharide or IFN-gamma reveals down-modulation of TNF-alpha, IL-6 and MCP-1, and up-regulation of IL-10. Prerequisite for this shift of the cytokine profile are parasite viability and contact with Kupffer cells, but not invasion. Whilst sporozoite-exposed Kupffer cells become TUNEL-positive and exhibit other signs of apoptotic death such as membrane blebbing, nuclear condensation and fragmentation, sporozoites remain intact and appear to transform to early exo-erythrocytic forms in Kupffer cell cultures. Together, the in vitro data indicate that Plasmodium possesses mechanisms to render Kupffer cells insensitive to pro-inflammatory stimuli and eventually eliminates these macrophages by forcing them into programmed cell death.


Subject(s)
Apoptosis/immunology , Cytokines/metabolism , Kupffer Cells/immunology , Malaria/immunology , Plasmodium yoelii/immunology , Sporozoites/physiology , 1-Methyl-3-isobutylxanthine/metabolism , Animals , Bucladesine/metabolism , Host-Parasite Interactions , Interferon-gamma/pharmacology , Interleukins/metabolism , Kupffer Cells/drug effects , Lipopolysaccharides/pharmacology , Liver/metabolism , Liver/pathology , Malaria/metabolism , Malaria/parasitology , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
18.
Subcell Biochem ; 47: 182-97, 2008.
Article in English | MEDLINE | ID: mdl-18512352

ABSTRACT

Malaria sporozoites must cross at least two cell barriers to reach their initial site of replication in the mammalian host. After transmission into the skin by an infected mosquito, they migrate towards small dermal capillaries, traverse the vascular endothelial layer, and rapidly home to the liver. To infect hepatocytes, the parasites must cross the sinusoidal cell layer, composed of specialized highly fenestrated sinusoidal endothelia and Kupffer cells, the resident macrophages of the liver (Fig. 1). The exact route Plasmodium sporozoites take to hepatocytes has been subject of controversial discussions for many years. Recent cell biological, microscopic, and genetic approaches have considerably enhanced our understanding of the initial events leading to the establishment of a malaria infection in the liver.


Subject(s)
Liver/parasitology , Plasmodium/pathogenicity , Sporozoites/physiology , Animals , Hepatocytes/immunology , Hepatocytes/parasitology , Humans , Kupffer Cells/immunology , Kupffer Cells/parasitology , Malaria/immunology , Malaria/parasitology , Models, Biological , Plasmodium/immunology , Plasmodium/physiology , Sporozoites/immunology
19.
Int J Parasitol ; 38(6): 655-72, 2008 May.
Article in English | MEDLINE | ID: mdl-18005972

ABSTRACT

Plasmodium gallinaceum typically causes sub-clinical disease with low mortality in its primary host, the Indian jungle fowl Gallus sonnerati. Domestic chickens of European origin, however, are highly susceptible to this avian malaria parasite. Here we describe the development of P. gallinaceum in young White Leghorn chicks with emphasis on the primary exoerythrocytic phase of the infection. Using various regimens for infection, we found that P. gallinaceum induced a transient primary exoerythrocytic infection followed by a fulminant lethal erythrocytic phase. Prerequisite for the appearance of secondary exoerythrocytic stages was the development of a certain level of parasitaemia. Once established, secondary exoerythrocytic stages could be propagated from bird to bird for several generations without causing fatalities. Infected brains contained large secondary exoerythrocytic stages in capillary endothelia, while in the liver primary and secondary erythrocytic stages developed primarily in Kupffer cells and remained smaller. At later stages, livers exhibited focal hepatocyte necrosis, Kupffer cell hyperplasia, stellate cell proliferation, inflammatory cell infiltration and granuloma formation. Because P. gallinaceum selectively infected Kupffer cells in the liver and caused a histopathology strikingly similar to mammalian species, this avian Plasmodium species represents an evolutionarily closely related model for studies on the hepatic phase of mammalian malaria.


Subject(s)
Chickens/parasitology , Plasmodium gallinaceum/growth & development , Poultry Diseases/parasitology , Animals , Brain/parasitology , Liver/parasitology , Liver/pathology , Malaria, Avian/immunology , Malaria, Avian/parasitology , Malaria, Avian/pathology , Microscopy, Electron
20.
PLoS Pathog ; 3(11): e171, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17997605

ABSTRACT

Plasmodium undergoes one round of multiplication in the liver prior to invading erythrocytes and initiating the symptomatic blood phase of the malaria infection. Productive hepatocyte infection by sporozoites leads to the generation of thousands of merozoites capable of erythrocyte invasion. Merozoites are released from infected hepatocytes as merosomes, packets of hundreds of parasites surrounded by host cell membrane. Intravital microscopy of green fluorescent protein-expressing P. yoelii parasites showed that the majority of merosomes exit the liver intact, adapt a relatively uniform size of 12-18 microm, and contain 100-200 merozoites. Merosomes survived the subsequent passage through the right heart undamaged and accumulated in the lungs. Merosomes were absent from blood harvested from the left ventricle and from tail vein blood, indicating that the lungs effectively cleared the blood from all large parasite aggregates. Accordingly, merosomes were not detectable in major organs such as brain, kidney, and spleen. The failure of annexin V to label merosomes collected from hepatic effluent indicates that phosphatidylserine is not exposed on the surface of the merosome membrane suggesting the infected hepatocyte did not undergo apoptosis prior to merosome release. Merosomal merozoites continued to express green fluorescent protein and did not incorporate propidium iodide or YO-PRO-1 indicating parasite viability and an intact merosome membrane. Evidence of merosomal merozoite infectivity was provided by hepatic effluent containing merosomes being significantly more infective than blood with an identical low-level parasitemia. Ex vivo analysis showed that merosomes eventually disintegrate inside pulmonary capillaries, thus liberating merozoites into the bloodstream. We conclude that merosome packaging protects hepatic merozoites from phagocytic attack by sinusoidal Kupffer cells, and that release into the lung microvasculature enhances the chance of successful erythrocyte invasion. We believe this previously unknown part of the plasmodial life cycle ensures an effective transition from the liver to the blood phase of the malaria infection.


Subject(s)
Liver/parasitology , Malaria/parasitology , Merozoites/physiology , Plasmodium yoelii/physiology , Pulmonary Circulation , Animals , Lung/blood supply , Lung/parasitology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Microscopy, Confocal
SELECTION OF CITATIONS
SEARCH DETAIL
...