Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 116(48): 24214-24220, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31723045

ABSTRACT

Although CRIg was originally identified as a macrophage receptor for binding complement C3b/iC3b in vitro, recent studies reveal that CRIg functions as a pattern recognition receptor in vivo for Kupffer cells (KCs) to directly bind bacterial pathogens in a complement-independent manner. This raises the critical question of whether CRIg captures circulating pathogens through interactions with complement in vivo under flow conditions. Furthermore, the role of CRIg during parasitic infection is unknown. Taking advantage of intravital microscopy and using African trypanosomes as a model, we studied the role of CRIg in intravascular clearance of bloodborne parasites. Complement C3 is required for intravascular clearance of African trypanosomes by KCs, preventing the early mortality of infected mice. Moreover, antibodies are essential for complement-mediated capture of circulating parasites by KCs. Interestingly, reduced antibody production was observed in the absence of complement C3 during infection. We further demonstrate that CRIg but not CR3 is critically involved in KC-mediated capture of circulating parasites, accounting for parasitemia control and host survival. Of note, CRIg cannot directly catch circulating parasites and antibody-induced complement activation is indispensable for CRIg-mediated parasite capture. Thus, we provide evidence that CRIg, by interacting with complement in vivo, plays an essential role in intravascular clearance of bloodborne parasites. Targeting CRIg may be considered as a therapeutic strategy.


Subject(s)
Complement C3b/metabolism , Host-Parasite Interactions/physiology , Parasitemia/parasitology , Receptors, Complement/physiology , Trypanosomiasis, African/blood , Animals , Complement C3b/immunology , Intravital Microscopy , Kupffer Cells/immunology , Kupffer Cells/parasitology , Macrophage-1 Antigen/metabolism , Macrophages/parasitology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/pathogenicity , Trypanosoma congolense/pathogenicity , Trypanosomiasis, African/mortality , Trypanosomiasis, African/parasitology
2.
Article in English | MEDLINE | ID: mdl-29127992

ABSTRACT

Kupffer cells (KC) are the liver macrophage population that resides in the hepatic sinusoids and efficiently phagocyte pathogens by establishing an intimate contact with circulating blood. KC constitute the liver host cells in Leishmania infection, nevertheless little is described about their role, apart from their notable contribution in granulomatous inflammation. The present study aims to investigate how canine KC sense and react to the presence of Leishmania infantum promastigotes and amastigotes by evaluating the gene expression of specific innate immune cell receptors and cytokines, as well as the induction of nitric oxide and urea production. Complementarily, the impact of a leishmanicidal drug - meglumine antimoniate (MgA) - in infected KC was also explored. KC revealed to be susceptible to both parasite forms and no major differences were found in the immune response generated. L. infantum parasites seem to interact with KC innate immune receptors and induce an anergic state, promoting immune tolerance and parasite survival. The addition of MgA to infected KC breaks the parasite imposed silence and increased gene expression of Toll-like receptors (TLR) 2 and TLR4, possibly activating downstream pathways. Understanding how KC sense and react to parasite presence could bring new insights into the control or even elimination of canine leishmaniasis.


Subject(s)
Antiprotozoal Agents/pharmacology , Dog Diseases/parasitology , Kupffer Cells/parasitology , Leishmania infantum/physiology , Leishmaniasis, Visceral/veterinary , Meglumine/pharmacology , Organometallic Compounds/pharmacology , Animals , Dog Diseases/immunology , Dog Diseases/metabolism , Dogs , Kupffer Cells/drug effects , Kupffer Cells/immunology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Meglumine Antimoniate , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
3.
J Exp Med ; 213(10): 2099-112, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27551151

ABSTRACT

Malaria transmission begins when an infected mosquito delivers Plasmodium sporozoites into the skin. The sporozoite subsequently enters the circulation and infects the liver by preferentially traversing Kupffer cells, a macrophage-like component of the liver sinusoidal lining. By screening a phage display library, we previously identified a peptide designated P39 that binds to CD68 on the surface of Kupffer cells and blocks sporozoite traversal. In this study, we show that the P39 peptide is a structural mimic of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) on the sporozoite surface and that GAPDH directly interacts with CD68 on the Kupffer cell surface. Importantly, an anti-P39 antibody significantly inhibits sporozoite liver invasion without cross-reacting with mammalian GAPDH. Therefore, Plasmodium-specific GAPDH epitopes may provide novel antigens for the development of a prehepatic vaccine.


Subject(s)
Cell Membrane/enzymology , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Liver/pathology , Liver/parasitology , Malaria/parasitology , Plasmodium berghei/pathogenicity , Sporozoites/enzymology , Amino Acid Sequence , Animals , Antibodies, Protozoan/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Line , Conserved Sequence , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Humans , Kupffer Cells/parasitology , Kupffer Cells/pathology , Ligands , Malaria/immunology , Malaria Vaccines/immunology , Mice, Knockout , Peptides/chemistry , Peptides/immunology , Plasmodium berghei/immunology , Protein Binding , Rats
4.
J Exp Med ; 212(9): 1391-403, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26216124

ABSTRACT

After being delivered by the bite from an infected mosquito, Plasmodium sporozoites enter the blood circulation and infect the liver. Previous evidence suggests that Kupffer cells, a macrophage-like component of the liver blood vessel lining, are traversed by sporozoites to initiate liver invasion. However, the molecular determinants of sporozoite-Kupffer cell interactions are unknown. Understanding the molecular basis for this specific recognition may lead to novel therapeutic strategies to control malaria. Using a phage display library screen, we identified a peptide, P39, that strongly binds to the Kupffer cell surface and, importantly, inhibits sporozoite Kupffer cell entry. Furthermore, we determined that P39 binds to CD68, a putative receptor for sporozoite invasion of Kupffer cells that acts as a gateway for malaria infection of the liver.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Kupffer Cells/metabolism , Malaria/metabolism , Plasmodium berghei/metabolism , Sporozoites/metabolism , Animals , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Kupffer Cells/parasitology , Kupffer Cells/pathology , Liver/metabolism , Liver/parasitology , Liver/pathology , Malaria/genetics , Malaria/pathology , Male , Mice , Mice, Knockout , Peptide Library , Rats , Rats, Sprague-Dawley
5.
Mediators Inflamm ; 2014: 362605, 2014.
Article in English | MEDLINE | ID: mdl-24891764

ABSTRACT

Malaria is a mosquito-borne infectious disease of humans. It begins with a bite from an infected female Anopheles mosquito and leads to the development of the pre-erythrocytic and blood stages. Blood-stage infection is the exclusive cause of clinical symptoms of malaria. In contrast, the pre-erythrocytic stage is clinically asymptomatic and could be an excellent target for preventive therapies. Although the robust host immune responses limit the development of the liver stage, malaria parasites have also evolved strategies to suppress host defenses at the pre-erythrocytic stage. This paper reviews the immune evasion strategies of malaria parasites at the pre-erythrocytic stage, which could provide us with potential targets to design prophylactic strategies against malaria.


Subject(s)
Erythrocytes/immunology , Immune Evasion , Malaria/immunology , Animals , Anopheles , Autophagy , Culicidae , Female , Hepatocytes/parasitology , Humans , Kupffer Cells/parasitology , Liver/parasitology , Phagocytes/parasitology , Proteoglycans/chemistry , Skin/parasitology , Sporozoites/physiology
6.
J Exp Med ; 210(5): 905-15, 2013 May 06.
Article in English | MEDLINE | ID: mdl-23610126

ABSTRACT

Malaria infection starts when the sporozoite stage of the Plasmodium parasite is injected into the skin by a mosquito. Sporozoites are known to traverse host cells before finally invading a hepatocyte and multiplying into erythrocyte-infecting forms, but how sporozoites reach hepatocytes in the liver and the role of host cell traversal (CT) remain unclear. We report the first quantitative imaging study of sporozoite liver infection in rodents. We show that sporozoites can cross the liver sinusoidal barrier by multiple mechanisms, targeting Kupffer cells (KC) or endothelial cells and associated or not with the parasite CT activity. We also show that the primary role of CT is to inhibit sporozoite clearance by KC during locomotion inside the sinusoid lumen, before crossing the barrier. By being involved in multiple steps of the sporozoite journey from the skin to the final hepatocyte, the parasite proteins mediating host CT emerge as ideal antibody targets for vaccination against the parasite.


Subject(s)
Cell Movement , Host-Parasite Interactions/immunology , Liver/pathology , Liver/parasitology , Malaria/pathology , Malaria/parasitology , Sporozoites/physiology , Animals , Anopheles/parasitology , Cell Death , Endothelial Cells/parasitology , Endothelial Cells/pathology , Female , Green Fluorescent Proteins/metabolism , Kupffer Cells/parasitology , Kupffer Cells/pathology , Male , Mice , Mice, Inbred C57BL , Plasmodium berghei/cytology , Plasmodium berghei/physiology , Sporozoites/cytology
7.
Biochim Biophys Acta ; 1810(7): 652-65, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21530614

ABSTRACT

BACKGROUND: Evidence in the literature suggests that down-regulation of nitric oxide (NO) is associated with the pathophysiological conditions during visceral leishmaniasis (VL). Here we have investigated the mechanism that leads to the down regulation of systemic NO in the infected condition. Moreover, we have determined whether down regulation of NO is associated with increased generation of reactive oxygen species (ROS) during this disease. Therapeutic strategy targeting signaling molecules of these events was evaluated. METHODS: Plasma protein-nitrotyrosine was examined by ELISA kit. Generation of superoxides and peroxynitrites was investigated by flow cytometry. NO bioavailability in endothelial cells was evaluated using DAF-2DA fluorescence. Ceramide contents were evaluated using FACS analysis, HPTLC and HPLC. RESULTS: L. donovani infected reticulo-endothelial cells regulated the activity of eNOS and NAD(P)H oxidase in the endothelial cells through the generation of intercellular messenger, ceramide. Activation of SMases played an important role in the generation of ceramide in animals during chronic infection. These events led to generation of ROS within endothelial cells. Modulation of redox status of plasma and accumulation of ROS in endothelial cells were critically involved in the regulation of NO bioavailability in plasma of the infected animal. Endothelial dysfunction and decline of NO were resulted from an increased production of superoxide where upregulation of eNOS expression appeared as an ineffective compensatory event. Inhibition of ceramide generation increased NO bioavailability, prevented endothelial dysfunction and concomitant oxidative stress. CONCLUSION AND GENERAL SIGNIFICANCE: Decreased NO bioavailability and endothelial dysfunction were the downstream of ceramide signaling cascade. ROS accumulation promoted peroxynitrite generation and reduced NO bioavailability. Inhibition of ceramide generation may be a potential therapeutic option in preventing the co-morbidity associated with VL.


Subject(s)
Endothelium, Vascular/physiopathology , Homeostasis/physiology , Leishmaniasis, Visceral/physiopathology , Animals , Blotting, Western , Cells, Cultured , Ceramides/blood , Ceramides/metabolism , Cricetinae , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Flow Cytometry , Glutathione/blood , Glutathione Disulfide/blood , Host-Parasite Interactions , Kupffer Cells/metabolism , Kupffer Cells/parasitology , Leishmania donovani/physiology , Leishmaniasis, Visceral/blood , Leishmaniasis, Visceral/parasitology , Male , Mesocricetus , NADPH Oxidases/metabolism , Nitric Oxide/blood , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidation-Reduction , Oxidoreductases/metabolism , Reactive Oxygen Species/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Superoxides/metabolism , Time Factors
8.
J R Soc Interface ; 8(58): 711-9, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21068031

ABSTRACT

Parasitized erythrocytes are ingested by murine hepatic macrophages during malaria infection. We non-invasively monitored how this altered the motion of intracellular phagosomes in Kupffer cells using magnetometry. Submicrometric γFe(2)O(3) particles were injected prior to malaria infection. They were cleared from the blood, primarily by Kupffer cells, and retained within their phagosomes. The mice were periodically magnetized. After removing this external magnet, the aligned iron particles created a remnant magnetic field (RMF) which then decayed (relaxation), reflecting the motion of particle-containing phagosomes. After baseline measurements of relaxation, the mice were injected intravenously with Plasmodium chabaudi-parasitized or normal murine red blood cells (RBCs). During the next 15 days, relaxation measurements, parasitaemia and haematocrit values were monitored. At 6 days post injection with 3 × 10(7) parasitized RBCs, relaxation rates had decreased. At this time, all mice had parasitaemias greater than 58 per cent and haematocrits less than 20 per cent. At day 7, while the parasitaemias were declining, the rate of relaxation continued to decrease. Throughout the experiment, relaxation remained constant in animals injected with normal RBCs. Electron microscopy revealed Kupffer cells filled with damaged and parasitized erythrocytes, and haemoglobin degradation pigment. We conclude that ingestion and metabolism of parasitized erythrocytes by liver macrophages during malaria infection decreases their organelle motion with likely consequences of compromised host defences.


Subject(s)
Erythrocytes/metabolism , Kupffer Cells/metabolism , Kupffer Cells/parasitology , Malaria/blood , Animals , Disease Models, Animal , Electromagnetic Fields , Female , Hemeproteins/chemistry , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Microscopy, Electron/methods , Parasitemia/blood , Phagocytosis , Phagosomes/parasitology , Plasmodium chabaudi/metabolism
9.
Exp Parasitol ; 126(2): 254-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20493849

ABSTRACT

Plasmodium sporozoites suppress the respiratory burst and antigen presentation of Kupffer cells, which are regarded as the portal of invasion into hepatocytes. It is not known whether immune modulation of Kupffer cells can affect the liver stage. In the present study, we found that sporozoites inoculated into Wistar rats could be detected in the liver, spleen, and lung; however, most sporozoites were arrested in the liver. Sporozoites were captured by Kupffer cells lined with endothelial cells in the liver sinusoid before hepatocyte invasion. Pretreatment with TLR3 agonist poly(I:C) and TLR2 agonist BCG primarily activated Kupffer cells, inhibiting the sporozoite development into the exoerythrocytic form, whereas Kupffer cell antagonists dexamethasone and cyclophosphamide promoted development of the liver stage. Our data suggests that sporozoite development into its exoerythrocytic form may be associated with Kupffer cell functional status. Immune modulation of Kupffer cells could be a promising strategy to prevent malaria parasite infection.


Subject(s)
Immunologic Factors/pharmacology , Liver/parasitology , Plasmodium yoelii/growth & development , Adjuvants, Immunologic/pharmacology , Animals , Anopheles , BCG Vaccine/pharmacology , Cryoelectron Microscopy , Cyclophosphamide/pharmacology , Dexamethasone/pharmacology , Female , Glucocorticoids/pharmacology , Immunosuppressive Agents/pharmacology , Interferon Inducers/pharmacology , Kupffer Cells/parasitology , Liver/ultrastructure , Lung/parasitology , Male , Microscopy, Electron, Scanning , Plasmodium yoelii/drug effects , Plasmodium yoelii/immunology , Poly I-C/pharmacology , Rats , Rats, Wistar , Spleen/parasitology
10.
PLoS Pathog ; 6(3): e1000805, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20300603

ABSTRACT

Kupffer cells (KCs) represent the major phagocytic population within the liver and provide an intracellular niche for the survival of a number of important human pathogens. Although KCs have been extensively studied in vitro, little is known of their in vivo response to infection and their capacity to directly interact with antigen-specific CD8(+) T cells. Here, using a combination of approaches including whole mount and thin section confocal microscopy, adoptive cell transfer and intra-vital 2-photon microscopy, we demonstrate that KCs represent the only detectable population of mononuclear phagocytes within granulomas induced by Leishmania donovani infection that are capable of presenting parasite-derived peptide to effector CD8(+) T cells. This restriction of antigen presentation to KCs within the Leishmania granuloma has important implications for the identification of new candidate vaccine antigens and for the design of novel immuno-therapeutic interventions.


Subject(s)
CD8-Positive T-Lymphocytes/parasitology , Granuloma/parasitology , Kupffer Cells/parasitology , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Animals , Antigen Presentation/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Granuloma/immunology , Kupffer Cells/immunology , Leishmania donovani/genetics , Leishmania donovani/growth & development , Leishmaniasis Vaccines/immunology , Leishmaniasis, Visceral/parasitology , Liver/cytology , Liver/immunology , Liver/parasitology , Macrophages/immunology , Macrophages/parasitology , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phagocytosis/immunology
11.
Parasitol Res ; 105(1): 227-36, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19294422

ABSTRACT

Leishmania (L.) infantum (syn. Leishmania chagasi) is a dimorphic protozoan parasite that lives in promastigote and amastigote form in its sandfly vector and mammalian hosts, respectively. Here, we describe an in vitro culture system for the generation of a pure population of L. infantum axenic amastigotes after only 4 days incubation in culture medium supplemented with fetal calf serum, human urine, L: -glutamine, and HEPES at 37 masculineC (pH 5.5). Ultrastrutural analysis and infection assays in two macrophage populations (Kupffer cells (KUP) and peritoneal macrophages (PM)) infected with axenic amastigotes demonstrated that they maintained morphological and biochemical (A2 expression) features and a similar infection pattern to tissue-derived L. infantum amastigotes. The susceptibility of the macrophage lines to axenic or tissue-derived amastigotes and promastigotes was investigated. We found a completely different susceptibility profile for KUP and PM. Liver macrophages, both KUP and immigrant macrophages, are intimately involved in the response to L. infantum infection; this difference in susceptibility is probably related to their capacity to eliminate these parasites. Our in vitro system was thus able to generate axenic amastigotes that resemble tissue-derived amastigotes both in morphology and infectivity pattern; this will help in further investigation of the biological characteristics of the host-parasite relationship as well as the process of pathogenesis.


Subject(s)
Kupffer Cells/parasitology , Leishmania infantum/growth & development , Leishmania infantum/pathogenicity , Macrophages, Peritoneal/parasitology , Animals , Cell Culture Techniques , Cells, Cultured , Culture Media/chemistry , Leishmania infantum/ultrastructure , Mice , Mice, Inbred BALB C
12.
Glycoconj J ; 26(6): 663-73, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19005752

ABSTRACT

As compared to cutaneous leishmaniasis, vaccination against visceral leishmaniasis (VL) has received limited attention. In this study, we demonstrate for the first time that an UDP-Galactose: N-acetylglucosamine beta 1-4 galactosyltransferase (GenBank Accession No. EF159943) expressing attenuated LD clonal population (A-LD) is able to confer protection against the experimental challenge with the virulent LD AG83 parasite. A-LD was also effective in established leishmania infection. The vaccinated animals showed both cell mediated (in vitro T-cell proliferation, and DTH response) and humoral responses (Th1 type). These results demonstrate the potential of the attenuated clones as an immunotherapeutic and immunoprophylactic agent against visceral leishmaniasis.


Subject(s)
Antigens, Protozoan/immunology , Galactosyltransferases/immunology , Leishmania donovani/enzymology , Leishmania donovani/immunology , Leishmaniasis, Visceral/prevention & control , Protozoan Vaccines/immunology , Animals , Antibody Formation/immunology , Erythrocytes/metabolism , Erythrocytes/parasitology , Immunity, Cellular/immunology , Kupffer Cells/metabolism , Kupffer Cells/parasitology , Leishmaniasis, Visceral/immunology , Liver/parasitology , Mice , Mice, Inbred BALB C , Nitric Oxide/metabolism , Rabbits , Reactive Oxygen Species/metabolism , Spleen/parasitology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
14.
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
15.
Mol Biochem Parasitol ; 156(1): 32-40, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17714805

ABSTRACT

Malaria sporozoites migrate through several cells prior to a productive invasion that involves the formation of a parasitophorous vacuole (PV) where sporozoites undergo transformation into Exo-erythorcytic forms (EEFs). The precise mechanism leading to sporozoite activation for invasion is unknown, but prior traversal of host cells is required. During cell migration sporozoites are exposed to large shifts in K(+) concentration. We report here that incubation of sporozoites to the intracellular K(+) concentration enhances 8-10 times the infectivity of Plasmodium berghei and 4-5 times the infectivity of Plasmodium yoelli sporozoites for a hepatocyte cell line, while simultaneously decreasing cell passage activity. The K(+) enhancing effect was time and concentration dependent, and was significantly decreased by K(+) channel inhibitors. Potassium-treated P. berghei sporozoites also showed enhanced numbers of EEFs in non-permissive cell lines. Treated sporozoites had reduced infectivity for mice, but infectivity was enhanced upon Kupffer cell depletion. Transcriptional analysis of K(+) treated and control sporozoites revealed a high degree of correlation in their levels of gene expression, indicating that the observed phenotypic changes are not due to radical changes in gene transcription. Only seven genes were upregulated by more than two-fold in K(+) treated sporozoites. The highest level was noted in PP2C, a phosphatase known to dephosphorylate the AKT potassium channel in plants.


Subject(s)
Plasmodium berghei/pathogenicity , Plasmodium yoelii/pathogenicity , Potassium/pharmacology , Sporozoites/drug effects , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Hepatocytes/parasitology , Host-Parasite Interactions , Humans , Kupffer Cells/parasitology , Mice , Mice, Inbred C57BL , Plasmodium berghei/drug effects , Plasmodium berghei/growth & development , Plasmodium yoelii/drug effects , Plasmodium yoelii/growth & development , Serial Passage , Sporozoites/growth & development , Sporozoites/physiology
16.
Cell Microbiol ; 9(2): 397-412, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16953803

ABSTRACT

Previous studies suggested Plasmodium sporozoites infect hepatocytes after passing through Kupffer cells, but proof has been elusive. Here we present new information strengthening that hypothesis. We used homozygous op/op mice known to have few Kupffer cells because they lack macrophage colony stimulating factor 1 required for macrophage maturation due to a deactivating point mutation in the osteopetrosis gene. We found these mice to have 77% fewer Kupffer cells and to exhibit reduced clearance of colloidal carbon particles compared with heterozygous phenotypically normal littermates. Using a novel quantitative reverse transcription polymerase chain reaction assay for P. yoelii 18S rRNA, we found liver infection of op/op mice to be decreased by 84% compared with controls. However, using another way of limiting Kupffer cells, treatment with liposome-encapsulated clodronate, infection of normal mice was enhanced seven- to 15-fold. This was explained by electron microscopy showing temporary gaps in the sinusoidal cell layer caused by this treatment. Thus, Kupffer cell deficiency in op/op mice decreases sporozoite infection by reducing the number of portals to the liver parenchyma, whereas clodronate increases sporozoite infection by opening portals and providing direct access to hepatocytes. Together these data provide strong support for the hypothesis that Kupffer cells are the portal for sporozoites to hepatocytes and critical for the onset of a malaria infection.


Subject(s)
Kupffer Cells/physiology , Liver/parasitology , Plasmodium yoelii/pathogenicity , Protozoan Proteins/physiology , Sporozoites/physiology , Animals , Kupffer Cells/parasitology , Liver/cytology , Liver/metabolism , Mice , Plasmodium yoelii/growth & development , Protozoan Proteins/biosynthesis
17.
Cell Microbiol ; 8(10): 1537-46, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16911567

ABSTRACT

The initial site of replication for Plasmodium parasites in mammalian hosts are hepatocytes, cells that offer unique advantages for the extensive parasite replication occurring prior to the erythrocytic phase of the life cycle. The liver is the metabolic centre of the body and has an unusual relationship to the immune system. However, to reach hepatocytes, sporozoites must cross the sinusoidal barrier, composed of specialized endothelia and Kupffer cells, the resident macrophages of the liver. Mounting evidence suggests that, instead of taking what would seem a safer route through endothelia, the parasites traverse Kupffer cells yet suffer no harm. Kupffer cells have a broad range of responses towards incoming microorganisms, toxins and antigens which depend on the nature of the intruder, the experimental conditions and the environmental circumstances. Kupffer cells may become activated or remain anergic, produce pro- or anti-inflammatory mediators. Consequently, outcomes are diverse and include development of immunity or tolerance, parenchymal necrosis or regeneration, chronic cirrhotic transformation or acute liver failure. Here we review data concerning the unique structural and functional characteristics of Kupffer cells and their interactions with Plasmodium sporozoites in the context of a model in which these hepatic macrophages function as the sporozoite gate to the liver.


Subject(s)
Kupffer Cells/parasitology , Malaria/parasitology , Plasmodium/physiology , Sporozoites/physiology , Animals , Humans , Immune Tolerance , Kupffer Cells/cytology , Kupffer Cells/immunology , Life Cycle Stages , Liver/cytology , Liver/immunology , Liver/parasitology , Malaria/immunology , Plasmodium/immunology , Plasmodium/metabolism , Portal Vein/immunology , Portal Vein/parasitology , Sporozoites/immunology , Sporozoites/metabolism
18.
J Gastroenterol Hepatol ; 21(1 Pt 2): 313-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16460493

ABSTRACT

BACKGROUND AND AIM: Previous studies using isolated perfused rat liver in vivo have suggested that during the erythrocytic phase of malaria infection, overall phagocytosis by Kupffer cells is enhanced. The aim of the present study was to further investigate the individual phagocytic capacity and prostaglandin E(2) (PGE(2)) secretion of isolated Kupffer cells in vitro, and the immunohistochemical characteristics of Kupffer cells in vivo. METHODS: Malaria was induced in male Sprague-Dawley rats (n = 12) by inoculation with parasitized red cells containing Plasmodium berghei. Kupffer cells were isolated by centrifugal elutriation. RESULTS: A significantly increased yield of Kupffer cells was obtained from malaria-infected livers compared to controls (36.7 +/- 4.5 vs 11.8 +/- 1.1 x10(6) cells, P < 0.0001, n = 12). There was an increased internalization by phagocytosis of [(3)H]-BSA latex microspheres after 60 min in malaria-infected Kupffer cells compared to controls (65.05 +/- 1.5 vs 48.6 +/- 0.7, P < 0.001, n = 12). PGE(2) secretion into the cell culture medium was significantly suppressed in malaria-infected Kupffer cells compared to controls (1167 +/- 88 vs 4537 +/- 383 pg per 10(6) cells, P < 0.001, n = 5). Staining of ED1, ED2 and PCNA was greater in malaria-infected livers compared to control. CONCLUSION: The results indicate that the number of Kupffer cells is significantly increased and their phagocytic activity on a cell-by-cell basis is enhanced during the erythrocytic stage of malaria.


Subject(s)
Kupffer Cells/physiology , Malaria/physiopathology , Phagocytosis , Plasmodium berghei , Animals , Cell Count , Dinoprostone/metabolism , Immunohistochemistry , In Vitro Techniques , Kupffer Cells/metabolism , Kupffer Cells/parasitology , Liver/metabolism , Liver/parasitology , Liver/pathology , Malaria/parasitology , Malaria/pathology , Male , Parasitemia/physiopathology , Rats , Rats, Sprague-Dawley
19.
Infect Immun ; 73(12): 8393-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16299337

ABSTRACT

In highly susceptible BALB/c mice infected with Trypanosoma congolense, the total number of Kupffer cells in the liver remains constant; however, their mean size increases fivefold towards the terminal stage. About 25% of Kupffer cells undergo apoptosis. We suggest that development of an impairment of the macrophage system might be a major mechanism for inefficient elimination of trypanosomes.


Subject(s)
Kupffer Cells/parasitology , Trypanosoma congolense , Trypanosomiasis, African/immunology , Animals , Apoptosis , Cell Count , Disease Susceptibility/immunology , Disease Susceptibility/parasitology , Kupffer Cells/immunology , Kupffer Cells/pathology , Liver/parasitology , Liver/pathology , Mice , Mice, Inbred BALB C , Trypanosomiasis, African/parasitology
20.
Cell Microbiol ; 7(2): 199-208, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15659064

ABSTRACT

Plasmodium sporozoites are injected into the mammalian host during mosquito blood feeding and carried by the blood stream to the liver, where they infect hepatocytes and develop into erythrocyte-invasive forms. To reach the hepatocytes, sporozoites must cross the liver sinusoidal cell layer, which separates the hepatocytes from the circulatory system. Little is known about the molecular mechanisms by which sporozoites breach this cellular barrier. Here we report that a protein with a membrane attack complex/perforin (MACPF)-related domain is involved in this step. This molecule is specifically expressed in liver-infective sporozoites and localized in micronemes, organelles engaged in host cell invasion. Gene disruption experiments revealed that this protein is essential for the membrane-wounding activity of the sporozoite and is involved in its traversal of the sinusoidal cell layer prior to hepatocyte-infection. Disruptants failed to leave the circulation, and most of them were eliminated from the blood by liver perfusion. Our results suggest that rupture of the host plasma membrane by the pore-forming activity of this molecule is essential for cell passage of the sporozoite. This report is the first to demonstrate an important role of a MACPF-related protein in host cell invasion by a pathogenic microorganism.


Subject(s)
Complement Membrane Attack Complex/chemistry , Hepatocytes/parasitology , Liver/parasitology , Plasmodium berghei/physiology , Protozoan Proteins/physiology , Amino Acid Sequence , Animals , Cell Membrane/parasitology , Expressed Sequence Tags , Gene Targeting , Humans , Kupffer Cells/parasitology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Molecular Sequence Data , Mutagenesis, Insertional , Perforin , Plasmodium berghei/genetics , Pore Forming Cytotoxic Proteins , Protein Structure, Tertiary/physiology , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Rats , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...