Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cell Mol Immunol ; 18(10): 2443-2454, 2021 10.
Article in English | MEDLINE | ID: mdl-33203936

ABSTRACT

Cerebral malaria (CM) is a life-threatening diffuse encephalopathy caused by Plasmodium falciparum, in which the destruction of the blood-brain barrier (BBB) is the main cause of death. However, increasing evidence has shown that antimalarial drugs, the current treatment for CM, do little to protect against CM-induced BBB damage. Therefore, a means to alleviate BBB dysfunction would be a promising adjuvant therapy for CM. The adhesion molecule CD146 has been reported to be expressed in both endothelial cells and proinflammatory immune cells and mediates neuroinflammation. Here, we demonstrate that CD146 expressed on BBB endothelial cells but not immune cells is a novel therapeutic target in a mouse model of experimental cerebral malaria (eCM). Endothelial CD146 is upregulated during eCM development and facilitates the sequestration of infected red blood cells (RBCs) and/or proinflammatory lymphocytes in CNS blood vessels, thereby promoting the disruption of BBB integrity. Mechanistic studies showed that the interaction of CD146 and Galectin-9 contributes to the aggregation of infected RBCs and lymphocytes. Deletion of endothelial CD146 or treatment with the anti-CD146 antibody AA98 prevents severe signs of eCM, such as limb paralysis, brain vascular leakage, and death. In addition, AA98 combined with the antiparasitic drug artemether improved the cognition and memory of mice with eCM. Taken together, our findings suggest that endothelial CD146 is a novel and promising target in combination with antiparasitic drugs for future CM therapies.


Subject(s)
Blood-Brain Barrier , CD146 Antigen , Malaria, Cerebral , Animals , Disease Models, Animal , Endothelial Cells , Galectins/therapeutic use , Malaria, Cerebral/drug therapy , Malaria, Cerebral/prevention & control , Mice
2.
PLoS One ; 14(3): e0213428, 2019.
Article in English | MEDLINE | ID: mdl-30849122

ABSTRACT

Clinical and model studies indicate that low nitric oxide (NO) bioavailability due in part to profound hypoargininemia contributes to cerebral malaria (CM) pathogenesis. Protection against CM pathogenesis may be achieved by altering the diet before infection with Plasmodium falciparum infection (nutraceutical) or by administering adjunctive therapy that decreases CM mortality (adjunctive therapy). This hypothesis was tested by administering citrulline or arginine in experimental CM (eCM). We report that citrulline injected as prophylaxis immediately post infection (PI) protected virtually all mice by ameliorating (i) hypoargininemia, (ii) urea cycle impairment, and (iii) disruption of blood brain barrier. Citrulline prophylaxis inhibited plasma arginase activity. Parasitemia was similar in citrulline- and vehicle control-groups, indicating that protection from pathogenesis was not due to decreased parasitemia. Both citrulline and arginine administered from day 1 PI in the drinking water significantly protected mice from eCM. These observations collectively indicate that increasing dietary citrulline or arginine decreases eCM mortality. Citrulline injected ip on day 4 PI with quinine-injected ip on day 6 PI partially protected mice from eCM; citrulline plus scavenging of superoxide with pegylated superoxide dismutase and pegylated catalase protected all recipients from eCM. These findings indicate that ameliorating hypoargininemia with citrulline plus superoxide scavenging decreases eCM mortality.


Subject(s)
Citrulline/pharmacology , Malaria, Cerebral/metabolism , Malaria, Cerebral/prevention & control , Animals , Arginase/blood , Arginine/administration & dosage , Arginine/blood , Arginine/deficiency , Blood-Brain Barrier/drug effects , Citrulline/administration & dosage , Dietary Supplements , Disease Models, Animal , Free Radical Scavengers/administration & dosage , Humans , Malaria, Cerebral/etiology , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Plasmodium berghei , Superoxides/metabolism , Urea/metabolism
3.
Sci Rep ; 7(1): 18052, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29273797

ABSTRACT

Bone marrow-derived mesenchymal stem cells (MSC) have been promoted for multiple therapeutic applications. Many beneficial effects of MSCs are paracrine, dependent on extracellular vesicles (EVs). Although MSC-derived EVs (mEVs) are beneficial for acute lung injury and pulmonary fibrosis, mechanisms of mEV uptake by lung fibroblasts and their effects on myofibroblastic differentiation have not been established. We demonstrate that mEVs, but not fibroblast EVs (fEVs), suppress TGFß1-induced myofibroblastic differentiation of normal and idiopathic pulmonary fibrosis (IPF) lung fibroblasts. MEVs display increased time- and dose-dependent cellular uptake compared to fEVs. Removal or blocking of Thy-1, or blocking Thy-1-beta integrin interactions, decreased mEV uptake and prevented suppression of myofibroblastic differentiation. MicroRNAs (miRs) 199a/b-3p, 21-5p, 630, 22-3p, 196a-5p, 199b-5p, 34a-5p and 148a-3p are selectively packaged in mEVs. In silico analyses indicated that IPF lung fibroblasts have increased expression of genes that are targets of mEV-enriched miRs. MiR-630 mimics blocked TGFß1 induction of CDH2 in normal and IPF fibroblasts, and antagomiR-630 abrogated the effect of mEV on CDH2 expression. These data suggest that the interaction of Thy-1 with beta integrins mediates mEV uptake by lung fibroblasts, which blocks myofibroblastic differentiation, and that mEVs are enriched for miRs that target profibrotic genes up-regulated in IPF fibroblasts.


Subject(s)
Cell Differentiation/physiology , Extracellular Vesicles/metabolism , Fibroblasts/cytology , Mesenchymal Stem Cells/cytology , Myofibroblasts/cytology , Thy-1 Antigens/metabolism , Fibroblasts/metabolism , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Lung/cytology , Lung/metabolism , Mesenchymal Stem Cells/metabolism , Myofibroblasts/metabolism , Transforming Growth Factor beta1/pharmacology
4.
Blood ; 129(12): 1669-1679, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28096086

ABSTRACT

Clinical studies indicate that thrombocytopenia correlates with the development of severe falciparum malaria, suggesting that platelets either contribute to control of parasite replication, possibly as innate parasite killer cells or function in eliciting pathogenesis. Removal of platelets by anti-CD41 mAb treatment, platelet inhibition by aspirin, and adoptive transfer of wild-type (WT) platelets to CD40-KO mice, which do not control parasite replication, resulted in similar parasitemia compared with control mice. Human platelets at a physiologic ratio of 1 platelet to 9 red blood cells (RBCs) did not inhibit the in vitro development or replication of blood-stage Plasmodium falciparum The percentage of Plasmodium-infected (iRBCs) with bound platelets during the ascending parasitemia in Plasmodium chabaudi- and Plasmodium berghei-infected mice and the 48-hour in vitro cycle of P falciparum was <10%. P chabaudi and P berghei iRBCs with apoptotic parasites (TdT+) exhibited minimal platelet binding (<5%), which was similar to nonapoptotic iRBCs. These findings collectively indicate platelets do not kill bloodstage Plasmodium at physiologically relevant effector-to-target ratios. P chabaudi primary and secondary parasitemia was similar in mice depleted of platelets by mAb-injection just before infection, indicating that activation of the protective immune response does not require platelets. In contrast to the lack of an effect on parasite replication, adoptive transfer of WT platelets to CD40-KO mice, which are resistant to experimental cerebral malaria, partially restored experimental cerebral malaria mortality and symptoms in CD40-KO recipients, indicating platelets elicit pathogenesis and platelet CD40 is a key molecule.


Subject(s)
Blood Platelets/physiology , Malaria/immunology , Animals , Blood Platelets/parasitology , CD40 Antigens , Cells, Cultured , Erythrocytes/parasitology , Humans , Immunity, Cellular , Malaria/blood , Malaria, Cerebral/etiology , Mice , Plasmodium chabaudi
5.
Sci Rep ; 3: 1687, 2013.
Article in English | MEDLINE | ID: mdl-23595028

ABSTRACT

The ability to selectively block the entry of leukocytes into the central nervous system (CNS) without compromising the immune system is an attractive therapeutic approach for treating multiple sclerosis (MS). Using endothelial CD146-deficienct mice as a MS model, we found that endothelial CD146 plays an active role in the CNS-directed extravasation of encephalitogenic T cells, including CD146(+) TH1 and TH17 lymphocytes. Moreover, treating both active and passive MS models with the anti-CD146 antibody AA98 significantly decreased the infiltrated lymphocytes in the CNS and decreased neuroinflammation. Interestingly, the ability of AA98 to inhibit the migration of CD146(+) lymphocytes was dependent on targeting endothelial CD146, but not lymphocytic CD146. These results suggest a key molecular target located on the blood-brain barrier endothelium that mediates the extravasation of inflammatory cells into the CNS. In addition, our data suggest that the AA98 is a promising candidate for treating MS and other CNS autoimmune diseases.


Subject(s)
Brain/immunology , Endothelium, Vascular/immunology , Lymphocytes/immunology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Animals , Brain/drug effects , Brain/pathology , CD146 Antigen/immunology , Encephalitis/drug therapy , Encephalitis/immunology , Encephalitis/pathology , Endothelium, Vascular/drug effects , Lymphocytes/drug effects , Mice , Mice, Knockout , Multiple Sclerosis/pathology
6.
Methods Mol Biol ; 699: 85-95, 2011.
Article in English | MEDLINE | ID: mdl-21116980

ABSTRACT

The Enzyme-Linked Immunosorbent Assay (ELISA) and Western Blot analysis have been workhorse techniques for the analysis of protein levels and state, such as phosphorylation. The ELISA is also useful for measuring the affinity of a molecule for its ligand. The disadvantage of these techniques is that only a single protein can be analyzed for ELISA and a few (up to three) proteins for Western Blotting. Exact quantification is difficult with Western Blotting and changes are often reported as fold differences. We present here protocols for using fluorescent microspheres coated with the selected capture molecule to perform in essence several hundred mini ELISAs with each microsphere representing an ELISA; this reduces the variability of the assay. In addition, it is possible to analyze up to 100 analytes simultaneously using microspheres because each fluorescent microsphere exhibits distinct fluorescence in the red and far red channels: the fluorescence intensity in the channels in the red and far red channels (up to ten different intensities for each channel leading to a 10 × 10 matrix of intensities) constitutes the address for each analyte.


Subject(s)
Flow Cytometry/methods , Animals , Antibodies/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescence , Humans , Microspheres , Staining and Labeling/methods
7.
Methods Mol Biol ; 699: 337-54, 2011.
Article in English | MEDLINE | ID: mdl-21116991

ABSTRACT

Cell-derived microparticles (MPs) are increasingly recognized as important cell-to-cell signaling mechanisms and may exhibit important functions in homeostasis but also in pathogenesis. Indeed, MPs are associated with a number of diseases inhibiting their production that protects against pathogenesis. MPs are distinct from exosomes and apoptotic bodies, often exhibiting the membrane proteins of the activated or apoptotic cell from which they are derived. Electron microscopic analyses have shown that MPs are produced by all cell types tested to date, and ELISA-based assays have established that increased numbers of MPs are produced following cell activation. These approaches do not, however, determine the exact number of MPs and distribution of functional proteins on their surface. Flow cytometry represents an obvious approach to analyze MPs, and we present here a method to assess the number and phenotype of MPs by using a conventional flow cytometer. We also present the caveats with this method and describe a new imaging flow cytometry approach that overcomes these limitations.


Subject(s)
Cell-Derived Microparticles/metabolism , Flow Cytometry/methods , Animals , Antibodies/metabolism , Blood Cells/metabolism , Endothelial Cells/metabolism , Fluorescent Dyes/metabolism , Humans , Image Processing, Computer-Assisted , Mice , Rats , Staining and Labeling
8.
Infect Immun ; 78(10): 4331-40, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20660608

ABSTRACT

Blood-stage Plasmodium chabaudi infections are suppressed by antibody-mediated immunity and/or cell-mediated immunity (CMI). To determine the contributions of NK cells and γδ T cells to protective immunity, C57BL/6 (wild-type [WT]) mice and B-cell-deficient (J(H(-/-))) mice were infected with P. chabaudi and depleted of NK cells or γδ T cells with monoclonal antibody. The time courses of parasitemia in NK-cell-depleted WT mice and J(H(-/-)) mice were similar to those of control mice, indicating that deficiencies in NK cells, NKT cells, or CD8(+) T cells had little effect on parasitemia. In contrast, high levels of noncuring parasitemia occurred in J(H(-/-)) mice depleted of γδ T cells. Depletion of γδ T cells during chronic parasitemia in B-cell-deficient J(H(-/-)) mice resulted in an immediate and marked exacerbation of parasitemia, suggesting that γδ T cells have a direct killing effect in vivo on blood-stage parasites. Cytokine analyses revealed that levels of interleukin-10, gamma interferon (IFN-γ), and macrophage chemoattractant protein 1 (MCP-1) in the sera of γδ T-cell-depleted mice were significantly (P < 0.05) decreased compared to hamster immunoglobulin-injected controls, but these cytokine levels were similar in NK-cell-depleted mice and their controls. The time courses of parasitemia in CCR2(-/-) and J(H(-/-)) × CCR2(-/-) mice and in their controls were nearly identical, indicating that MCP-1 is not required for the control of parasitemia. Collectively, these data indicate that the suppression of acute P. chabaudi infection by CMI is γδ T cell dependent, is independent of NK cells, and may be attributed to the deficient IFN-γ response seen early in γδ T-cell-depleted mice.


Subject(s)
Killer Cells, Natural/physiology , Malaria/immunology , Plasmodium chabaudi , T-Lymphocyte Subsets/physiology , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cricetinae , Female , Gene Expression Regulation/immunology , Immunity, Cellular , Immunoglobulin G/pharmacology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Malaria/parasitology , Male , Mice , Mice, Knockout , Parasitemia/immunology , Time Factors
9.
Biol Blood Marrow Transplant ; 13(5): 530-42, 2007 May.
Article in English | MEDLINE | ID: mdl-17448912

ABSTRACT

Cyclic AMP (cAMP) is an important negative regulator of T cell activation, and an increased level of cAMP is associated with T cell hyporesponsiveness in vitro. We sought to determine whether elevating intracellular cAMP levels ex vivo in alloreactive T cells during primary mixed lymphocyte reactions (MLR) is sufficient to induce alloantigen-specific tolerance and prevent graft-versus-host disease (GVHD). Primary MLRs were treated with exogenous (8)Br-cAMP and IBMX, a compound that increases intracellular cAMP levels by inhibition of phosphodiesterases. T cell proliferation and IL-2 responsiveness in the treated primary MLR cultures were greatly reduced, and viable T cells recovered on day 8 also had impaired responses to restimulation with alloantigen compared to control-treated cells, but without an impairment to nonspecific mitogens. Labeling experiments showed that cAMP/IBMX inhibited alloreactive T cell proliferation by limiting the number of cell divisions, increasing susceptibility to apoptosis, and rendering nondeleted alloreactive T cells hyporesponsive to alloantigen restimulation. cAMP/IBMX-treated CD4(+) T cells had a markedly reduced capacity for GVHD lethality in major histocompatibility complex class II disparate recipients, but maintained the capacity to mediate other CD4(+) T cell responses in vivo. Thus, our results provide the first preclinical evidence of using cAMP-elevating pharmaceutical reagents to achieve long-term alloantigen-specific T cell tolerance that is sufficient to prevent GVHD.


Subject(s)
CD4-Positive T-Lymphocytes/transplantation , Cyclic AMP/metabolism , Graft vs Host Disease/prevention & control , Phosphodiesterase Inhibitors/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Transplantation , Cyclic AMP/analogs & derivatives , Graft vs Host Disease/immunology , Immune Tolerance , Isoantibodies/analysis , Isoantibodies/immunology , Lymphocyte Culture Test, Mixed/methods , Mice , Mice, Transgenic , Survival Analysis
10.
Am J Pathol ; 170(2): 505-17, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17255319

ABSTRACT

Microcirculatory changes and tissue oxygenation were investigated during Plasmodium berghei-induced severe malaria in the hamster window chamber model, which allows chronic, noninvasive investigation of the microvasculature in an awake animal. The main finding was that functional capillary density, a parameter reflecting tissue viability independent of tissue oxygenation, was reduced early during the course of disease and continued to decline to approximately 20% of baseline of uninfected controls on day 10 after infection. Parasitized red blood cells and leukocytes adhered to arterioles and venules but did not affect overall blood flow, and there was little evidence of complete obstruction of blood flow. According to the sequestration hypothesis, obstruction of blood flow by adherent parasitized erythrocytes is the cause of tissue hypoxia and, eventually, cell death in severe malaria. Tissue oxygen tensions were lower on day 10 of infection when the animals were moribund compared with uninfected controls, but this level was markedly higher than the lethal threshold. No necrotic cells labeled with propidium iodide were detected in moribund animals on day 10 after infection. We therefore conclude that loss of functional capillaries rather than tissue hypoxia is a major lethal event in severe malaria.


Subject(s)
Malaria/physiopathology , Oxygen Consumption , Plasmodium berghei , Animals , Capillaries/parasitology , Capillaries/physiopathology , Cricetinae , Erythrocytes/parasitology , Hypoxia/parasitology , Hypoxia/physiopathology , Leukocytes , Malaria/parasitology , Mesocricetus
11.
Cytometry A ; 71(4): 242-50, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17252581

ABSTRACT

BACKGROUND: Measuring antibody production in response to antigen exposure or vaccination is key to disease prevention and treatment. Our understanding of the mechanisms involved in the antibody response is limited by a lack of sensitive analysis methods. We address this limitation using multiplexed microsphere arrays for the semi -quantitative analysis of antibody production in response to malaria infection. METHODS: We used microspheres as solid supports on which to capture and analyze circulating antibodies. Antigen immobilized on beads captured antigen-specific antibodies for semi- quantitative analysis using fluorescent secondary antibodies. Anti-immunoglobulin antibodies on beads captured specific antibody isotypes for affinity estimation using fluorescent antigen. RESULTS: Antigen-mediated capture of plasma antibodies enables determination of antigen-specific antibody "titer," a semi-quantitative parameter describing a convolution of antibody abundance and avidity, as well as parameters describing numbers of antibodies bound/bead at saturation and the plasma concentration-dependent approach to saturation. Results were identical in single-plex and multiplex assays, and in qualitative agreement with similar parameters derived from ELISA-based assays. Isotype-specific antibody-mediated capture of plasma antibodies allowed the estimation of the affinity of antibody for antigen. CONCLUSION: Analysis of antibody responses using microspheres and flow cytometry offer significant advantages in speed, sample size, and quantification over standard ELISA-based titer methods.


Subject(s)
Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Malaria/immunology , Animals , Antibodies, Protozoan/immunology , Antigens, Protozoan/blood , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunoglobulin Isotypes/blood , Immunoglobulin Isotypes/immunology , Malaria/metabolism , Membrane Proteins/blood , Membrane Proteins/immunology , Merozoite Surface Protein 1/immunology , Mice , Mice, Inbred C57BL , Microspheres , Models, Animal , Models, Biological , Plasmodium chabaudi/immunology , Protozoan Proteins/blood , Protozoan Proteins/immunology
12.
Nat Med ; 12(12): 1417-22, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17099710

ABSTRACT

The role of nitric oxide (NO) in the genesis of cerebral malaria is controversial. Most investigators propose that the unfortunate consequence of the high concentrations of NO produced to kill the parasite is the development of cerebral malaria. Here we have tested this high NO bioavailability hypothesis in the setting of experimental cerebral malaria (ECM), but find instead that low NO bioavailability contributes to the genesis of ECM. Specifically, mice deficient in vascular NO synthase showed parasitemia and mortality similar to that observed in control mice. Exogenous NO did not affect parasitemia but provided marked protection against ECM; in fact, mice treated with exogenous NO were clinically indistinguishable from uninfected mice at a stage when control infected mice were moribund. Administration of exogenous NO restored NO-mediated signaling in the brain, decreased proinflammatory biomarkers in the blood, and markedly reduced vascular leak and petechial hemorrhage into the brain. Low NO bioavailability in the vasculature during ECM was caused in part by an increase in NO-scavenging free hemoglobin in the blood, by hypoargininemia, and by low blood and erythrocyte nitrite concentrations. Exogenous NO inactivated NO-scavenging free hemoglobin in the plasma and restored nitrite to concentrations observed in uninfected mice. We therefore conclude that low rather than high NO bioavailability contributes to the genesis of ECM.


Subject(s)
Malaria, Cerebral/etiology , Nitric Oxide/metabolism , Alkenes/administration & dosage , Alkenes/blood , Animals , Arginine/blood , Blood Pressure/drug effects , Capillary Permeability/drug effects , Cyclic GMP/metabolism , Hemoglobins/analysis , Malaria, Cerebral/metabolism , Malaria, Cerebral/mortality , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Models, Biological , Nitric Oxide/blood , Nitric Oxide Synthase Type II/genetics , Nitrites/blood , Plasmodium berghei , Tissue Distribution
13.
Trends Parasitol ; 22(11): 503-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16979941

ABSTRACT

A unifying hypothesis for the genesis of cerebral malaria proposes that parasite antigens (released by replication in blood, surface molecules on parasitized erythrocytes, or merozoites) activate platelets that, in turn, contribute to the activation of the inflammatory response and increased levels of endothelial cell adhesion molecules (eCAMs). Increased levels of eCAMs result in further parasitized-erythrocyte sequestration and marked local inflammation that might disrupt the brain microvasculature, which cannot be repaired by the hemostasis system because of its procoagulant state. Disruption of the brain microvasculature can result in vascular leak and/or hemorrhaging into the brain; similar processes can occur in other vascular beds, including the lung. The blockage of functional capillaries by parasitized and/or unparasitized erythrocytes with decreased deformability or rosettes is also a key interaction between hemostasis and mechanical obstruction leading to pathogenesis. The events resulting in the development of cerebral malaria complications are multi-factorial, encompassing a dynamic interaction between three processes, thereby explaining the complexity of this deadly syndrome.


Subject(s)
Brain/blood supply , Cell Adhesion Molecules/metabolism , Cerebrovascular Circulation , Erythrocytes/parasitology , Malaria, Cerebral/pathology , Malaria, Falciparum/pathology , Animals , Brain/parasitology , Cell Adhesion Molecules/analysis , Endothelium, Vascular , Humans , Inflammation/complications , Inflammation/etiology , Malaria, Cerebral/parasitology , Malaria, Falciparum/parasitology , Plasmodium falciparum/pathogenicity
14.
J Immunol ; 175(11): 7699-707, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16301680

ABSTRACT

Experimental malarial thrombocytopenia can reach life-threatening levels and is believed to be due to Abs targeting platelets for destruction by the reticuloendothelial system. However, we report that Abs account for at most 15% of platelet destruction as Plasmodium berghei-infected B cell-deficient mice exhibited profound thrombocytopenia (83%) as did C57BL/6 controls (98%). Further, no significant increase in Abs bound to intact platelets was observed during infection. P. berghei infection can enhance the activity of anti-platelet Abs as indicated by a significantly (p < 0.005) increased thrombocytopenia on day 4 of infection in mice that were administered a low dose anti-CD41 mAb compared with rat IgG1-injected controls. RAG1-/- and CD4- plus CD8-deficient mice were markedly protected from thrombocytopenia (p < 0.005) and malarial pathogenesis. CD8- or TCRgammadelta-deficient mice were not protected from thrombocytopenia and CD4-deficient mice were modestly protected. RAG1-/- mice exhibited significantly (p < 0.05) lower levels of plasma TNF, IFN-gamma, and IL-12 during infection. IFNgamma-/- and IL-12-/- mice exhibited increased survival but similar thrombocytopenia to C57BL/6 controls. Collectively, these data indicate that thrombocytopenia is necessary but not sufficient for malarial pathogenesis and Abs are not the major contributors to malarial thrombocytopenia. Rather, we propose that both CD4+ and CD8+ T cell populations play key roles in malarial thrombocytopenia; a complex bidirectional interaction between cell-mediated immunity and platelets exists during experimental severe malaria that regulates both responses.


Subject(s)
Antibody Formation , Immunity, Cellular , Malaria/immunology , Plasmodium berghei/immunology , Thrombocytopenia/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Flow Cytometry , Interferon-gamma/blood , Interleukin-12/blood , Lymphocyte Depletion , Malaria/complications , Mice , Platelet Membrane Glycoprotein IIb/immunology , Thrombocytopenia/etiology , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/immunology
15.
Infect Immun ; 73(10): 6704-10, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16177347

ABSTRACT

Reactive oxygen species (ROS) are widely believed to kill malarial parasites. C57BL/6 mice injected with P. berghei inocula incubated with supraphysiological doses of NO (< or =150 microM) or with peroxynitrite (220 microM), however, exhibited parasitemia similar to that seen with those given control inocula, and there was no difference in disease development. Only treatment of inocula with NO doses nearing saturation (> or =1.2 mM) resulted in no detectable parasitemia in the recipients; flow cytometric analysis with a vital dye (hydroethidine) indicated that 1.5 mM NO lysed the erythrocytes rather than killing the parasites. The hemoglobin level in the inocula was about 8 muM; the hemoglobin was mainly oxyhemoglobin (oxyHb) (96%), which was converted to methemoglobin (>95%) after treatment with 150 microM NO. The concentrations of 150 microM of NO and 220 microM of peroxynitrite were far in excess of the hemoglobin concentration (approximately 8 microM), and yet no parasite killing was detected. We therefore conclude that hemoglobin protects Plasmodium parasites from ROS, but the parasite likely possesses intrinsic defense mechanisms against ROS.


Subject(s)
Malaria/parasitology , Nitric Oxide/pharmacology , Oxidative Stress , Plasmodium berghei/drug effects , Reactive Oxygen Species/pharmacology , Animals , Erythrocytes/drug effects , Hemoglobins/analysis , Hemolysis , Mice , Mice, Inbred C57BL , Nitric Oxide/administration & dosage , Parasitemia/parasitology , Peroxynitrous Acid/administration & dosage , Peroxynitrous Acid/pharmacology , Plasmodium berghei/pathogenicity , Reactive Oxygen Species/administration & dosage
16.
J Investig Med ; 53(5): 246-52, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16042958

ABSTRACT

Our understanding of how the host immune response kills Plasmodium, the causative agent of malaria, is limited and controversial. One widely held belief is that reactive oxygen species are crucial for controlling parasite replication. One of the hallmarks of blood-stage malaria is the cyclic rupture of erythrocytes by the parasite, which releases free hemoglobin into the circulation. We propose that this free hemoglobin, as well as the hemoglobin within the erythrocyte and surrounding the parasite, effectively shields Plasmodium from reactive oxygen species well in excess of those achievable in vivo.


Subject(s)
Free Radical Scavengers/metabolism , Hemoglobins/metabolism , Nitric Oxide/metabolism , Plasmodium/physiology , Reactive Oxygen Species/metabolism , Animals , Erythrocytes/parasitology , Host-Parasite Interactions
17.
Trends Parasitol ; 21(9): 415-22, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16039159

ABSTRACT

Rational development of adjunct or anti-disease therapy for severe Plasmodium falciparum malaria requires cellular and molecular definition of malarial pathogenesis. Nitric oxide (NO) is a potential target for such therapy but its role during malaria is controversial. It has been proposed that NO is produced at high levels to kill Plasmodium parasites, although the unfortunate consequence of elevated NO levels might be impaired neuronal signaling, oxidant damage and red blood cell damage that leads to anemia. In this case, inhibitors of NO production or NO scavengers might be an effective adjunct therapy. However, increasing amounts of evidence support the alternate hypothesis that NO production is limited during malaria. Furthermore, the well-documented NO scavenging by cell-free plasma hemoglobin and superoxide, the levels of which are elevated during malaria, has not been considered. Low NO bioavailability in the vasculature during malaria might contribute to pathologic activation of the immune system, the endothelium and the coagulation system: factors required for malarial pathogenesis. Therefore, restoring NO bioavailability might represent an effective anti-disease therapy.


Subject(s)
Malaria, Falciparum/parasitology , Nitric Oxide/metabolism , Plasmodium falciparum/metabolism , Anemia/blood , Anemia/parasitology , Animals , Biological Availability , Hemoglobins/metabolism , Humans , Malaria, Falciparum/blood , Malaria, Falciparum/metabolism , Malaria, Falciparum/prevention & control , Nitric Oxide/biosynthesis , Nitric Oxide/physiology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Superoxides/metabolism
18.
Blood ; 105(5): 1956-63, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15494426

ABSTRACT

Accumulating evidence indicates that platelets play a critical role in the pathogenesis of experimental severe malaria (ESM) elicited by infection with Plasmodium berghei. Mice injected on day 1 of P berghei infection (early) with either anti-CD41 or anti-CD61 monoclonal antibodies (mAbs) exhibited significantly (P<.001) increased survival from ESM compared with infection controls, indicating that platelets function early in the disease. In contrast, groups of mice treated on days 4, 5, and 6 (late) with anti-CD41 mAb exhibited similar mortality as controls. Because platelet depletion by anti-CD41 mAb on day 4 of infection did not protect mice, and platelet adherence occurs on day 6, platelet adherence to endothelium is not required to mediate malarial pathogenesis. Few platelet microparticles were detected in the blood during the course of malaria, but large numbers of erythrocyte vesicles, microparticles, and debris were detected. The protective effect of early anti-CD41 mAb treatment was independent of the number of platelets, platelet microparticles, erythrocyte-platelet conjugates, and erythrocyte vesicles. Mice treated early with anti-CD41 mAb exhibited markedly altered cytokine production on day 4 of P berghei infection (increased interleukin 10 [IL-10], IL-1alpha, IL-6, interferon-gamma [IFN-gamma], and tumor necrosis factor alpha [TNF-alpha]; decreased IL-2) but no decline in coagulation factors compared with rat immunoglobulin G (IgG)-treated controls, indicating that platelets regulate the levels of pathogenic cytokines.


Subject(s)
Antibodies, Monoclonal/pharmacology , Blood Platelets/physiology , Malaria/etiology , Platelet Membrane Glycoprotein IIb/physiology , Animals , Antibodies, Monoclonal/administration & dosage , Blood Platelets/drug effects , Cell Adhesion , Cytokines/biosynthesis , Disease Models, Animal , Female , Integrin beta3/immunology , Malaria/blood , Malaria/drug therapy , Mice , Mice, Inbred C57BL , Particle Size , Plasmodium berghei , Platelet Adhesiveness , Platelet Count , Platelet Membrane Glycoprotein IIb/immunology , Time
19.
Blood ; 101(12): 5076-83, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12609834

ABSTRACT

We previously reported that interleukin-10 (IL-10) and transforming growth factor (TGF)-beta treatment of primary mixed lymphocyte reaction (MLR) cultures resulted in secondary alloantigen-specific hyporesponsiveness and protection from graft-versus-host disease (GVHD) lethality. Here, we report that CD4+ T cells recovered from the IL-10- and TGF-beta-treated primary MLR cultures have immunoregulatory function. Tolerized cells significantly inhibited proliferation of naive alloreactive CD4+ T cells in a primary MLR. Inhibition of the naive alloresponse was observed with as few as 1 tolerized cell to 10 naive responder cells. Tolerized cells were able to significantly reduce GVHD lethality when injected with naive alloreactive CD4+ T cells into major histocombatibility class (MHC) II disparate recipients. Rigorous CD25 depletion of the primary MLR had no effect on generation of a regulatory capacity, suggesting that the regulatory cells likely originated from CD4+CD25- T cells. Immune suppression was mediated independently of IL-10 and TGF-beta production, as neutralizing antibodies for IL-10, IL-10R, and TGF-beta were unable to revert suppression, and IL-10- deficient CD4+ T cells were able to mediate in vitro and in vivo suppression. The generation of immunoregulatory cells from a CD4+CD25- population during tolerization with IL-10 and TGF-beta provides an additional mechanism to prevent GVHD lethality by T cells that may escape full tolerance induction.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immune Tolerance , Interleukin-10/pharmacology , Isoantigens/immunology , Receptors, Interleukin-2/analysis , Transforming Growth Factor beta/pharmacology , Animals , Antibodies/pharmacology , Flow Cytometry , Graft vs Host Disease/mortality , Graft vs Host Disease/prevention & control , Interleukin-10/immunology , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred C57BL , Receptors, Interleukin/immunology , Receptors, Interleukin-10 , Transforming Growth Factor beta/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...