31 research outputs found

    HlSRB, a Class B Scavenger Receptor, Is Key to the Granulocyte-Mediated Microbial Phagocytosis in Ticks

    Get PDF
    Ixodid ticks transmit various pathogens of deadly diseases to humans and animals. However, the specific molecule that functions in the recognition and control of pathogens inside ticks is not yet to be identified. Class B scavenger receptor CD36 (SRB) participates in internalization of apoptotic cells, certain bacterial and fungal pathogens, and modified low-density lipoproteins. Recently, we have reported on recombinant HlSRB, a 50-kDa protein with one hydrophobic SRB domain from the hard tick, Haemaphysalis longicornis. Here, we show that HlSRB plays vital roles in granulocyte-mediated phagocytosis to invading Escherichia coli and contributes to the first-line host defense against various pathogens. Data clearly revealed that granulocytes that up-regulated the expression of cell surface HlSRB are almost exclusively involved in hemocyte-mediated phagocytosis for E. coli in ticks, and post-transcriptional silencing of the HlSRB-specific gene ablated the granulocytes' ability to phagocytose E. coli and resulted in the mortality of ticks due to high bacteremia. This is the first report demonstrating that a scavenger receptor molecule contributes to hemocyte-mediated phagocytosis against exogenous pathogens, isolated and characterized from hematophagous arthropods

    Potential of Vitamin E Deficiency, Induced by Inhibition of α-Tocopherol Efflux, in Murine Malaria Infection

    No full text
    Although epidemiological and experimental studies have suggested beneficial effects of vitamin E deficiency on malaria infection, it has not been clinically applicable for the treatment of malaria owing to the significant content of vitamin E in our daily food. However, since α-tocopherol transfer protein (α-TTP) has been shown to be a determinant of vitamin E level in circulation, manipulation of α-tocopherol levels by α-TTP inhibition was considered as a potential therapeutic strategy for malaria. Knockout studies in mice indicated that inhibition of α-TTP confers resistance against malaria infections in murines, accompanied by oxidative stress-induced DNA damage in the parasite, arising from vitamin E deficiency. Combination therapy with chloroquine and α-TTP inhibition significantly improved the survival rates in murines with malaria. Thus, clinical application of α-tocopherol deficiency could be possible, provided that α-tocopherol concentration in circulation is reduced. Probucol, a recently found drug, induced α-tocopherol deficiency in circulation and was effective against murine malaria. Currently, treatment of malaria relies on the artemisinin-based combination therapy (ACT); however, when mice infected with malarial parasites were treated with probucol and dihydroartemisinin, the beneficial effect of ACT was pronounced. Protective effects of vitamin E deficiency might be extended to manage other parasites in future

    ローデントマラリアに対するプロブコールの効果

    Get PDF
    博士(獣医学)岐阜大学(Gifu University

    Probucol-Induced α-Tocopherol Deficiency Protects Mice against Malaria Infection

    No full text
    <div><p>The emergence of malaria pathogens having resistance against antimalarials implies the necessity for the development of new drugs. Recently, we have demonstrated a resistance against malaria infection of α-tocopherol transfer protein knockout mice showing undetectable plasma levels of α-tocopherol, a lipid-soluble antioxidant. However, dietary restriction induced α-tocopherol deficiency is difficult to be applied as a clinical antimalarial therapy. Here, we report on a new strategy to potentially treat malaria by using probucol, a drug that can reduce the plasma α-tocopherol concentration. Probucol pre-treatment for 2 weeks and treatment throughout the infection rescued from death of mice infected with <i>Plasmodium yoelii</i> XL-17 or <i>P</i>. <i>berghei</i> ANKA. In addition, survival was extended when the treatment started immediately after parasite inoculation. The ratio of lipid peroxidation products to parent lipids increased in plasma after 2 weeks treatment of probucol. This indicates that the protective effect of probucol might be mediated by the oxidative stressful environment induced by α-tocopherol deficiency. Probucol in combination with dihydroartemisin suppressed the proliferation of <i>P</i>. <i>yoelii</i> XL-17. These results indicated that probucol might be a candidate for a drug against malaria infection by inducing α-tocopherol deficiency without dietary α-tocopherol restriction.</p></div

    Plasma concentrations of α-tocopherol and cholesterol decreased after probucol treatment.

    No full text
    <p>A, Experimental design for α-tocopherol deficiency induction using 1% w/w probucol in the diet. Six-week-old C57BL/6J mice were treated with 1% w/w probucol for 2 weeks. Then, mice were withdrawn from the probucol diet and changed to a standard diet for 2 weeks. Plasma and erythrocyte samples were obtained at day 0, 1, 2, 4, 7, and 14 after starting probucol treatment (n = 5 per group) and 1 or 2 weeks post-withdrawal (n = 3 per group). In addition, the plasma of eight-week-old α-tocopherol transfer protein knockout (α-ttp<sup>Δ</sup>) mice fed a standard diet was obtained (n = 3). Plasma α-tocopherol concentrations were measured after probucol treatment (B) and after withdrawal (C). The levels of α-tocopherol in erythrocytes were normalized with the protein concentration (n = 5 per group) (D). Plasma cholesterol concentrations were measured by using the cholesterol E-test after 2 weeks of probucol treatment and after withdrawal (n = 3) (E). All data are expressed as mean ± SE. Statistical analysis was carried out by analysis of variance (ANOVA; multiple comparisons Tukey’s test). *p < 0.05.</p
    corecore