12 research outputs found

    Curcumin-Arteether Combination Therapy of Plasmodium berghei-Infected Mice Prevents Recrudescence Through Immunomodulation

    Get PDF
    Earlier studies in this laboratory have shown the potential of artemisinin-curcumin combination therapy in experimental malaria. In a parasite recrudescence model in mice infected with Plasmodium berghei (ANKA), a single dose of alpha,beta-arteether (ART) with three oral doses of curcumin prevented recrudescence, providing almost 95% protection. The parasites were completely cleared in blood with ART-alone (AE) or ART+curcumin (AC) treatments in the short-term, although the clearance was faster in the latter case involving increased ROS generation. But, parasites in liver and spleen were not cleared in AE or AC treatments, perhaps, serving as a reservoir for recrudescence. Parasitemia in blood reached up to 60% in AE-treated mice during the recrudescence phase, leading to death of animals. A transient increase of up to 2–3% parasitemia was observed in AC-treatment, leading to protection and reversal of splenomegaly. A striking increase in spleen mRNA levels for TLR2, IL-10 and IgG-subclass antibodies but a decrease in those for INFγ and IL-12 was observed in AC-treatment. There was a striking increase in IL-10 and IgG subclass antibody levels but a decrease in INFγ levels in sera leading to protection against recrudescence. AC-treatment failed to protect against recrudescence in TLR2−/− and IL-10−/− animals. IL-10 injection to AE-treated wild type mice and AC-treated TLR2−/− mice was able to prolong survival. Blood from the recrudescence phase in AE-treatment, but not from AC-treatment, was able to reinfect and kill naïve animals. Sera from the recrudescence phase of AC-treated animals reacted with several parasite proteins compared to that from AE-treated animals. It is proposed that activation of TLR2-mediated innate immune response leading to enhanced IL-10 production and generation of anti-parasite antibodies contribute to protective immunity in AC-treated mice. These results indicate a potential for curcumin-based combination therapy to be tested for prevention of recrudescence in falciparum and relapse in vivax malaria

    Nanocurcumin is superior to native curcumin in preventing degenerative changes in Experimental Cerebral Malaria

    No full text
    Curcumin has many pharmacological activities despite its poor bioavailability and in vivo stability. Here, we show that a nanoformulated curcumin (PLGA-curcumin) has better therapeutic index than native curcumin in preventing the onset of neurological symptoms and delaying the death of mice in experimental cerebral malaria. Oral PLGA-curcumin was at least as effective as native curcumin at a 15-fold lower concentration in preventing the breakdown of blood-brain barrier and inhibition of brain mRNAs for inflammatory cytokines, chemokine receptor CXCR3 and its ligand CXCL10, with an increase in the anti-inflammatory cytokine IL-10. This was also reflected in serum cytokine and chemokine levels. At equivalent concentrations, a single oral dose of PLGA-curcumin was more effective in inhibiting serum IFN gamma levels and enhancing IL-10 levels than native curcumin. Even at low concentrations, PLGA-curcumin was superior to native curcumin in inhibiting the sequestration of parasitized-RBCs and CD8(+) T cells in the brain. A single oral dose of 5 mg PLGA-curcumin containing 350 mu g of curcumin resulted in 3-4 fold higher concentration and prolonged presence of curcumin in the brain than that obtained with 5 mg of native curcumin, indicating better bioavailability of PLGA-curcumin. PLGA-curcumin has potential as an adjunct drug to treat human cerebral malaria

    Simultaneously targeting inflammatory response and parasite sequestration in brain to treat Experimental Cerebral Malaria

    No full text
    Malaria afflicts around 200 million people annually, with a mortality number close to 600,000. The mortality rate in Human Cerebral Malaria (HCM) is unacceptably high (15-20%), despite the availability of artemisinin-based therapy. An effective adjunct therapy is urgently needed. Experimental Cerebral Malaria (ECM) in mice manifests many of the neurological features of HCM. Migration of T cells and parasite-infected RBCs (pRBCs) into the brain are both necessary to precipitate the disease. We have been able to simultaneously target both these parameters of ECM. Curcumin alone was able to reverse all the parameters investigated in this study that govern inflammatory responses, CD8(+) T cell and pRBC sequestration into the brain and blood brain barrier (BBB) breakdown. But the animals eventually died of anemia due to parasite build-up in blood. However, arteether-curcumin (AC) combination therapy even after the onset of symptoms provided complete cure. AC treatment is a promising therapeutic option for HCM

    Preexisting helminth challenge exacerbates infection and reactivation of gammaherpesvirus in tissue resident macrophages.

    No full text
    Even though gammaherpesvirus and parasitic infections are endemic in parts of the world, there is a lack of understanding about the outcome of coinfection. In humans, coinfections usually occur sequentially, with fluctuating order and timing in different hosts. However, experimental studies in mice generally do not address the variables of order and timing of coinfections. We sought to examine the variable of coinfection order in a system of gammaherpesvirus-helminth coinfection. Our previous work demonstrated that infection with the intestinal parasite, Heligmosomoides polygyrus, induced transient reactivation from latency of murine gammaherpesvirus-68 (MHV68). In this report, we reverse the order of coinfection, infecting with H. polygyrus first, followed by MHV68, and examined the effects of preexisting parasite infection on MHV68 acute and latent infection. We found that preexisting parasite infection increased the propensity of MHV68 to reactivate from latency. However, when we examined the mechanism for reactivation, we found that preexisting parasite infection increased the ability of MHV68 to reactivate in a vitamin A dependent manner, a distinct mechanism to what we found previously with parasite-induced reactivation after latency establishment. We determined that H. polygyrus infection increased both acute and latent MHV68 infection in a population of tissue resident macrophages, called large peritoneal macrophages. We demonstrate that this population of macrophages and vitamin A are required for increased acute and latent infection during parasite coinfection

    Effect of AE and AC- treatments on parasitemia in mice infected with <i>P.berghei</i> for 72 hr.

    No full text
    <p>(A) Changes in parasite load in blood at different time periods through real-time PCR analysis of parasite 18S rRNA after drug treatment. UT, untreated; C, curcumin, AE, ART alone; AC, ART+CUR. D, died; N, not detectable. Data provided represent Mean ± S.D. from three animals. The whole series was repeated thrice (total of 9 animals in each group) and number of animals surviving on day 30 in each group were as follows: UT, 0; C, 0; AE, 1; AC, 9. (B) Semiquantitative RT-PCR analysis of parasite 18S rRNA in blood, liver and spleen on day 10 and 15 after infection. GAPDH was used as a control. *, positive control. (C) Effect of injection of blood from recrudescing animals after AE and AC-treatments into naïve animals. Five animals were used in each group.</p

    Effect of AE and AC-treatments on changes in serum cytokine levels and survival response of TLR2<sup>−/−</sup> and IL-10<sup>−/−</sup> mice to <i>P.berghei</i>-infection.

    No full text
    <p>(A) Changes in the serum levels of INFγ, IL-10 and IL-12. The data represent Mean + S.D. from three sera preparations and collected during day 6 (D6) to day 23 (D23) at intervals. The day 0 values correspond to those of uninfected animals. The values (pg/ml) corresponding to infected animals, which all died on day 6, are as follows: INFγ, 274±22; IL-10, 422±29; IL-12, 510±39. The values corresponding to infected animals treated with curcumin alone and which all died between, 7–8 days are as follows: INFγ, 243±22; IL-10, 310±33; IL-12, 377±39. (B) Effect of IL-10 injection in AE-treated animals. IL-10 was injected on alternate days from day 10 in five doses ranging from 10 ng to 200 ng. Alternate i.p. and i.v. routes were used. (C) Survival response of <i>P.berghei</i>infected C57BL/6 mice to AE and AC-treatments. The data are from two experiments with five animals in each group. (D) Survival response of TLR<sup>−/−</sup> animals with and without IL-10 injection (100 ng per animal for 5 days). (E) Survival response of IL-10<sup>−/−</sup> animals. Four animals were used in each group for the experiments in (B), (D) and (E). The knock-out animals were in C57BL/6 background. While, the animals were injected with around 10<sup>4</sup> parasites in general, the IL 10<sup>−/−</sup> animals received around 10<sup>3</sup> parasites. UI, unifected; I, infected; UT, untreated; C, curcumin; AE, ART alone; AC, ART+CUR.</p

    Effect of AE and AC-treatments on changes in spleen mass and mRNA levels of cytokines, TLRs and IgG-sub-class antibodies in <i>P.berghei</i>-infected mice.

    No full text
    <p>(A) Changes in spleen weight through out the course of infection and drug treatments. Data provided represent Mean ± S.D. from five animals. *, infected animals died 5/6 day (B) Semi-quantitative RT-PCR analysis of spleen RNA on day 14. UI, unifected; I, infected; AE, ART alone; AC, ART+CUR.</p

    Effect of AE and AC-treatments on hemozoin content and ROS generation in <i>P.berghei</i>infected mice.

    No full text
    <p>(A). Hemozoin content in parasite 8 hr and 12 hr after a single injection of ART and one oral dose of curcumin. The data represent Mean ± S.D. from three animals. (B) FACS analysis of <i>P.berghi</i>–infected red blood cells for ROS measurement 12 hr after drug treatment as per (A).</p

    Effect of AE and AC-treatments on changes in serum total IgG and IgG-subclass antibody levels and Western analysis of parasite proteins with such sera in <i>P.berghei</i>-infected mice.

    No full text
    <p>Antibody levels were quantified in sera by ELISA using microtitre plates coated with parasite lysate from day 6 (D6) onwards. (A) Changes in the serum levels of total IgG and IgG-subclass antibodies against parasite lysate. The data represent Mean ± S.D. from three sera preparations (pooled from two animals each) and collected during day 6 (D6) to day 23 (D23) at intervals. The day 0 values correspond to those of uninfected animals. The values (A<sub>405</sub>) corresponding to infected animals, which all died on day 6, are as follows: IgG, 0.07±0.002; IgG1, 0.03±0.005; IgG2a, not detectable; IgG2b, not detectable; IgG3, not detectable; IgM, 0.22±0.007. (B) Anamnestic response of AC-treated animals. The animals were injected with fresh parasitized blood 10 days before the day mentioned in the Figure. The antibody titre (IgG) was measured before and after challenge. The data represent an average from two sera preparations. (C) Western blot analysis of parasite proteins with the different sera preparations. 1, uninfected; 2, day 5- infected; 3, AE -day 22; 4, AC day-22; 5 and 6, AC - day 75, before and after challenge; 7 and 8; AC - day 180, before and after challenge.</p
    corecore