Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-28983468

ABSTRACT

A major constraint in the study of Plasmodium falciparum malaria, including vaccine development, lies on the parasite's strict human host specificity and therefore the shortage of animal experimental models able to harbor human plasmodia. The best experimental models are neo-tropical primates of the genus Saimiri and Aotus, but they require splenectomy to reduce innate defenses for achieving high and consistent parasitemias, an important limitation. Clodronate-liposomes (CL) have been successfully used to deplete monocytes/macrophages in several experimental models. We investigated whether a reduction in the numbers of phagocytic cells by CL would improve the development of P. falciparum parasitemia in non-splenectomized Saimiri sciureus monkeys. Depletion of S. sciureus splenocytes after in vitro incubation with CL was quantified using anti-CD14 antibodies and flow cytometry. Non-infected and P. falciparum-infected S. sciureus were injected intravenously twice a week with either CL at either 0.5 or 1 mL (5 mg/mL) or phosphate buffered saline (PBS). Animals were monitored during infection and treated with mefloquine. After treatment and euthanasia, spleen and liver were collected for histological analysis. In vitro CL depleted S. sciureus splenic monocyte/macrophage population in a dose- and time-dependent manner. In vivo, half of P. falciparum-infected S. sciureus treated with CL 0.5 mL, and two-thirds of those treated with CL 1 mL developed high parasitemias requiring mefloquine treatment, whereas all control animals were able to self-control parasitemia without the need for antimalarial treatment. CL-treated infected S. sciureus showed a marked decrease in the degree of splenomegaly despite higher parasitemias, compared to PBS-treated animals. Histological evidence of partial monocyte/macrophage depletion, decreased hemozoin phagocytosis and decreased iron recycling was observed in both the spleen and liver of CL-treated infected S. sciureus. CL is capable of promoting higher parasitemia in P. falciparum-infected S. sciureus, associated with evidence of partial macrophage depletion in the spleen and liver. Macrophage depletion by CL is therefore a practical and viable alternative to surgical splenectomy in this experimental model.


Subject(s)
Clodronic Acid/pharmacology , Macrophages/drug effects , Malaria, Falciparum/parasitology , Monocytes/drug effects , Parasitemia/chemically induced , Plasmodium falciparum/growth & development , Animals , Clodronic Acid/administration & dosage , Disease Models, Animal , Female , Haplorhini , Humans , Liposomes , Liver/cytology , Male , Phagocytosis/drug effects , Saimiri , Spleen/cytology , Time Factors
2.
Cochrane Database Syst Rev ; 2: CD006589, 2016 Feb 27.
Article in English | MEDLINE | ID: mdl-26921618

ABSTRACT

BACKGROUND: Iron-deficiency anaemia is common during childhood. Iron administration has been claimed to increase the risk of malaria. OBJECTIVES: To evaluate the effects and safety of iron supplementation, with or without folic acid, in children living in areas with hyperendemic or holoendemic malaria transmission. SEARCH METHODS: We searched the Cochrane Infectious Diseases Group Specialized Register; the Cochrane Central Register of Controlled Trials (CENTRAL), published in the Cochrane Library, MEDLINE (up to August 2015) and LILACS (up to February 2015). We also checked the metaRegister of Controlled Trials (mRCT) and World Health Organization International Clinical Trials Registry Platform (WHO ICTRP) up to February 2015. We contacted the primary investigators of all included trials, ongoing trials, and those awaiting assessment to ask for unpublished data and further trials. We scanned references of included trials, pertinent reviews, and previous meta-analyses for additional references. SELECTION CRITERIA: We included individually randomized controlled trials (RCTs) and cluster RCTs conducted in hyperendemic and holoendemic malaria regions or that reported on any malaria-related outcomes that included children younger than 18 years of age. We included trials that compared orally administered iron, iron with folic acid, and iron with antimalarial treatment versus placebo or no treatment. We included trials of iron supplementation or fortification interventions if they provided at least 80% of the Recommended Dietary Allowance (RDA) for prevention of anaemia by age. Antihelminthics could be administered to either group, and micronutrients had to be administered equally to both groups. DATA COLLECTION AND ANALYSIS: The primary outcomes were clinical malaria, severe malaria, and death from any cause. We assessed the risk of bias in included trials with domain-based evaluation and assessed the quality of the evidence using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. We performed a fixed-effect meta-analysis for all outcomes and random-effects meta-analysis for hematological outcomes, and adjusted analyses for cluster RCTs. We based the subgroup analyses for anaemia at baseline, age, and malaria prevention or management services on trial-level data. MAIN RESULTS: Thirty-five trials (31,955 children) met the inclusion criteria. Overall, iron does not cause an excess of clinical malaria (risk ratio (RR) 0.93, 95% confidence intervals (CI) 0.87 to 1.00; 14 trials, 7168 children, high quality evidence). Iron probably does not cause an excess of clinical malaria in both populations where anaemia is common and those in which anaemia is uncommon. In areas where there are prevention and management services for malaria, iron (with or without folic acid) may reduce clinical malaria (RR 0.91, 95% CI 0.84 to 0.97; seven trials, 5586 participants, low quality evidence), while in areas where such services are unavailable, iron (with or without folic acid) may increase the incidence of malaria, although the lower CIs indicate no difference (RR 1.16, 95% CI 1.02 to 1.31; nine trials, 19,086 participants, low quality evidence). Iron supplementation does not cause an excess of severe malaria (RR 0.90, 95% CI 0.81 to 0.98; 6 trials, 3421 children, high quality evidence). We did not observe any differences for deaths (control event rate 1%, low quality evidence). Iron and antimalarial treatment reduced clinical malaria (RR 0.54, 95% CI 0.43 to 0.67; three trials, 728 children, high quality evidence). Overall, iron resulted in fewer anaemic children at follow up, and the end average change in haemoglobin from base line was higher with iron. AUTHORS' CONCLUSIONS: Iron treatment does not increase the risk of clinical malaria when regular malaria prevention or management services are provided. Where resources are limited, iron can be administered without screening for anaemia or for iron deficiency, as long as malaria prevention or management services are provided efficiently.


Subject(s)
Anemia, Iron-Deficiency/prevention & control , Endemic Diseases , Iron/administration & dosage , Malaria/complications , Adolescent , Anemia, Iron-Deficiency/etiology , Antimalarials/administration & dosage , Child , Child, Preschool , Dietary Supplements/adverse effects , Folic Acid/adverse effects , Humans , Iron/adverse effects , Malaria/chemically induced , Parasitemia/chemically induced , Parasitemia/complications , Randomized Controlled Trials as Topic
3.
Exp Parasitol ; 156: 1-11, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26005191

ABSTRACT

Phenylhydrazine (PHZ) treatment is generally used to enhance parasitemia in infected mice models. Transient reticulocytosis is commonly observed in iron-deficient anemic hosts after treatment with iron supplementation, and is also associated with short-term hemolysis caused by PHZ treatment. In this study, we investigated the relationship between reticulocytosis and cerebral malaria (CM) in a murine model induced by PHZ administration before Plasmodium berghei ANKA (PbA) infection. Mortality and parasitemia were checked daily. Pro-inflammatory cytokines and IL-10 were quantified by ELISA. The expression of CXCL9, CXCL10, CCL5, and CXCR3 mRNAs was determined by real-time PCR. Brain sequestration of CD4(+) and CD8(+) T cells and populations of splenic Th1 CD4(+) T cells, dendritic cells (DCs), CD11b(+) Gr1(+) cells, and regulatory T cells (Tregs) were assessed by FACS. PHZ administration dramatically increased parasitemia from day 3 to day 5 post infection (p.i.) compared with the untreated control infected mice group; also, CM developed at day 5 p.i., compared with day 7 p.i. in untreated control infected mice, as well as significantly decreased blood-brain barrier function (P < 0.001). PHZ administration during PbA infection significantly increased the expression of CXCL9 (P <0.05) and VCAM-1 (P <0.001) in the brain, increased the expression of CXCL10, CCL5 and CXCR3, and significantly increased the recruitment of CD4(+) and CD8(+) T cells (P <0.001 and P <0.01, respectively) as well as CD11b(+) Gr1(+) cells to the brain. In addition, PHZ administration significantly increased the numbers of IL-12-secreting DCs at days 3 and 5 p.i. compared to those of untreated control infected mice (P <0.001 and P <0.01, respectively). Consequently, the activation of CD4(+) T cells, especially the expansion of the Th1 subset (P <0.05), was significantly and dramatically enhanced and was accompanied by marked increases in the production of protein and/or mRNA of the Th1-type pro-inflammatory mediators, IFN-γ and TNF-α (P <0.01 for both for protein; P <0.05 for TNF-α mRNA). Our results suggest that, compared to healthy individuals, people suffering from reticulocytosis may be more susceptible to severe malaria infection in malaria endemic areas. This has implications for the most appropriate selection of treatment, which may also cause reticulocytosis in patients living in such areas.


Subject(s)
Malaria, Cerebral/chemically induced , Oxidants/adverse effects , Parasitemia/chemically induced , Phenylhydrazines/adverse effects , Plasmodium berghei/drug effects , Reticulocytosis/drug effects , Animals , Blood-Brain Barrier/metabolism , Erythrocyte Count , Erythrocyte Indices , Female , Hemoglobins/analysis , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Plasmodium berghei/pathogenicity , Random Allocation , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Reticulocytes/cytology , Reticulocytes/drug effects , Reticulocytosis/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes/drug effects , Up-Regulation
4.
Cochrane Database Syst Rev ; (10): CD006589, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21975754

ABSTRACT

BACKGROUND: Iron-deficiency anaemia is common during childhood. Iron supplementation has been claimed to increase the risk of malaria. OBJECTIVES: To assess the effect of iron on malaria and deaths. SEARCH STRATEGY: We searched The Cochrane Library, PUBMED, MEDLINE, LILACS; and trial registry databases, all up to June 2011. We scanned references of included trials. SELECTION CRITERIA: Individually and cluster randomized controlled trials conducted in hypoendemic to holoendemic malaria regions and including children below 18 years of age. We included trials comparing orally administered iron, iron with antimalarial treatment, or iron with folic acid versus placebo or no treatment. Iron fortification was excluded. Antihelminthics could be administered to either group. Additional micronutrients had to be administered equally to both groups. DATA COLLECTION AND ANALYSIS: The primary outcomes were clinical (symptomatic) malaria, severe malaria, and death. Two authors independently selected the studies and extracted the data. We assessed heterogeneity and conducted subgroup analyses by the presence of anaemia at baseline, age, and malaria endemicity. We assessed risk of bias using domain-based evaluation. We performed a fixed-effect meta-analysis for all outcomes and random-effects meta-analysis for hematological outcomes. We adjusted analyses for cluster randomized trials. MAIN RESULTS: Seventy-one trials (45,353 children) were included. For clinical malaria, no significant difference between iron alone and placebo was detected, (risk ratio (RR) 0.99, 95% confidence intervals (CI) 0.90 to 1.09, 13 trials). The results were similar in the subgroups of non-anaemic children and children below 2 years of age. There was no significant difference in deaths in hyper- and holoendemic areas, risk difference +1.93 per 1000 children (95% CI -1.78 to 5.64, 13 trials, 17,898 children). Iron administered for treatment of anaemia resulted in a larger increase in haemoglobin than iron given for prevention, and the benefit was similar in hyper- or holoendemic and lower endemicity settings. Iron and folic acid supplementation resulted in mixed results for severe malaria. Overall, the risk for clinical malaria was higher with iron or with iron plus folic acid in trials where services did not provide for malaria surveillance and treatment. Iron with antimalarial treatment significantly reduced malaria. Iron supplementation during an acute attack of malaria did not increase the risk for parasitological failure, (RR 0.96, 95% CI 0.74 to 1.24, three trials) or deaths. AUTHORS' CONCLUSIONS: Iron alone or with antimalaria treatment does not increase the risk of clinical malaria or death when regular malaria surveillance and treatment services are provided. There is no need to screen for anaemia prior to iron supplementation.


Subject(s)
Anemia, Iron-Deficiency/prevention & control , Endemic Diseases , Iron/administration & dosage , Malaria/complications , Adolescent , Anemia, Iron-Deficiency/etiology , Antimalarials/administration & dosage , Child , Child, Preschool , Dietary Supplements/adverse effects , Folic Acid/adverse effects , Humans , Iron/adverse effects , Malaria/chemically induced , Parasitemia/chemically induced , Parasitemia/complications , Randomized Controlled Trials as Topic
5.
Cochrane Database Syst Rev ; (3): CD006589, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19588399

ABSTRACT

BACKGROUND: Iron-deficiency anaemia is common during childhood. Iron supplementation has been claimed to increase the risk of malaria. OBJECTIVES: To assess the effect of iron on malaria and deaths. SEARCH STRATEGY: We searched The Cochrane Library (2009, issue 1); MEDLINE; EMBASE; LILACS and metaRegister of Controlled Trials, all up to March 2009. We scanned references of included trials. SELECTION CRITERIA: Individually and cluster-randomized controlled trials conducted in hypoendemic to holoendemic malaria regions and including children < 18 years. We included trials comparing orally administered iron with or without folic acid vs. placebo or no treatment. Iron fortification was excluded. Antimalarials and/or antiparasitics could be administered to either group. Additional micronutrients could only be administered equally to both groups. DATA COLLECTION AND ANALYSIS: The primary outcomes were malaria-related events and deaths. Secondary outcomes included haemoglobin, anaemia, other infections, growth, hospitalizations, and clinic visits. We assessed risk of bias using domain-based evaluation. Two authors independently selected studies and extracted data. We contacted authors for missing data. We assessed heterogeneity. We performed fixed-effect meta-analysis and presented random-effects results when heterogeneity was present. We present pooled risk ratios (RR) with 95% confidence intervals (CIs). We used adjusted analyses for cluster-randomized trials. MAIN RESULTS: Sixty-eight trials (42,981 children) fulfilled the inclusion criteria. Iron supplementation did not increase the risk of clinical malaria (RR 1.00, 95% CI 0.88 to 1.13; 22,724 children, 14 trials, random-effects model). The risk was similar among children who were non-anaemic at baseline (RR 0.96, 95% CI 0.85 to 1.09). An increased risk of malaria with iron was observed in trials that did not provide malaria surveillance and treatment. The risk of malaria parasitaemia was higher with iron (RR 1.13, 95% CI 1.01 to 1.26), but there was no difference in adequately concealed trials. Iron + antimalarial was protective for malaria (four trials). Iron did not increase the risk of parasitological failure when given during malaria (three trials). There was no increased risk of death across all trials comparing iron versus placebo (RR 1.11, 95% CI 0.91 to 1.36; 21,272 children, 12 trials). Iron supplementation increased haemoglobin, with significant heterogeneity, and malaria endemicity did not affect this effect. Growth and other infections were mostly not affected by iron supplementation. AUTHORS' CONCLUSIONS: Iron does not increase the risk of clinical malaria or death, when regular malaria surveillance and treatment services are provided. There is no need to screen for anaemia prior to iron supplementation.


Subject(s)
Anemia, Iron-Deficiency/prevention & control , Endemic Diseases , Iron/adverse effects , Malaria/complications , Anemia, Iron-Deficiency/etiology , Antimalarials/administration & dosage , Child , Child, Preschool , Dietary Supplements/adverse effects , Humans , Iron/therapeutic use , Malaria/chemically induced , Parasitemia/chemically induced , Parasitemia/complications , Randomized Controlled Trials as Topic
6.
Parasitol. latinoam ; 61(1/2): 3-11, jun. 2006. ilus, tab, graf
Article in English | LILACS | ID: lil-432842

ABSTRACT

This study has been done to evaluate the central nervous system (CNS) of mice infected with Trypanosoma cruzi and its relationships with the irreversible decrease of motor activity of the rear limbs during acute Chagas´disease. The course of the present study shows the in vivo behaviour of three parasites strains which were isolated from different sources and geographical areas, with the purpose of explaining the parasitemia, mortality rate, clinical, pathological and histopathological changes in the CNS of infected mice. The mice were injected intraperitoneally with 5.103 bloodstreams of different T. cruzi strains. The mice infected with PR and ASM strains from Venezuela, showed low parasitemia and high mortality, while the Y strain produced higher parasitemia levels. At the 30th day post-infection both left parietal brain cortex (LPC) and spinal cord (SC) were sectioned, stained with hematoxilin and eosin (H-E) and examined by means of confocal ligth microscopy. At this time, the pathology of the CNS exhibited focal infiltrates of monocytes, lymphocytes, plasmocytes, polymorphonuclear cells and loss of neuronas and motoneurons. The sections of LPC of infected mice with ASM strain, showed loss neuronal, parasites and abundant T. cruzi antigen deposits in the proximity of the swollen neurons. The sections of SC stained with Enolase-Avidin-Biotin-Peroxidase showed a reduction in the average number of neurons of the cervical region (CR) of the infected mice with PR, ASM and Y strains. Sections stained with Propidium Ioduro (IP) showed a reduction of the number of motoneurons in all regions of the SC, with a significant difference between groups infected with different T. cruzi strains and control uninfected mice (P < 0.05). This study established a correlation between the parasitism in the proximity to inflammatory cells, together the appearance of T. cruzi antigen and neuronal destruction in the brain. Therefore it can be concluded that the changes in CNS may be attributed to early parasitism in nervous tissue, which occur in a few days, involving clinico-pathological manifestations, which produced alterations of the mobility with paralysis of the rear limbs and death in 100% of mice with acute infection produced by PR and ASM-T. cruzi strains from Venezuela.


Subject(s)
Animals , Rats , Central Nervous System Parasitic Infections , Chagas Disease/complications , Central Nervous System/parasitology , Central Nervous System/pathology , Trypanosoma cruzi , Acute Disease , Chagas Disease/immunology , Neurodegenerative Diseases/parasitology , Neurons/parasitology , Parasitemia/chemically induced , Venezuela
7.
Trop Med Int Health ; 10(11): 1099-107, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16262734

ABSTRACT

OBJECTIVE: Mild viral illness, including that following immunization with live attenuated measles virus (LAMV), has been associated with transient decreases in haemoglobin (Hb) and cellular immune response that may persist for several weeks. In areas of intense malaria transmission, such as western Kenya, infants experience a progressive drop in Hb until age 9-10 months and one-third may have Hb < 8 g/dl. These children may be at risk of developing severe anaemia with further haematological insult. The objective of this paper was to determine if immunization with LAMV was associated with increased risk of transient anaemia and malaria infection. METHODS: Data from previous cross-sectional surveys (n = 5970) and one cohort study (n = 546) conducted among pre-school children were analyzed retrospectively. RESULTS: Measles vaccination coverage between 12 and 23 months of age ranged from 44.8% to 62.7%. Hb concentrations in children aged 6-23 months with documented measles immunization within the previous 14 or 30 days (n = 103) were similar to those with no history of measles immunization in the previous 90 days (n = 996); mean differences [95% confidence interval (CI)] by 30 days were: in cross-sectional surveys, -0.49 g/dl (-1.12, 0.14); in the cohort study, -0.032 g/dl (-0.52, 0.46). Similarly, the risk of malaria parasitemia or severe to moderate anaemia did not differ. CONCLUSION: These data do not suggest that the transient decrease in Hb and cellular immune response after immunization with LAMV results in clinically significant changes in the risk of subsequent severe to moderate anaemia or malaria in young children living in malaria-endemic regions.


Subject(s)
Anemia/etiology , Malaria/etiology , Measles Vaccine/adverse effects , Measles/prevention & control , Anemia/epidemiology , Cell Size/drug effects , Female , Hemoglobins/analysis , Humans , Infant , Kenya/epidemiology , Malaria/epidemiology , Male , Measles/epidemiology , Parasitemia/chemically induced , Retrospective Studies , Vaccination/adverse effects , Vaccines, Attenuated/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL