Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
1.
Nat Commun ; 15(1): 2021, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38448421

ABSTRACT

In Bandiagara, Mali, children experience on average two clinical malaria episodes per year. However, even in the same transmission area, the number of uncomplicated symptomatic infections, and their parasitemia, can vary dramatically among children. We simultaneously characterize host and parasite gene expression profiles from 136 Malian children with symptomatic falciparum malaria and examine differences in the relative proportion of immune cells and parasite stages, as well as in gene expression, associated with infection and or patient characteristics. Parasitemia explains much of the variation in host and parasite gene expression, and infections with higher parasitemia display proportionally more neutrophils and fewer T cells, suggesting parasitemia-dependent neutrophil recruitment and/or T cell extravasation to secondary lymphoid organs. The child's age also strongly correlates with variations in gene expression: Plasmodium falciparum genes associated with age suggest that older children carry more male gametocytes, while variations in host gene expression indicate a stronger innate response in younger children and stronger adaptive response in older children. These analyses highlight the variability in host responses and parasite regulation during P. falciparum symptomatic infections and emphasize the importance of considering the children's age when studying and treating malaria infections.


Subject(s)
Malaria, Falciparum , Malaria , Child , Humans , Male , Adolescent , Parasitemia/genetics , Gene Expression Profiling , Malaria, Falciparum/genetics , Cell Movement
2.
PLoS One ; 18(7): e0287668, 2023.
Article in English | MEDLINE | ID: mdl-37471337

ABSTRACT

Plasmodium vivax Duffy Binding Protein (PvDBP) is essential for interacting with Duffy antigen receptor for chemokines (DARC) on the surface of red blood cells to allow invasion. Earlier whole genome sequence analyses provided evidence for the duplications of PvDBP. It is unclear whether PvDBP duplications play a role in recent increase of P. vivax in Sudan and in Duffy-negative individuals. In this study, the prevalence and type of PvDBP duplications, and its relationship to demographic and clinical features were investigated. A total of 200 malaria-suspected blood samples were collected from health facilities in Khartoum, River Nile, and Al-Obied. Among them, 145 were confirmed to be P. vivax, and 43 (29.7%) had more than one PvDBP copies with up to four copies being detected. Both the Malagasy and Cambodian types of PvDBP duplication were detected. No significant difference was observed between the two types of duplications between Duffy groups. Parasitemia was significantly higher in samples with the Malagasy-type than those without duplications. No significant difference was observed in PvDBP duplication prevalence and copy number among study sites. The functional significance of PvDBP duplications, especially those Malagasy-type that associated with higher parasitemia, merit further investigations.


Subject(s)
Malaria, Vivax , Plasmodium vivax , Humans , Gene Duplication , Sudan/epidemiology , Parasitemia/genetics , Prevalence , Antigens, Protozoan , Protozoan Proteins/metabolism , Malaria, Vivax/epidemiology , Malaria, Vivax/genetics , Duffy Blood-Group System/genetics , Duffy Blood-Group System/metabolism , Erythrocytes/metabolism
3.
Innate Immun ; 29(5): 83-94, 2023 07.
Article in English | MEDLINE | ID: mdl-37306242

ABSTRACT

Malaria is often characterized by a complicated disease course due to multifaceted intrinsic genetic factors of the host and the parasite. This study aimed to investigate the role of interleukin-27 (IL-27) gene polymorphisms in Plasmodium falciparum malaria infection in a Saudi Arabian cohort. This case-control study obtained blood samples from 250 malaria patients with P. falciparum and 200 randomly identified healthy control subjects from the Malaria Center in the Jazan area. Malaria patients were grouped into three cohorts as follow: low (<500 parasites/µl of blood), moderate (500-1000 parasites/µl of blood), and high (>1000 parasites/µl of blood) parasitemia. The results show that the IL-27 variant rs181209 was significantly associated with malaria patients (P = 0.026). Similarly, the homozygous GG genotype of rs26528 was also associated with risk of developing P. falciparum malaria (P = 0.032). The minor allele C of variant rs181206 exhibited an association with low to moderate parasitemia (P = 0.046). Furthermore, the rs181209 AA genotype was statistically significant in age group 1-5 years (P = 0.049). In conclusion, this study suggests that variant rs181209 and rs26528 could be associated with the risk of malaria infection by P. falciparum in the population studied.


Subject(s)
Interleukin-27 , Malaria, Falciparum , Malaria , Humans , Infant , Child, Preschool , Interleukin-27/genetics , Plasmodium falciparum/genetics , Parasitemia/genetics , Parasitemia/epidemiology , Case-Control Studies , Saudi Arabia , Malaria, Falciparum/genetics , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Polymorphism, Genetic
4.
J Infect Dis ; 228(2): 202-211, 2023 07 14.
Article in English | MEDLINE | ID: mdl-36961831

ABSTRACT

BACKGROUND: TP53 has been shown to play a role in inflammatory processes, including malaria. We previously found that p53 attenuates parasite-induced inflammation and predicts clinical protection to Plasmodium falciparum infection in Malian children. Here, we investigated whether p53 codon 47 and 72 polymorphisms are associated with differential risk of P. falciparum infection and uncomplicated malaria in a prospective cohort study of malaria immunity. METHODS: p53 codon 47 and 72 polymorphisms were determined by sequencing TP53 exon 4 in 631 Malian children and adults enrolled in the Kalifabougou cohort study. The effects of these polymorphisms on the prospective risk of febrile malaria, incident parasitemia, and time to fever after incident parasitemia over 6 months of intense malaria transmission were assessed using Cox proportional hazards models. RESULTS: Confounders of malaria risk, including age and hemoglobin S or C, were similar between individuals with or without p53 S47 and R72 polymorphisms. Relative to their respective common variants, neither S47 nor R72 was associated with differences in prospective risk of febrile malaria, incident parasitemia, or febrile malaria after parasitemia. CONCLUSIONS: These findings indicate that p53 codon 47 and 72 polymorphisms are not associated with protection against incident P. falciparum parasitemia or uncomplicated febrile malaria.


Subject(s)
Malaria, Falciparum , Malaria , Child , Adult , Humans , Cohort Studies , Prospective Studies , Parasitemia/genetics , Tumor Suppressor Protein p53/genetics , Plasmodium falciparum/genetics , Malaria/complications , Malaria, Falciparum/epidemiology , Malaria, Falciparum/genetics , Malaria, Falciparum/complications , Fever/etiology
5.
Microbiol Spectr ; 10(5): e0262822, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36190410

ABSTRACT

Trypanosoma cruzi infection has expanded globally through human migration. In Spain, the mother-to-child route is the mode of transmission contributing to autochthonous Chagas disease (CD); however, most people acquired the infection in their country of origin and were diagnosed in the chronic phase (imported chronic CD). In this context, we assessed the quantitative potential of the Loopamp Trypanosoma cruzi detection kit (Sat-TcLAMP) based on satellite DNA (Sat-DNA) to determine parasitemia levels compared to those detected by real-time quantitative PCRs (qPCRs) targeting Sat-DNA (Sat-qPCR) and kinetoplast DNA minicircles (kDNA-qPCR). This study included 173 specimens from 39 autochthonous congenital and 116 imported chronic CD cases diagnosed in Spain. kDNA-qPCR showed higher sensitivity than Sat-qPCR and Sat-TcLAMP. According to all quantitative approaches, parasitemia levels were significantly higher in congenital infection than in chronic CD (1 × 10-1 to 5 × 105 versus >1 × 10-1 to 6 × 103 parasite equivalents/mL, respectively [P < 0.001]). Sat-TcLAMP, Sat-qPCR, and kDNA-qPCR results were equivalent at high levels of parasitemia (P = 0.381). Discrepancies were significant for low levels of parasitemia and older individuals. Differences between Sat-TcLAMP and Sat-qPCR were not qualitatively significant, but estimations of parasitemia using Sat-TcLAMP were closer to those by kDNA-qPCR. Parasitemia changes were assessed in 6 individual cases in follow-up, in which trends showed similar patterns by all quantitative approaches. At high levels of parasitemia, Sat-TcLAMP, Sat-qPCR, and kDNA-qPCR worked similarly, but significant differences were found for the low levels characteristic of late chronic CD. A suitable harmonization strategy needs to be developed for low-level parasitemia detection using Sat-DNA- and kDNA-based tests. IMPORTANCE Currently, molecular equipment has been introduced into many health care centers, even in low-income countries. PCR, qPCR, and loop-mediated isothermal amplification (LAMP) are becoming more accessible for the diagnosis of neglected infectious diseases. Chagas disease (CD) is spreading worldwide, and in countries where the disease is not endemic, such as Spain, the parasite Trypanosoma cruzi is transmitted from mother to child (congenital CD). Here, we explore why LAMP, aimed at detecting T. cruzi parasite DNA, is a reliable option for the diagnosis of congenital CD and the early detection of reactivation in chronic infection. When the parasite load is high, LAMP is equivalent to any qPCR. In addition, the estimations of T. cruzi parasitemia in patients living in Spain, a country where the disease is not endemic, resemble natural evolution in areas of endemicity. If molecular tests are introduced into the diagnostic algorithm for congenital infection, early diagnosis and timely treatment would be accomplished, so the interruption of vertical transmission can be an achievable goal.


Subject(s)
Chagas Disease , Trypanosoma cruzi , Female , Humans , DNA, Kinetoplast/genetics , Parasitemia/diagnosis , Parasitemia/epidemiology , Parasitemia/genetics , DNA, Satellite , Spain/epidemiology , DNA, Protozoan/genetics , DNA, Protozoan/analysis , Infectious Disease Transmission, Vertical , Chagas Disease/diagnosis , Chagas Disease/epidemiology , Chagas Disease/genetics , Trypanosoma cruzi/genetics , Real-Time Polymerase Chain Reaction/methods
6.
Front Immunol ; 12: 650028, 2021.
Article in English | MEDLINE | ID: mdl-33815410

ABSTRACT

Variation within the HLA locus been shown to play an important role in the susceptibility to and outcomes of numerous infections, but its influence on immunity to P. falciparum malaria is unclear. Increasing evidence indicates that acquired immunity to P. falciparum is mediated in part by the cellular immune response, including NK cells, CD4 and CD8 T cells, and semi-invariant γδ T cells. HLA molecules expressed by these lymphocytes influence the epitopes recognized by P. falciparum-specific T cells, and class I HLA molecules also serve as ligands for inhibitory receptors including KIR. Here we assessed the relationship of HLA class I and II alleles to the risk of P. falciparum infection and symptomatic malaria in a cohort of 892 Ugandan children and adults followed prospectively via both active and passive surveillance. We identified two HLA class I alleles, HLA-B*53:01 and HLA-C*06:02, that were associated with a higher prevalence of P. falciparum infection. Notably, no class I or II HLA alleles were found to be associated with protection from P. falciparum parasitemia or symptomatic malaria. These findings suggest that class I HLA plays a role in the ability to restrict parasitemia, supporting an essential role for the cellular immune response in P. falciparum immunity. Our findings underscore the need for better tools to enable mechanistic studies of the T cell response to P. falciparum at the epitope level and suggest that further study of the role of HLA in regulating pre-erythrocytic stages of the P. falciparum life cycle is warranted.


Subject(s)
HLA Antigens/genetics , HLA-C Antigens/genetics , Malaria, Falciparum/epidemiology , Parasitemia/epidemiology , Plasmodium falciparum/immunology , Adult , Alleles , Antigens, Protozoan/immunology , Child , Child, Preschool , Epitopes, T-Lymphocyte/immunology , Female , Follow-Up Studies , Genetic Predisposition to Disease , Genotyping Techniques , HLA Antigens/metabolism , HLA-C Antigens/metabolism , Humans , Incidence , Infant , Malaria, Falciparum/blood , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Male , Parasitemia/blood , Parasitemia/genetics , Parasitemia/parasitology , Plasmodium falciparum/isolation & purification , Prospective Studies , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Uganda/epidemiology
7.
Front Immunol ; 11: 566476, 2020.
Article in English | MEDLINE | ID: mdl-33329529

ABSTRACT

Background: Considering the complexity of the factors involved in the immunopathology of Chagas disease, which influence the Chagas' disease pathogenesis, anti-T. cruzi immune response, and chemotherapy outcome, further studies are needed to improve our understanding about these relationships. On this way, in this article we analyzed the host genetic influence on hematological, histopathological and immunological aspects after T. cruzi infection. Methods: BALB/c and A mice were intragastrically infected with T. cruzi SC2005 strain, isolated from a patient of an outbreak of Chagas disease. Parameters such as parasite load, survival rates, cytokines production, macrophages, T and B cell frequencies, and histopathology analysis were carried out. Results: BALB/c mice presented higher parasitemia and mortality rates than A mice. Both mouse lineages exhibited hematological alterations suggestive of microcytic hypochromic anemia and histopathological alterations in stomach, heart and liver. The increase of CD8+ T cells, in heart, liver and blood, and the increase of CD19+ B cells, in liver, associated with a high level of proinflammatory cytokines (IL-6, TNF-α, IFN-γ), confer a resistance profile to the host. Although BALB/c animals exhibited the same findings observed in A mice, the response to infection occurred later, after a considerable parasitemia increase. By developing an early response to the infection, A mice were found to be less susceptible to T. cruzi SC2005 infection. Conclusions: Host genetics background shaping the response to infection. The early development of a cytotoxic cellular response profile with the production of proinflammatory cytokines is important to lead a less severe manifestation of Chagas disease.


Subject(s)
Chagas Disease , Animals , Chagas Disease/genetics , Chagas Disease/immunology , Chagas Disease/parasitology , Chagas Disease/pathology , Cytokines/immunology , Female , Heart/parasitology , Liver/parasitology , Liver/pathology , Mice, Inbred Strains , Myocardium/pathology , Parasite Load , Parasitemia/genetics , Parasitemia/immunology , Parasitemia/pathology , Species Specificity , Stomach/parasitology , Stomach/pathology
8.
Front Immunol ; 11: 521409, 2020.
Article in English | MEDLINE | ID: mdl-33193300

ABSTRACT

Background: Chagas disease caused by Trypanosoma cruzi (T. cruzi) affects approximately six million individuals worldwide. Clinical manifestations are expected to occur due to the parasite persistence and host immune response. Herein we investigated potential associations between IL1B, IL6, IL17A, or IL18 polymorphism profiles and cardiomyopathy or T. cruzi parasitemia, as well as the impact of HIV infection on cardiopathy. Methods: Two hundred twenty-six patients and 90 control individuals were analyzed. IL1B rs1143627 T>C, IL6 rs1800795 C>G, IL17A rs2275913 G>A, IL18 rs187238 C>G, and IL18 rs1946518 C>A SNVs were analyzed by real-time PCR and T. cruzi parasitemia by PCR. Results: Our data revealed association between a cytokine gene polymorphism and parasitemia never previously reported. The IL6 rs1800795 CG genotype lowered the risk of positive parasitemia (OR = 0.45, 95% CI 0.24-0.86, P = 0.015). Original findings included associations between IL17A rs2275913 AA and IL18 s1946518 AA genotypes with decreased risk of developing cardiomyopathy (OR = 0.27, 95% CI 0.07-0.97, P = 0.044; and OR = 0.35, 95% CI 0.14-0.87, P = 0.023, respectively). IL18 rs1946518 AA and IL1B rs1143627 TC were associated with reduced risk for cardiomyopathy severity, including NYHA (New York Heart Association) class ≥ 2 (OR = 0.21, 95% CI 0.06-0.68, P = 0.009; and OR = 0.48, 95% CI 0.24-0.95, P = 0.036, respectively) and LVEF (left ventricular ejection fraction) <45% for IL18 rs1946518 AA (OR = 0.22, 95% CI 0.05-0.89, P = 0.034). A novel, unexpected protective effect of HIV infection against development/progression of cardiomyopathy was identified, based on a lower risk of developing cardiopathy (OR = 0.48, 95% CI 0.23-0.96, P = 0.039), NYHA class ≥ 2 (OR = 0.15, 95% CI 0.06-0.39, P < 0.001), and LVEF < 45% (OR = 0.03, 95% CI 0.00-0.25, P = 0.001). Digestive involvement was negatively associated with NYHA ≥ 2 and LVEF < 45% (OR = 0.20, 95% CI 0.09-0.47, P < 0.001; and OR = 0.24, 95% CI 0.09-0.62, P = 0.004, respectively). Conclusions: Our data support a protective role of IL17A AA, IL18 AA, and IL1B TC genotypes against development/progression of cardiomyopathy and a modulatory effect of the IL6 CG genotype on the risk of parasitemia in Chagas disease. Notably, HIV infection was shown to protect against development/progression of cardiopathy, potentially associated with a synergistic effect of HIV and highly active antiretroviral therapy (HAART), attenuating a Th1-mediated response in the myocardium. This proposed hypothesis requires confirmation, however, in larger and more comprehensive future studies.


Subject(s)
Chagas Disease , Genotype , Interleukin-17 , Interleukin-18 , Interleukin-1beta , Interleukin-6 , Parasitemia , Polymorphism, Genetic , Trypanosoma cruzi/immunology , Adult , Chagas Disease/genetics , Chagas Disease/immunology , Female , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Male , Middle Aged , Parasitemia/genetics , Parasitemia/immunology
9.
Nat Commun ; 11(1): 5093, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33037226

ABSTRACT

The mechanisms behind the ability of Plasmodium falciparum to evade host immune system are poorly understood and are a major roadblock in achieving malaria elimination. Here, we use integrative genomic profiling and a longitudinal pediatric cohort in Burkina Faso to demonstrate the role of post-transcriptional regulation in host immune response in malaria. We report a strong signature of miRNA expression differentiation associated with P. falciparum infection (127 out of 320 miRNAs, B-H FDR 5%) and parasitemia (72 miRNAs, B-H FDR 5%). Integrative miRNA-mRNA analysis implicates several infection-responsive miRNAs (e.g., miR-16-5p, miR-15a-5p and miR-181c-5p) promoting lymphocyte cell death. miRNA cis-eQTL analysis using whole-genome sequencing data identified 1,376 genetic variants associated with the expression of 34 miRNAs (B-H FDR 5%). We report a protective effect of rs114136945 minor allele on parasitemia mediated through miR-598-3p expression. These results highlight the impact of post-transcriptional regulation, immune cell death processes and host genetic regulatory control in malaria.


Subject(s)
Immune Evasion/genetics , Malaria, Falciparum/genetics , Malaria, Falciparum/immunology , MicroRNAs/genetics , Plasmodium falciparum/pathogenicity , Burkina Faso , Child , Child, Preschool , Gene Expression Regulation , Genome, Human , Humans , Longitudinal Studies , Parasitemia/genetics , Parasitemia/immunology , Plasmodium falciparum/immunology , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , Whole Genome Sequencing
10.
Malar J ; 19(1): 21, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31941490

ABSTRACT

BACKGROUND: Multiple red blood cell (RBC) variants appear to offer protection against the most severe forms of Plasmodium falciparum malaria. Associations between these variants and uncomplicated malaria are less clear. METHODS: Data from a longitudinal cohort study conducted in 3 sub-counties in Uganda was used to quantify associations between three red blood cell variants Hb [AA, AS, S (rs334)], alpha thalassaemia 3.7 kb deletion, and glucose-6-phosphate dehydrogenase deficiency A-(G6PD 202A genotype) and malaria incidence, parasite prevalence, parasite density (a measure of anti-parasite immunity) and body temperature adjusted for parasite density (a measure of anti-disease immunity). All analyses were adjusted for age, average household entomological inoculation rate, and study site. Results for all variants were compared to those for wild type genotypes. RESULTS: In children, HbAS was associated, compared to wild type, with a lower incidence of malaria (IRR = 0.78, 95% CI 0.66-0.92, p = 0.003), lower parasite density upon infection (PR = 0.66, 95% CI 0.51-0.85, p = 0.001), and lower body temperature for any given parasite density (- 0.13 â„ƒ, 95% CI - 0.21, - 0.05, p = 0.002). In children, HbSS was associated with a lower incidence of malaria (IRR = 0.17, 95% CI 0.04-0.71, p = 0.02) and lower parasite density upon infection (PR = 0.31, 95% CI 0.18-0.54, p < 0.001). α-/αα thalassaemia, was associated with higher parasite prevalence in both children and adults (RR = 1.23, 95% CI 1.06-1.43, p = 0.008 and RR = 1.52, 95% CI 1.04-2.23, p = 0.03, respectively). G6PD deficiency was associated with lower body temperature for any given parasite density only among male hemizygote children (- 0.19 â„ƒ, 95% CI - 0.31, - 0.06, p = 0.003). CONCLUSION: RBC variants were associated with non-severe malaria outcomes. Elucidation of the mechanisms by which they confer protection will improve understanding of genetic protection against malaria.


Subject(s)
Erythrocytes/cytology , Malaria/blood , Adult , Age Factors , Binomial Distribution , Caregivers , Child , Child, Preschool , Cohort Studies , Erythrocytes/chemistry , Erythrocytes/classification , Female , Humans , Incidence , Infant , Longitudinal Studies , Malaria/epidemiology , Malaria/genetics , Malaria/parasitology , Male , Parasitemia/blood , Parasitemia/epidemiology , Parasitemia/genetics , Parasitemia/parasitology , Prevalence , Prospective Studies , Sex Factors , Uganda/epidemiology , Young Adult
11.
PLoS Pathog ; 15(9): e1007974, 2019 09.
Article in English | MEDLINE | ID: mdl-31536608

ABSTRACT

Plasmodium relapses are attributed to the activation of dormant liver-stage parasites and are responsible for a significant number of recurring malaria blood-stage infections. While characteristic of human infections caused by P. vivax and P. ovale, their relative contribution to malaria disease burden and transmission remains poorly understood. This is largely because it is difficult to identify 'bona fide' relapse infections due to ongoing transmission in most endemic areas. Here, we use the P. cynomolgi-rhesus macaque model of relapsing malaria to demonstrate that clinical immunity can form after a single sporozoite-initiated blood-stage infection and prevent illness during relapses and homologous reinfections. By integrating data from whole blood RNA-sequencing, flow cytometry, P. cynomolgi-specific ELISAs, and opsonic phagocytosis assays, we demonstrate that this immunity is associated with a rapid recall response by memory B cells that expand and produce anti-parasite IgG1 that can mediate parasite clearance of relapsing parasites. The reduction in parasitemia during relapses was mirrored by a reduction in the total number of circulating gametocytes, but importantly, the cumulative proportion of gametocytes increased during relapses. Overall, this study reveals that P. cynomolgi relapse infections can be clinically silent in macaques due to rapid memory B cell responses that help to clear asexual-stage parasites but still carry gametocytes.


Subject(s)
Immunity, Humoral , Malaria/immunology , Malaria/parasitology , Plasmodium cynomolgi/immunology , Plasmodium cynomolgi/pathogenicity , Animals , Antibodies, Protozoan/blood , B-Lymphocytes/immunology , Gene Expression Profiling , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology , Humans , Immunity, Humoral/genetics , Immunoglobulin G/blood , Immunologic Memory/genetics , Macaca mulatta , Malaria/genetics , Malaria, Vivax/genetics , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Male , Parasitemia/genetics , Parasitemia/immunology , Parasitemia/parasitology , Plasmodium vivax/immunology , Plasmodium vivax/pathogenicity , Recurrence , Sporozoites/immunology , Sporozoites/pathogenicity
12.
Front Immunol ; 10: 1345, 2019.
Article in English | MEDLINE | ID: mdl-31316497

ABSTRACT

T cells play significant roles during Plasmodium falciparum infections. Their regulation of the immune response in symptomatic children with malaria has been deemed necessary to prevent immune associated pathology. In this study, we phenotypically characterized the expression of T cell inhibitory(PD-1, CTLA-4) and senescent markers (CD28(-), CD57) from children with symptomatic malaria, asymptomatic malaria and healthy controls using flow cytometry. We observed increased expression of T cell exhaustion and senescence markers in the symptomatic children compared to the asymptomatic and healthy controls. T cell senescence markers were more highly expressed on CD8 T cells than on CD4 T cells. Asymptomatically infected children had comparable levels of these markers with healthy controls except for CD8+ PD-1+ T cells which were significantly elevated in the asymptomatic children. Also, using multivariate regression analysis, CTLA-4 was the only marker that could predict parasitaemia level. The results suggest that the upregulation of immune exhaustion and senescence markers during symptomatic malaria may affect the effector function of T cells leading to inefficient clearance of parasites, hence the inability to develop sterile immunity to malaria.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cellular Senescence/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Asymptomatic Infections , CD28 Antigens/genetics , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD57 Antigens/genetics , CD57 Antigens/immunology , CD57 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/parasitology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Cells, Cultured , Cellular Senescence/genetics , Child , Child, Preschool , Female , Gene Expression Profiling/methods , Humans , Immunophenotyping , Malaria, Falciparum/parasitology , Male , Parasitemia/genetics , Parasitemia/immunology , Parasitemia/metabolism , Plasmodium falciparum/physiology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism
13.
Free Radic Biol Med ; 129: 227-236, 2018 12.
Article in English | MEDLINE | ID: mdl-30248443

ABSTRACT

Elderly organisms are more susceptible to infectious diseases. However, the impact of aging on antiparasitic mechanisms, especially the nitric oxide pathway, is poorly understood. Using an integrated in vivo and in vitro model, we compared the severity of Trypanosoma cruzi infection in young and elderly (8 or 72 weeks old) mice. Forty C57BL/6 mice were randomized into four groups: Y-inf, young infected; Yn-inf, young uninfected; A-inf, aged infected; An-inf, aged uninfected. Parasitemia was measured daily, and animals were euthanized after 15 days of infection. Trypanosoma cruzi-induced inflammatory processes were analyzed in blood and heart samples, as well as in bone marrow-derived macrophages (BMDMs) co-cultured with splenocytes isolated from young or elderly mice. Our results indicated upregulated IgG2b and IL-17 production in elderly animals, which was not sufficient to reduce parasitemia, parasitic load and myocarditis to levels observed in young animals. The higher susceptibility of elderly mice to T. cruzi infection was accompanied by reduced cardiac inducible nitric oxide synthase (iNOS) gene expression, nitric oxide (NO) and IFN-γ levels, as well as an antagonistic upregulation of arginase-1 expression and arginase activity. The same responses were observed when BMDMs co-cultured with splenocytes from elderly mice were stimulated with T. cruzi antigens. Our findings indicate that elderly mice were more susceptible to T. cruzi infection, which was potentially related to an attenuated response to antigenic stimulation, inhibition of iNOS gene expression and NO production, and antagonistic upregulation of arginase gene expression and activity, which created favorable conditions for heart parasitism and myocarditis development.


Subject(s)
Aging/genetics , Arginase/genetics , Chagas Cardiomyopathy/genetics , Chagas Disease/genetics , Nitric Oxide Synthase Type II/genetics , Parasitemia/genetics , Trypanosoma cruzi/pathogenicity , Aging/immunology , Animals , Antigens, Protozoan/pharmacology , Arginase/blood , Chagas Cardiomyopathy/immunology , Chagas Cardiomyopathy/parasitology , Chagas Disease/immunology , Chagas Disease/parasitology , Coculture Techniques , Gene Expression Regulation , Heart/parasitology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunoglobulin G/blood , Immunoglobulin G/genetics , Interferon-gamma/blood , Interferon-gamma/genetics , Interleukin-10/blood , Interleukin-10/genetics , Interleukin-17/blood , Interleukin-17/genetics , Macrophages/drug effects , Macrophages/immunology , Macrophages/parasitology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/blood , Parasitemia/immunology , Severity of Illness Index , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/parasitology , Trypanosoma cruzi/immunology
14.
Sci Transl Med ; 10(447)2018 06 27.
Article in English | MEDLINE | ID: mdl-29950443

ABSTRACT

The pathogenesis of infectious diseases depends on the interaction of host and pathogen. In Plasmodium falciparum malaria, host and parasite processes can be assessed by dual RNA sequencing of blood from infected patients. We performed dual transcriptome analyses on samples from 46 malaria-infected Gambian children to reveal mechanisms driving the systemic pathophysiology of severe malaria. Integrating these transcriptomic data with estimates of parasite load and detailed clinical information allowed consideration of potentially confounding effects due to differing leukocyte proportions in blood, parasite developmental stage, and whole-body pathogen load. We report hundreds of human and parasite genes differentially expressed between severe and uncomplicated malaria, with distinct profiles associated with coma, hyperlactatemia, and thrombocytopenia. High expression of neutrophil granule-related genes was consistently associated with all severe malaria phenotypes. We observed severity-associated variation in the expression of parasite genes, which determine cytoadhesion to vascular endothelium, rigidity of infected erythrocytes, and parasite growth rate. Up to 99% of human differential gene expression in severe malaria was driven by differences in parasite load, whereas parasite gene expression showed little association with parasite load. Coexpression analyses revealed interactions between human and P. falciparum, with prominent co-regulation of translation genes in severe malaria between host and parasite. Multivariate analyses suggested that increased expression of granulopoiesis and interferon-γ-related genes, together with inadequate suppression of type 1 interferon signaling, best explained severity of infection. These findings provide a framework for understanding the contributions of host and parasite to the pathogenesis of severe malaria and identifying new treatments.


Subject(s)
Gene Expression Profiling , Host-Pathogen Interactions/genetics , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Child , Child, Preschool , Female , Gene Expression Regulation , Gene Regulatory Networks , Humans , Malaria, Falciparum/blood , Male , Neutrophils/metabolism , Parasitemia/blood , Parasitemia/genetics , Plasmodium falciparum/genetics , Sequence Analysis, RNA , Species Specificity
15.
PLoS One ; 13(6): e0199392, 2018.
Article in English | MEDLINE | ID: mdl-29920562

ABSTRACT

Malaria continues to be one of mankind's most devastating diseases despite the many and varied efforts to combat it. Indispensable for malaria elimination and eventual eradication is the development of effective vaccines. Controlled human malaria infection (CHMI) is an invaluable tool for vaccine efficacy assessment and investigation of early immunological and molecular responses against Plasmodium falciparum infection. Here, we investigated gene expression changes following CHMI using RNA-Seq. Peripheral blood samples were collected in Bagamoyo, Tanzania, from ten adults who were injected intradermally (ID) with 2.5x104 aseptic, purified, cryopreserved P. falciparum sporozoites (Sanaria® PfSPZ Challenge). A total of 2,758 genes were identified as differentially expressed following CHMI. Transcriptional changes were most pronounced on day 5 after inoculation, during the clinically silent liver phase. A secondary analysis, grouping the volunteers according to their prepatent period duration, identified 265 genes whose expression levels were linked to time of blood stage parasitemia detection. Gene modules associated with these 265 genes were linked to regulation of transcription, cell cycle, phosphatidylinositol signaling and erythrocyte development. Our study showed that in malaria pre-exposed volunteers, parasite prepatent period in each individual is linked to magnitude and timing of early gene expression changes after ID CHMI.


Subject(s)
Malaria, Falciparum/genetics , Parasitemia/blood , Plasmodium falciparum/isolation & purification , Transcriptome/genetics , Blood Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation/genetics , Humans , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Parasitemia/genetics , Plasmodium falciparum/pathogenicity , Volunteers
16.
Glycobiology ; 28(7): 534-541, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29718295

ABSTRACT

The HS3ST3A1/B1 genes encode two homologous 3-O-sulfotransferases involved in the late modification step during heparan sulfate (HS) biosynthesis. In addition to the single nucleotide polymorphisms (SNPs) rs28470223 (C > T) in the promoter region of both HS3ST3A1 and rs62636623 (Gly/Arg) in the stem region of HS3ST3B1, three missense mutations (rs62056073, rs61729712 and rs9906590) located within the catalytic sulfotransferase domain of 3-OST-B1 are linked and associated to Plasmodium falciparum parasitaemia. To ascertain the functional effects of these SNP associations, we investigated the regulatory effect of rs28470223 and characterized the enzymatic activity of the missense SNP rs61729712 (Ser279Asn) localized at proximity of the substrate binding cleft. The SNP rs28470223 results in decreased promoter activity of HS3ST3A1 in K562 cells, suggesting a reduced in vivo transcription activity of the target gene. A comparative kinetic analysis of wt HS3ST3B1 and the Ser269Asn variant (rs61729712) using a HS-derived oligosaccharide substrate reveals a slightly higher catalytic activity for the SNP variant. These genetic and enzymatic studies suggest that genetic variations in enzymes responsible of HS 3-O-sulfation can modulate their promoter and enzymatic activities and may influence P. falciparum parasitaemia.


Subject(s)
Parasitemia/genetics , Plasmodium falciparum , Polymorphism, Single Nucleotide , Sulfotransferases/genetics , Binding Sites , Cell Line, Tumor , Heparitin Sulfate/metabolism , Humans , Mutation, Missense , Protein Binding , Sulfotransferases/chemistry , Sulfotransferases/metabolism
17.
Infect Genet Evol ; 66: 399-409, 2018 12.
Article in English | MEDLINE | ID: mdl-28579526

ABSTRACT

Malaria is a life-threatening blood disease caused by the protozoan Plasmodium. Infection may lead to several different patterns of symptoms in the host: asymptomatic state, uncomplicated disease or severe disease. Severe malaria occurs mostly in young children and is a major cause of death. Disease is thought to result from the sequestration of parasites in the small blood vessels of the brain and the deregulation of key immune system elements. The cellular and molecular regulatory mechanisms underlying the pathogenesis of disease are however not fully understood. What is known it is that the genetic determinants of the host play an important role in the severity of the disease and the outcome of infection. Here we review the most convincing results obtained through genetic epidemiology studies concerning the genetic control of malaria in human caused by Plasmodium falciparum infection. The identification of genes conferring susceptibility or resistance to malaria might improve diagnosis and treatment.


Subject(s)
Genetic Predisposition to Disease , Host-Parasite Interactions/genetics , Malaria/genetics , Malaria/parasitology , Biomarkers , Genetic Linkage , Genome-Wide Association Study , Host-Parasite Interactions/immunology , Humans , Malaria/diagnosis , Malaria/epidemiology , Malaria, Falciparum/diagnosis , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Parasite Load , Parasitemia/diagnosis , Parasitemia/genetics , Parasitemia/parasitology , Plasmodium falciparum , Quantitative Trait Loci , Severity of Illness Index
18.
Pharmacogenomics ; 18(15): 1393-1400, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28975866

ABSTRACT

AIM: The association of transporters gene polymorphisms with chloroquine/primaquine malaria treatment response was investigated in a Brazilian population. PATIENTS & METHODS: Totally, 164 Plasmodium vivax malaria infected patients were included. Generalized estimating equations were performed to determine gene influences on parasitemia and/or gametocytemia clearance over treatment time. RESULTS: Significant interaction between SLCO2B1 genotypes and treatment over time for parasitemia clearance rate on day 2 were observed (p FDR = 0.002). SLCO1A2 and SLCO1B1 gene treatment over time interactions were associated with gametocytemia clearance rate (p FDR = 0.018 and p FDR = 0.024). ABCB1, ABCC4 and SLCO1B3 were not associated with treatment response. CONCLUSION: The present work presents the first pharmacogenetic report of an association between chloroquine/primaquine responses with OATP transporters.


Subject(s)
Chloroquine/therapeutic use , Liver-Specific Organic Anion Transporter 1/genetics , Malaria, Vivax/genetics , Organic Anion Transporters/genetics , Polymorphism, Genetic/genetics , Primaquine/therapeutic use , Adult , Antimalarials/therapeutic use , Brazil , Drug Therapy, Combination/methods , Female , Genotype , Humans , Malaria, Vivax/drug therapy , Male , Parasitemia/drug therapy , Parasitemia/genetics , Plasmodium vivax/drug effects , Treatment Outcome
19.
Sci Rep ; 7(1): 13835, 2017 10 23.
Article in English | MEDLINE | ID: mdl-29062028

ABSTRACT

Malaria is a global disease associated with considerable mortality and morbidity. An appropriately balanced immune response is crucial in determining the outcome of malarial infection. The glucocorticoid (GC) metabolising enzyme, 11ß-hydroxysteroid dehydrogenase-1 (11ß-HSD1) converts intrinsically inert GCs into active GCs. 11ß-HSD1 shapes endogenous GC action and is immunomodulatory. We investigated the role of 11ß-HSD1 in two mouse models of malaria. 11ß-HSD1 deficiency did not affect survival after malaria infection, but it increased disease severity and parasitemia in mice infected with Plasmodium chabaudi AS. In contrast, 11ß-HSD1 deficiency rather decreased parasitemia in mice infected with the reticulocyte-restricted parasite Plasmodium berghei NK65 1556Cl1. Malaria-induced antibody production and pathology were unaltered by 11ß-HSD1 deficiency though plasma levels of IL-4, IL-6 and TNF-α were slightly affected by 11ß-HSD1 deficiency, dependent on the infecting parasite. These data suggest that 11ß-HSD1 is not crucial for survival of experimental malaria, but alters its progression in a parasite strain-specific manner.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/deficiency , Malaria/metabolism , Parasitemia/metabolism , Plasmodium chabaudi/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Female , Malaria/genetics , Male , Mice , Mice, Mutant Strains , Parasitemia/genetics , Species Specificity
20.
Nat Commun ; 8(1): 223, 2017 08 09.
Article in English | MEDLINE | ID: mdl-28790316

ABSTRACT

Infection of mice with strains of Plasmodium yoelii parasites can result in different pathology, but molecular mechanisms to explain this variation are unclear. Here we show that a P. yoelii gene encoding a HECT-like E3 ubiquitin ligase (Pyheul) influences parasitemia and host mortality. We genetically cross two lethal parasites with distinct disease phenotypes, and identify 43 genetically diverse progeny by typing with microsatellites and 9230 single-nucleotide polymorphisms. A genome-wide quantitative trait loci scan links parasite growth and host mortality to two major loci on chromosomes 1 and 7 with LOD (logarithm of the odds) scores = 6.1 and 8.1, respectively. Allelic exchange of partial sequences of Pyheul in the chromosome 7 locus and modification of the gene expression alter parasite growth and host mortality. This study identifies a gene that may have a function in parasite growth, virulence, and host-parasite interaction, and therefore could be a target for drug or vaccine development.Many strains of Plasmodium differ in virulence, but factors that control these distinctions are not known. Here the authors comparatively map virulence loci using the offspring from a P. yoelii YM and N67 genetic cross, and identify a putative HECT E3 ubiquitin ligase that may explain the variance.


Subject(s)
Host-Parasite Interactions/genetics , Malaria/parasitology , Plasmodium yoelii/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Crosses, Genetic , Female , Mice, Inbred BALB C , Mice, Inbred C57BL , Parasitemia/genetics , Parasitemia/parasitology , Plasmodium yoelii/growth & development , Plasmodium yoelii/pathogenicity , Virulence/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...