Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Brief Bioinform ; 18(3): 394-402, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-27178992

RESUMO

The era of genome-wide association studies (GWAS) has led to the discovery of numerous genetic variants associated with disease. Better understanding of whether these or other variants interact leading to differential risk compared with individual marker effects will increase our understanding of the genetic architecture of disease, which may be investigated using the family-based study design. We present M-TDT (the multi-locus transmission disequilibrium test), a tool for detecting family-based multi-locus multi-allelic effects for qualitative or quantitative traits, extended from the original transmission disequilibrium test (TDT). Tests to handle the comparison between additive and epistatic models, lack of independence between markers and multiple offspring are described. Performance of M-TDT is compared with a multifactor dimensionality reduction (MDR) approach designed for investigating families in the hypothesis-free genome-wide setting (the multifactor dimensionality reduction pedigree disequilibrium test, MDR-PDT). Other methods derived from the TDT or MDR to investigate genetic interaction in the family-based design are also discussed. The case of three independent biallelic loci is illustrated using simulations for one- to three-locus alternative hypotheses. M-TDT identified joint-locus effects and distinguished effectively between additive and epistatic models. We showed a practical example of M-TDT based on three genes already known to be implicated in malaria susceptibility. Our findings demonstrate the value of M-TDT in a hypothesis-driven context to test for multi-way epistasis underlying common disease etiology, whereas MDR-PDT-based methods are more appropriate in a hypothesis-free genome-wide setting.


Assuntos
Epistasia Genética , Genoma , Estudo de Associação Genômica Ampla , Humanos , Desequilíbrio de Ligação , Modelos Genéticos , Linhagem
2.
J Infect Dis ; 217(11): 1690-1698, 2018 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-29490079

RESUMO

Background: Early detection of severe dengue can improve patient care and survival. To date, no reliable single-gene biomarker exists. We hypothesized that robust multigene signatures exist. Methods: We performed a prospective study on Cambodian dengue patients aged 4 to 22 years. Peripheral blood mononuclear cells (PBMCs) were obtained at hospital admission. We analyzed 42 transcriptomic profiles of patients with secondary dengue infected with dengue serotype 1. Our novel signature discovery approach controls the number of included genes and captures nonlinear relationships between transcript concentrations and severity. We evaluated the signature on secondary cases infected with different serotypes using 2 datasets: 22 PBMC samples from additional patients in our cohort and 32 whole blood samples from an independent cohort. Results: We identified an 18-gene signature for detecting severe dengue in patients with secondary infection upon hospital admission with a sensitivity of 0.93 (95% confidence interval [CI], .82-.98), specificity of 0.67 (95% CI, .53-.80), and area under the receiver operating characteristic curve (AUC) of 0.86 (95% CI, .75-.97). At validation, the signature had empirical AUCs of 0.85 (95% CI, .69-1.00) and 0.83 (95% CI, .68-.98) for the PBMCs and whole blood datasets, respectively. Conclusions: The signature could detect severe dengue in secondary-infected patients upon hospital admission. Its genes offer new insights into the pathogenesis of severe dengue.


Assuntos
RNA/sangue , Dengue Grave/sangue , Dengue Grave/diagnóstico , Adolescente , Adulto , Criança , Pré-Escolar , Coinfecção/sangue , Coinfecção/diagnóstico , Coinfecção/virologia , Vírus da Dengue/genética , Feminino , Marcadores Genéticos/genética , Hospitalização , Hospitais , Humanos , Leucócitos Mononucleares/virologia , Masculino , Estudos Prospectivos , Curva ROC , Sensibilidade e Especificidade , Sorogrupo , Transcriptoma/genética , Adulto Jovem
3.
J Infect Dis ; 212(12): 2011-20, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26063222

RESUMO

Dengue is a mosquito-borne viral disease that afflicts millions of individuals worldwide every year. Infection by any of the 4 dengue virus (DENV) serotypes can result in a spectrum of disease severity. We investigated the impact of variants of interferon-regulated innate immunity genes with a potent antiviral effect on the outcome of DENV infection. We compared the effect of OAS gene family variants on 2 DENV serotypes in cell culture. While both OAS1-p42 and p46 showed antiviral activity against DENV-2, only OAS1-p42 presented anti-DENV-1 activity. Conversely, whereas both OAS3_S381 and R381 variants were able to block DENV-1 infection, the anti-DENV-2 activity observed for OAS3_S381 was largely lost for the R381 variant. By means of an allelic association study of a cohort of 740 patients with dengue, we found a protective effect of OAS3_R381 against shock (odds ratio [OR], 0.37; P < .001). This effect was due to DENV-2 infections (OR, 0.13; P = .007) but was absent for DENV-1, in accordance with the serotype-dependent OAS3 activity found in the functional study. Severe dengue has long been associated with a cytokine storm of unclear origin. This work identifies an early innate immunity process that could lead to the immune overreaction observed in severe dengue and could be triggered by a specific host genotype-pathogen genotype interaction.


Assuntos
2',5'-Oligoadenilato Sintetase/genética , Vírus da Dengue/imunologia , Dengue/patologia , Predisposição Genética para Doença , 2',5'-Oligoadenilato Sintetase/metabolismo , Adolescente , Adulto , Células Cultivadas , Criança , Pré-Escolar , Dengue/genética , Dengue/imunologia , Feminino , Estudos de Associação Genética , Humanos , Lactente , Masculino , Adulto Jovem
5.
Nat Genet ; 37(5): 507-13, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15838506

RESUMO

Dengue fever and dengue hemorrhagic fever are mosquito-borne viral diseases. Dendritic cell-specific ICAM-3 grabbing nonintegrin (DC-SIGN1, encoded by CD209), an attachment receptor of dengue virus, is essential for productive infection of dendritic cells. Here, we report strong association between a promoter variant of CD209, DCSIGN1-336, and risk of dengue fever compared with dengue hemorrhagic fever or population controls. The G allele of the variant DCSIGN1-336 was associated with strong protection against dengue fever in three independent cohorts from Thailand, with a carrier frequency of 4.7% in individuals with dengue fever compared with 22.4% in individuals with dengue hemorrhagic fever (odds ratio for risk of dengue hemorrhagic fever versus dengue fever: 5.84, P = 1.4 x 10(-7)) and 19.5% in controls (odds ratio for protection: 4.90, P = 2 x 10(-6)). This variant affects an Sp1-like binding site and transcriptional activity in vitro. These results indicate that CD209 has a crucial role in dengue pathogenesis, which discriminates between severe dengue fever and dengue hemorrhagic fever. This may have consequences for therapeutic and preventive strategies.


Assuntos
Moléculas de Adesão Celular/genética , Dengue/genética , Lectinas Tipo C/genética , Regiões Promotoras Genéticas , Receptores de Superfície Celular/genética , Índice de Gravidade de Doença , Dengue/fisiopatologia , Humanos , Polimorfismo Genético
6.
Infect Immun ; 80(6): 2240-6, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22451520

RESUMO

An immunomodulatory role of arthropod saliva has been well documented, but evidence for an effect on Plasmodium sp. infectiousness remains controversial. Mosquito saliva may orient the immune response toward a Th2 profile, thereby priming a Th2 response against subsequent antigens, including Plasmodium. Orientation toward a Th1 versus a Th2 profile promotes IgG and IgE proliferation, respectively, where the former is crucial for the development of an efficient antiparasite immune response. Here we assessed the direct effect of mosquito bites on the density of Plasmodium falciparum asexual parasites and the prevalence of gametocytes in chronic, asymptomatic infections in a longitudinal cohort study of seasonal transmission. We additionally correlated these parasitological measures with IgE and IgG antiparasite and anti-salivary gland extract titers. The mosquito biting density was positively correlated with the asexual parasite density but not asexual parasite prevalence and was negatively correlated with gametocyte prevalence. Individual anti-salivary gland IgE titers were also negatively correlated with gametocyte carriage and were strongly positively correlated with antiparasite IgE titers, consistent with the hypothesis that mosquito bites predispose individuals to develop an IgE antiparasite response. We provide evidence that mosquito bites have an impact on asymptomatic infections and differentially so for the production of asexual and sexual parasites. An increased research focus on the immunological impact of mosquito bites during asymptomatic infections is warranted, to establish whether strategies targeting the immune response to saliva can reduce the duration of infection and the onward transmission of the parasite.


Assuntos
Culicidae/fisiologia , Imunoglobulina E/sangue , Imunoglobulina G/sangue , Mordeduras e Picadas de Insetos/complicações , Malária Falciparum/parasitologia , Plasmodium falciparum/fisiologia , Animais , Anticorpos Antiprotozoários/sangue , Doença Crônica , Estudos de Coortes , Culicidae/imunologia , Família , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Humanos , Mordeduras e Picadas de Insetos/imunologia , Malária Falciparum/sangue , Malária Falciparum/epidemiologia , Malária Falciparum/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Senegal/epidemiologia
7.
Sci Rep ; 9(1): 6319, 2019 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-31004099

RESUMO

Mosquitoes are colonized by a little-studied natural virome. Like the bacterial microbiome, the virome also probably influences the biology and immunity of mosquito vector populations, but tractable experimental models are lacking. We recently discovered two novel viruses in the virome of wild Anopheles and in colonies of the malaria vector Anopheles coluzzii: Anopheles C virus and Anopheles cypovirus. Here, we describe biological interactions between these two viruses and An. coluzzii mosquitoes. Viral abundance varies reproducibly during mosquito development. DNA forms of these viruses were not detected, and thus viral persistence is likely based on vertical transmission of RNA genomes. At least Anopheles C virus is vertically transmitted by an intraembryonic route. Relative abundance of the two viruses is inversely correlated in individual mosquitoes. One possible mechanism for this could be interactions with host immunity, and functional genomic analysis indicated differential influence of at least the Toll and JAK/STAT immune signaling pathways upon the viruses. The nonrandom distributions and interactions with host immunity suggest that these and other members of the natural virome may constitute a source of unrecognized heterogeneity in mosquito vector populations.


Assuntos
Anopheles , Genoma Viral , Mosquitos Vetores , Vírus de RNA/metabolismo , Animais , Anopheles/embriologia , Anopheles/virologia , Malária , Mosquitos Vetores/embriologia , Mosquitos Vetores/virologia , Vírus de RNA/genética
8.
Front Immunol ; 10: 1424, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31293584

RESUMO

Dengue virus (DENV) induces strong T and B cell responses upon infection. Hence, it is difficult to determine the contribution of cell-mediated immunity alone in the long lasting protection against DENV infection and disease. Numerous CD4+ and CD8+ T cell epitopes have been identified, mainly in the non-structural proteins of DENV. Taking into account the immunogenicity and peptide sequence conservation among the different DENV serotypes, a minimal DENV antigen, called DENV1-NS, has been designed. This antigen is enriched in conserved and highly antigenic epitopes located in the NS3, NS4B, and NS5 regions of DENV1. To evaluate the ability of the DENV1-NS poly-epitope to express the antigenic peptides in the context of different HLA class I molecules, we established its in vivo immunogenicity by measuring, after DNA immunization and electroporation, the activation of DENV-specific CD8 T cells in transgenic mice expressing the human HLA-A*0201, -A*2402, -B*0702, and -B*3502 class I alleles. We then engineered a lipid nanoparticle (LNP) encapsulated modified mRNA vaccine encoding DENV1-NS and tested immunogenicity and protection in these human HLA class I transgenic mice, after transient blockade of the interferon (IFN) type I receptor. Significant protection was observed, after two injections of the mRNA vaccine. Collectively, these data strongly support the development of T cell-based vaccines targeting immunodominant T cell epitopes that generate potent virus-specific T cell responses conferring immunity against DENV infection.


Assuntos
Antígenos Virais/imunologia , Vacinas contra Dengue/imunologia , Dengue/imunologia , Epitopos de Linfócito T/imunologia , Epitopos Imunodominantes/imunologia , Animais , Vírus da Dengue/imunologia , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Camundongos , Camundongos Transgênicos , RNA Mensageiro
9.
Infect Genet Evol ; 62: 60-72, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29673983

RESUMO

Population genetics theory predicted that rare frequent markers would be the main contributors for heritability of complex diseases, but meta-analyses of genome-wide association studies are revealing otherwise common markers, present in all population groups, as the identified candidate genes. In this work, we applied a population-genetics informed meta-analysis to 10 markers located in seven genes said to be associated with dengue fever disease. Seven markers (in PLCE1, CD32, CD209, OAS1 and OAS3 genes) have high-frequency and the other three (in MICB and TNFA genes) have intermediate frequency. Most of these markers have high discriminatory power between population groups, but their frequencies follow the rules of genetic drift, and seem to have not been under strong selective pressure. There was a good agreement in directional consistency across trans-ethnic association signals, in East Asian and Latin American cohorts, with heterogeneity generated by randomness between studies and especially by low sample sizes. This led to confirm the following significant associations: with DF, odds ratio of 0.67 for TNFA-rs1800629-A; with DHF, 0.82 for CD32-rs1801274-G; with DSS, 0.55 for OAS3-rs2285933-G, 0.80 for PLCE1-rs2274223-G and 1.32 for MICB-rs3132468-C. The overall genetic risks confirmed sub-Saharan African populations and descendants as the best protected against the severer forms of the disease, while Southeast and Northeast Asians are the least protected ones. European and close neighbours are the best protected against dengue fever, while, again, Southeast and Northeast Asians are the least protected ones. These risk scores provide important predictive information for the largely naïve European and North American regions, as well as for Africa where misdiagnosis with other hemorrhagic diseases is of concern.


Assuntos
Dengue/genética , Predisposição Genética para Doença , Regulação da Expressão Gênica , Marcadores Genéticos , Genótipo , Humanos , Polimorfismo de Nucleotídeo Único
10.
PLoS Negl Trop Dis ; 12(2): e0006202, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29447178

RESUMO

Ethnic diversity has been long considered as one of the factors explaining why the severe forms of dengue are more prevalent in Southeast Asia than anywhere else. Here we take advantage of the admixed profile of Southeast Asians to perform coupled association-admixture analyses in Thai cohorts. For dengue shock syndrome (DSS), the significant haplotypes are located in genes coding for phospholipase C members (PLCB4 added to previously reported PLCE1), related to inflammation of blood vessels. For dengue fever (DF), we found evidence of significant association with CHST10, AHRR, PPP2R5E and GRIP1 genes, which participate in the xenobiotic metabolism signaling pathway. We conducted functional analyses for PPP2R5E, revealing by immunofluorescence imaging that the coded protein co-localizes with both DENV1 and DENV2 NS5 proteins. Interestingly, only DENV2-NS5 migrated to the nucleus, and a deletion of the predicted top-linking motif in NS5 abolished the nuclear transfer. These observations support the existence of differences between serotypes in their cellular dynamics, which may contribute to differential infection outcome risk. The contribution of the identified genes to the genetic risk render Southeast and Northeast Asian populations more susceptible to both phenotypes, while African populations are best protected against DSS and intermediately protected against DF, and Europeans the best protected against DF but the most susceptible against DSS.


Assuntos
Povo Asiático/genética , Vírus da Dengue/genética , Dengue/genética , Genoma Viral/genética , Estudo de Associação Genômica Ampla , Dengue Grave/genética , Adolescente , Adulto , Sudeste Asiático , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Transporte/genética , Linhagem Celular , Núcleo Celular/virologia , Pré-Escolar , Estudos de Coortes , Dengue/virologia , Feminino , Expressão Gênica , Predisposição Genética para Doença , Genótipo , Humanos , Lactente , Masculino , Proteínas do Tecido Nervoso/genética , Razão de Chances , Proteína Fosfatase 2/genética , Proteínas Repressoras/genética , Sorogrupo , Dengue Grave/etnologia , Sulfotransferases , Tailândia , Fosfolipases Tipo C/genética , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética , Adulto Jovem
11.
Genome Biol Evol ; 9(11): 3202-3213, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29165566

RESUMO

The majority of bat rabies cases in Europe are attributed to European bat 1 lyssavirus (EBLV-1), circulating mainly in serotine bats (Eptesicus serotinus). Two subtypes have been defined (EBLV-1a and EBLV-1b), each associated with a different geographical distribution. In this study, we undertake a comprehensive sequence analysis based on 80 newly obtained EBLV-1 nearly complete genome sequences from nine European countries over a 45-year period to infer selection pressures, rates of nucleotide substitution, and evolutionary time scale of these two subtypes in Europe. Our results suggest that the current lineage of EBLV-1 arose in Europe ∼600 years ago and the virus has evolved at an estimated average substitution rate of ∼4.19×10-5 subs/site/year, which is among the lowest recorded for RNA viruses. In parallel, we investigate the genetic structure of French serotine bats at both the nuclear and mitochondrial level and find that they constitute a single genetic cluster. Furthermore, Mantel tests based on interindividual distances reveal the absence of correlation between genetic distances estimated between viruses and between host individuals. Taken together, this indicates that the genetic diversity observed in our E. serotinus samples does not account for EBLV-1a and -1b segregation and dispersal in Europe.


Assuntos
Evolução Biológica , Quirópteros/genética , Quirópteros/virologia , Lyssavirus/genética , Animais , Núcleo Celular/genética , DNA Mitocondrial/genética , Europa (Continente) , Genoma Viral , Interações Hospedeiro-Patógeno , Lyssavirus/classificação , Lyssavirus/isolamento & purificação , Repetições de Microssatélites , Seleção Genética
12.
Sci Transl Med ; 9(405)2017 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-28855396

RESUMO

Clinical symptoms of dengue virus (DENV) infection, the most prevalent arthropod-borne viral disease, range from classical mild dengue fever to severe, life-threatening dengue shock syndrome. However, most DENV infections cause few or no symptoms. Asymptomatic DENV-infected patients provide a unique opportunity to decipher the host immune responses leading to virus elimination without negative impact on an individual's health. We used an integrated approach of transcriptional profiling and immunological analysis to compare a Cambodian population of strictly asymptomatic viremic individuals with clinical dengue patients. Whereas inflammatory pathways and innate immune response pathways were similar between asymptomatic individuals and clinical dengue patients, expression of proteins related to antigen presentation and subsequent T cell and B cell activation pathways was differentially regulated, independent of viral load and previous DENV infection history. Feedback mechanisms controlled the immune response in asymptomatic viremic individuals, as demonstrated by increased activation of T cell apoptosis-related pathways and FcγRIIB (Fcγ receptor IIB) signaling associated with decreased anti-DENV-specific antibody concentrations. Together, our data illustrate that symptom-free DENV infection in children is associated with increased activation of the adaptive immune compartment and proper control mechanisms, leading to elimination of viral infection without excessive immune activation, with implications for novel vaccine development strategies.


Assuntos
Imunidade Adaptativa , Dengue/imunologia , Retroalimentação Fisiológica , Doença Aguda , Imunidade Adaptativa/genética , Anticorpos Antivirais/metabolismo , Apresentação de Antígeno/imunologia , Apoptose , Diferenciação Celular , Criança , Citocinas/sangue , Demografia , Dengue/sangue , Dengue/genética , Regulação da Expressão Gênica , Humanos , Imunidade Inata/genética , Inflamação/patologia , Ativação Linfocitária/imunologia , Plasmócitos/metabolismo , Linfócitos T/imunologia , Transcrição Gênica , Resultado do Tratamento , Carga Viral , Viremia/imunologia
13.
PLoS One ; 5(6): e11358, 2010 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-20613877

RESUMO

BACKGROUND: Studies on human genetic factors associated with malaria have hitherto concentrated on their role in susceptibility to and protection from disease. In contrast, virtually no attention has been paid to the role of human genetics in eliciting the production of parasite transmission stages, the gametocytes, and thus enhancing the spread of disease. METHODS AND FINDINGS: We analysed four longitudinal family-based cohort studies from Senegal and Thailand followed for 2-8 years and evaluated the relative impact of the human genetic and non-genetic factors on gametocyte production in infections of Plasmodium falciparum or P. vivax. Prevalence and density of gametocyte carriage were evaluated in asymptomatic and symptomatic infections by examination of Giemsa-stained blood smears and/or RT-PCR (for falciparum in one site). A significant human genetic contribution was found to be associated with gametocyte prevalence in asymptomatic P. falciparum infections. By contrast, there was no heritability associated with the production of gametocytes for P. falciparum or P. vivax symptomatic infections. Sickle cell mutation, HbS, was associated with increased gametocyte prevalence but its contribution was small. CONCLUSIONS: The existence of a significant human genetic contribution to gametocyte prevalence in asymptomatic infections suggests that candidate gene and genome wide association approaches may be usefully applied to explore the underlying human genetics. Prospective epidemiological studies will provide an opportunity to generate novel and perhaps more epidemiologically pertinent gametocyte data with which similar analyses can be performed and the role of human genetics in parasite transmission ascertained.


Assuntos
Reservatórios de Doenças , Plasmodium falciparum/patogenicidade , Plasmodium vivax/patogenicidade , Anemia Falciforme/parasitologia , Animais , Estudos de Coortes , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Talassemia alfa/parasitologia
14.
Virology ; 384(1): 216-22, 2009 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19056102

RESUMO

Chikungunya virus (CHIKV) becomes one of the most important mosquito-borne alphavirus in the medical field. CHIKV is highly sensitive to antiviral activity of Type-I interferons (IFN-alpha/beta). Here, we investigated the role of IFN-induced 2',5'-Oligoadenylate Synthetase (OAS) family in innate immunity to CHIKV. We established inducible human epithelial HeLa cell lines expressing either the large form of human OAS, OAS3, or the genetic variant OAS3-R844X which is predicted to lack about 20% of the OAS3 protein from the carboxy terminus. HeLa cells respond to ectopic OAS3 expression by efficiently inhibiting CHIKV growth. The characteristic of the antiviral effect was a blockade in early stages of virus replication. Thus, OAS3 pathway may represent a novel antialphaviral mechanism by which IFN-alpha/beta controls CHIKV growth. HeLa cells expressing the truncated form of OAS3 were less resistant to CHIKV infection, raising the question on the involvement of OAS3 genetic polymorphism in human susceptibility to alphavirus infection.


Assuntos
2',5'-Oligoadenilato Sintetase/farmacologia , Infecções por Alphavirus/tratamento farmacológico , Antivirais/farmacologia , Vírus Chikungunya/efeitos dos fármacos , 2',5'-Oligoadenilato Sintetase/genética , Infecções por Alphavirus/transmissão , Animais , Culicidae/virologia , Células HeLa/efeitos dos fármacos , Células HeLa/enzimologia , Células HeLa/virologia , Humanos , Interferon Tipo I/uso terapêutico , Transfecção
15.
Science ; 326(5959): 1546-9, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-20007901

RESUMO

Glucose-6-phosphate dehydrogenase (G6PD) deficiency--the most common known enzymopathy--is associated with neonatal jaundice and hemolytic anemia usually after exposure to certain infections, foods, or medications. Although G6PD-deficient alleles appear to confer a protective effect against malaria, the link with clinical protection from Plasmodium infection remains unclear. We investigated the effect of a common G6PD deficiency variant in Southeast Asia--the G6PD-Mahidol(487A) variant--on human survival related to vivax and falciparum malaria. Our results show that strong and recent positive selection has targeted the Mahidol variant over the past 1500 years. We found that the G6PD-Mahidol(487A) variant reduces vivax, but not falciparum, parasite density in humans, which indicates that Plasmodium vivax has been a driving force behind the strong selective advantage conferred by this mutation.


Assuntos
Deficiência de Glucosefosfato Desidrogenase/genética , Glucosefosfato Desidrogenase/genética , Malária Vivax/parasitologia , Mutação , Plasmodium vivax/fisiologia , Seleção Genética , Envelhecimento , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Feminino , Dosagem de Genes , Estudos de Associação Genética , Predisposição Genética para Doença , Genótipo , Deficiência de Glucosefosfato Desidrogenase/sangue , Deficiência de Glucosefosfato Desidrogenase/complicações , Haplótipos , Humanos , Imunidade Inata , Malária Falciparum/complicações , Malária Falciparum/genética , Malária Falciparum/parasitologia , Malária Vivax/complicações , Malária Vivax/genética , Masculino , Plasmodium falciparum/fisiologia , Polimorfismo de Nucleotídeo Único , Tailândia
16.
PLoS One ; 3(12): e3887, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19060954

RESUMO

The majority of studies concerning malaria host genetics have focused on individual genes that confer protection against rather than susceptibility to malaria. Establishing the relative impact of genetic versus non-genetic factors on malaria infection and disease is essential to focus effort on key determinant factors. This relative contribution has rarely been evaluated for Plasmodium falciparum and almost never for Plasmodium vivax. We conducted a longitudinal cohort study in a Karen population of 3,484 individuals in a region of mesoendemic malaria, Thailand from 1998 to 2005. The number of P. falciparum and P. vivax clinical cases and the parasite density per person were determined. Statistical analyses were performed to account for the influence of environmental factors and the genetic heritability of the phenotypes was calculated using the pedigree-based variance components model. The genetic contribution to the number of clinical episodes resulting from P. falciparum and P. vivax were 10% and 19% respectively. There was also moderate genetic contribution to the maximum and overall parasite trophozoite density phenotypes for both P. falciparum (16%&16%) and P. vivax (15%&13%). These values, for P. falciparum, were similar to those previously observed in a region of much higher transmission intensity in Senegal, West Africa. Although environmental factors play an important role in acquiring an infection, genetics plays a determinant role in the outcome of an infection with either malaria parasite species prior to the development of immunity.


Assuntos
Etnicidade/genética , Padrões de Herança/genética , Malária Falciparum/genética , Malária Vivax/genética , Plasmodium falciparum/genética , Plasmodium vivax/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Tipagem e Reações Cruzadas Sanguíneas , Criança , Pré-Escolar , Sistema do Grupo Sanguíneo Duffy , Meio Ambiente , Feminino , Humanos , Lactente , Recém-Nascido , Malária Falciparum/sangue , Malária Falciparum/epidemiologia , Malária Vivax/sangue , Malária Vivax/epidemiologia , Masculino , Pessoa de Meia-Idade , Fenótipo , Tailândia/epidemiologia
17.
PLoS One ; 3(4): e2000, 2008 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-18431485

RESUMO

Plasmodium falciparum malaria episodes may vary considerably in their severity and clinical manifestations. There is good evidence that host genetic factors contribute to this variability. To date, most genetic studies aiming at the identification of these genes have used a case/control study design for severe malaria, exploring specific candidate genes. Here, we performed a family-based genetic study of falciparum malaria related phenotypes in two independent longitudinal survey cohorts, as a first step towards the identification of genes and mechanisms involved in the outcome of infection. We studied two Senegalese villages, Dielmo and Ndiop that differ in ethnicity, malaria transmission and endemicity. We performed genome-scan linkage analysis of several malaria-related phenotypes both during clinical attacks and asymptomatic infection. We show evidence for a strong genetic contribution to both the number of clinical falciparum malaria attacks and the asymptomatic parasite density. The asymptomatic parasite density showed linkage to chromosome 5q31 (LOD = 2.26, empirical p = 0.0014, Dielmo), confirming previous findings in other studies. Suggestive linkage values were also obtained at three additional chromosome regions: the number of clinical malaria attacks on chromosome 5p15 (LOD = 2.57, empirical p = 0.001, Dielmo) and 13q13 (LOD = 2.37, empirical p = 0.0014 Dielmo), and the maximum parasite density during asymptomatic infection on chromosome 12q21 (LOD = 3.1, empirical p<10(-4), Ndiop). While regions of linkage show little overlap with genes known to be involved in severe malaria, the four regions appear to overlap with regions linked to asthma or atopy related traits, suggesting that common immune related pathways may be involved.


Assuntos
Mapeamento Cromossômico , Malária Falciparum/genética , Característica Quantitativa Herdável , Animais , Etnicidade/genética , Família , Genoma Humano , Humanos , Fenótipo , Análise de Regressão , População Rural , Senegal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA