Acessibilidade / Reportar erro

Molecular genotyping of G6PD mutations and Duffy blood group in Afro-descendant communities from Brazilian Amazon

Abstract

Glucose-6-phosphate dehydrogenase deficiency (G6PDd) and Duffy-negative blood group are two red blood cells variants that confer protection against malaria. In this study, the distribution of the most common G6PD variants (G6PD*A-, GGPD*A and G6PD Mediterranean) and the major alleles of the Duffy blood group (FY*A, FY*B and FY*BES) were investigated in an Afro-descendant population from state of Pará, Brazilian Amazon. G6PD variants and Duffy blood group alleles were determined by TaqMan SNP genotyping assay. Overall, molecular genotyping revealed the presence of G6PD variants in 126 (24%) of the individuals studied (5% male and 19% female), and frequencies of the G6PD*A- and G6PD*A alleles were 0.061 and 0.104, respectively. Duffy blood group genotyping showed that 24.3% of people were Duffy-negative and 41.3% were heterozygous for FY*BES. The frequency of allele FY*BES was 41.0%. The results emphasize the need to monitor G6PD deficiency for the use of primaquine in the routine care of the Afro-descendant communities of the Trombetas, Erepecuru and Cumná rivers, evaluating the risks of hemolytic crisis in case of recurrence of malaria in the region. In addition, the possible greater protection against malaria conferred by these erythrocyte polymorphisms deserves to be better investigated and explored among these Afro-descendants.

Keywords:
G6PD mutations; Duffy blood group; Afro-descendants; Brazilian Amazon

Introduction

Deficiency of glucose-6-phosphate dehydrogenase (G6PDd) and Duffy-negative blood group are two red blood cells variants that exhibit pattern of population distribution strongly influenced by natural selection by malaria. These variants are two well-known hereditary alterations, in addition to hemoglobin disorders and immunogenic variants, conferring protection against malaria, commonly found worldwide in areas inhabited by populations historically exposed to endemic malaria, including Africa, Mediterranean Europe, Southeast Asia and Latin America (review in Monteiro et al., 2014).

G6PDd is primarily associated with protection against falciparum malaria (Ruwende et al., 1995Ruwende C, Khoo SC, Snow RW, Yates SNR, Kwiatkowski D, Gupta A, Warn P, Allsopp CEM, Gilbert SC, Peschu N et al. (1995) Natural protection of hemi- and heterozygotes for G6PD deficiency in Africa by resistance to severe malaria. Nature 376:246–249), but a protective effect of G6PDd has also been shown against malaria episodes caused by Plasmodium vivax (Leslie et al., 2010Leslie T, Briceño M, Mayan I, Mohammed N, Klinkenberg E, Sibley CH, Whitty CJM and Rowland M (2010) The impact of phenotypic and genotypic G6PD deficiency on risk of Plasmodium vivax infection: A case–control study amongst Afghan refugees in Pakistan. PLoS Med 7:e1000283., Santana et al., 2013Santana MS, Monteiro WM, Siqueira AM, Costa MF, Sampaio V, Lacerda MV and Alecrim MG (2013) Glucose-6-phosphate dehydrogenase deficient variants are associated with reduced susceptibility to malaria in the Brazilian Amazon. Trans R Soc Trop Med Hyg 107:301-306.). Besides the protective effects against malaria infection, hemolysis induced by antimalarial drugs (primaquine) in G6PDd individuals is of major public health importance. The gene encoding G6PD, situated near the telomeric region of the long arm of the X chromosome (band Xq28), consists of 13 axons with a length of 18 kb (Pai et al., 1980Pai GS, Sprenkle JA, Do TT, Mareni CE and Migeon BR (1980) Localization of loci for hypoxanthine phosphoribosyltransferase and glucose-6-phosphate dehydrogenase and biochemical evidence of nonrandom X chromosome expression from studies of a human X-autosome translocation. Proc Natl Acad Sci USA 77:2810–2813.). Over 400 biochemical variants have been described and at least 217 mutations were identified in the G6PD gene. Not all mutations are polymorphic and of public health importance, and many appear only sporadically within populations. Almost half of the mutations identified are associated with the most serious clinical phenotypes and are very rare (Howes et al., 2013Howes RE, Battle KE, Satyagraha AW, Baird JK and Hay SI (2013) G6PD deficiency: Global distribution, genetic variants and primaquine therapy. Adv Parasitol 81:133-201.). The variants are subdivided into five classes based on the functional severity of the deficiency: Class I, deficient variants associated with non-spherocytic hemolytic anemia; Class II, variants with severe deficiency (less than 10% of activity); Class III, variants with moderate deficiency (10% to 60% of activity); Class IV, variants with normal or slightly decreased activity (60% to 100%); Class V, variants with increased enzymatic activity. Clinically important variants are those of classes II and III because they are more common than those of class I, which are sporadic (Yoshida et al., 1971Yoshida A, Beutler E and Motulsky AG (1971) Human glucose-6-phosphate dehydrogenase variants. Bull World Health Organ 45:243–253.; WHO Working Group, 1989WHO Working Group (1989) Glucose-6-phosphate dehydrogenase deficiency. Bull World Health Organ 67:601–611.).

Among all the known deficiencies, the G6PD*A and G6PD*Med (G6PD Mediterranean) variants are the most frequent in certain populations and are the main responsible for the occurrence of haemolytic events. The G6PD*A- variant predominates in sub-Saharan Africa where it affects 15 to 20% of the African population (review in Howes et al., 2013Howes RE, Battle KE, Satyagraha AW, Baird JK and Hay SI (2013) G6PD deficiency: Global distribution, genetic variants and primaquine therapy. Adv Parasitol 81:133-201.). Until recently the G6PD*A- variant, a Class III type, was considered homogeneous when studied biochemically, but with the advent of molecular biology methods, DNA-level investigation has shown that this variant is heterogeneous. All types of G6PD*A- have in common the rs1050829 (G6PD c.376T>C mutation), that characterizes G6PD*A variant (Takizawa et al., 1987Takizawa T, Yoneyama Y, Miwa S and Yoshida A (1987) A single nucleotide base transition is the basis of the common human glucose-6-phosphate dehydrogenase variant A(+). Genomics 1:228-231.), in addition to a second mutation, more commonly the rs1050828 (G6PD c.202C>T) (Hirono and Beutler, 1988Hirono A and Beutler E (1988) Molecular cloning and nucleotide sequence of cDNA for human glucose-6-phosphate dehydrogenase variant A (-). Proc Natl Acad Sci USA 85:3951-3954.), or rs137852328 (c.680G>T) (Beutler et al., 1989Beutler E, Kuhl W, Vives-Corrons JL and Prchal JY (1989) Molecular heterogeneity of glucose-6-phosphate dehydrogenase A-. Blood 74:2550-2555.), or rs76723693 (c.968T>C) (Beutler et al., 1989Beutler E, Kuhl W, Vives-Corrons JL and Prchal JY (1989) Molecular heterogeneity of glucose-6-phosphate dehydrogenase A-. Blood 74:2550-2555.). The African variant G6PD*A (c.376T>C) has electrophoretic mobility similar to that of the A- variant, and normal or very-mild deficiency (Class IV) enzymatic activity (Takizawa et al., 1987Takizawa T, Yoneyama Y, Miwa S and Yoshida A (1987) A single nucleotide base transition is the basis of the common human glucose-6-phosphate dehydrogenase variant A(+). Genomics 1:228-231.). This variant occurs in 20-30% of Africans (Beutler, 1996Beutler E (1996) G6PD: Population genetics and clinical manifestations. Blood Rev 10:45-52.). The Mediterranean variant, rs5030868 (c.563C>T transition), a Class II variant (Vulliamy et al., 1988Vulliamy TJ, D’Urso M, Battistuzzi G, Estrada M, Foulkes NS, Martini G, Calabro V, Poggi V, Giordano R, Town M et al. (1988) Diverse point mutations in the human glucose 6-phosphate dehydrogenase gene cause enzyme deficiency and mild or severe hemolytic anemia. Proc Natl Acad Sci USA 85:5171-5175.), is found in southern Europe, the Middle East and in the Indian subcontinent (Beutler, 1996Beutler E (1996) G6PD: Population genetics and clinical manifestations. Blood Rev 10:45-52.; Howes et al., 2013Howes RE, Battle KE, Satyagraha AW, Baird JK and Hay SI (2013) G6PD deficiency: Global distribution, genetic variants and primaquine therapy. Adv Parasitol 81:133-201.).

In Latin America, low prevalence rates of G6PDd are documented in Argentina, Bolivia, Mexico, Peru and Uruguay, but studies from Curaçao, Ecuador, Jamaica, Saint Lucia, Suriname and Trinidad, as well as some areas of Brazil, Colombia and Cuba, have shown a high prevalence (>10%) of G6PDd (review in Howes et al., 2013Howes RE, Battle KE, Satyagraha AW, Baird JK and Hay SI (2013) G6PD deficiency: Global distribution, genetic variants and primaquine therapy. Adv Parasitol 81:133-201.; Monteiro et al., 2014Monteiro WM, Val FF, Siqueira AM, Franca GP, Sampaio VS, Melo GC, Almeida AC, Brito MA, Peixoto HM, Fuller D et al. (2014) G6PD deficiency in Latin America: Systematic review on prevalence and variants. Mem Inst Oswaldo Cruz 109:553-68.; Gómez-Manzo et al., 2016Gómez-Manzo S, Marcial-Quino J, Vanoye-Carlo A, Serrano-Posada H, Ortega-Cuellar D, González-Valdez A, Castillo-Rodríguez RA, Hernández-Ochoa B, Sierra-Palacios E, Rodríguez-Bustamante E et al. (2016) Glucose-6-phosphate dehydrogenase: Update and analysis of new mutations around the World. Int J Mol Sci 17:2069.). G6PD*A– (376G/202A) is the variant most broadly distributed across Latin America, found in 81% of the deficient individuals surveyed (review in Monteiro et al., 2014Monteiro WM, Val FF, Siqueira AM, Franca GP, Sampaio VS, Melo GC, Almeida AC, Brito MA, Peixoto HM, Fuller D et al. (2014) G6PD deficiency in Latin America: Systematic review on prevalence and variants. Mem Inst Oswaldo Cruz 109:553-68.; Gómez-Manzo et al., 2016Gómez-Manzo S, Marcial-Quino J, Vanoye-Carlo A, Serrano-Posada H, Ortega-Cuellar D, González-Valdez A, Castillo-Rodríguez RA, Hernández-Ochoa B, Sierra-Palacios E, Rodríguez-Bustamante E et al. (2016) Glucose-6-phosphate dehydrogenase: Update and analysis of new mutations around the World. Int J Mol Sci 17:2069.).

The Duffy glycoprotein, also known as the Duffy antigen receptor for chemokines (DARC), is a transmembrane glycoprotein that functions as a chemokine transporter. It is also a receptor for Plasmodium vivax and Plamodium knowlesi and expresses the Duffy blood group antigens (Fy). The Duffy blood group locus, at position q21–q25 on chromosome 1 (Donahue et al., 1968Donahue RP, Bias WB, Renwick JH and McKusick VA (1968) Probable assignment of the duffy blood group locus to chromosome in man. Proc Natl Acad Sci USA 61:949-955.), is characterized by three main alleles: FY*A, FY*B and FY*BES. The FY*A and FY*B alleles are distinguished by a missense mutation, which results in a single amino acid difference and gives the common Fy(a-b-), Fy(a-b+) and Fy(a+b+) phenotypes (Chaudhuri et al., 1995Chaudhuri A, Polyakova J, Zbrzezna V and Pogo AO (1995) The coding sequence of Duffy blood group gene in humans and simians: Restriction fragment length polymorphism, antibody and malarial parasite specificities, and expression in no erythroid tissues in Duffy-negative individuals. Blood 85:615–621.; Iwamoto et al., 1995Iwamoto S, Omi T, Kajii E and Ikemoto S (1995) Genomic organization of the glycoprotein D gene: Duffy blood group Fya/Fyb alloantigen system is associated with a polymorphism at the 44-amino acid residue. Blood 85:622–626.; Mallinson et al., 1995Mallinson G, Soo KS, Schall TJ, Pisacka M and Anstee DJ (1995) Mutation in the erythrocyte chemokine receptor (Duffy) gene: The molecular basis of the Fya/Fyb antigens and identification of a deletion in the Duffy gene of an apparently healthy individual with the Fy(a-b-) phenotype. Brit J Hematol 90:823–829.; Tournamille et al., 1995aTournamille C, Le Van Kim C, Gane P, Cartón PJ and Colin Y (1995a) Molecular basis and PCR- typing of the FY*A/FY*B blood group polymorphism. Hum Genet 95:407-410.). The FY*BES allele, which corresponds to the Fy(a-b-) serological phenotype (i.e., the absence of Fy antigen), is due to a T-33C point mutation on the FY*B gene promoter, which abolishes the erythroid gene expression by disrupting a binding site for the GATA-1 erythroid transcription factor. This mutation results in the elimination of the transcription of FY mRNA in red blood cells (RBCs), but not in other cell types (Tournamille et al., 1995bTournamille C, Colin Y, Cartron JP and Kim CLV (1995b) Disruption of a GATA motif in the Duffy gene promoter abolishes erythroid gene expression in Duffy-negative individual. Nat Genet 10:224–228.; Pogo and Chaudhuri, 2000Pogo AO and Chaudhuri A (2000) The Duffy protein: A malarial and chemokine receptor. Semin Hematol 37:122–129.). The same mutation associated with the FY*A variant (FY*AES allele) was already identified at low frequencies in individuals living in a P. vivax-endemic region of Papua New Guinea (Zimmerman et al., 1999Zimmerman PA, Woolley I, Masinde GL, Miller SM, McNamara DT, Hazlett F, Mgone CS, Alpers MP, Genton B and Kazura JW (1999) Emergence of FY*A (null) in a Plasmodium vivax-endemic region of Papua New Guinea. Proc Natl Acad Sci USA 96:13973–13977.; Kasehagen et al., 2007Kasehagen LJ, Mueller I, Kiniboro B, Bockarie MJ, Reeder JC, Kazura JW, Kastens W, McNamara DT, King CH, Whalen CC et al. (2007) Reduced Plasmodium vivax erythrocyte infection in PNG Duffy-negative heterozygotes. PLoS One 2:e336.).

The distribution of Duffy alleles reveal strong geographic patterns, particularly the distribution of the silent FY*BES allele across sub-Saharan Africa. The FY*BES allele is at or near fixation in most sub-Saharan African populations. This allele is also found at high frequencies across Madagascar and through the Arabian Peninsula (above 80% and 50%, respectively), and at median frequencies (5–20%) across India and up to 11% in South-East Asia. However, it is very rare outside Africa, the lowest frequencies of the FY*BES allele being found in the Americas (review in Howes et al., 2011Howes RE, Patil AP, Piel FB, Nyangiri OA, Kabaria CW, Gething PW, Zimmerman PA, Barnadas C, Beall CM, Gebremedhin A et al. (2011) The global distribution of the Duffy blood group. Nat Commun 2:266.). The pattern of allele frequencies at FY locus has been attributed to a positive natural selection, based on the observation that individuals homozygous for the FY*BES allele are highly resistant to vivax malaria, since P. vivax requires the presence of Duffy antigen receptor for chemokines on the RBC surface to be able to invade cells and cause disease (Miller et al., 1976Miller LH, Mason SJ, Clyde DF and McGinniss MH (1976) The resistance factor to Plasmodium vivax in blacks: The Duffy-blood-group genotype, FyFy. N Engl J Med 295:302-304., Livingstone, 1984Livingstone FB (1984) The Duffy blood groups, vivax malaria, and malaria selection in human populations: A review. Hum Biol 56:413–425.; Tournamille et al., 1995bTournamille C, Colin Y, Cartron JP and Kim CLV (1995b) Disruption of a GATA motif in the Duffy gene promoter abolishes erythroid gene expression in Duffy-negative individual. Nat Genet 10:224–228.; Hadley and Peiper, 1997Hadley TJ and Peiper SC (1997) From malaria to chemokine receptor: The emerging physiologic role of the Duffy blood group antigen. Blood 89:3077-3091.).

In this study, the distribution of the most common variants of glucose-6-phosphate dehydrogenase, G6PD*A- (G202A and A376G alleles), G6PD*A (A376G allele) and G6PD*Med (C563T allele), and the major alleles of the blood system Duffy (FY*A, FY*B and FY*BES), RBC variants that confer protection against malaria, were investigated in Afro-descendant communities along the Trombetas, Erepecuru and Cuminá rivers, in the rural area of the municipality of Oriximiná, in the northeast of the state of Pará, in the Brazilian Amazon. Until recently, this region was considered endemic for malaria but, as in the state of Pará and Brazil in general, there was a substantial reduction in the incidence of microscopically confirmed malaria cases between 2000 and 2016, due to the intensification of malaria control and prevention through the National Malaria Control Program (PNCM), Brazilian Ministry of Health. (Sivep-Malária, 2017Sivep-Malária (2017) Serviço de Vigilância Epidemiológica da Malária – SIVEP – Malária; http://portalweb04.saude.gov.br/sivep_malaria.
http://portalweb04.saude.gov.br/sivep_ma...
). Based on this scenario, the present study has bioanthropological purposes, addressing the genetic variability associated to genetic variants considered as markers of African ancestry. At the same time, knowledge of the frequencies of these variants will allow assessing the risks of hemolytic crisis triggered by the use of primaquine in the treatment of malaria, as well as contribute to clarify aspects of the epidemiology of transmission of this pathology, with a description of biological factors of the human host that participate in malaria protection mechanisms.

Subjects and Methods

Sample collection

Study protocol and sample collection were approved by the Ethics Committee on Human Research at the Institute of Health Sciences of the Federal University of Pará. This study was carried out in Afro-descendant communities along the Trombetas, Erepecuru and Cuminá rivers, a rural area in the municipality of Oriximiná, in the northeast of the state of Pará, in the Brazilian Amazon (Figure 1). Venous blood samples were collected in tubes containing EDTA, after informed consent, in a cross-sectional study conducted in these communities in 2011. Blood samples were collected from 594 individuals, aged between 15 and 70 years (mean age 35 years), 54.7% females. Two hundred and seventeen infividuals were from communities along the Trombetas River (Arancuan, Tapagem, Abuí and Cachoeira Porteira), a population estimated at 870 people, and 377 were from communities along the Erepecuru and Cuminá Rivers (Serrinha, Jauari, Araçá de Fora, Jarauacá and Boa Vista do Cuminá), with a total population of about 1320 people. These communities, known in the region as the “Blacks of the Trombetas”, are remnants of a quilombo created in 1820 under the leadership of the slave Atanásio, and by 1823 it had a population of about 2,000 runaway slaves when it was destroyed by the Portuguese (Klein, 1986Klein HS (1986) African slavery in Latin America and the Caribbean. Oxford University Press, New York, 311 p.).

Figure 1
Geographical locations of Afro-descendant communities in the municipality of Oriximiná, northeast of the state of Pará.

DNA extraction

DNA was extracted from 300 mL of EDTA-treated blood using the NeoIsoColumn kit (One Lambda Inc., San Diego, CA, USA) according to the manufacturer’s instructions. DNA was eluted in 200 μL of elution buffer (provided with the kit).

G6PD genotyping

The single nucleotide polymorphisms (SNPs) rs1050829 (G6PD c.376T>C), rs1050828 (G6PD c.202C>T) and rs5030868 (c.563C>T) were investigated by a TaqMan SNP genotyping assay (Applied BioSystems, Foster City, CA, USA) according to the manufacturer’s instructions. Pre-designed probes were ordered for the genotyping analyses. About 10–50 ng of DNA were amplified with 5 μL of 2X TaqMan Universal PCR master mix, 0.5 μL of 40X primer and TaqMan probe dye mix. Cycling conditions were 10 min at 95 °C, followed by 40 cycles of 15 s at 92 °C and 1 min at 60 °C. Allelic discrimination was performed on an Applied BioSystems Real Time-PCR system.

Duffy blood group genotyping

The samples were genotyped using primers (forward and reverse) and TaqMan fluorescence-labeled probes for real-time PCR designed using File Builder 3.1, Applied Biosystems, to genotype FY*A (G125A, rs2814778), FY*B (G125A, rs12075) and FY*BES (T-33C, rs863002) alleles at the Duffy blood group locus. Allelic discrimination was also performed on a real-time PCR platform (Applied Biosystems 7500 analytical PCR system (SDS version 1.7).

Double heterozygotes in the promoter region (T-33C) and in the coding sequence (G125A) were also genotyped using an allele-specific PCR technique described by Olsson et al. (1998)Olsson ML, Hansson C, Avent ND, Akesson IE, Green CA and Daniels GL (1998) A clinically applicable method for determining the Duffy (Fy) blood group locus using polymerase chain reaction with allele-specific primers. Transfusion 38:168-173.. Amplification was performed for each subject with sense primers corresponding to the normal and GATA-1-mutated promoter sequence combined with antisense primers that discriminate the FY*A and FY*B alleles in four different combinations of primers. PCR products were separated electrophoretically in 1.5% agarose gels at 150 V for 30 min and visualized with SYBR® Safe DNA gel stain under UV excitation. G6PD allele frequencies were calculated by direct counting and expected Hardy-Weinberg values were estimated using Chi-squared method.

Results

G6PD genotypes and allele frequencies

The observed G6PD genotypes distribution and the estimated G6PD allele frequencies are shown in Table 1. The G6PD*A- (376G/202A) variant was identified in 10 (1.9%) hemizygous males and in 36 (6.5%) females (32 heterozygous and 4 homozygous). The G6PD*A (376G) variant was found in 17 (3.2%) hemizygous males and in 63 (11.6%) females (55 heterozygous and 8 homozygous). The Mediterranean variant (G6PD c.563C>T), was not detected. Overall, 5% of the males and 19% of the females presented a G6PD variant, and the global frequencies of the G6PD*B, G6PD*A- and G6PD*A alleles were 0.835, 0.061 and 0.104, respectively. No deviation from Hardy–Weinberg equilibrium was observed in the sample studied (p=0.22).

Table 1
G6PD allele frequencies in Afro-descendant communities from Brazilian Amazon.

When the distribution of the G6PD variants in the communities was analyzed according to geographical location (Trombetas and Erepecuru rivers), it was possible to observe that in the Trombetas communities the G6PD*A- variant is more common than the G6PD*A variant (10%, 4% and 7.5%, respectively), contrary to what was observed in the Erepecuru communities where the G6PD*A variant predominates. Overall, the communities of Erepecuru exhibited a lower frequency of the A- variant (3.3% versus 10.2%) and a higher frequency of variant A (12.1% versus 7.5%) when compared to the Trombetas communities. No deviations from Hardy-Weinberg equilibrium were observed in the studied communities (Erepecuru, p=0.07; Trombetas, p=0.12).

Duffy blood group genotypes and allele frequencies

Overall, 20.4% (121/594) individuals were genotyped as Duffy-negative (FY*BES/*BES) and 41,3% (254/594) were heterozygous for the FY*B ES allele (26.0% FYA/FYBES and 15.3% FYB/FYBES). In the total sample, the frequency of allele FY*BES was the highest (41.0%) (Table 2). The genotype frequencies observed in the communities grouped by geographical location revealed a similar profile, with predominance of the FY*BES/FY*BES genotype in both regions (17.9% in the Erepecuru river and 25.1% in the Trombetas river) and high heterozygote frequencies for the FY*BES allele (40.4% in the Erepecuru River and 41.3% in the Trombetas River). The FY*BES allele was also the most common one in the communities of both regions, but the frequency observed in the Trombetas River (47.5%) was higher than that found in the Erepecuru River (37.6%). Allele frequencies were in Hardy-Weinberg equilibrium both in the global sample and in the communities grouped by geographical location (Trombetas and Erepecuru), with p=0.22.

Table 2
Duffy allele frequencies in Afro-descendant communities from Brazilian Amazon.

Discusssion

G6PD variants

The results obtained in this study with Afro-descendants of Pará in the Brazilian Amazon, the “Blacks of the Trombetas”, can be considered as expected for a population of predominantly African origin, with some degree of local differentiation and admixture with people of Caucasian and Amerindian ancestry. The G6PD A- (376 A>G and 202 G>A) and A (376 A>G mutation) variants, found here at frequencies of 0.061 and 0.104, respectively, are the most common variants in Africa, particularly in sub-Saharan Africa, where they reach frequencies between 15% and 30%. The frequencies observed for these variants among the Afrodescendants were lower than those described for most of the sub-Saharan African populations (Carter et al., 2011Carter N, Pamba A, Duparc S and Waitumbi JN (2011) Frequency of glucose-6-phosphate dehydrogenase deficiency in malaria patients from six African countries enrolled in two randomized anti-malarial clinical trials. Malar J 10:241.; Howes et al., 2013Howes RE, Battle KE, Satyagraha AW, Baird JK and Hay SI (2013) G6PD deficiency: Global distribution, genetic variants and primaquine therapy. Adv Parasitol 81:133-201.), but are similar to those found by Millimono et al. (2012)Millimono TS, Loua KM, Rath SL, Relvas L, Bento C, Diakite M, Jarvis M, Daries N, Ribeiro LM, Manco L and Kaeda JS (2012) High prevalence of hemoglobin disorders and glucose-6-phosphate dehydrogenase (G6PD) deficiency in the Republic of Guinea (West Africa). Hemoglobin 36:25-37. in the Republic of Guinea, West Africa, as follows: G6PD*A- (A376G/G202A), 5.7% and G6PD*A (A376G), 17.6%.

Comparisons of the distribution of the G6PD variants in Afro-descendants studied here with those observed in other Brazilian populations can be made with other studies carried out in Brazil using the same approach, namely the use of molecular biology methods for genotyping, regardless of previous enzymatic activity screening. The frequency of the G6PD*A- variant among the Afro-descendants studied here (6.1%) is somewhat higher than that observed by Santana et al. (2013)Santana MS, Monteiro WM, Siqueira AM, Costa MF, Sampaio V, Lacerda MV and Alecrim MG (2013) Glucose-6-phosphate dehydrogenase deficient variants are associated with reduced susceptibility to malaria in the Brazilian Amazon. Trans R Soc Trop Med Hyg 107:301-306. among males from the population of Manaus, state of Amazonas (3.8%) and by Dombrowski et al. (2017)Dombrowski JG, Souza ST, Curry J, Hinton L, Silva NRM, Grignard L, Gonçalves LA, Gomes AR, Epiphanio S, Drakeley C et al. (2017) G6PD deficiency alleles in a malaria-endemic region in the Western Brazilian Amazon. Malar J 16:253-262. in males from the Juruá valley, state of Acre, northern Brazil. It is also higher than the frequency found by Castro et al. (2007)Castro SM, Weber R, Matte U and Giugliani R (2007) Molecular characterization of glucose-6-phosphate dehydrogenase deficiency in patients from the southern Brazilian city of Porto Alegre, RS. Genet Mol Biol 30:10-13. in newborns from Porto Alegre, southern Brazil (2.9%), but is similar to those found in Campinas (6.1%) and São Paulo (5.8%), southeastern Brazil, by Mezzacappa et al. (2010)Mezzacappa MA, Facchini FP, Pinto AC, Cassone AEL, Souza DS, Bezerra MAC, Albuquerque DM, Saad STO and Costa FF (2010) Clinical and genetic risk factors for moderate hyperbilirubinemia in Brazilian newborn infants. J Perinatol 30:819-826. and Oliveira et al. (2009)Oliveira RAG, Oshiro M, Hirata MH, Hirata RD, Ribeiro GS, Medeiros T and Barretto OCDO (2009) A novel point mutation in a class IV glucose-6-phosphate dehydrogenase variant (G6PD São Paulo) and polymorphic G6PD variants in São Paulo State, Brazil. Genet Mol Biol 32:251-254., respectively. On the other hand, the frequency of G6PD*A- in the current study is lower than the frequency of 8.2% reported by Moura-Neto et al. (2008)Moura-Neto JP, Dourado MV, Reis MG and Gonçalves MS (2008) A novel c.197T>A variant among Brazilian neonates with glucose-6-phosphate dehydrogenase deficiency. Genet Mol Biol 31:33-35. for the population of Salvador, Bahia, northeastern Brazil, a population of predominantly African origin (Santos et al., 2016Santos HC, Horimoto AV, Tarazona-Santos E, Rodrigues-Soares F, Barreto ML, Horta BL, Lima-Costa MF, Gouveia MH, Machado M, Silva TM et al. (2016) A minimum set of ancestry informative markers for determining admixture proportions in a mixed American population: The Brazilian set. 1. Eur J Hum Genet 24:725-731.).

The other variant found among Afro-descendants, the African variant G6PD*A, which exhibited an unexpected frequency of 10.4%, was found in the population of Salvador, Bahia, with a frequency of 3.1% (Moura-Neto et al., 2008Moura-Neto JP, Dourado MV, Reis MG and Gonçalves MS (2008) A novel c.197T>A variant among Brazilian neonates with glucose-6-phosphate dehydrogenase deficiency. Genet Mol Biol 31:33-35.) and in a sample of Black males from Rio Grande do Sul, with a frequency of 8% (Weimer et al., 1981Weimer TA, Salzano FM and Hutz MH (1981) Erythrocyte isozymes and hemoglobin types in a southern Brazilian population. J Hum Evol 10:319-322.). This finding may be a particular feature of the Afro-descendant population studied here, particularly those from Erepecuru River, reflecting the origin of the African slaves who founded these communities. Alternatively, the absence of the G6PD*A variant in other populations may be attributed to the fact that most studies investigating G6PD mutations are carried out only in subjects with enzyme deficiency, and so the variant would be less likely to be detect since it exhibits normal activity or a very-mild deficiency. Therefore, the search for G6PD variants only in individuals with enzyme deficiency appears to hide a genetic variability revealed only in population-based studies. On the other hand, the absence of the Mediterranean variant among the Afro-descendants studied is an expected result, given the predominantly African origin of the communities, and the absence of evidence of miscegenation with individuals of Italian, Greek, Arab or Jewish origin, in whom this allele is more common. In Brazil, this variant was found in the cities of Campinas (Saad et al., 1997Saad ST, Salles TS, Carvalho MH and Costa FF (1997) Molecular characterization of glucose-6-phosphate dehydrogenase in Brazil. Hum Hered 4:17-21.) and Araraquara (Ferreira et al.. 2014Ferreira PI, Leite F and Leite A (2014) Polimorfismo genético da glicose-6-fosfato desidrogenase na população da região de Araraquara, Estado de São Paulo. Rev Ciênc Farm Básica Apl 35:469–72.), both in the state of São Paulo, southeastern Brazil, with frequencies of 3.0%, and in Caucasian males from Porto Alegre, state of Rio Grande do Sul, with a frequency of 8.0% (Weimer et al., 1981Weimer TA, Salzano FM and Hutz MH (1981) Erythrocyte isozymes and hemoglobin types in a southern Brazilian population. J Hum Evol 10:319-322.). The variant was also found in the population of Manaus with a frequency of 1.0% (Santana et al., 2013Santana MS, Monteiro WM, Siqueira AM, Costa MF, Sampaio V, Lacerda MV and Alecrim MG (2013) Glucose-6-phosphate dehydrogenase deficient variants are associated with reduced susceptibility to malaria in the Brazilian Amazon. Trans R Soc Trop Med Hyg 107:301-306.) attributed to the contribution of Arabs and Jews in the population of the city of Manaus in the state of Amazonas.

Intracontinental comparisons (Latin America) show that the results observed in the “Blacks of the Trombetas” are more similar to those reported by Petit et al. (2016)Petit F, Bailly P, Chiaroni J and Mazières S (2016) Sub-Saharan red cell antigen phenotypes and glucose-6-phosphate dehydrogenase deficiency variants in French Guiana. Malar J 15:310. in people from French Guiana, where the variants G6PD*A- and G6PD*A were found at frequencies of 0.11 and 0.10, respectively. The frequency of G6PD*A- found in the current study (0.06) is similar to the ones found in Colombia and Honduras (0.09), but is higher than that reported for Venezuelans (0.017). The African G6PD*A variant, however, has been found at lower frequencies in these three Latin American countries: 0.02 in Colombia, 0.03 in Honduras, and 0.002 in Venezuela (Valencia et al., 2016Valencia SH, Ocampo ID, Arce-Plata MI, Recht J and Arévalo-Herrera M (2016) Glucose-6-phosphate dehydrogenase deficiency prevalence and genetic variants in malaria endemic areas of Colombia. Malar J 15:291., Zuñiga et al., 2015Zúñiga MA, Mejía RE, Sánchez AL, Sosa-Ochoa WH and Fontecha GA (2015) Glucose-6-phosphate dehydrogenase deficiency among malaria patients of Honduras: A descriptive study of archival blood samples. Malar J 14:308., Vizzi et al., 2016Vizzi E, Bastidas G, Hidalgo M, Colman L and Pérez HA (2016) Prevalence and molecular characterization of G6PD deficiency in two Plasmodium vivax endemic areas in Venezuela: Predominance of the African A-202A/376G variant. Malar J 15:19.).

The results obtained in the present study emphasize the need to use appropriate G6PDd diagnostic methods in the routine care of vivax malaria in the Amazon region, especially in Afro-descendant communities, to avoid hemolysis induced by treatment with primaquine in individuals genetically deficient for G6PD. Primaquine (PQ) is an 8-aminoquinoline considered the most effective drug against the latent hepatic stages of P. vivax, preventing clinical relapses, but is also highly active against gametocytes of all species of human malaria, preventing further transmission of parasites for mosquitoes (WHO, 2015World Health Organization (2015) Guidelines for the treatment of malaria. 3rd edition. World Health Organization, Geneva, 313 p.). Therefore, primaquine is used for both P. vivax and P. falciparum treatment, but the dosage for P. vivax malaria is much higher than the single low dose recommended by WHO (0.25 mg base/kg) to block transmission of P. falciparum malaria, being associated with a considerably lower risk of haemolytic toxicity. The test for the reduction of methemoglobin (Brewer et al., 1962Brewer GJ, Tarlov AR and Alving AS (1962) The methemoglobin reduction test for primaquine -type sensitivity of erythrocytes. JAMA 180:386-388.), due to its high sensitivity and low cost, may also be used for monitoring the presence of G6PDd, although a rapid diagnostic test (RDT) to detect G6PDd in the Brazilian Amazon is already available and is a good cost-effective strategy for diagnosing G6PDd. Based on this monitoring, it is recommended that in patients with a diagnosis of G6PDd, treatment with primaquine should be adjusted for intermittent administration of 0.75 mg/kg weekly for eight weeks under medical supervision in order to reduce the risk of hemolysis (Silva et al., 2004Silva MC, Santos EB, Costal EG, Filho MG, Guerreiro JF and Póvoa MM (2004) Clinical and laboratorial alterations in Plasmodium vivax malaria patients and glucose-6-phosphate dehydrogenase deficiency treated with primaquine at 0.50 mg/kg/day. Rev Soc Bras Med Trop 37:215-217.; Brito et al., 2016Brito MA, Peixoto HM, Almeida AC, Oliveira MR, Romero GA, Moura-Neto JP, Singh N, Monteiro WM and Lacerda MV (2016) Validation of the rapid test Carestart(tm) G6PD among malaria vivax-infected subjects in the Brazilian Amazon. Rev Soc Bras Med Trop 49:446-55.; Peixoto et al., 2016Peixoto HM, Brito MA, Romero GA, Monteiro WM, de Lacerda MV and de Oliveira MR (2016) Cost-effectiveness analysis of rapid diagnostic tests for G6PD deficiency in patients with Plasmodium vivax malaria in the Brazilian Amazon. Malar J 15:82.).

Duffy blood group alleles

The distributions of Duffy blood group genotypes and alleles among the “Blacks of the Trombetas” were also consistent with the predominantly African origin of the population. The observed frequencies of the Duffy-negative genotype (20%) and FY*BES allele (41%) were slightly different from those previously described in Afro-descendant communities from the states of Pará and Amapá (Perna et al., 2007Perna SJQ, Cardoso GL and Guerreiro JF (2007) Duffy blood group genotypes among African-Brazilian communities of the Amazon region. Genet Mol Res 6:166–172.), where frequencies of the Duffy-negative genotype ranged from 32.3% to 58.8%, and frequencies of the FY*BES allele ranged from 56.4% to 72.2%. On the other hand, the frequencies found in Afro-descendants were higher than those reported for Amazonian riverine communities, where frequencies of the Duffy-negative genotype are usually low, varying from 3% to 8%, and the mean frequency of FY*BES is around 5% (Cavasini et al., 2007Cavasini CE, Mattos LC, Couto AA, Bonini-Domingos CR, Valencia SH, Neiras WC, Alves RT, Rossit AR, Castilho L and Machado RL (2007) Duffy blood group gene polymorphisms among malaria vivax patients in four areas of the Brazilian Amazon region. Malar J 6:167.).

Considering that the Duffy-negative blood group is a well-documented protection factor against P. vivax malaria and that there is also evidence that G6PDd protects against P. vivax (Louicharoen et al., 2009Louicharoen C, Patin E, Paul R, Nuchprayoon I, Witoonpanich B, Peerapit-tayamongkol C, Casademont I, Sura T, Laird NM, Singhasivanon P et al. (2009) Positively selected G6PD-Mahidol mutation reduces Plasmodium vivax density in Southeast Asians. Science 326:1546–1549.; Leslie et al., 2010Leslie T, Briceño M, Mayan I, Mohammed N, Klinkenberg E, Sibley CH, Whitty CJM and Rowland M (2010) The impact of phenotypic and genotypic G6PD deficiency on risk of Plasmodium vivax infection: A case–control study amongst Afghan refugees in Pakistan. PLoS Med 7:e1000283.; Santana et al., 2013Santana MS, Monteiro WM, Siqueira AM, Costa MF, Sampaio V, Lacerda MV and Alecrim MG (2013) Glucose-6-phosphate dehydrogenase deficient variants are associated with reduced susceptibility to malaria in the Brazilian Amazon. Trans R Soc Trop Med Hyg 107:301-306.), it is possible to consider that the co-occurrence of the G6PD variant and the FY*BES allele in polymorphic frequencies in the “Trombetas Blacks may confer greater protection against malaria in this population. Indeed, 24% of the individuals presented a G6PD variant (1.9% hemizygous, 0.9% homozygous and 5.7% heterozygous), and 61.7% presented the FY*BES allele (41.3% heterozygotes and 20, 4% homozygotes). In addition, an earlier study in these communities identified the HBB*S gene, which codes for hemoglobin S, being the first of the structural variants of hemoglobin associated with malaria protection, with a frequency of 6% (Schneider et al., 1987).

The greater protection against malaria conferred by these erythrocyte polymorphisms deserves to be better investigated and explored. Until the 1990s the region of Trombetas, where quilombolas and riverine people live, was considered endemic for malaria, accounting for most cases of malaria recorded in the municipality of Oriximiná. From the year 2000, however, there was a significant decrease in the number of cases in the region. This was initially due to the implementation of a malaria control project in the Trombetas river region, by Mineração Rio do Norte S.A., in collaboration with the Endemism Sector of the Municipality of Oriximiná, and later with support from the Brazilian Ministry of Health through the National Program for Malaria Control (PNCM) that started in 2003. With these actions, the municipality of Oriximiná, which was considered a medium risk region for malaria, is now considered low risk, based on the annual parasite index (IPA) (Sousa et al., 2015Sousa JR, Santos ACF, Almeida WS, Albarado KVP, Magno LD, Rocha JAM and Pimentel ZMS (2015) Malaria situation in the Lower Amazon Region, Pará State, Brazil, from 2009 to 2013: An epidemiological approach. Rev Pan-Amaz Saude 6:39-47.). However, data from the Brazilian Ministry of Health indicate that the number of malaria cases has increased again in Brazil, after years of decline, particularly in the state of Pará, where the number of cases in 2017 has more than doubled compared to 2016. Most likely, there will also be an increase in the number of malaria cases in the region where Afro-descendants live (Trombetas, Erepecuru and Cuminá rivers), and procedures for prompt diagnosis and treatment of the disease will become necessary again. Thus, monitoring of G6PDd in cases of primaquine use should play an important role in the routine care of Afro-descendant communities to assess the risks of hemolytic crisis in the case of malaria recurrence.

References

  • Beutler E (1996) G6PD: Population genetics and clinical manifestations. Blood Rev 10:45-52.
  • Beutler E, Kuhl W, Vives-Corrons JL and Prchal JY (1989) Molecular heterogeneity of glucose-6-phosphate dehydrogenase A-. Blood 74:2550-2555.
  • Brewer GJ, Tarlov AR and Alving AS (1962) The methemoglobin reduction test for primaquine -type sensitivity of erythrocytes. JAMA 180:386-388.
  • Brito MA, Peixoto HM, Almeida AC, Oliveira MR, Romero GA, Moura-Neto JP, Singh N, Monteiro WM and Lacerda MV (2016) Validation of the rapid test Carestart(tm) G6PD among malaria vivax-infected subjects in the Brazilian Amazon. Rev Soc Bras Med Trop 49:446-55.
  • Carter N, Pamba A, Duparc S and Waitumbi JN (2011) Frequency of glucose-6-phosphate dehydrogenase deficiency in malaria patients from six African countries enrolled in two randomized anti-malarial clinical trials. Malar J 10:241.
  • Castro SM, Weber R, Matte U and Giugliani R (2007) Molecular characterization of glucose-6-phosphate dehydrogenase deficiency in patients from the southern Brazilian city of Porto Alegre, RS. Genet Mol Biol 30:10-13.
  • Cavasini CE, Mattos LC, Couto AA, Bonini-Domingos CR, Valencia SH, Neiras WC, Alves RT, Rossit AR, Castilho L and Machado RL (2007) Duffy blood group gene polymorphisms among malaria vivax patients in four areas of the Brazilian Amazon region. Malar J 6:167.
  • Chaudhuri A, Polyakova J, Zbrzezna V and Pogo AO (1995) The coding sequence of Duffy blood group gene in humans and simians: Restriction fragment length polymorphism, antibody and malarial parasite specificities, and expression in no erythroid tissues in Duffy-negative individuals. Blood 85:615–621.
  • Dombrowski JG, Souza ST, Curry J, Hinton L, Silva NRM, Grignard L, Gonçalves LA, Gomes AR, Epiphanio S, Drakeley C et al. (2017) G6PD deficiency alleles in a malaria-endemic region in the Western Brazilian Amazon. Malar J 16:253-262.
  • Donahue RP, Bias WB, Renwick JH and McKusick VA (1968) Probable assignment of the duffy blood group locus to chromosome in man. Proc Natl Acad Sci USA 61:949-955.
  • Ferreira PI, Leite F and Leite A (2014) Polimorfismo genético da glicose-6-fosfato desidrogenase na população da região de Araraquara, Estado de São Paulo. Rev Ciênc Farm Básica Apl 35:469–72.
  • Gómez-Manzo S, Marcial-Quino J, Vanoye-Carlo A, Serrano-Posada H, Ortega-Cuellar D, González-Valdez A, Castillo-Rodríguez RA, Hernández-Ochoa B, Sierra-Palacios E, Rodríguez-Bustamante E et al. (2016) Glucose-6-phosphate dehydrogenase: Update and analysis of new mutations around the World. Int J Mol Sci 17:2069.
  • WHO Working Group (1989) Glucose-6-phosphate dehydrogenase deficiency. Bull World Health Organ 67:601–611.
  • Hadley TJ and Peiper SC (1997) From malaria to chemokine receptor: The emerging physiologic role of the Duffy blood group antigen. Blood 89:3077-3091.
  • Hirono A and Beutler E (1988) Molecular cloning and nucleotide sequence of cDNA for human glucose-6-phosphate dehydrogenase variant A (-). Proc Natl Acad Sci USA 85:3951-3954.
  • Howes RE, Patil AP, Piel FB, Nyangiri OA, Kabaria CW, Gething PW, Zimmerman PA, Barnadas C, Beall CM, Gebremedhin A et al. (2011) The global distribution of the Duffy blood group. Nat Commun 2:266.
  • Howes RE, Battle KE, Satyagraha AW, Baird JK and Hay SI (2013) G6PD deficiency: Global distribution, genetic variants and primaquine therapy. Adv Parasitol 81:133-201.
  • Iwamoto S, Omi T, Kajii E and Ikemoto S (1995) Genomic organization of the glycoprotein D gene: Duffy blood group Fya/Fyb alloantigen system is associated with a polymorphism at the 44-amino acid residue. Blood 85:622–626.
  • Kasehagen LJ, Mueller I, Kiniboro B, Bockarie MJ, Reeder JC, Kazura JW, Kastens W, McNamara DT, King CH, Whalen CC et al. (2007) Reduced Plasmodium vivax erythrocyte infection in PNG Duffy-negative heterozygotes. PLoS One 2:e336.
  • Klein HS (1986) African slavery in Latin America and the Caribbean. Oxford University Press, New York, 311 p.
  • Leslie T, Briceño M, Mayan I, Mohammed N, Klinkenberg E, Sibley CH, Whitty CJM and Rowland M (2010) The impact of phenotypic and genotypic G6PD deficiency on risk of Plasmodium vivax infection: A case–control study amongst Afghan refugees in Pakistan. PLoS Med 7:e1000283.
  • Livingstone FB (1984) The Duffy blood groups, vivax malaria, and malaria selection in human populations: A review. Hum Biol 56:413–425.
  • Louicharoen C, Patin E, Paul R, Nuchprayoon I, Witoonpanich B, Peerapit-tayamongkol C, Casademont I, Sura T, Laird NM, Singhasivanon P et al. (2009) Positively selected G6PD-Mahidol mutation reduces Plasmodium vivax density in Southeast Asians. Science 326:1546–1549.
  • Mallinson G, Soo KS, Schall TJ, Pisacka M and Anstee DJ (1995) Mutation in the erythrocyte chemokine receptor (Duffy) gene: The molecular basis of the Fya/Fyb antigens and identification of a deletion in the Duffy gene of an apparently healthy individual with the Fy(a-b-) phenotype. Brit J Hematol 90:823–829.
  • Mezzacappa MA, Facchini FP, Pinto AC, Cassone AEL, Souza DS, Bezerra MAC, Albuquerque DM, Saad STO and Costa FF (2010) Clinical and genetic risk factors for moderate hyperbilirubinemia in Brazilian newborn infants. J Perinatol 30:819-826.
  • Millimono TS, Loua KM, Rath SL, Relvas L, Bento C, Diakite M, Jarvis M, Daries N, Ribeiro LM, Manco L and Kaeda JS (2012) High prevalence of hemoglobin disorders and glucose-6-phosphate dehydrogenase (G6PD) deficiency in the Republic of Guinea (West Africa). Hemoglobin 36:25-37.
  • Miller LH, Mason SJ, Clyde DF and McGinniss MH (1976) The resistance factor to Plasmodium vivax in blacks: The Duffy-blood-group genotype, FyFy. N Engl J Med 295:302-304.
  • Monteiro WM, Val FF, Siqueira AM, Franca GP, Sampaio VS, Melo GC, Almeida AC, Brito MA, Peixoto HM, Fuller D et al. (2014) G6PD deficiency in Latin America: Systematic review on prevalence and variants. Mem Inst Oswaldo Cruz 109:553-68.
  • Moura-Neto JP, Dourado MV, Reis MG and Gonçalves MS (2008) A novel c.197T>A variant among Brazilian neonates with glucose-6-phosphate dehydrogenase deficiency. Genet Mol Biol 31:33-35.
  • Oliveira RAG, Oshiro M, Hirata MH, Hirata RD, Ribeiro GS, Medeiros T and Barretto OCDO (2009) A novel point mutation in a class IV glucose-6-phosphate dehydrogenase variant (G6PD São Paulo) and polymorphic G6PD variants in São Paulo State, Brazil. Genet Mol Biol 32:251-254.
  • Olsson ML, Hansson C, Avent ND, Akesson IE, Green CA and Daniels GL (1998) A clinically applicable method for determining the Duffy (Fy) blood group locus using polymerase chain reaction with allele-specific primers. Transfusion 38:168-173.
  • Pai GS, Sprenkle JA, Do TT, Mareni CE and Migeon BR (1980) Localization of loci for hypoxanthine phosphoribosyltransferase and glucose-6-phosphate dehydrogenase and biochemical evidence of nonrandom X chromosome expression from studies of a human X-autosome translocation. Proc Natl Acad Sci USA 77:2810–2813.
  • Peixoto HM, Brito MA, Romero GA, Monteiro WM, de Lacerda MV and de Oliveira MR (2016) Cost-effectiveness analysis of rapid diagnostic tests for G6PD deficiency in patients with Plasmodium vivax malaria in the Brazilian Amazon. Malar J 15:82.
  • Perna SJQ, Cardoso GL and Guerreiro JF (2007) Duffy blood group genotypes among African-Brazilian communities of the Amazon region. Genet Mol Res 6:166–172.
  • Petit F, Bailly P, Chiaroni J and Mazières S (2016) Sub-Saharan red cell antigen phenotypes and glucose-6-phosphate dehydrogenase deficiency variants in French Guiana. Malar J 15:310.
  • Pogo AO and Chaudhuri A (2000) The Duffy protein: A malarial and chemokine receptor. Semin Hematol 37:122–129.
  • Ruwende C, Khoo SC, Snow RW, Yates SNR, Kwiatkowski D, Gupta A, Warn P, Allsopp CEM, Gilbert SC, Peschu N et al. (1995) Natural protection of hemi- and heterozygotes for G6PD deficiency in Africa by resistance to severe malaria. Nature 376:246–249
  • Saad ST, Salles TS, Carvalho MH and Costa FF (1997) Molecular characterization of glucose-6-phosphate dehydrogenase in Brazil. Hum Hered 4:17-21.
  • Santana MS, Monteiro WM, Siqueira AM, Costa MF, Sampaio V, Lacerda MV and Alecrim MG (2013) Glucose-6-phosphate dehydrogenase deficient variants are associated with reduced susceptibility to malaria in the Brazilian Amazon. Trans R Soc Trop Med Hyg 107:301-306.
  • Santos HC, Horimoto AV, Tarazona-Santos E, Rodrigues-Soares F, Barreto ML, Horta BL, Lima-Costa MF, Gouveia MH, Machado M, Silva TM et al. (2016) A minimum set of ancestry informative markers for determining admixture proportions in a mixed American population: The Brazilian set. 1. Eur J Hum Genet 24:725-731.
  • Silva MC, Santos EB, Costal EG, Filho MG, Guerreiro JF and Póvoa MM (2004) Clinical and laboratorial alterations in Plasmodium vivax malaria patients and glucose-6-phosphate dehydrogenase deficiency treated with primaquine at 0.50 mg/kg/day. Rev Soc Bras Med Trop 37:215-217.
  • Sousa JR, Santos ACF, Almeida WS, Albarado KVP, Magno LD, Rocha JAM and Pimentel ZMS (2015) Malaria situation in the Lower Amazon Region, Pará State, Brazil, from 2009 to 2013: An epidemiological approach. Rev Pan-Amaz Saude 6:39-47.
  • Takizawa T, Yoneyama Y, Miwa S and Yoshida A (1987) A single nucleotide base transition is the basis of the common human glucose-6-phosphate dehydrogenase variant A(+). Genomics 1:228-231.
  • Tournamille C, Colin Y, Cartron JP and Kim CLV (1995b) Disruption of a GATA motif in the Duffy gene promoter abolishes erythroid gene expression in Duffy-negative individual. Nat Genet 10:224–228.
  • Tournamille C, Le Van Kim C, Gane P, Cartón PJ and Colin Y (1995a) Molecular basis and PCR- typing of the FY*A/FY*B blood group polymorphism. Hum Genet 95:407-410.
  • Valencia SH, Ocampo ID, Arce-Plata MI, Recht J and Arévalo-Herrera M (2016) Glucose-6-phosphate dehydrogenase deficiency prevalence and genetic variants in malaria endemic areas of Colombia. Malar J 15:291.
  • Vizzi E, Bastidas G, Hidalgo M, Colman L and Pérez HA (2016) Prevalence and molecular characterization of G6PD deficiency in two Plasmodium vivax endemic areas in Venezuela: Predominance of the African A-202A/376G variant. Malar J 15:19.
  • Vulliamy TJ, D’Urso M, Battistuzzi G, Estrada M, Foulkes NS, Martini G, Calabro V, Poggi V, Giordano R, Town M et al. (1988) Diverse point mutations in the human glucose 6-phosphate dehydrogenase gene cause enzyme deficiency and mild or severe hemolytic anemia. Proc Natl Acad Sci USA 85:5171-5175.
  • Weimer TA, Salzano FM and Hutz MH (1981) Erythrocyte isozymes and hemoglobin types in a southern Brazilian population. J Hum Evol 10:319-322.
  • World Health Organization (2015) Guidelines for the treatment of malaria. 3rd edition. World Health Organization, Geneva, 313 p.
  • Yoshida A, Beutler E and Motulsky AG (1971) Human glucose-6-phosphate dehydrogenase variants. Bull World Health Organ 45:243–253.
  • Zimmerman PA, Woolley I, Masinde GL, Miller SM, McNamara DT, Hazlett F, Mgone CS, Alpers MP, Genton B and Kazura JW (1999) Emergence of FY*A (null) in a Plasmodium vivax-endemic region of Papua New Guinea. Proc Natl Acad Sci USA 96:13973–13977.
  • Zúñiga MA, Mejía RE, Sánchez AL, Sosa-Ochoa WH and Fontecha GA (2015) Glucose-6-phosphate dehydrogenase deficiency among malaria patients of Honduras: A descriptive study of archival blood samples. Malar J 14:308.

Internet Resources

  • Associate Editor: Francisco Mauro Salzano

Publication Dates

  • Publication in this collection
    29 Nov 2018
  • Date of issue
    Oct-Dec 2018

History

  • Received
    16 Aug 2017
  • Accepted
    15 Feb 2018
Sociedade Brasileira de Genética Rua Cap. Adelmio Norberto da Silva, 736, 14025-670 Ribeirão Preto SP Brazil, Tel.: (55 16) 3911-4130 / Fax.: (55 16) 3621-3552 - Ribeirão Preto - SP - Brazil
E-mail: editor@gmb.org.br