Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
J Infect Dis ; 227(3): 371-380, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36314635

RESUMEN

BACKGROUND: Evaluating the performance of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) serological assays and clearly articulating the utility of selected antigens, isotypes, and thresholds is crucial to understanding the prevalence of infection within selected communities. METHODS: This cross-sectional study, implemented in 2020, screened PCRconfirmed coronavirus disease 2019 patients (n 86), banked prepandemic and negative samples (n 96), healthcare workers and family members (n 552), and university employees (n 327) for antiSARS-CoV-2 receptor-binding domain, trimeric spike protein, and nucleocapsid protein immunoglobulin (Ig)G and IgA antibodies with a laboratory-developed enzyme-linked immunosorbent assay and tested how antigen, isotype and threshold choices affected the seroprevalence outcomes. The following threshold methods were evaluated: (i) mean 3 standard deviations of the negative controls; (ii) 100 specificity for each antigen-isotype combination; and (iii) the maximal Youden index. RESULTS: We found vastly different seroprevalence estimates depending on selected antigens and isotypes and the applied threshold method, ranging from 0.0 to 85.4. Subsequently, we maximized specificity and reported a seroprevalence, based on more than one antigen, ranging from 9.3 to 25.9. CONCLUSIONS: This study revealed the importance of evaluating serosurvey tools for antigen-, isotype-, and threshold-specific sensitivity and specificity, to interpret qualitative serosurvey outcomes reliably and consistently across studies.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/epidemiología , Estudios Seroepidemiológicos , Estudios Transversales , Proteínas de la Nucleocápside , Ensayo de Inmunoadsorción Enzimática/métodos , Sensibilidad y Especificidad , Inmunoglobulina G , Anticuerpos Antivirales , Glicoproteína de la Espiga del Coronavirus
2.
Int J Mol Sci ; 24(7)2023 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-37047683

RESUMEN

The seroprevalence of Kaposi sarcoma-associated herpesvirus (KSHV) and the incidence of endemic Kaposi sarcoma (KS) overlap with regions of malaria endemicity in sub-Saharan Africa. Multiple studies have shown an increased risk of KSHV seroconversion in children from high malaria compared to low malaria regions; however, the impact of acute episodes of Plasmodium falciparum (P. falciparum) malaria on KSHV's biphasic life cycle and lytic reactivation has not been determined. Here, we examined KSHV serological profiles and viral loads in 134 children with acute malaria and 221 healthy children from high malaria regions in Kisumu, as well as 77 healthy children from low malaria regions in Nandi. We assayed KSHV, Epstein-Barr virus (EBV), and P. falciparum malaria antibody responses in these three by multiplexed Luminex assay. We confirmed that KSHV seroprevalence was significantly associated with malaria endemicity (OR = 1.95, 1.18-3.24 95% CI, p = 0.01) with 71-77% seropositivity in high-malaria (Kisumu) compared to 28% in low-malaria (Nandi) regions. Furthermore, KSHV serological profiles during acute malaria episodes were distinct from age-matched non-malaria-infected children from the same region. Paired IgG levels also varied after malaria treatment, with significantly higher anti-ORF59 at day 0 but elevated ORF38, ORF73, and K8.1 at day 3. Acute malaria episodes is characterized by perturbation of KSHV latency in seropositive children, providing further evidence that malaria endemicity contributes to the observed increase in endemic KS incidence in sub-Saharan Africa.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 8 , Malaria Falciparum , Sarcoma de Kaposi , Niño , Humanos , Estudios Seroepidemiológicos , Herpesvirus Humano 4 , Malaria Falciparum/epidemiología
3.
Virol J ; 19(1): 208, 2022 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-36482473

RESUMEN

BACKGROUND: Endemic Burkitt lymphoma (eBL) is potentiated through the interplay of Epstein Barr virus (EBV) and holoendemic Plasmodium falciparum malaria. To better understand EBV's biology and role in eBL, we characterized genome-wide recombination sites and patterns as a source of genetic diversity in EBV genomes in our well-defined population of eBL cases and controls from Western Kenya. METHODS: EBV genomes representing 54 eBL cases and 32 healthy children from the same geographic region in Western Kenya that we previously sequenced were analyzed. Whole-genome multiple sequence alignment, recombination analyses, and phylogenetic inference were made using multiple alignment with fast Fourier transform, recombination detection program 4, and molecular evolutionary genetics analysis. RESULTS: We identified 28 different recombination events and 71 (82.6%) of the 86 EBV genomes analyzed contained evidence of one or more recombinant segments. Associated recombination breakpoints were found to occur in a total of 42 different genes, with only 7 (16.67%) being latent genes. Recombination events were major drivers of clustering within genome-wide phylogenetic trees. The occurrence of recombination segments was comparable between genomes from male and female participants and across age groups. More recombinant segments were found in EBV type 1 genomes (p = 6.4e - 06) and the genomes from the eBLs (p = 0.037). Two recombination events were enriched in the eBLs; event 47 (OR = 4.07, p = 0.038) and event 50 (OR = 14.24, p = 0.012). CONCLUSIONS: EBV genomes have extensive evidence of recombination likely acquired progressively and cumulatively over time. Recombination patterns display a heterogeneous occurrence rate across the genome with enrichment in lytic genes. Overall, recombination appears to be a major evolutionary force impacting EBV diversity and genome structure with evidence of the association of specific recombinants with eBL.


Asunto(s)
Linfoma de Burkitt , Infecciones por Virus de Epstein-Barr , Niño , Humanos , Linfoma de Burkitt/genética , Herpesvirus Humano 4/genética , Filogenia , Kenia/epidemiología
4.
J Virol ; 94(17)2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32581102

RESUMEN

Endemic Burkitt lymphoma (eBL), the most prevalent pediatric cancer in sub-Saharan Africa, is distinguished by its inclusion of Epstein-Barr virus (EBV). In order to better understand the impact of EBV variation in eBL tumorigenesis, we improved viral DNA enrichment methods and generated a total of 98 new EBV genomes from both eBL cases (n = 58) and healthy controls (n = 40) residing in the same geographic region in Kenya. Using our unbiased methods, we found that EBV type 1 was significantly more prevalent in eBL patients (74.5%) than in healthy children (47.5%) (odds ratio = 3.24, 95% confidence interval = 1.36 to 7.71, P = 0.007), as opposed to similar proportions in both groups. Controlling for EBV type, we also performed a genome-wide association study identifying six nonsynonymous variants in the genes EBNA1, EBNA2, BcLF1, and BARF1 that were enriched in eBL patients. In addition, viruses isolated from plasma of eBL patients were identical to their tumor counterparts consistent with circulating viral DNA originating from the tumor. We also detected three intertypic recombinants carrying type 1 EBNA2 and type 2 EBNA3 regions, as well as one novel genome with a 20-kb deletion, resulting in the loss of multiple lytic and virion genes. Comparing EBV types, viral genes displayed differential variation rates as type 1 appeared to be more divergent, while type 2 demonstrated novel substructures. Overall, our findings highlight the complexities of the EBV population structure and provide new insight into viral variation, potentially deepening our understanding of eBL oncogenesis.IMPORTANCE Improved viral enrichment methods conclusively demonstrate EBV type 1 to be more prevalent in eBL patients than in geographically matched healthy controls, which previously underrepresented the prevalence of EBV type 2. Genome-wide association analysis between cases and controls identifies six eBL-associated nonsynonymous variants in EBNA1, EBNA2, BcLF1, and BARF1 genes. Analysis of population structure reveals that EBV type 2 exists as two genomic subgroups and was more commonly found in female than in male eBL patients.


Asunto(s)
Linfoma de Burkitt/genética , Linfoma de Burkitt/virología , Infecciones por Virus de Epstein-Barr/virología , Genoma Viral , Herpesvirus Humano 4/genética , Adolescente , Niño , Preescolar , ADN Viral , Infecciones por Virus de Epstein-Barr/epidemiología , Antígenos Nucleares del Virus de Epstein-Barr/genética , Femenino , Variación Genética , Estudio de Asociación del Genoma Completo , Humanos , Lactante , Kenia/epidemiología , Masculino , Oportunidad Relativa , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Análisis de Secuencia de ADN , Proteínas Virales/genética
5.
Blood ; 134(19): 1598-1607, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31558468

RESUMEN

Burkitt lymphoma (BL) is an aggressive, MYC-driven lymphoma comprising 3 distinct clinical subtypes: sporadic BLs that occur worldwide, endemic BLs that occur predominantly in sub-Saharan Africa, and immunodeficiency-associated BLs that occur primarily in the setting of HIV. In this study, we comprehensively delineated the genomic basis of BL through whole-genome sequencing (WGS) of 101 tumors representing all 3 subtypes of BL to identify 72 driver genes. These data were additionally informed by CRISPR screens in BL cell lines to functionally annotate the role of oncogenic drivers. Nearly every driver gene was found to have both coding and non-coding mutations, highlighting the importance of WGS for identifying driver events. Our data implicate coding and non-coding mutations in IGLL5, BACH2, SIN3A, and DNMT1. Epstein-Barr virus (EBV) infection was associated with higher mutation load, with type 1 EBV showing a higher mutational burden than type 2 EBV. Although sporadic and immunodeficiency-associated BLs had similar genetic profiles, endemic BLs manifested more frequent mutations in BCL7A and BCL6 and fewer genetic alterations in DNMT1, SNTB2, and CTCF. Silencing mutations in ID3 were a common feature of all 3 subtypes of BL. In vitro, mass spectrometry-based proteomics demonstrated that the ID3 protein binds primarily to TCF3 and TCF4. In vivo knockout of ID3 potentiated the effects of MYC, leading to rapid tumorigenesis and tumor phenotypes consistent with those observed in the human disease.


Asunto(s)
Linfoma de Burkitt/genética , Secuenciación Completa del Genoma/métodos , Animales , Humanos , Ratones
6.
J Infect Dis ; 222(1): 111-120, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32072172

RESUMEN

BACKGROUND: Endemic Burkitt lymphoma (eBL) is associated with Epstein-Barr virus (EBV) and Plasmodium falciparum malaria coinfections. However, the role of Kaposi sarcoma-associated herpesvirus (KSHV), also endemic in Africa, has not been evaluated as a cofactor in eBL pathogenesis. METHODS: Multiplexed seroprofiles for EBV, malaria, and KSHV were generated for 266 eBL patients, 78 non-eBL cancers, and 202 healthy children. KSHV and EBV loads were quantified by PCR. RESULTS: KSHV seroprevalence did not differ by study group but was associated with age. Seropositivity, defined by K8.1/LANA or in combination with 5 other KSHV antigens (ORF59, ORF65, ORF61, ORF38, and K5) was associated with antimalarial antibody levels to AMA1 (odds ratio [OR], 2.41, P < .001; OR, 2.07, P < .001) and MSP1 (OR, 2.41, P = .0006; OR, 5.78, P < .001), respectively. KSHV loads did not correlate with antibody levels nor differ across groups but were significantly lower in children with detectable EBV viremia (P = .014). CONCLUSIONS: Although KSHV-EBV dual infection does not increase eBL risk, EBV appears to suppress reactivation of KSHV while malaria exposure is associated with KSHV infection and/or reactivation. Both EBV and malaria should, therefore, be considered as potential effect modifiers for KSHV-associated cancers in sub-Saharan Africa.


Asunto(s)
Linfoma de Burkitt/etiología , Linfoma de Burkitt/genética , Infecciones por Herpesviridae/etiología , Infecciones por Herpesviridae/genética , Herpesviridae/genética , Sarcoma de Kaposi/complicaciones , Sarcoma de Kaposi/genética , Adolescente , Factores de Edad , Linfoma de Burkitt/epidemiología , Linfoma de Burkitt/fisiopatología , Niño , Preescolar , Coinfección , Femenino , Infecciones por Herpesviridae/epidemiología , Infecciones por Herpesviridae/fisiopatología , Humanos , Lactante , Kenia/epidemiología , Masculino , Sarcoma de Kaposi/epidemiología , Sarcoma de Kaposi/fisiopatología , Estudios Seroepidemiológicos
7.
Parasite Immunol ; 41(8): e12628, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30972776

RESUMEN

The past decade has witnessed dramatic decreases in malaria-associated mortality and morbidity around the world. This progress has largely been due to intensified malaria control measures, implementation of rapid diagnostics and establishing a network to anticipate and mitigate antimalarial drug resistance. However, the ultimate tool for malaria prevention is the development and implementation of an effective vaccine. To date, malaria vaccine efforts have focused on determining which of the thousands of antigens expressed by Plasmodium falciparum are instrumental targets of protective immunity. The antigenic variation and antigenic polymorphisms arising in parasite genes under immune selection present a daunting challenge for target antigen selection and prioritization, and is a given caveat when interpreting immune recall responses or results from monovalent vaccine trials. Other immune evasion strategies executed by the parasite highlight the myriad of ways in which it can become a recurrent infection. This review provides an update on immune effector mechanisms in malaria and focuses on our improved ability to interrogate the complexity of human immune system, accelerated by recent methodological advances. Appreciating how the human immune landscape influences the effectiveness and longevity of antimalarial immunity will help explain which conditions are necessary for immune effector mechanisms to prevail.


Asunto(s)
Sistema Inmunológico/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Animales , Antimaláricos/uso terapéutico , Humanos , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/inmunología , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Malaria Falciparum/prevención & control , Plasmodium falciparum/genética , Plasmodium falciparum/fisiología , Vacunación
8.
BMC Cancer ; 17(1): 761, 2017 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-29132323

RESUMEN

BACKGROUND: Burkitt lymphoma (BL) is characterized by overexpression of the c-myc oncogene, which in the vast majority of cases is a consequence of an IGH/MYC translocation. While myc is the seminal event, BL is a complex amalgam of genetic and epigenetic changes causing dysregulation of both coding and non-coding transcripts. Emerging evidence suggest that abnormal modulation of mRNA transcription via miRNAs might be a significant factor in lymphomagenesis. However, the alterations in these miRNAs and their correlations to their putative mRNA targets have not been extensively studied relative to normal germinal center (GC) B cells. METHODS: Using more sensitive and specific transcriptome deep sequencing, we compared previously published small miRNA and long mRNA of a set of GC B cells and eBL tumors. MiRWalk2.0 was used to identify the validated target genes for the deregulated miRNAs, which would be important for understanding the regulatory networks associated with eBL development. RESULTS: We found 211 differentially expressed (DE) genes (79 upregulated and 132 downregulated) and 49 DE miRNAs (22 up-regulated and 27 down-regulated). Gene Set enrichment analysis identified the enrichment of a set of MYC regulated genes. Network propagation-based method and correlated miRNA-mRNA expression analysis identified dysregulated miRNAs, including miR-17~95 cluster members and their target genes, which have diverse oncogenic properties to be critical to eBL lymphomagenesis. Central to all these findings, we observed the downregulation of ATM and NLK genes, which represent important regulators in response to DNA damage in eBL tumor cells. These tumor suppressors were targeted by multiple upregulated miRNAs (miR-19b-3p, miR-26a-5p, miR-30b-5p, miR-92a-5p and miR-27b-3p) which could account for their aberrant expression in eBL. CONCLUSION: Combined loss of p53 induction and function due to miRNA-mediated regulation of ATM and NLK, together with the upregulation of TFAP4, may be a central role for human miRNAs in eBL oncogenesis. This facilitates survival of eBL tumor cells with the IGH/MYC chromosomal translocation and promotes MYC-induced cell cycle progression, initiating eBL lymphomagenesis. This characterization of miRNA-mRNA interactions in eBL relative to GC B cells provides new insights on miRNA-mediated transcript regulation in eBL, which are potentially useful for new improved therapeutic strategies.


Asunto(s)
Linfoma de Burkitt/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , ARN Mensajero/genética , Linfoma de Burkitt/diagnóstico , Linfoma de Burkitt/epidemiología , Linfoma de Burkitt/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Niño , Preescolar , Biología Computacional/métodos , Femenino , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Modelos Biológicos , Interferencia de ARN , Transducción de Señal
9.
J Infect Dis ; 213(6): 985-91, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26531246

RESUMEN

Endemic Burkitt lymphoma is associated with Epstein-Barr virus (EBV) and Plasmodium falciparum coinfection, although how P. falciparum exposure affects the dynamics of EBV infection is unclear. We have used a modeling approach to study EBV infection kinetics in a longitudinal cohort of children living in regions of high and low malaria transmission in Kenya. Residence in an area of high malaria transmission was associated with a higher rate of EBV expansion during primary EBV infection in infants and during subsequent episodes of EBV DNA detection, as well as with longer episodes of EBV DNA detection and shorter intervals between subsequent episodes of EBV DNA detection. In addition, we found that concurrent P. falciparum parasitemia also increases the likelihood of the first and subsequent peaks of EBV in peripheral blood. This suggests that P. falciparum infection is associated with increased EBV growth and contributes to endemic Burkitt lymphoma pathogenesis.


Asunto(s)
Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/fisiología , Malaria Falciparum/complicaciones , Plasmodium falciparum , Replicación Viral/fisiología , Área Bajo la Curva , Infecciones por Virus de Epstein-Barr/epidemiología , Humanos , Lactante , Recién Nacido , Kenia/epidemiología , Malaria Falciparum/epidemiología , Modelos de Riesgos Proporcionales , Carga Viral
10.
J Infect Dis ; 214(9): 1390-1398, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27571902

RESUMEN

The combination of Epstein-Barr virus (EBV) infection and high malaria exposure are risk factors for endemic Burkitt lymphoma, and evidence suggests that infants in regions of high malaria exposure have earlier EBV infection and increased EBV reactivation. In this study we analyzed the longitudinal antibody response to EBV in Kenyan infants with different levels of malaria exposure. We found that high malaria exposure was associated with a faster decline of maternally derived immunoglobulin G antibody to both the EBV viral capsid antigen and EBV nuclear antigen, followed by a more rapid rise in antibody response to EBV antigens in children from the high-malaria-transmission region. We also observed the long-term persistence of anti-viral capsid antigen immunoglobulin M responses in children from the high-malaria region. More rapid decay of maternal antibodies was a major predictor of EBV infection outcome, because decay predicted time to EBV DNA detection, independent of high or low malaria exposure.


Asunto(s)
Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , Infecciones por Virus de Epstein-Barr/etiología , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/inmunología , Malaria/complicaciones , Malaria/inmunología , Antígenos Virales/inmunología , Linfoma de Burkitt/etiología , Linfoma de Burkitt/inmunología , Proteínas de la Cápside/inmunología , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Lactante , Recién Nacido , Kenia , Carga Viral/inmunología
11.
Int J Cancer ; 139(6): 1231-40, 2016 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-27136063

RESUMEN

Discovering how to improve survival and establishing clinical reference points for children diagnosed with endemic Burkitt lymphoma (eBL) in resource-constrained settings has recaptured international attention. Using multivariate analyses, we evaluated 428 children with eBL in Kenya for age, gender, tumor stage, nutritional status, hemoglobin, lactate dehydrogenase (LDH), Epstein-Barr virus (EBV) and Plasmodium falciparum prior to induction of chemotherapy (cyclophosphamide, vincristine, methotrexate and doxorubicin) to identify predictive and prognostic biomarkers of survival. During this 10 year prospective study period, 22% died in-hospital and 78% completed six-courses of chemotherapy. Of those, 16% relapsed or died later; 31% achieved event-free-survival; and 31% were lost to follow-up; the overall one-year survival was 45%. After adjusting for covariates, low hemoglobin (<8 g/dL) and high LDH (>400 mU/ml) were associated with increased risk of death (adjusted Hazard Ratio (aHR) = 1.57 [0.97-2.41]) and aHR = 1.84, [0.91-3.69], respectively). Anemic children with malaria were 3.55 times more likely to die [1.10-11.44] compared to patients without anemia or malarial infection. EBV load did not differ by tumor stage nor was it associated with survival. System-level factors can also contribute to poor outcomes. Children were more likely to die when inadvertently overdosed by more than 115% of the correct dose of cyclophosphamide (a HR = 1.43 [0.84-2.43]) or doxorubicin (a HR = 1.25, [0.66-2.35]), compared with those receiving accurate doses of the respective agent in this setting. This study codifies risk factors associated with poor outcomes for eBL patients in Africa and provides a benchmark by which to assess improvements in survival for new chemotherapeutic approaches.


Asunto(s)
Linfoma de Burkitt/epidemiología , Tasa de Supervivencia , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biopsia , Linfoma de Burkitt/diagnóstico , Linfoma de Burkitt/historia , Linfoma de Burkitt/mortalidad , Niño , Preescolar , Estudios de Cohortes , Femenino , Historia del Siglo XXI , Mortalidad Hospitalaria , Humanos , Lactante , Estimación de Kaplan-Meier , Kenia/epidemiología , Masculino , Estadificación de Neoplasias , Vigilancia de la Población , Factores de Riesgo
12.
Curr Top Microbiol Immunol ; 390(Pt 1): 267-85, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26424650

RESUMEN

Endemic Burkitt's lymphoma (BL) remains the most prevalent pediatric cancer in sub-Saharan Africa even though it was the first human cancer with a viral etiology described over 50 years ago. Epstein-Barr virus (EBV) was discovered in a BL tumor in 1964 and has since been implicated in other malignancies. The etiology of endemic BL has been linked to EBV and Plasmodium falciparum malaria co-infection. While epidemiologic studies have yielded insight into EBV infection and the etiology of endemic BL, the modulation of viral persistence in children by malaria and deficits in EBV immunosurveillance has more recently been reified. Renewed efforts to design prophylactic and therapeutic EBV vaccines provide hope of preventing EBV-associated BL as well as increasing the ability to cure this cancer.


Asunto(s)
Linfoma de Burkitt/virología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/fisiología , Animales , Linfoma de Burkitt/historia , Linfoma de Burkitt/inmunología , Infecciones por Virus de Epstein-Barr/historia , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/genética , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Monitorización Inmunológica
13.
J Infect Dis ; 212(9): 1429-38, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25883384

RESUMEN

BACKGROUND: Immunoglobulin G antibodies (Abs) to Plasmodium falciparum antigens have been associated with naturally acquired immunity to symptomatic malaria. METHODS: We probed protein microarrays covering 824 unique P. falciparum protein features with plasma from residents of a community in Kenya monitored for 12 weeks for (re)infection and symptomatic malaria after administration of antimalarial drugs. P. falciparum proteins recognized by Abs from 88 children (aged 1-14 years) and 86 adults (aged ≥ 18 years), measured at the beginning of the observation period, were ranked by Ab signal intensity. RESULTS: Abs from immune adults reacted with a total 163 of 824 P. falciparum proteins. Children gradually acquired Abs to the full repertoire of antigens recognized by adults. Abs to some antigens showed high seroconversion rates, reaching maximal levels early in childhood, whereas others did not reach adult levels until adolescence. No correlation between Ab signal intensity and time to (re)infection was observed. In contrast, Ab levels to 106 antigens were significantly higher in children who were protected from symptomatic malaria compared with those who were not. Abs to antigens predictive of protection included P. falciparum erythrocyte membrane protein 1, merozoite surface protein (MSP) 10, MSP2, liver-stage antigen 3, PF70, MSP7, and Plasmodium helical interspersed subtelomeric domain protein. CONCLUSIONS: Protein microarrays may be useful in the search for malaria antigens associated with protective immunity.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Malaria/inmunología , Plasmodium falciparum/inmunología , Análisis por Matrices de Proteínas , Adolescente , Adulto , Anciano , Animales , Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/sangre , Antimaláricos/uso terapéutico , Niño , Preescolar , Femenino , Humanos , Inmunidad Innata , Inmunoglobulina G/sangre , Lactante , Kenia , Malaria/sangre , Malaria/tratamiento farmacológico , Proteínas de la Membrana/inmunología , Merozoítos/inmunología , Ratones , Persona de Mediana Edad , Plasmodium falciparum/aislamiento & purificación , Modelos de Riesgos Proporcionales , Proteínas Protozoarias/sangre , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Adulto Joven
14.
J Infect Dis ; 211(5): 680-8, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25180240

RESUMEN

Plasmodium falciparum parasites that are resistant to artemisinins have been detected in Southeast Asia. Resistance is associated with several polymorphisms in the parasite's K13-propeller gene. The molecular epidemiology of these artemisinin resistance genotypes in African parasite populations is unknown. We developed an assay to quantify rare polymorphisms in parasite populations that uses a pooled deep-sequencing approach to score allele frequencies, validated it by evaluating mixtures of laboratory parasite strains, and then used it to screen P. falciparum parasites from >1100 African infections collected since 2002 from 14 sites across sub-Saharan Africa. We found no mutations in African parasite populations that are associated with artemisinin resistance in Southeast Asian parasites. However, we observed 15 coding mutations, including 12 novel mutations, and limited allele sharing between parasite populations, consistent with a large reservoir of naturally occurring K13-propeller variation. Although polymorphisms associated with artemisinin resistance in P. falciparum in Southeast Asia are not prevalent in sub-Saharan Africa, numerous K13-propeller coding polymorphisms circulate in Africa. Although their distributions do not support a widespread selective sweep for an artemisinin-resistant phenotype, the impact of these mutations on artemisinin susceptibility is unknown and will require further characterization. Rapid, scalable molecular surveillance offers a useful adjunct in tracking and containing artemisinin resistance.


Asunto(s)
Antimaláricos/farmacología , Artemisininas/farmacología , Malaria Falciparum/parasitología , Mutación , Plasmodium falciparum/efectos de los fármacos , Adulto , África del Sur del Sahara/epidemiología , Niño , Preescolar , Femenino , Frecuencia de los Genes , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Epidemiología Molecular , Plasmodium falciparum/aislamiento & purificación , Polimorfismo Genético , Embarazo , Prevalencia
15.
BMC Med ; 13: 19, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25633459

RESUMEN

BACKGROUND: The identification of protective immune responses to P. falciparum infection is an important goal for the development of a vaccine for malaria. This requires the identification of susceptible and resistant individuals, so that their immune responses may be studied. Time-to-infection studies are one method for identifying putative susceptible individuals (infected early) versus resistant individuals (infected late). However, the timing of infection is dependent on random factors, such as whether the subject was bitten by an infected mosquito, as well as individual factors, such as their level of immunity. It is important to understand how much of the observed variation in infection is simply due to chance. METHODS: We analyse previously published data from a treatment-time-to-infection study of 201 individuals aged 0.5 to 78 years living in Western Kenya. We use a mathematical modelling approach to investigate the role of immunity versus random factors in determining time-to-infection in this cohort. We extend this analysis using a modelling approach to understand what factors might increase or decrease the utility of these studies for identifying susceptible and resistant individuals. RESULTS: We find that, under most circumstances, the observed distribution of time-to-infection is consistent with this simply being a random process. We find that age, method for detection of infection (PCR versus microscopy), and underlying force of infection are all factors in determining whether time-to-infection is a useful correlate of immunity. CONCLUSIONS: Many epidemiological studies of P. falciparum infection assume that the observed variation in infection outcomes, such as time-to-infection or presence or absence of infection, is determined by host resistance or susceptibility. However, under most circumstances, this distribution appears largely due to the random timing of infection, particularly in children. More direct measurements, such as parasite growth rate, may be more useful than time-to-infection in segregating patients based on their level of immunity.


Asunto(s)
Malaria Falciparum/inmunología , Modelos Biológicos , Plasmodium falciparum , Adolescente , Factores de Edad , Anciano , Niño , Preescolar , Femenino , Humanos , Lactante , Kenia , Malaria Falciparum/diagnóstico , Masculino , Microscopía , Persona de Mediana Edad , Plasmodium falciparum/genética , Plasmodium falciparum/aislamiento & purificación , Reacción en Cadena de la Polimerasa , Factores de Tiempo , Adulto Joven
16.
J Infect Dis ; 209(7): 1136-43, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24265441

RESUMEN

In malaria holoendemic settings, decreased parasitemia and clinical disease is associated with age and cumulative exposure. The relative contribution of acquired immunity against various stages of the parasite life cycle is not well understood. In particular, it is not known whether changes in infection dynamics can be best explained by decreasing rates of infection, or by decreased growth rates of parasites in blood. Here, we analyze the dynamics of Plasmodium falciparum infection after treatment in a cohort of 197 healthy study participants of different ages. We use both polymerase chain reaction (PCR) and microscopy detection of parasitemia in order to understand parasite growth rates and infection rates over time. The more sensitive PCR assay detects parasites earlier than microscopy, and demonstrates a higher overall prevalence of infection than microscopy alone. The delay between PCR and microscopy detection is significantly longer in adults compared with children, consistent with slower parasite growth with age. We estimated the parasite multiplication rate from delay to PCR and microscopy detections of parasitemia. We find that both the delay between PCR and microscopy infection as well as the differing reinfection dynamics in different age groups are best explained by a slowing of parasite growth with age.


Asunto(s)
Sangre/parasitología , Malaria Falciparum/epidemiología , Malaria Falciparum/inmunología , Parasitemia/epidemiología , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/inmunología , Adolescente , Adulto , Factores de Edad , Animales , Antimaláricos/administración & dosificación , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Lactante , Malaria Falciparum/tratamiento farmacológico , Masculino , Microscopía , Reacción en Cadena de la Polimerasa , Adulto Joven
17.
Int J Cancer ; 134(3): 645-53, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23832374

RESUMEN

Endemic Burkitt lymphoma (eBL) is associated with Epstein-Barr virus (EBV) and Plasmodium falciparum coinfections. Malaria appears to dysregulate immunity that would otherwise control EBV, thereby contributing to eBL etiology. Juxtaposed to human genetic variants associated with protection from malaria, it has been hypothesized that such variants could decrease eBL susceptibility, historically referred to as "the protective hypothesis." Past studies attempting to link sickle cell trait (HbAS), which is known to be protective against malaria, with protection from eBL were contradictory and underpowered. Therefore, using a case-control study design, we examined HbAS frequency in 306 Kenyan children diagnosed with eBL compared to 537 geographically defined and ethnically matched controls. We found 23.8% HbAS for eBL patients, which was not significantly different compared to 27.0% HbAS for controls [odds ratio (OR) = 0.85; 95% confidence interval (CI) 0.61-1.17; p-value = 0.33]. Even though cellular EBV titers, indicative of the number of latently infected B cells, were significantly higher (p-value < 0.0003) in children residing in malaria holoendemic compared to hypoendemic areas, levels were not associated with HbAS genotype. Combined, this suggests that although HbAS protects against severe malaria and hyperparasitemia, it is not associated with viral control or eBL protection. However, based on receiver operating characteristic curves factors that enable the establishment of EBV persistence, in contrast to those involved in EBV lytic reactivation, may have utility as an eBL precursor biomarker. This has implications for future human genetic association studies to consider variants influencing control over EBV in addition to malaria as risk factors for eBL.


Asunto(s)
Biomarcadores/sangre , Linfoma de Burkitt/complicaciones , Etnicidad , Herpesvirus Humano 4/aislamiento & purificación , Malaria/complicaciones , Rasgo Drepanocítico/complicaciones , Adolescente , Secuencia de Bases , Linfoma de Burkitt/epidemiología , Estudios de Casos y Controles , Niño , Preescolar , Cartilla de ADN , Femenino , Genotipo , Humanos , Malaria/epidemiología , Masculino , Reacción en Cadena de la Polimerasa , Rasgo Drepanocítico/genética
18.
BMC Med ; 12: 134, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-25285918

RESUMEN

Determining an immunologic correlate of protection against Plasmodium falciparum malaria has been the holy grail of natural infection studies, and sought after as an endpoint for malaria vaccine trials. An in vitro assay that provides an accurate and precise assessment of protective immunity to malaria would make smaller, short-duration studies feasible, rather than the currently powered study designs that use morbidity or mortality as outcomes. Such a biomarker would be especially desirable in situations where malaria control measures that result in decreases in clinical endpoints and putatively waning protective immunity have been implemented. In an article published in BMC Medicine, Osier and colleagues addressed this problem, and demonstrated that antibodies promoting opsonic phagocytosis of merozoites provide a functional link between antigen-specific responses and protection. Understanding the mechanisms conferring protection against malaria not only improves our knowledge of basic human immunology, but promises to help in the design of an effective malaria vaccine.


Asunto(s)
Anticuerpos Antiprotozoarios/análisis , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Animales , Antígenos de Protozoos/inmunología , Humanos , Plasmodium falciparum/inmunología
19.
J Virol ; 87(3): 1779-88, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23175378

RESUMEN

Coinfection with Plasmodium falciparum malaria and Epstein-Barr virus (EBV) is a major risk factor for endemic Burkitt lymphoma (eBL), still one of the most prevalent pediatric cancers in equatorial Africa. Although malaria infection has been associated with immunosuppression, the precise mechanisms that contribute to EBV-associated lymphomagenesis remain unclear. In this study, we used polychromatic flow cytometry to characterize CD8(+) T-cell subsets specific for EBV-derived lytic (BMFL1 and BRLF1) and latent (LMP1, LMP2, and EBNA3C) antigens in individuals with divergent malaria exposure. No malaria-associated differences in EBV-specific CD8(+) T-cell frequencies were observed. However, based on a multidimensional analysis of CD45RO, CD27, CCR7, CD127, CD57, and PD-1 expression, we found that individuals living in regions with intense and perennial (holoendemic) malaria transmission harbored more differentiated EBV-specific CD8(+) T-cell populations that contained fewer central memory cells than individuals living in regions with little or no (hypoendemic) malaria. This profile shift was most marked for EBV-specific CD8(+) T-cell populations that targeted latent antigens. Importantly, malaria exposure did not skew the phenotypic properties of either cytomegalovirus (CMV)-specific CD8(+) T cells or the global CD8(+) memory T-cell pool. These observations define a malaria-associated aberration localized to the EBV-specific CD8(+) T-cell compartment that illuminates the etiology of eBL.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Coinfección/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/patogenicidad , Malaria Falciparum/epidemiología , Malaria Falciparum/inmunología , Plasmodium falciparum/patogenicidad , África/epidemiología , Niño , Preescolar , Infecciones por Virus de Epstein-Barr/complicaciones , Citometría de Flujo , Humanos , Lactante , Malaria Falciparum/complicaciones , Subgrupos de Linfocitos T/inmunología
20.
J Infect Dis ; 208(1): 149-58, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23539744

RESUMEN

BACKGROUND: Acquired immunity to malaria develops with increasing age and repeated infections. Understanding immune correlates of protection from malaria would facilitate vaccine development and identification of biomarkers that reflect changes in susceptibility resulting from ongoing malaria control efforts. METHODS: The relationship between immunoglobulin G (IgG) antibody and both interferon γ (IFN-γ) and interleukin 10 (IL-10) responses to the 42-kD C-terminal fragment of Plasmodium falciparum merozoite surface protein 1 (MSP142) and the risk of (re)infection were examined following drug-mediated clearance of parasitemia in 94 adults and 95 children in an area of holoendemicity of western Kenya. RESULTS: Positive IFN-γ enzyme-linked immunosorbent assay (ELISA) and enzyme-linked immunosorbent spot assay (ELISPOT) responses to MSP142 3D7 were associated with delayed time to (re)infection, whereas high-titer IgG antibodies to MSP142 3D7 or FVO alleles were not independently predictive of the risk of (re)infection. When IFN-γ and IL-10 responses were both present, the protective effect of IFN-γ was abrogated. A Cox proportional hazard model including IFN-γ, IL-10, MSP142 3D7 IgG antibody responses, hemoglobin S genotype, age, and infection status at baseline showed that the time to blood-stage infection correlated positively with IFN-γ responses and negatively with IL-10 responses, younger age, and asymptomatic parasitemia. CONCLUSIONS: Evaluating combined allele-specific cellular and humoral immunity elicited by malaria provides a more informative measure of protection relative to evaluation of either measure alone.


Asunto(s)
Malaria Falciparum/inmunología , Proteína 1 de Superficie de Merozoito/inmunología , Plasmodium falciparum/inmunología , Adolescente , Adulto , Anciano , Niño , Preescolar , Ensayo de Inmunoadsorción Enzimática , Femenino , Genotipo , Hemoglobina Falciforme/genética , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Inmunoglobulina G/inmunología , Lactante , Interferón gamma/fisiología , Interleucina-10/fisiología , Kenia , Malaria Falciparum/prevención & control , Masculino , Persona de Mediana Edad , Parasitemia/inmunología , Parasitemia/parasitología , Adulto Joven
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda