1
|
Shahin T, Jurkovic J, Dieng MM, Manikandan V, Abdrabou W, Alamad B, Bayaraa O, Diawara A, Sermé SS, Henry NB, Sombie S, Almojil D, Arnoux M, Drou N, Soulama I, Idaghdour Y. Single-cell transcriptomics reveals inter-ethnic variation in immune response to Falciparum malaria. Am J Hum Genet 2025; 112:709-723. [PMID: 39970911 PMCID: PMC11947175 DOI: 10.1016/j.ajhg.2025.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/21/2025] Open
Abstract
Africa's environmental, cultural, and genetic diversity can profoundly shape population responses to infectious diseases, including malaria caused by Plasmodium falciparum. Differences in malaria susceptibility among populations are documented, but the underlying mechanisms remain poorly understood. Notably, the Fulani ethnic group in Africa is less susceptible to malaria compared to other sympatric groups, such as the Mossi. They exhibit lower disease rates and parasite load as well as enhanced serological protection. However, elucidating the molecular and cellular basis of this protection has been challenging in part due to limited immunological characterization at the cellular level. To address this question, we performed single-cell transcriptomic profiling of peripheral blood mononuclear cells from 126 infected and non-infected Fulani and Mossi children in rural Burkina Faso. This analysis generated over 70,000 single-cell transcriptomes and identified 30 distinct cell subtypes. We report a profound effect of ethnicity on the transcriptional landscape, particularly within monocyte populations. Differential expression analysis across cell subtypes revealed ethnic-specific immune signatures under both infected and non-infected states. Specifically, monocytes and T cell subtypes of the Fulani exhibited reduced pro-inflammatory responses, while their B cell subtypes displayed stronger activation and inflammatory profiles. Furthermore, single-cell expression quantitative trait locus (eQTL) analysis in monocytes of infected children revealed several significant regulatory variants with ethnicity-specific effects on immune-related genes, including CD36 and MT2A. Overall, we identify ethnic, cell-type-specific, and genetic regulatory effects on host immune responses to malaria and provide valuable single-cell eQTL and transcriptomic datasets from under-represented populations.
Collapse
Affiliation(s)
- Tala Shahin
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Jakub Jurkovic
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Mame Massar Dieng
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Vinu Manikandan
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Wael Abdrabou
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Bana Alamad
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Odmaa Bayaraa
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Aïssatou Diawara
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Samuel Sindié Sermé
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Noëlie Béré Henry
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Salif Sombie
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Dareen Almojil
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Marc Arnoux
- Core Technology Platforms, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Nizar Drou
- Center for Genomics and Systems Biology, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Issiaka Soulama
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso; Institut de Recherche en Sciences de la Santé (IRSS)/CNRST, 03 BP 7192, Ouagadougou, Burkina Faso
| | - Youssef Idaghdour
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates; Center for Genomics and Systems Biology, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
2
|
Alemu R, Sharew NT, Arsano YY, Ahmed M, Tekola-Ayele F, Mersha TB, Amare AT. Multi-omics approaches for understanding gene-environment interactions in noncommunicable diseases: techniques, translation, and equity issues. Hum Genomics 2025; 19:8. [PMID: 39891174 PMCID: PMC11786457 DOI: 10.1186/s40246-025-00718-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/16/2025] [Indexed: 02/03/2025] Open
Abstract
Non-communicable diseases (NCDs) such as cardiovascular diseases, chronic respiratory diseases, cancers, diabetes, and mental health disorders pose a significant global health challenge, accounting for the majority of fatalities and disability-adjusted life years worldwide. These diseases arise from the complex interactions between genetic, behavioral, and environmental factors, necessitating a thorough understanding of these dynamics to identify effective diagnostic strategies and interventions. Although recent advances in multi-omics technologies have greatly enhanced our ability to explore these interactions, several challenges remain. These challenges include the inherent complexity and heterogeneity of multi-omic datasets, limitations in analytical approaches, and severe underrepresentation of non-European genetic ancestries in most omics datasets, which restricts the generalizability of findings and exacerbates health disparities. This scoping review evaluates the global landscape of multi-omics data related to NCDs from 2000 to 2024, focusing on recent advancements in multi-omics data integration, translational applications, and equity considerations. We highlight the need for standardized protocols, harmonized data-sharing policies, and advanced approaches such as artificial intelligence/machine learning to integrate multi-omics data and study gene-environment interactions. We also explore challenges and opportunities in translating insights from gene-environment (GxE) research into precision medicine strategies. We underscore the potential of global multi-omics research in advancing our understanding of NCDs and enhancing patient outcomes across diverse and underserved populations, emphasizing the need for equity and fairness-centered research and strategic investments to build local capacities in underrepresented populations and regions.
Collapse
Affiliation(s)
- Robel Alemu
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Anderson School of Management, University of California Los Angeles, Los Angeles, CA, USA.
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| | - Nigussie T Sharew
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Yodit Y Arsano
- Alpert Medical School, Lifespan Health Systems, Brown University, WarrenProvidence, Rhode Island, USA
| | - Muktar Ahmed
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Tesfaye B Mersha
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Azmeraw T Amare
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
3
|
Sharma JR, Fokkens H, Laubscher R, Apalata TR, Nomatshila SC, Alomatu SY, Strijdom H, Johnson R. No Association Between AGT Gene Polymorphisms with Hypertension in a South African Population. Diabetes Metab Syndr Obes 2024; 17:1853-1865. [PMID: 38706806 PMCID: PMC11069114 DOI: 10.2147/dmso.s452272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/16/2024] [Indexed: 05/07/2024] Open
Abstract
Purpose Hypertension is a leading cause of cardiovascular-related morbidity and mortality worldwide, with a prevalence increasing at an alarming rate in both middle- and low-income countries. Various environmental and genetic factors have been attributed to play a significant role in the increasing prevalence of hypertension. Single nucleotide polymorphisms (SNPs) in the angiotensinogen (AGT) gene are reported to have a significant association with hypertension; however, there are limited studies done on South African populations. Therefore, this case-control study aimed to investigate the association between AGT SNPs (rs2004776, rs3789678, rs5051 and rs7079) with hypertension in a study population of isiXhosa-speaking participants from the Eastern Cape Province in South Africa. Materials and Methods The SNPs were genotyped in 250 hypertensive cases and 237 normotensive controls, using TaqMan genotyping assays. Results For the SNP rs2004776, the frequency of CC genotype (18.4%) and C allele (44%) in hypertensive cases showed no significant differences (p = 0.52, χ2 = 1.32), when compared to the normotensive control group (CC: 19.8% and C allele: 43%). Similar results were obtained for the genotypic and allelic frequencies between hypertensive cases and normotensive controls for rs3789678 (p = 0.88, χ2=0.26) and rs5051 (p = 0.57, χ2=1.12), and rs7079 (p = 0.33, χ2=2.23). These findings demonstrate that there were no significant associations between the SNPs rs2004776, rs3789678, rs7079, rs5051 with hypertension in our study population. Conclusion These findings suggest that AGT gene polymorphisms are not associated with the development of hypertension in the studied population. The present study represents the first genetic report to investigate the AGT gene polymorphisms with hypertension in an isiXhosa-speaking South African population.
Collapse
Affiliation(s)
- Jyoti Rajan Sharma
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
| | - Hannah Fokkens
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
- Centre for Cardiometabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Ria Laubscher
- Biostatistics Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Teke Ruffin Apalata
- Department of Laboratory-Medicine and Pathology, Faculty of Health Sciences, Walter Sisulu University and National Health Laboratory Services, Mthatha, South Africa
| | - Sibusiso Cyprian Nomatshila
- Department of Public Health, Faculty of Medicine and Health Sciences, Walter Sisulu University, Mthatha, South Africa
| | - Samuel Yao Alomatu
- Department of Internal Medicine, Nelson Mandela Central Hospital and Walter Sisulu University, Mthatha, South Africa
| | - Hans Strijdom
- Centre for Cardiometabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
- Centre for Cardiometabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
4
|
Yeyeodu S, Hanafi D, Webb K, Laurie NA, Kimbro KS. Population-enriched innate immune variants may identify candidate gene targets at the intersection of cancer and cardio-metabolic disease. Front Endocrinol (Lausanne) 2024; 14:1286979. [PMID: 38577257 PMCID: PMC10991756 DOI: 10.3389/fendo.2023.1286979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/07/2023] [Indexed: 04/06/2024] Open
Abstract
Both cancer and cardio-metabolic disease disparities exist among specific populations in the US. For example, African Americans experience the highest rates of breast and prostate cancer mortality and the highest incidence of obesity. Native and Hispanic Americans experience the highest rates of liver cancer mortality. At the same time, Pacific Islanders have the highest death rate attributed to type 2 diabetes (T2D), and Asian Americans experience the highest incidence of non-alcoholic fatty liver disease (NAFLD) and cancers induced by infectious agents. Notably, the pathologic progression of both cancer and cardio-metabolic diseases involves innate immunity and mechanisms of inflammation. Innate immunity in individuals is established through genetic inheritance and external stimuli to respond to environmental threats and stresses such as pathogen exposure. Further, individual genomes contain characteristic genetic markers associated with one or more geographic ancestries (ethnic groups), including protective innate immune genetic programming optimized for survival in their corresponding ancestral environment(s). This perspective explores evidence related to our working hypothesis that genetic variations in innate immune genes, particularly those that are commonly found but unevenly distributed between populations, are associated with disparities between populations in both cancer and cardio-metabolic diseases. Identifying conventional and unconventional innate immune genes that fit this profile may provide critical insights into the underlying mechanisms that connect these two families of complex diseases and offer novel targets for precision-based treatment of cancer and/or cardio-metabolic disease.
Collapse
Affiliation(s)
- Susan Yeyeodu
- Julius L Chambers Biomedical/Biotechnology Institute (JLC-BBRI), North Carolina Central University, Durham, NC, United States
- Charles River Discovery Services, Morrisville, NC, United States
| | - Donia Hanafi
- Julius L Chambers Biomedical/Biotechnology Institute (JLC-BBRI), North Carolina Central University, Durham, NC, United States
| | - Kenisha Webb
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Nikia A. Laurie
- Julius L Chambers Biomedical/Biotechnology Institute (JLC-BBRI), North Carolina Central University, Durham, NC, United States
| | - K. Sean Kimbro
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
5
|
Ankunda V, Katende JS, Oluka GK, Sembera J, Baine C, Odoch G, Ejou P, Kato L, Kaleebu P, Serwanga J. The subdued post-boost spike-directed secondary IgG antibody response in Ugandan recipients of the Pfizer-BioNTech BNT162b2 vaccine has implications for local vaccination policies. Front Immunol 2024; 15:1325387. [PMID: 38469296 PMCID: PMC10926532 DOI: 10.3389/fimmu.2024.1325387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/31/2024] [Indexed: 03/13/2024] Open
Abstract
Introduction This study aimed to delineate longitudinal antibody responses to the Pfizer-BioNTech BNT162b2 COVID-19 vaccine within the Ugandan subset of the Sub-Saharan African (SSA) demographic, filling a significant gap in global datasets. Methods We enrolled 48 participants and collected 320 specimens over 12 months after the primary vaccination dose. A validated enzyme-linked immunosorbent assay (ELISA) was used to quantify SARS-CoV-2-specific IgG, IgM, and IgA antibody concentrations (ng/ml) and optical densities (ODs). Statistical analyses included box plots, diverging bar graphs, and the Wilcoxon test with Bonferroni correction. Results We noted a robust S-IgG response within 14 days of the primary vaccine dose, which was consistent with global data. There was no significant surge in S-IgG levels after the booster dose, contrasting trends in other global populations. The S-IgM response was transient and predominantly below established thresholds for this population, which reflects its typical early emergence and rapid decline. S-IgA levels rose after the initial dose then decreased after six months, aligning with the temporal patterns of mucosal immunity. Eleven breakthrough infections were noted, and all were asymptomatic, regardless of the participants' initial S-IgG serostatus, which suggests a protective effect from vaccination. Discussion The Pfizer-BioNTech BNT162b2 COVID-19 vaccine elicited strong S-IgG responses in the SSA demographic. The antibody dynamics distinctly differed from global data highlighting the significance of region-specific research and the necessity for customised vaccination strategies.
Collapse
Affiliation(s)
- Violet Ankunda
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Joseph Ssebwana Katende
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Gerald Kevin Oluka
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Jackson Sembera
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Claire Baine
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Geoffrey Odoch
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Peter Ejou
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Laban Kato
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Pontiano Kaleebu
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Jennifer Serwanga
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| |
Collapse
|
6
|
Scigliuolo GM, Boukouaci W, Cappelli B, Volt F, Rivera Franco MM, Dhédin N, de Latour RP, Devalck C, Dalle J, Castelle M, Hermine O, Chardin MO, Poiré X, Brichard B, Paillard C, Rafii H, Kenzey C, Wu C, Bouassida J, Robin M, Raus N, Rocha V, Ruggeri A, Gluckman E, Tamouza R. HLA haplotype frequencies and diversity in patients with hemoglobinopathies. EJHAEM 2023; 4:963-969. [PMID: 38024588 PMCID: PMC10660433 DOI: 10.1002/jha2.763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/10/2023] [Accepted: 07/26/2023] [Indexed: 12/01/2023]
Abstract
The genetic diversity of the human leukocyte antigen (HLA) system was shaped by evolutionary constraints exerted by environmental factors. Analyzing HLA diversity may allow understanding of the underlying pathways and offer useful tools in transplant setting. The aim of this study was to investigate the HLA haplotype diversity in patients with sickle cell disease (SCD, N = 282) or β-thalassemia (β-Thal, N = 60), who received hematopoietic cell transplantation (HCT) reported to Eurocord and the Société Francophone de Greffe de Moelle et de Thérapie Cellulaire (SFGM-TC). We identified 405 different HLA-A-B-DRB1 haplotypes in SCD and 108 in β-Thal patients. Using data from African and European populations of the "1000 Genomes Project" for comparison with SCD and β-Thal, respectively, we found that the haplotypes HLA-A*30-B*14-DRB1*15 (OR 7.87, 95% CI: 1.66-37.3, p b = 0.035), HLA-A*23-B*08 (OR 6.59, 95% CI: 1.8-24.13, p b = 0.023), and HLA-B*14-DRB1*15 (OR 10.74, 95% CI: 3.66-31.57, p b = 0.000) were associated with SCD, and the partial haplotypes HLA-A*30-B*13 and HLA-A*68-B*53 were associated with β-Thal (OR 4.810, 95% CI: 1.55-14.91, p b = 0.033, and OR 17.52, 95% CI: 2.81-184.95, p b = 0.011). Our results confirm the extreme HLA genetic diversity in SCD patients likely due to their African ancestry. This diversity seems less accentuated in patients with β-Thal. Our findings emphasize the need to expand inclusion of donors of African descent in HCT donor registries and cord blood banks.
Collapse
Affiliation(s)
- Graziana M. Scigliuolo
- Eurocord, Hôpital Saint‐Louis APHPInstitut de Recherche de Saint‐Louis (IRSL) EA3518Université de Paris CitéParisFrance
- Monacord, Centre Scientifique de MonacoMonacoMonaco
| | - Wahid Boukouaci
- Laboratoire Neuro‐Psychiatrie TranslationnelleINSERM U955, IMRB, et APHPHôpital Henri MondorCréteilFrance
| | - Barbara Cappelli
- Eurocord, Hôpital Saint‐Louis APHPInstitut de Recherche de Saint‐Louis (IRSL) EA3518Université de Paris CitéParisFrance
- Monacord, Centre Scientifique de MonacoMonacoMonaco
| | - Fernanda Volt
- Eurocord, Hôpital Saint‐Louis APHPInstitut de Recherche de Saint‐Louis (IRSL) EA3518Université de Paris CitéParisFrance
| | - Monica M. Rivera Franco
- Eurocord, Hôpital Saint‐Louis APHPInstitut de Recherche de Saint‐Louis (IRSL) EA3518Université de Paris CitéParisFrance
| | - Nathalie Dhédin
- Service d'hématologie Adolescents Jeunes AdultesHôpital Saint LouisAPHPParisFrance
| | | | - Christine Devalck
- HUDERF(Hôpital Universitaire des Enfants Reine Fabiola)Department of Hemato‐OncologyUniversité Libre de BruxellesBruxellesBelgium
| | | | | | - Olivier Hermine
- AP‐HP, Department of Adult HematologyHôpital NeckerUniversity of ParisParisFrance
| | | | - Xavier Poiré
- Service d'hématologie, Cliniques Universitaires St‐LucUniversité Catholique de LouvainBrusselsBelgium
| | - Bénédicte Brichard
- Department of Paediatric Haematology and OncologyCliniques Universitaires Saint LucBrusselsBelgium
| | - Catherine Paillard
- Department of Pediatric Hemato‐oncology and Bone Marrow Transplantation UnitHopital de HautepierreStrasbourgFrance
| | - Hanadi Rafii
- Eurocord, Hôpital Saint‐Louis APHPInstitut de Recherche de Saint‐Louis (IRSL) EA3518Université de Paris CitéParisFrance
| | - Chantal Kenzey
- Eurocord, Hôpital Saint‐Louis APHPInstitut de Recherche de Saint‐Louis (IRSL) EA3518Université de Paris CitéParisFrance
| | - Ching‐Lien Wu
- Laboratoire Neuro‐Psychiatrie TranslationnelleINSERM U955, IMRB, et APHPHôpital Henri MondorCréteilFrance
| | - Jihène Bouassida
- Laboratoire Neuro‐Psychiatrie TranslationnelleINSERM U955, IMRB, et APHPHôpital Henri MondorCréteilFrance
| | - Marie Robin
- Service d'Hématologie‐GreffeHôpital Saint‐Louis, APHPUniversité de Paris‐CitéParisFrance
- La Société Francophone de Greffe de Moelle et de Thérapie CellulaireLyonFrance
| | - Nicole Raus
- La Société Francophone de Greffe de Moelle et de Thérapie CellulaireLyonFrance
| | - Vanderson Rocha
- Eurocord, Hôpital Saint‐Louis APHPInstitut de Recherche de Saint‐Louis (IRSL) EA3518Université de Paris CitéParisFrance
- Faculty of MedicineHospital das ClínicasSão Paulo UniversitySão PauloBrazil
| | - Annalisa Ruggeri
- Eurocord, Hôpital Saint‐Louis APHPInstitut de Recherche de Saint‐Louis (IRSL) EA3518Université de Paris CitéParisFrance
- Hematology and Bone Marrow Transplant UnitIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Eliane Gluckman
- Eurocord, Hôpital Saint‐Louis APHPInstitut de Recherche de Saint‐Louis (IRSL) EA3518Université de Paris CitéParisFrance
- Monacord, Centre Scientifique de MonacoMonacoMonaco
| | - Ryad Tamouza
- Eurocord, Hôpital Saint‐Louis APHPInstitut de Recherche de Saint‐Louis (IRSL) EA3518Université de Paris CitéParisFrance
- Laboratoire Neuro‐Psychiatrie TranslationnelleINSERM U955, IMRB, et APHPHôpital Henri MondorCréteilFrance
| | | |
Collapse
|
7
|
Tukwasibwe S, Mboowa G, Sserwadda I, Nankabirwa JI, Arinaitwe E, Ssewanyana I, Taremwa Y, Tumusiime G, Kamya MR, Jagannathan P, Nakimuli A. Impact of high human genetic diversity in Africa on immunogenicity and efficacy of RTS,S/AS01 vaccine. Immunogenetics 2023; 75:207-214. [PMID: 37084013 PMCID: PMC10119520 DOI: 10.1007/s00251-023-01306-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 04/06/2023] [Indexed: 04/22/2023]
Abstract
In modern medicine, vaccination is one of the most effective public health strategies to prevent infectious diseases. Indisputably, vaccines have saved millions of lives by reducing the burden of many serious infections such as polio, tuberculosis, measles, pneumonia, and tetanus. Despite the recent recommendation by the World Health Organization (WHO) to roll out RTS,S/AS01, this malaria vaccine still faces major challenges of variability in its efficacy partly due to high genetic variation in humans and malaria parasites. Immune responses to malaria vary between individuals and populations. Human genetic variation in immune system genes is the probable cause for this heterogeneity. In this review, we will focus on human genetic factors that determine variable responses to vaccination and how variation in immune system genes affect the immunogenicity and efficacy of the RTS,S/AS01 vaccine.
Collapse
Affiliation(s)
- Stephen Tukwasibwe
- Infectious Diseases Research Collaboration, Kampala, Uganda.
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda.
- School of Medicine, Uganda Christian University, Kampala, Uganda.
| | - Gerald Mboowa
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Ivan Sserwadda
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | | | | | - Yoweri Taremwa
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Gerald Tumusiime
- School of Medicine, Uganda Christian University, Kampala, Uganda
| | - Moses R Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda
- School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | - Annettee Nakimuli
- School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
8
|
Hussein BK, Ibrahium OM, Alamin MF, Ahmed LAM, Abuswar SAE, Abdelraheem MH, Ibrahim ME. The Spike Protein of SARS-coV2 19B (S) Clade Mirrors Critical Features of Viral Adaptation and Coevolution. Microorganisms 2022; 10:2017. [PMID: 36296293 PMCID: PMC9609303 DOI: 10.3390/microorganisms10102017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 09/27/2022] [Accepted: 10/02/2022] [Indexed: 12/01/2022] Open
Abstract
Pathogens including viruses evolve in tandem with diversity in their animal and human hosts. For SARS-coV2, the focus is generally for understanding such coevolution on the virus spike protein, since it demonstrates high mutation rates compared to other genome regions, particularly in the receptor-binding domain (RBD). Viral sequences of the SARS-coV2 19B (S) clade and variants of concern from different continents were investigated, with a focus on the A.29 lineage, which presented with different mutational patterns within the 19B (S) lineages in order to learn more about how SARS-coV2 may have evolved and adapted to widely diverse populations globally. Results indicated that SARS-coV2 went through evolutionary constrains and intense selective pressure, particularly in Africa. This was manifested in a departure from neutrality with excess nonsynonymous mutations and a negative Tajima D consistent with rapid expansion and directional selection as well as deletion and deletion-frameshifts in the N-terminal domain (NTD region) of the spike protein. In conclusion, we hypothesize that viral transmission during epidemics through populations of diverse genomic structures and marked complexity may be a significant factor for the virus to acquire distinct patterns of mutations within these populations in order to ensure its survival and fitness, explaining the emergence of novel variants and strains.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Muntaser E. Ibrahim
- Unit of Disease and Diversity, Department of Molecular Biology, Institute of Endemic Diseases, Khartoum University, Khartoum P.O Box 102, Sudan
| |
Collapse
|
9
|
Mpoulimari I, Zintzaras E. Synthesis of genetic association studies on autism spectrum disorders using a genetic model-free approach. Psychiatr Genet 2022; 32:91-104. [PMID: 35353796 DOI: 10.1097/ypg.0000000000000316] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a clinically and genetically heterogeneous group of neurodevelopmental disorders. Despite the extensive efforts of scientists, the etiology of ASD is far from completely elucidated. In an effort to enlighten the genetic architecture of ASDs, a meta-analysis of all available genetic association studies (GAS) was conducted. METHODS We searched in the Human Genome Epidemiology Navigator (HuGE Navigator) and PubMed for available case-control GAS of ASDs. The threshold for meta-analysis was two studies per genetic variant. The association between genotype distribution and ASDs was examined using the generalized linear odds ratio (ORG). For variants with available allele frequencies, the examined model was the allele contrast. RESULTS Overall, 57 candidate genes and 128 polymorphisms were investigated in 159 articles. In total 28 genetic polymorphisms have been shown to be associated with ASDs, that are harbored in 19 genes. Statistically significant results were revealed for the variants of the following genes adenosine deaminase (ADA), bone marrow stromal cell antigen-1 (CD157/BST1), Dopamine receptor D1 (DRD1), engrailed homolog 2 (EN2), met proto-oncogene (MET), methylenetetrahydrofolate reductase (MTHFR), solute carrier family 6 member 4 (SLC6A4), Synaptosomal-associated protein, 25kDa (SNAP-25) and vitamin D receptor (VDR). In the allele contrast model of cases versus healthy controls, significant associations were observed for Adrenoceptor Alpha 1B (ADRA1B), acetyl serotonin O - methyltransferase (ASMT), complement component 4B (C4B), dopamine receptor D3 (DRD3), met proto-oncogene (MET), neuroligin 4, X-linked (NLGN4), neurexin 1 (NRXN1), oxytocin receptor (OXTR), Serine/Threonine-Protein Kinase PFTAIRE-1 (PFTK1), Reelin (RELN) and Ras-like without CAAX 2 (RIT2). CONCLUSION These significant findings provide further evidence for genetic factors' implication in ASDs offering new perspectives in means of prevention and prognosis.
Collapse
Affiliation(s)
- Ioanna Mpoulimari
- Department of Biomathematics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Elias Zintzaras
- Department of Biomathematics, Faculty of Medicine, University of Thessaly, Larissa, Greece
- Department of Medicine, The Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
10
|
African Genomic Medicine Portal: A Web Portal for Biomedical Applications. J Pers Med 2022; 12:jpm12020265. [PMID: 35207753 PMCID: PMC8879570 DOI: 10.3390/jpm12020265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/22/2022] [Accepted: 01/26/2022] [Indexed: 11/17/2022] Open
Abstract
Genomics data are currently being produced at unprecedented rates, resulting in increased knowledge discovery and submission to public data repositories. Despite these advances, genomic information on African-ancestry populations remains significantly low compared with European- and Asian-ancestry populations. This information is typically segmented across several different biomedical data repositories, which often lack sufficient fine-grained structure and annotation to account for the diversity of African populations, leading to many challenges related to the retrieval, representation and findability of such information. To overcome these challenges, we developed the African Genomic Medicine Portal (AGMP), a database that contains metadata on genomic medicine studies conducted on African-ancestry populations. The metadata is curated from two public databases related to genomic medicine, PharmGKB and DisGeNET. The metadata retrieved from these source databases were limited to genomic variants that were associated with disease aetiology or treatment in the context of African-ancestry populations. Over 2000 variants relevant to populations of African ancestry were retrieved. Subsequently, domain experts curated and annotated additional information associated with the studies that reported the variants, including geographical origin, ethnolinguistic group, level of association significance and other relevant study information, such as study design and sample size, where available. The AGMP functions as a dedicated resource through which to access African-specific information on genomics as applied to health research, through querying variants, genes, diseases and drugs. The portal and its corresponding technical documentation, implementation code and content are publicly available.
Collapse
|
11
|
Anyigba CA, Awandare GA, Paemka L. Breast cancer in sub-Saharan Africa: The current state and uncertain future. Exp Biol Med (Maywood) 2021; 246:1377-1387. [PMID: 33926257 DOI: 10.1177/15353702211006047] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the commonest cause of global cancer-related deaths in women and a public health burden in sub-Saharan Africa (SSA). Although the disease incidence in SSA seems lower, mortality rates are disproportionately high in comparison to high-income countries. The global disease burden is growing, with SSA reporting the majority of cases; however, the dearth of information results in insufficient data which is barely representative of the actual disease burden in this population. Future incidence predictions assign the subregion with a majority of the cases and associated deaths. Breast cancer presents with racial and ethnic variations, and available evidence suggests geographical diversity and persistent risk factors that have barely been explored in SSA. Breast cancer is a complex genetic disease, but the genetic risk factors in the extant African population, which is the most genetically diverse population, is scant and of low quality. This review focuses on the burden, prevalence, detection, treatment, survival, biology, as well as risk factors, and reinforces the need for breast cancer-associated risk factor investigation and population-specific studies in SSA.
Collapse
Affiliation(s)
- Claudia A Anyigba
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, 58835University of Ghana, Accra, GH 00233, Ghana
| | - Gordon A Awandare
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, 58835University of Ghana, Accra, GH 00233, Ghana
| | - Lily Paemka
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, 58835University of Ghana, Accra, GH 00233, Ghana
| |
Collapse
|
12
|
Alosaimi S, van Biljon N, Awany D, Thami PK, Defo J, Mugo JW, Bope CD, Mazandu GK, Mulder NJ, Chimusa ER. Simulation of African and non-African low and high coverage whole genome sequence data to assess variant calling approaches. Brief Bioinform 2020; 22:6042242. [PMID: 33341897 DOI: 10.1093/bib/bbaa366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/14/2020] [Accepted: 01/08/2020] [Indexed: 12/15/2022] Open
Abstract
Current variant calling (VC) approaches have been designed to leverage populations of long-range haplotypes and were benchmarked using populations of European descent, whereas most genetic diversity is found in non-European such as Africa populations. Working with these genetically diverse populations, VC tools may produce false positive and false negative results, which may produce misleading conclusions in prioritization of mutations, clinical relevancy and actionability of genes. The most prominent question is which tool or pipeline has a high rate of sensitivity and precision when analysing African data with either low or high sequence coverage, given the high genetic diversity and heterogeneity of this data. Here, a total of 100 synthetic Whole Genome Sequencing (WGS) samples, mimicking the genetics profile of African and European subjects for different specific coverage levels (high/low), have been generated to assess the performance of nine different VC tools on these contrasting datasets. The performances of these tools were assessed in false positive and false negative call rates by comparing the simulated golden variants to the variants identified by each VC tool. Combining our results on sensitivity and positive predictive value (PPV), VarDict [PPV = 0.999 and Matthews correlation coefficient (MCC) = 0.832] and BCFtools (PPV = 0.999 and MCC = 0.813) perform best when using African population data on high and low coverage data. Overall, current VC tools produce high false positive and false negative rates when analysing African compared with European data. This highlights the need for development of VC approaches with high sensitivity and precision tailored for populations characterized by high genetic variations and low linkage disequilibrium.
Collapse
Affiliation(s)
- Shatha Alosaimi
- Faculty of Health Sciences, Division of Human Genetics, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Noëlle van Biljon
- Department of Statistical Sciences, University of Cape Town, Cape Town, South Africa
| | - Denis Awany
- Faculty of Health Sciences, Division of Human Genetics, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Prisca K Thami
- Faculty of Health Sciences, Division of Human Genetics, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Joel Defo
- Faculty of Health Sciences, Division of Human Genetics, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Jacquiline W Mugo
- Faculty of Health Sciences, Division of Computational Biology, Department of Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Christian D Bope
- Faculty of Sciences, Department of Mathematics and Computer Science, University of Kinshasa, Kinshasa, DRC
| | - Gaston K Mazandu
- Faculty of Health Sciences, Division of Human Genetics, Department of Pathology, University of Cape Town, Cape Town, South Africa.,Faculty of Health Sciences, Division of Computational Biology, Department of Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Nicola J Mulder
- Faculty of Health Sciences, Division of Computational Biology, Department of Biomedical Sciences, University of Cape Town, Cape Town, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| | - Emile R Chimusa
- Faculty of Health Sciences, Division of Human Genetics, Department of Pathology, University of Cape Town, Cape Town, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| |
Collapse
|
13
|
Taiwo RO, Ipadeola J, Yusuf T, Fagbohunlu F, Jenfa G, Adebamowo SN, Adebamowo CA. Qualitative study of comprehension of heritability in genomics studies among the Yoruba in Nigeria. BMC Med Ethics 2020; 21:124. [PMID: 33298068 PMCID: PMC7726892 DOI: 10.1186/s12910-020-00567-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/03/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND With growth of genomics research in Africa, concern has arisen about comprehension and adequacy of informed consent given the highly technical terms used in this field. We therefore decided to study whether there are linguistic and cultural concepts used to communicate heritability of characters, traits and diseases in an indigenous African population. METHODS We conducted Focus Group Discussions among 115 participants stratified by sex, age and socio-economic status and Key Informant Interviews among 25 stakeholders and Key Opinion Leaders among Yoruba living in Ibadan, Nigeria. We used Atlas-ti v.8.3.17 software to analyze the data, using thematic approach. RESULTS The study participants identified several linguistic and cultural concepts including words, proverbs, and aphorisms that are used to describe heritable characters, traits and diseases in their local dialect. These included words that can be appropriated to describe dominant and recessive traits, variations in penetrance and dilution of strength of heritable characteristics by time and inter-marriage. They also suggested that these traits are transmitted by "blood", and specific partner's blood may be stronger than the other regardless of sex. CONCLUSIONS Indigenous Yoruba populations have words and linguistic concepts that describe the heritability of characters, traits and diseases which can be appropriated to improve comprehension and adequacy of informed consent in genomics research. Our methods are openly available and can be used by genomic researchers in other African communities.
Collapse
Affiliation(s)
- Rasheed O Taiwo
- Division of Research Ethics, Center for Bioethics and Research, Ibadan, Nigeria
| | - John Ipadeola
- Division of Research Ethics, Center for Bioethics and Research, Ibadan, Nigeria
| | - Temilola Yusuf
- Division of Research Ethics, Center for Bioethics and Research, Ibadan, Nigeria
| | - Faith Fagbohunlu
- Division of Research Ethics, Center for Bioethics and Research, Ibadan, Nigeria
| | - Gbemisola Jenfa
- Division of Research Ethics, Center for Bioethics and Research, Ibadan, Nigeria
| | - Sally N Adebamowo
- Division of Research Ethics, Center for Bioethics and Research, Ibadan, Nigeria.,Department of Epidemiology and Public Health, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Clement A Adebamowo
- Division of Research Ethics, Center for Bioethics and Research, Ibadan, Nigeria. .,Department of Epidemiology and Public Health, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA. .,Institute of Human Virology, Abuja, Nigeria. .,Institute of Human Virology Building, School of Medicine, University of Maryland, 725 West Lombard Street, Baltimore, MD, 21201, USA.
| | | |
Collapse
|
14
|
Kashangura C. Enhanced education on vaccines can reduce the scourge of vaccine rejection and hesitation. S AFR J SCI 2020. [DOI: 10.17159/sajs.2020/8143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
15
|
N-Acetyltransferase 2 Genotypes among Zulu-Speaking South Africans and Isoniazid and N-Acetyl-Isoniazid Pharmacokinetics during Antituberculosis Treatment. Antimicrob Agents Chemother 2020; 64:AAC.02376-19. [PMID: 31964788 PMCID: PMC7179278 DOI: 10.1128/aac.02376-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 12/23/2019] [Indexed: 01/06/2023] Open
Abstract
The distribution of N-acetyltransferase 2 gene (NAT2) polymorphisms varies considerably among different ethnic groups. Information on NAT2 single-nucleotide polymorphisms in the South African population is limited. We investigated NAT2 polymorphisms and their effect on isoniazid pharmacokinetics (PK) in Zulu black HIV-infected South Africans in Durban, South Africa. HIV-infected participants with culture-confirmed pulmonary tuberculosis (TB) were enrolled from two unrelated studies. Participants with culture-confirmed pulmonary TB were genotyped for the NAT2 polymorphisms 282C>T, 341T>C, 481C>T, 857G>A, 590G>A, and 803A>G using Life Technologies prevalidated TaqMan assays (Life Technologies, Paisley, UK). Participants underwent sampling for determination of plasma isoniazid and N-acetyl-isoniazid concentrations. Among the 120 patients, 63/120 (52.5%) were slow metabolizers (NAT2*5/*5), 43/120 (35.8%) had an intermediate metabolism genotype (NAT2*5/12), and 12/120 (11.7%) had a rapid metabolism genotype (NAT2*4/*11, NAT2*11/12, and NAT2*12/12). The NAT2 alleles evaluated in this study were *4, *5C, *5D, *5E, *5J, *5K, *5KA, *5T, *11A, *12A/12C, and *12M. NAT2*5 was the most frequent allele (70.4%), followed by NAT2*12 (27.9%). Fifty-eight of 60 participants in study 1 had PK results. The median area under the concentration-time curve from 0 to infinity (AUC0-∞) was 5.53 (interquartile range [IQR], 3.63 to 9.12 μg h/ml), and the maximum concentration (C max) was 1.47 μg/ml (IQR, 1.14 to 1.89 μg/ml). Thirty-four of 40 participants in study 2 had both PK results and NAT2 genotyping results. The median AUC0-∞ was 10.76 μg·h/ml (IQR, 8.24 to 28.96 μg·h/ml), and the C max was 3.14 μg/ml (IQR, 2.39 to 4.34 μg/ml). Individual polymorphisms were not equally distributed, with some being represented in small numbers. The genotype did not correlate with the phenotype, with those with a rapid acetylator genotype showing higher AUC0-∞ values than those with a slow acetylator genotype, but the difference was not significant (P = 0.43). There was a high prevalence of slow acetylator genotypes, followed by intermediate and then rapid acetylator genotypes. The poor concordance between genotype and phenotype suggests that other factors or genetic loci influence isoniazid metabolism, and these warrant further investigation in this population.
Collapse
|
16
|
Mayne ES, Louw S. Good Fences Make Good Neighbors: Human Immunodeficiency Virus and Vascular Disease. Open Forum Infect Dis 2019; 6:ofz303. [PMID: 31737735 PMCID: PMC6847507 DOI: 10.1093/ofid/ofz303] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/25/2019] [Indexed: 01/08/2023] Open
Abstract
Cardiovascular disease, venous thrombosis, and microvascular disease in people with HIV (PWH) is predicted to increase in an aging HIV-infected population. Endothelial damage and dysfunction is a risk factor for cardiovascular events in PWH and is characterized by impaired vascular relaxation and decreased nitric oxide availability. Vascular disease has been attributed to direct viral effects, opportunistic infections, chronic inflammation, effects of antiretroviral therapy, and underlying comorbid conditions, like hypertension and use of tobacco. Although biomarkers have been examined to predict and prognosticate thrombotic and cardiovascular disease in this population, more comprehensive validation of risk factors is necessary to ensure patients are managed appropriately. This review examines the pathogenesis of vascular disease in PWH and summarizes the biomarkers used to predict vascular disease in this population.
Collapse
Affiliation(s)
- Elizabeth S Mayne
- Department of Immunology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service
| | - Susan Louw
- Department of Molecular Medicine Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
17
|
Mitropoulos K, Cooper DN, Mitropoulou C, Agathos S, Reichardt JKV, Al-Maskari F, Chantratita W, Wonkam A, Dandara C, Katsila T, Lopez-Correa C, Ali BR, Patrinos GP. Genomic Medicine Without Borders: Which Strategies Should Developing Countries Employ to Invest in Precision Medicine? A New "Fast-Second Winner" Strategy. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 21:647-657. [PMID: 29140767 DOI: 10.1089/omi.2017.0141] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Genomic medicine has greatly matured in terms of its technical capabilities, but the diffusion of genomic innovations worldwide faces significant barriers beyond mere access to technology. New global development strategies are sorely needed for biotechnologies such as genomics and their applications toward precision medicine without borders. Moreover, diffusion of genomic medicine globally cannot adhere to a "one-size-fits-all-countries" development strategy, in the same way that drug treatments should be customized. This begs a timely, difficult but crucial question: How should developing countries, and the resource-limited regions of developed countries, invest in genomic medicine? Although a full-scale investment in infrastructure from discovery to the translational implementation of genomic science is ideal, this may not always be feasible in all countries at all times. A simple "transplantation of genomics" from developed to developing countries is unlikely to be feasible. Nor should developing countries be seen as simple recipients and beneficiaries of genomic medicine developed elsewhere because important advances in genomic medicine have materialized in developing countries as well. There are several noteworthy examples of genomic medicine success stories involving resource-limited settings that are contextualized and described in this global genomic medicine innovation analysis. In addition, we outline here a new long-term development strategy for global genomic medicine in a way that recognizes the individual country's pressing public health priorities and disease burdens. We term this approach the "Fast-Second Winner" model of innovation that supports innovation commencing not only "upstream" of discovery science but also "mid-stream," building on emerging highly promising biomarker and diagnostic candidates from the global science discovery pipeline, based on the unique needs of each country. A mid-stream entry into innovation can enhance collective learning from other innovators' mistakes upstream in discovery science and boost the probability of success for translation and implementation when resources are limited. This à la carte model of global innovation and development strategy offers multiple entry points into the global genomics innovation ecosystem for developing countries, whether or not extensive and expensive discovery infrastructures are already in place. Ultimately, broadening our thinking beyond the linear model of innovation will help us to enable the vision and practice of genomics without borders in both developed and resource-limited settings.
Collapse
Affiliation(s)
| | - David N Cooper
- 2 Institute of Medical Genetics, School of Medicine, Cardiff University , Cardiff, United Kingdom
| | | | - Spiros Agathos
- 4 Yachay Tech University , San Miguel de Urcuquí, Ecuador
| | | | - Fatima Al-Maskari
- 5 Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University , Al-Ain, United Arab Emirates .,6 Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University , Al-Ain, United Arab Emirates
| | - Wasun Chantratita
- 7 Department of Pathology, Medical Genomic Center, Ramathibodi Hospital, Faculty of Medicine, Mahidol University , Bangkok, Thailand
| | - Ambroise Wonkam
- 8 Division of Human Genetics, Department of Medicine and Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Collet Dandara
- 8 Division of Human Genetics, Department of Medicine and Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Theodora Katsila
- 9 Department of Pharmacy, School of Health Sciences, University of Patras , Patras, Greece
| | | | - Bassam R Ali
- 5 Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University , Al-Ain, United Arab Emirates .,6 Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University , Al-Ain, United Arab Emirates
| | - George P Patrinos
- 5 Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University , Al-Ain, United Arab Emirates .,6 Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University , Al-Ain, United Arab Emirates .,9 Department of Pharmacy, School of Health Sciences, University of Patras , Patras, Greece
| |
Collapse
|
18
|
Population genetic evidence for positive and purifying selection acting at the human IFN-γ locus in Africa. Genes Immun 2018; 20:143-157. [PMID: 29599512 DOI: 10.1038/s41435-018-0016-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 01/09/2023]
Abstract
Despite its critical role in the defense against microbial infection and tumor development, little is known about the range of nucleotide and haplotype variation at IFN-γ, or the evolutionary forces that have shaped patterns of diversity at this locus. To address this gap in knowledge, we examined sequence data from the IFN-γ gene in 1461 individuals from 15 worldwide populations. Our analyses uncovered novel patterns of variation in distinct African populations, including an excess of high frequency-derived alleles, unusually long haplotype structure surrounding the IFN-γ gene, and a "star-like" genealogy of African-specific haplotypes carrying variants previously associated with infectious disease. We also inferred a deep time to coalescence of variation at IFN-γ (~ 0.8 million years ago) and ancient ages for common polymorphisms predating the evolution of modern humans. Taken together, these results are congruent with a model of positive selection on standing variation in African populations. Furthermore, we inferred that common variants in intron 3 of IFN-γ are the likely targets of selection. In addition, we observed a paucity of non-synonymous substitutions relative to synonymous changes in the exons of IFN-γ in African and non-African populations, suggestive of strong purifying selection. Therefore, we contend that positive and purifying selection have influenced levels of diversity in different regions of IFN-γ, implying that these distinct genic regions are, or have been, functionally important. Overall, this study provides additional insights into the evolutionary events that have contributed to the frequency and distribution of alleles having a role in human health and disease.
Collapse
|
19
|
Mekonnen E, Bekele E, Stein CM. Novel polymorphisms in TICAM2 and NOD1 associated with tuberculosis progression phenotypes in Ethiopian populations. Glob Health Epidemiol Genom 2018; 3:e1. [PMID: 29868226 PMCID: PMC5870410 DOI: 10.1017/gheg.2017.17] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 12/04/2017] [Accepted: 12/08/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Infection by Mycobacterium tuberculosis (Mtb) is a necessary but not sufficient cause for tuberculosis (TB). Although numerous studies suggest human genetic variation may influence TB pathogenesis, there is a conspicuous lack of replication, likely due to imprecise phenotype definition. We aimed to replicate novel findings from a Ugandan cohort in Ethiopian populations. METHOD We ascertained TB cases and household controls (n = 292) from three different ethnic groups. Latent Mtb infection was determined using Quantiferon to develop reliable TB progression phenotypes. We sequenced exonic regions of TICAM2 and NOD1. RESULT Significant novel associations were observed between two variants in NOD1 and TB: rs751770147 [unadjusted p = 7.28 × 10-5] and chr7:30477156(T), a novel variant, [unadjusted p = 1.04 × 10-4]. Two SNPs in TICAM2 were nominally associated with TB, including rs2288384 [unadjusted p = 0.003]. Haplotype-based association tests supported the SNP-based results. CONCLUSION We replicated the association of TICAM2 and NOD1 with TB and identified novel genetic associations with TB in Ethiopian populations.
Collapse
Affiliation(s)
- E. Mekonnen
- Microbial, Cellular, Molecular Biology Department, Addis Ababa University, P.O.Box:17087, Addis Ababa, Ethiopia
- Health Biotechnology Department, Institute of Biotechnology, Addis Ababa University, P.O.Box:17087, Addis Ababa, Ethiopia
| | - E. Bekele
- Microbial, Cellular, Molecular Biology Department, Addis Ababa University, Ethiopia
| | - C. M. Stein
- Department of Population & Quantitative Health Sciences, Center for Proteomics & Bioinformatics, and Tuberculosis Research Unit, Case Western Reserve University, USA
| |
Collapse
|
20
|
Davison GM, Hendrickse HL, Matsha TE. The relationship between immunogenic red blood cell antigens and Human Immunodeficiency Virus infection. Transfus Apher Sci 2017; 57:58-62. [PMID: 29269123 DOI: 10.1016/j.transci.2017.11.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/21/2017] [Accepted: 11/28/2017] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Evidence suggests that red cell antigens may act as receptors for viruses and bacteria and therefore could be associated with HIV infection. Previous studies have been controversial and therefore the aim of this exploratory study was to analyse the expression of immunogenic red cell antigens in HIV-seropositive individuals and to compare the results to negative donors from South Africa. METHODS The expression of ABO, Rh, Kell and Duffy antigens from 119 HIV-seropositive patients was compared to 317 HIV-seronegative blood donors. Nucleic acid amplification testing and PCR were used to determine the HIV status and the ID-Gel Card Technology was used to determine the blood group antigen profile. RESULTS There was no significant difference in the expression of A, B, AB, Duffy or Kel antigens between the two groups but significantly lower numbers of HIV+ individuals were O Rh Negative (p = ,0.0001). Analysis of those with a Duffy null phenotype revealed a significantly higher incidence of blood type A RH1-Positive, Dce/R0r and B RH1-Positive, DcEe/R2r within the HIV-seropositive group (p = < 0.05). None of the HIV-seropositive individuals were O RH1-Negative, dce/rr. CONCLUSION In conclusion these initial findings have demonstrated a decreased incidence of blood type O Rh1-negative in HIV + individuals which suggests that red blood cell antigens may play an important role in susceptibility to HIV infection. The relationship between red cell antigens and HIV infection however remains complex and therefore larger studies are required to confirm these results.
Collapse
Affiliation(s)
- Glenda M Davison
- Department of Biomedical sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa.
| | - Heather L Hendrickse
- Department of Biomedical sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| | - Tandi E Matsha
- Department of Biomedical sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| |
Collapse
|
21
|
Mekonnen E, Bekele E. An ancestral human genetic variant linked to an ancient disease: A novel association of FMO2 polymorphisms with tuberculosis (TB) in Ethiopian populations provides new insight into the differential ethno-geographic distribution of FMO2*1. PLoS One 2017; 12:e0184931. [PMID: 28981537 PMCID: PMC5628799 DOI: 10.1371/journal.pone.0184931] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 09/01/2017] [Indexed: 11/19/2022] Open
Abstract
The human FMO2 (flavin-containing monooxygenase 2) gene has been shown to be involved in innate immunity against microbial infections, including tuberculosis (TB), via the modulation of oxidative stress levels. It has also been found to possess a curious loss-of-function mutation (FMO2*1/FMO2*2) that demonstrates a distinctive differentiation in expression, function and ethno-geographic distribution. However, despite evidences of ethnic-specific genetic associations in the inflammatory profile of TB, no studies were done to investigate whether these patterns of variations correlate with evidences for the involvement of FMO2 in antimicrobial immune responses and ethnic differences in the distribution of FMO2 polymorphisms except for some pharmacogenetic data that suggest a potentially deleterious role for the functional variant (FMO2*1). This genetic epidemiological study was designed to investigate whether there is an association between FMO2 polymorphisms and TB, an ancient malady that remains a modern global health concern, in a sub-Saharan Africa setting where there is not only a relatively high co-prevalence of the disease and the ancestral FMO2*1 variant but also where both Mycobcaterium and Homo sapiens are considered to have originated and co-evolved. Blood samples and TB related clinical data were collected from ascertained TB cases and unrelated household controls (n = 292) from 3 different ethnic groups in Ethiopia. Latent Mtb infection was determined using Quantiferon to develop reliable TB progression phenotypes. We sequenced exonic regions of FMO2.We identified for the first time an association between FMO2 and TB both at the SNP and haplotype level. Two novel SNPs achieved a study-wide significance [chr1:171181877(A), p = 3.15E-07, OR = 4.644 and chr1:171165749(T), p = 3.32E-06, OR = 6.825] while multiple SNPs (22) showed nominal signals. The pattern of association suggested a protective effect of FMO2 against both active and latent TB with distinct genetic variants underlying the TB-progression pathway. The results were robust for population stratification. Haplotype-based tests confirmed the SNP-based results with a single haplotype bearing the ancestral-and-functional FMO2*1 "C" allele ("AGCTCTACAATCCCCTCGTTGCGC") explaining the overall association (haplotype-specific-p = 0.000103). Strikingly, not only was FMO2*1 nominally associated with reduced risk to "Active TB" (p = 0.0118, OR = 0.496) but it also does not co-segregate with the 5'-3' flanking top high-TB-risk alleles. The study provides an evidence for the existence of an evolutionary adaptation to an ancient disease based on an ancestral genetic variant acting in a haplotypic framework in Ethiopian populations.
Collapse
Affiliation(s)
- Ephrem Mekonnen
- Department of Microbial, Cellular, Molecular Biology, Addis Ababa University, Addis Ababa, Ethiopia
- Department of Health Biotechnology, Institute of Biotechnology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Endashaw Bekele
- Department of Microbial, Cellular, Molecular Biology, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
22
|
Nyirenda MJ. Non-communicable diseases in sub-Saharan Africa: understanding the drivers of the epidemic to inform intervention strategies. Int Health 2017; 8:157-8. [PMID: 27178673 DOI: 10.1093/inthealth/ihw021] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 04/12/2016] [Indexed: 11/13/2022] Open
Abstract
Sub-Saharan Africa is experiencing a rapidly increasing epidemic of non-communicable diseases (NCDs), while it continues to face longstanding challenges from infectious diseases. This double burden of disease could have a devastating impact on a continent that already has significant resource constraints, emphasizing the urgent need for appropriate interventions in the region. However, it is crucial to have a careful understanding of the local drivers of NCDs. This should not only include study of the traditional lifestyle risk factors, but also an examination of emerging risk factors (such as the influence of insults early in life or interactions with infection), which may be of particular importance in Africa.
Collapse
Affiliation(s)
- Moffat J Nyirenda
- London School of Hygiene & Tropical Medicine, Department of Non-Communicable Disease Epidemiology, Keppel Street, London, WC1E 7HT, UK
| |
Collapse
|
23
|
Roberts L, Ratnapriya R, du Plessis M, Chaitankar V, Ramesar RS, Swaroop A. Molecular Diagnosis of Inherited Retinal Diseases in Indigenous African Populations by Whole-Exome Sequencing. Invest Ophthalmol Vis Sci 2017; 57:6374-6381. [PMID: 27898983 PMCID: PMC5132076 DOI: 10.1167/iovs.16-19785] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose A majority of genes associated with inherited retinal diseases (IRDs) have been identified in patients of European origin. Indigenous African populations exhibit rich genomic diversity, and evaluation of reported genetic mutations has yielded low returns so far. Our goal was to perform whole-exome sequencing (WES) to examine variants in known IRD genes in underrepresented African cohorts. Methods Whole-exome sequencing was performed on 56 samples from 16 families with diverse IRD phenotypes that had remained undiagnosed after screening for known mutations using genotyping-based microarrays (Asper Ophthalmics). Variants in reported IRD genes were identified using WES and validated by Sanger sequencing. Custom TaqMan assays were used to screen for identified mutations in 193 unrelated indigenous Africans with IRDs. Results A total of 3494 variants were identified in 217 known IRD genes, leading to the identification of seven different mutations (including six novel) in six genes (RHO, PRPF3, PRPF31, ABCA4, CERKL, and PDE6B) in six distinct families. TaqMan screening in additional probands revealed identical homozygous CERKL and PDE6B variants in four more patients. Conclusions This is the first report of WES of patients with IRDs in indigenous African populations. Our study identified genetic defects in almost 40% of the families analyzed, significantly enhancing the molecular diagnosis of IRD in South Africa. Thus, WES of understudied cohorts seems to present an effective strategy for determining novel mutations in heterogeneous retinal diseases.
Collapse
Affiliation(s)
- Lisa Roberts
- University of Cape Town/MRC Human Genetics Research Unit, Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rinki Ratnapriya
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Morné du Plessis
- University of Cape Town/MRC Human Genetics Research Unit, Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Vijender Chaitankar
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Raj S Ramesar
- University of Cape Town/MRC Human Genetics Research Unit, Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Anand Swaroop
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
24
|
Disease burden and the role of pharmacogenomics in African populations. GLOBAL HEALTH EPIDEMIOLOGY AND GENOMICS 2017; 2:e1. [PMID: 29868213 PMCID: PMC5870420 DOI: 10.1017/gheg.2016.21] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 11/22/2016] [Accepted: 11/24/2016] [Indexed: 12/15/2022]
Abstract
Background The burden of communicable and non-communicable diseases in Sub-Saharan Africa poses a challenge in achieving quality healthcare. Although therapeutic drugs have generally improved health, their efficacy differs from individual to individual. Variability in treatment response is mainly because of genetic variants that affect the pharmacokinetics and pharmacodynamics of drugs. Method The intersection of disease burden and therapeutic intervention is reviewed, and the status of pharmacogenomics knowledge in African populations is explored. Results The most commonly studied variants with pharmacogenomics relevance are discussed, especially in genes coding for enzymes that affect the response to drugs used for HIV, malaria, sickle cell disease and cardiovascular diseases. Conclusions The genetically diverse African population is likely to benefit from a pharmacogenomics-based healthcare approach, especially with respect to reduction of drug side effects, and separation of responders and non-responders leading to optimized drug choices and doses for each patient.
Collapse
|
25
|
Is there a role of pharmacogenomics in Africa. GLOBAL HEALTH EPIDEMIOLOGY AND GENOMICS 2016; 1:e9. [PMID: 29868201 PMCID: PMC5870419 DOI: 10.1017/gheg.2016.4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 03/31/2016] [Accepted: 04/09/2016] [Indexed: 12/21/2022]
Abstract
Pharmacogenomics has the potential of transforming clinical research and improving healthcare in sub-Saharan Africa (SSA). The role of African genome diversity and the opportunities for pharmacogenomics research are highlighted and will enable discovery of novel genetic mechanisms and validation of established markers. African genomics and biobank consortia will play an important role in building capacity for pharmacogenomics in SSA.
Collapse
|
26
|
Variability at the FCGR locus: characterization in Black South Africans and evidence for ethnic variation in and out of Africa. Genes Immun 2015; 17:93-104. [PMID: 26673965 DOI: 10.1038/gene.2015.60] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 01/07/2023]
Abstract
This study set out to comprehensively investigate all known functional FcγR variants in South African Black and Caucasian individuals. Population diversity was further assessed using data from the 1000 Genomes Project. In our cohort, Black South Africans neither possessed the haplotypes previously associated with increased surface densities of FcγRIIb and FcγRIIIa nor the FCGR2C haplotype recently associated with increased vaccine efficacy in the RV144 HIV-1 vaccine trial (despite 48.7% bearing the c.134-96T tag allele). Moreover, Africans (South Africans, Luhya Kenyans and Yoruba Nigerians) lack the FCGR2C c.798+1G splice-site allele required for the expression of functional FcγRIIc. Although the presence or absence of surface FcγRIIc did not affect natural killer cell-mediated antibody-dependent cellular cytotoxicity capability, this may be significant for other FcγRIIc-mediated functions. Overall, allele distribution and linkage disequilibrium in Africans and Caucasians differed in a manner that would suggest a differentially maintained balance of FcγR-mediated cell activation in these populations. Finally, significant variation observed among different African populations precludes the use of any one African population as a proxy for FcγR diversity in Africans. In conclusion, the findings of this study highlight further ethnic variation at the FCGR gene locus, in particular for FCGR2C, a gene with increasingly recognized clinical significance.
Collapse
|
27
|
Mulder NJ, Adebiyi E, Alami R, Benkahla A, Brandful J, Doumbia S, Everett D, Fadlelmola FM, Gaboun F, Gaseitsiwe S, Ghazal H, Hazelhurst S, Hide W, Ibrahimi A, Jaufeerally Fakim Y, Jongeneel CV, Joubert F, Kassim S, Kayondo J, Kumuthini J, Lyantagaye S, Makani J, Mansour Alzohairy A, Masiga D, Moussa A, Nash O, Ouwe Missi Oukem-Boyer O, Owusu-Dabo E, Panji S, Patterton H, Radouani F, Sadki K, Seghrouchni F, Tastan Bishop Ö, Tiffin N, Ulenga N. H3ABioNet, a sustainable pan-African bioinformatics network for human heredity and health in Africa. Genome Res 2015; 26:271-7. [PMID: 26627985 PMCID: PMC4728379 DOI: 10.1101/gr.196295.115] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/25/2015] [Indexed: 11/24/2022]
Abstract
The application of genomics technologies to medicine and biomedical research is increasing in popularity, made possible by new high-throughput genotyping and sequencing technologies and improved data analysis capabilities. Some of the greatest genetic diversity among humans, animals, plants, and microbiota occurs in Africa, yet genomic research outputs from the continent are limited. The Human Heredity and Health in Africa (H3Africa) initiative was established to drive the development of genomic research for human health in Africa, and through recognition of the critical role of bioinformatics in this process, spurred the establishment of H3ABioNet, a pan-African bioinformatics network for H3Africa. The limitations in bioinformatics capacity on the continent have been a major contributory factor to the lack of notable outputs in high-throughput biology research. Although pockets of high-quality bioinformatics teams have existed previously, the majority of research institutions lack experienced faculty who can train and supervise bioinformatics students. H3ABioNet aims to address this dire need, specifically in the area of human genetics and genomics, but knock-on effects are ensuring this extends to other areas of bioinformatics. Here, we describe the emergence of genomics research and the development of bioinformatics in Africa through H3ABioNet.
Collapse
Affiliation(s)
- Nicola J Mulder
- Computational Biology Group, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa 7925
| | - Ezekiel Adebiyi
- Covenant University Bioinformatics Research (CUBRe) and Department of Computer and Information Sciences, Covenant University, Ota, Ogun State, Nigeria, P.M.B. 1023
| | - Raouf Alami
- Centre National de Transfusion Sanguine, Rabat, Morocco 10100
| | | | - James Brandful
- Noguchi Memorial Institute for Medical Research, University of Ghana, Ghana, LG
| | - Seydou Doumbia
- University of Sciences, Techniques and Technology of Bamako, Bamako, Mali BPE 3206
| | - Dean Everett
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi, 3/Institute of Infection and Global Health, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Faisal M Fadlelmola
- Centre for Bioinformatics and Systems Biology, Faculty of Science, University of Khartoum/Future University of Sudan, Khartoum, Sudan 11115
| | - Fatima Gaboun
- Institut National de Recherche Agronomique, Rabat, Morocco 10000
| | | | | | - Scott Hazelhurst
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa 2193
| | - Winston Hide
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA/Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield S10 2HQ, United Kingdom
| | - Azeddine Ibrahimi
- Faculté de Médecine et de Pharmacie de Rabat, Université Mohammed V Souissi, Rabat, Morocco 10100
| | | | - C Victor Jongeneel
- National Center for Supercomputing Applications and Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Fourie Joubert
- Department of Biochemistry, University of Pretoria, Pretoria, South Africa 0083
| | - Samar Kassim
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt 11566
| | | | - Judit Kumuthini
- Centre for Proteomic and Genomic Research, Cape Town, South Africa 7925
| | | | - Julie Makani
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania 00255
| | | | - Daniel Masiga
- International Centre of Insect Physiology and Ecology, Nairobi, Kenya 00100
| | - Ahmed Moussa
- Abdelmalek Essaadi University, ENSA, Tangier, Morocco 90000
| | - Oyekanmi Nash
- National Biotechnology Development Agency, Abuja, Nigeria 10099
| | | | - Ellis Owusu-Dabo
- Kumasi Centre for Collaborative Research in Tropical Medicine/Kwame Nkrumah University of Science and Technology, Kumasi, Ghana, PMB
| | - Sumir Panji
- Computational Biology Group, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa 7925
| | - Hugh Patterton
- University of the Free State, Bloemfontein, South Africa 9300
| | | | - Khalid Sadki
- Faculty of Sciences of Rabat, University Mohammed V of Rabat, Rabat, Morocco 10000
| | | | - Özlem Tastan Bishop
- Research Unit in Bioinformatics, Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, South Africa 6140
| | - Nicki Tiffin
- South African National Bioinformatics Institute/Medical Research Council of South Africa Bioinformatics Unit, University of the Western Cape, Cape Town, South Africa 7530
| | - Nzovu Ulenga
- Management and Development for Health, Dar es Salaam, Tanzania, 61
| | | |
Collapse
|
28
|
Renin-Angiotensin System Genes Polymorphisms and Essential Hypertension in Burkina Faso, West Africa. Int J Hypertens 2015; 2015:979631. [PMID: 26351579 PMCID: PMC4553326 DOI: 10.1155/2015/979631] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/08/2015] [Indexed: 11/18/2022] Open
Abstract
Objective. This study aimed to investigate the association between three polymorphisms of renin-angiotensin system and the essential hypertension in the population of Burkina Faso. Methodology. This was a case-control study including 202 cases and 204 matched controls subjects. The polymorphisms were identified by a classical and a real-time PCR. Results. The AGT 235M/T and AT1R 1166A/C polymorphisms were not associated with the hypertension while the genotype frequencies of the ACE I/D polymorphism between patients and controls (DD: 66.83% and 35.78%, ID: 28.22% and 50.98%, II: 4.95% and 13.24%, resp.) were significantly different (p < 10−4). The genotype DD of ACE gene (OR = 3.40, p < 0.0001), the increasing age (OR = 3.83, p < 0.0001), obesity (OR = 4.84, p < 0.0001), dyslipidemia (OR = 3.43, p = 0.021), and alcohol intake (OR = 2.76, p < 0.0001) were identified as the independent risk factors for hypertension by multinomial logistic regression. Conclusion. The DD genotype of the ACE gene is involved in susceptibility to hypertension. Further investigations are needed to better monitor and provide individualized care for hypertensive patients.
Collapse
|
29
|
Mitropoulos K, Al Jaibeji H, Forero DA, Laissue P, Wonkam A, Lopez-Correa C, Mohamed Z, Chantratita W, Lee MTM, Llerena A, Brand A, Ali BR, Patrinos GP. Success stories in genomic medicine from resource-limited countries. Hum Genomics 2015; 9:11. [PMID: 26081768 PMCID: PMC4485996 DOI: 10.1186/s40246-015-0033-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 06/09/2015] [Indexed: 02/08/2023] Open
Abstract
In recent years, the translation of genomic discoveries into mainstream medical practice and public health has gained momentum, facilitated by the advent of new technologies. However, there are often major discrepancies in the pace of implementation of genomic medicine between developed and developing/resource-limited countries. The main reason does not only lie in the limitation of resources but also in the slow pace of adoption of the new findings and the poor understanding of the potential that this new discipline offers to rationalize medical diagnosis and treatment. Here, we present and critically discuss examples from the successful implementation of genomic medicine in resource-limited countries, focusing on pharmacogenomics, genome informatics, and public health genomics, emphasizing in the latter case genomic education, stakeholder analysis, and economics in pharmacogenomics. These examples can be considered as model cases and be readily replicated for the wide implementation of pharmacogenomics and genomic medicine in other resource-limited environments.
Collapse
Affiliation(s)
| | - Hayat Al Jaibeji
- University of Maastricht, Maastricht, The Netherlands. .,Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.
| | - Diego A Forero
- Laboratory of NeuroPsychiatric Genetics, Biomedical Sciences Research Group, School of Medicine, Universidad Antonio Nariño, Bogotá, Colombia.
| | - Paul Laissue
- Unidad de Genética, Grupo GENIUROS, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia.
| | - Ambroise Wonkam
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| | | | - Zahurin Mohamed
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Wasun Chantratita
- Department of Pathology, Medical Genomic Center, Ramathibodi Hospital, Faculty of Medicine, Mahidol University, Bangkok, Thailand.
| | - Ming Ta Michael Lee
- Laboratory for International Alliance on Genomic Research, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan. .,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| | - Adrian Llerena
- CICAB Clinical Research Center, Extremadura University Hospital and Medical School, Badajoz, Spain.
| | - Angela Brand
- University of Maastricht, Maastricht, The Netherlands.
| | - Bassam R Ali
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.
| | - George P Patrinos
- Department of Pharmacy, University of Patras School of Health Sciences, Patras, Greece.
| |
Collapse
|
30
|
Chin'ombe N, Ruhanya V. HIV/AIDS vaccines for Africa: scientific opportunities, challenges and strategies. Pan Afr Med J 2015; 20:386. [PMID: 26185576 PMCID: PMC4499268 DOI: 10.11604/pamj.2015.20.386.4660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 04/13/2015] [Indexed: 01/01/2023] Open
Abstract
More than decades have already elapsed since human immunodeficiency virus (HIV) was identified as the causative agent of acquired immunodeficiency syndrome (AIDS). The HIV has since spread to all parts of the world with devastating effects. In sub-saharan Africa, the HIV/AIDS epidemic has reached unprecedented proportions. Safe, effective and affordable HIV/AIDS vaccines for Africans are therefore urgently needed to contain this public health problem. Although, there are challenges, there are also scientific opportunities and strategies that can be exploited in the development of HIV/AIDS vaccines for Africa. The recent RV144 Phase III trial in Thailand has demonstrated that it is possible to develop a vaccine that can potentially elicit modest protective immunity against HIV infection. The main objective of this review is to outline the key scientific opportunities, challenges and strategies in HIV/AIDS vaccine development in Africa.
Collapse
Affiliation(s)
- Nyasha Chin'ombe
- Department of Medical Microbiology, College of Health Sciences, University of Zimbabwe, P O Box A178, Avondale, Harare, Zimbabwe
| | - Vurayai Ruhanya
- Department of Medical Microbiology, College of Health Sciences, University of Zimbabwe, P O Box A178, Avondale, Harare, Zimbabwe
| |
Collapse
|
31
|
Tindana P, Molyneux CS, Bull S, Parker M. Ethical issues in the export, storage and reuse of human biological samples in biomedical research: perspectives of key stakeholders in Ghana and Kenya. BMC Med Ethics 2014; 15:76. [PMID: 25326753 PMCID: PMC4210627 DOI: 10.1186/1472-6939-15-76] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/09/2014] [Indexed: 11/25/2022] Open
Abstract
Background For many decades, access to human biological samples, such as cells, tissues, organs, blood, and sub-cellular materials such as DNA, for use in biomedical research, has been central in understanding the nature and transmission of diseases across the globe. However, the limitations of current ethical and regulatory frameworks in sub-Saharan Africa to govern the collection, export, storage and reuse of these samples have resulted in inconsistencies in practice and a number of ethical concerns for sample donors, researchers and research ethics committees. This paper examines stakeholders’ perspectives of and responses to the ethical issues arising from these research practices. Methods We employed a qualitative strategy of inquiry for this research including in-depth interviews and focus group discussions with key research stakeholders in Kenya (Nairobi and Kilifi), and Ghana (Accra and Navrongo). Results The stakeholders interviewed emphasised the compelling scientific importance of sample export, storage and reuse, and acknowledged the existence of some structures governing these research practices, but they also highlighted the pressing need for a number of practical ethical concerns to be addressed in order to ensure high standards of practice and to maintain public confidence in international research collaborations. These concerns relate to obtaining culturally appropriate consent for sample export and reuse, understanding cultural sensitivities around the use of blood samples, facilitating a degree of local control of samples and sustainable scientific capacity building. Conclusion Drawing on these findings and existing literature, we argue that the ethical issues arising in practice need to be understood in the context of the interactions between host research institutions and local communities and between collaborating institutions. We propose a set of ‘key points-to-consider’ for research institutions, ethics committees and funding agencies to address these issues.
Collapse
Affiliation(s)
- Paulina Tindana
- Navrongo Health Research Centre, Ghana Health Service, P,O, Box 114, Navrongo, Ghana.
| | | | | | | |
Collapse
|
32
|
Alessandrini M, Pepper MS. Priority pharmacogenetics for the African continent: focus on CYP450. Pharmacogenomics 2014; 15:385-400. [PMID: 24533717 DOI: 10.2217/pgs.13.252] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Countries in Africa have a high burden of communicable disease, and are experiencing an increase in noncommunicable diseases due to the effects of globalization, industrialization and urbanization. The costs incurred through adverse drug reactions and nonresponsiveness to therapy further aggravates the situation, and the application of pharmacogenetic principles is likely to provide some relief. Having undertaken an extensive evaluation of CYP450 reports in Africa, our objective was to map out areas of need based on regional disease burdens. The data confirms a paucity of CYP450 reports and illustrates large regions for which no population information exists. There is a dire need to address the health problems of Africa, and wide-scale pharmacogenetic profiling of these populations will add significantly to improving patient care on the continent. Priority pharmacogenetics for the African continent gives precedence to the profiling of clinically relevant pharmacogenetic biomarkers, and defines the immediate need in the context of disease burden.
Collapse
Affiliation(s)
- Marco Alessandrini
- Department of Immunology & The Institute for Cellular & Molecular Medicine, Faculty of Health Sciences, University of Pretoria, South Africa
| | | |
Collapse
|
33
|
Osthoff M, Irungu E, Ngure K, Mugo N, Thomas KK, Baeten JM, Eisen DP. Mannose-binding lectin and ficolin-2 do not influence humoral immune response to hepatitis B vaccine. Vaccine 2014; 32:4772-7. [PMID: 25024112 PMCID: PMC4374143 DOI: 10.1016/j.vaccine.2014.06.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/17/2014] [Accepted: 06/06/2014] [Indexed: 01/05/2023]
Abstract
BACKGROUND Host genetics appear to be an important factor in the failure to generate a protective immune response after hepatitis B (HBV) vaccination. Mannose-binding lectin (MBL) and ficolin-2 (FCN2), two pattern recognition receptors of the lectin pathway of complement, influence the clinical outcome of HBV, and MBL deficiency has been shown to augment the humoral response to HBV vaccination in several experimental models. Here, we investigated the association of MBL and FCN2 with the humoral response to HBV vaccination in a candidate gene and functional study. PATIENTS AND METHODS A post hoc analysis of a prospective, interventional HBV vaccination study among human immunodeficiency virus type 1 (HIV-1) uninfected individuals in Kenya was conducted. Serum levels and polymorphisms of MBL and FCN2 were analysed in relation to the immune response to HBV vaccination. RESULTS Protective hepatitis B surface antibody levels (≥ 10 mI U/mL) were evident in 251/293 (85.7%) individuals. Median MBL and FCN2 levels were similar in responders vs. non-responders with a weak trend towards lower median MBL levels in non-responders (1.0 vs. 1.6μg/mL, p=0.1). Similarly, there was no difference in four MBL and six FCN2 polymorphisms analysed in the two groups with the exception of an increased frequency of a homozygous MBL codon 57 mutation in non-responders (4 (9.5%) vs. 8 (3.2%), p=0.05) corresponding to lower MBL levels. Results were similar after adjusting for age and sex. CONCLUSIONS Our study does not support a prominent role of the lectin pathway of complement in general and MBL and FCN2 in particular in the humoral immune response to HBV vaccination in African adults.
Collapse
Affiliation(s)
- Michael Osthoff
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Parkville, VIC, Australia; Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | | | - Kenneth Ngure
- Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya; Department of Global Health, University of Washington, Seattle, WA, USA
| | - Nelly Mugo
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | | | - Jared M Baeten
- Department of Epidemiology, University of Washington, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA; Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Damon P Eisen
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Parkville, VIC, Australia; Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
34
|
White MJ, Tacconelli A, Chen JS, Wejse C, Hill PC, Gomes VF, Velez-Edwards DR, Østergaard LJ, Hu T, Moore JH, Novelli G, Scott WK, Williams SM, Sirugo G. Epiregulin (EREG) and human V-ATPase (TCIRG1): genetic variation, ethnicity and pulmonary tuberculosis susceptibility in Guinea-Bissau and The Gambia. Genes Immun 2014; 15:370-7. [PMID: 24898387 DOI: 10.1038/gene.2014.28] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/23/2014] [Accepted: 04/24/2014] [Indexed: 02/07/2023]
Abstract
We analyzed two West African samples (Guinea-Bissau: n=289 cases and 322 controls; The Gambia: n=240 cases and 248 controls) to evaluate single-nucleotide polymorphisms (SNPs) in Epiregulin (EREG) and V-ATPase (T-cell immune regulator 1 (TCIRG1)) using single and multilocus analyses to determine whether previously described associations with pulmonary tuberculosis (PTB) in Vietnamese and Italians would replicate in African populations. We did not detect any significant single locus or haplotype associations in either sample. We also performed exploratory pairwise interaction analyses using Visualization of Statistical Epistasis Networks (ViSEN), a novel method to detect only interactions among multiple variables, to elucidate possible interaction effects between SNPs and demographic factors. Although we found no strong evidence of marginal effects, there were several significant pairwise interactions that were identified in either the Guinea-Bissau or the Gambian samples, two of which replicated across populations. Our results indicate that the effects of EREG and TCIRG1 variants on PTB susceptibility, to the extent that they exist, are dependent on gene-gene interactions in West African populations as detected with ViSEN. In addition, epistatic effects are likely to be influenced by inter- and intra-population differences in genetic or environmental context and/or the mycobacterial lineages causing disease.
Collapse
Affiliation(s)
- M J White
- 1] Center for Human Genetics Research, Vanderbilt University, Nashville, TN, USA [2] Department of Genetics and Institute of Quantitative Biomedical Sciences, Dartmouth College, Hanover, NH, USA
| | - A Tacconelli
- Centro di Ricerca, Ospedale San Pietro Fatebenefratelli, Rome, Italy
| | - J S Chen
- Department of Genetics and Institute of Quantitative Biomedical Sciences, Dartmouth College, Hanover, NH, USA
| | - C Wejse
- 1] Bandim Health Project, Danish Epidemiology Science Centre and Statens Serum Institute, Bissau, Guinea-Bissau [2] Department of Infectious Diseases, Aarhus University Hospital, Skejby, Denmark [3] Center for Global Health, School of Public Health, Aarhus University, Skejby, Denmark
| | - P C Hill
- 1] Centre for International Health, University of Otago School of Medicine, Dunedin, New Zealand [2] MRC Laboratories, Fajara, The Gambia
| | - V F Gomes
- Bandim Health Project, Danish Epidemiology Science Centre and Statens Serum Institute, Bissau, Guinea-Bissau
| | - D R Velez-Edwards
- 1] Vanderbilt Epidemiology Center, Vanderbilt University, Nashville, TN, USA [2] Institute for Medicine and Public Health, Vanderbilt University, Nashville, TN, USA [3] Center for Human Genetics Research, Vanderbilt University, Nashville, TN, USA [4] Department of Obstetrics and Gynecology, Vanderbilt University, Nashville, TN, USA
| | - L J Østergaard
- Department of Infectious Diseases, Aarhus University Hospital, Skejby, Denmark
| | - T Hu
- Department of Genetics and Institute of Quantitative Biomedical Sciences, Dartmouth College, Hanover, NH, USA
| | - J H Moore
- Department of Genetics and Institute of Quantitative Biomedical Sciences, Dartmouth College, Hanover, NH, USA
| | - G Novelli
- 1] Centro di Ricerca, Ospedale San Pietro Fatebenefratelli, Rome, Italy [2] Dipartimento di Biomedicina e Prevenzione, Sezione di Genetica, Università di Roma 'Tor Vergata', Rome, Italy
| | - W K Scott
- Dr John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - S M Williams
- Department of Genetics and Institute of Quantitative Biomedical Sciences, Dartmouth College, Hanover, NH, USA
| | - G Sirugo
- Centro di Ricerca, Ospedale San Pietro Fatebenefratelli, Rome, Italy
| |
Collapse
|
35
|
Ferrer-Admetlla A, Liang M, Korneliussen T, Nielsen R. On detecting incomplete soft or hard selective sweeps using haplotype structure. Mol Biol Evol 2014; 31:1275-91. [PMID: 24554778 PMCID: PMC3995338 DOI: 10.1093/molbev/msu077] [Citation(s) in RCA: 244] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We present a new haplotype-based statistic (nSL) for detecting both soft and hard sweeps in population genomic data from a single population. We compare our new method with classic single-population haplotype and site frequency spectrum (SFS)-based methods and show that it is more robust, particularly to recombination rate variation. However, all statistics show some sensitivity to the assumptions of the demographic model. Additionally, we show that nSL has at least as much power as other methods under a number of different selection scenarios, most notably in the cases of sweeps from standing variation and incomplete sweeps. This conclusion holds up under a variety of demographic models. In many aspects, our new method is similar to the iHS statistic; however, it is generally more robust and does not require a genetic map. To illustrate the utility of our new method, we apply it to HapMap3 data and show that in the Yoruban population, there is strong evidence of selection on genes relating to lipid metabolism. This observation could be related to the known differences in cholesterol levels, and lipid metabolism more generally, between African Americans and other populations. We propose that the underlying causes for the selection on these genes are pleiotropic effects relating to blood parasites rather than their role in lipid metabolism.
Collapse
Affiliation(s)
- Anna Ferrer-Admetlla
- Department of Integrative Biology, University of California at Berkeley, Berkeley
| | - Mason Liang
- Department of Integrative Biology, University of California at Berkeley, Berkeley
| | - Thorfinn Korneliussen
- Centre for GeoGenetics, Natural History Museum of Denmark, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Nielsen
- Department of Integrative Biology, University of California at Berkeley, Berkeley
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Department of Bioinformatics, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Dandara C, Swart M, Mpeta B, Wonkam A, Masimirembwa C. Cytochrome P450 pharmacogenetics in African populations: implications for public health. Expert Opin Drug Metab Toxicol 2014; 10:769-85. [PMID: 24588448 DOI: 10.1517/17425255.2014.894020] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Africa harbors a disproportionate burden of disease when taking into account the triple challenge caused by HIV/AIDS, tuberculosis (TB) and malaria, against a backdrop of an increasing burden of noncommunicable diseases. More than 80% of therapeutic drugs used in the management of these diseases/conditions are metabolized by CYP enzymes that exhibit genetic polymorphisms. AREAS COVERED There is variability in the expression and activities of CYPs resulting in interindividual differences in the response to standard doses of therapeutic drugs, due to genetic polymorphisms, which exhibit both quantitative and qualitative differences between racial and between ethnic groups. The review aims to evaluate the implications of the genetic variation in CYPs on the public health of Africans. The CYPs reviewed here metabolize most of the commonly used therapeutic drugs and include CYP1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 3A4 and 3A5. Allele frequencies are compared between African ethnic groups and among populations of African, Asian and European origin. Data are obtained from our own studies and literature. EXPERT OPINION The variability in the pattern of genetic variation between populations translates into differences in drug response. Understanding CYP variability improves rational drug use and has public health significance.
Collapse
Affiliation(s)
- Collet Dandara
- University of Cape Town, Faculty of Health Sciences, Pharmacogenetics and Cancer Research Group, Division of Human Genetics, Department of Clinical Laboratory Sciences , Anzio Road Observatory, 7925, Cape Town , South Africa +27 21 406 6506 ;
| | | | | | | | | |
Collapse
|
37
|
Peprah E, Wonkam A. Biomedical research, a tool to address the health issues that affect African populations. Global Health 2013; 9:50. [PMID: 24143865 PMCID: PMC4015770 DOI: 10.1186/1744-8603-9-50] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 08/05/2013] [Indexed: 12/22/2022] Open
Abstract
Traditionally, biomedical research endeavors in low to middle resources countries have focused on communicable diseases. However, data collected over the past 20 years by the World Health Organization (WHO) show a significant increase in the number of people suffering from non-communicable diseases (e.g. heart disease, diabetes, cancer and pulmonary diseases). Within the coming years, WHO predicts significant decreases in communicable diseases while non-communicable diseases are expected to double in low and middle income countries in sub-Saharan Africa. The predicted increase in the non-communicable diseases population could be economically burdensome for the basic healthcare infrastructure of countries that lack resources to address this emerging disease burden. Biomedical research could stimulate development of healthcare and biomedical infrastructure. If this development is sustainable, it provides an opportunity to alleviate the burden of both communicable and non-communicable diseases through diagnosis, prevention and treatment. In this paper, we discuss how research using biomedical technology, especially genomics, has produced data that enhances the understanding and treatment of both communicable and non-communicable diseases in sub-Saharan Africa. We further discuss how scientific development can provide opportunities to pursue research areas responsive to the African populations. We limit our discussion to biomedical research in the areas of genomics due to its substantial impact on the scientific community in recent years however, we also recognize that targeted investments in other scientific disciplines could also foster further development in African countries.
Collapse
Affiliation(s)
- Emmanuel Peprah
- Current address: National Institutes of Health, Building 1, RM 256A, Bethesda MD 20892, USA
| | - Ambroise Wonkam
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Anzio Road-7925, Observatory, Cape Town, South Africa
| |
Collapse
|
38
|
Zhang Y, Shen X, Pan W. Adjusting for population stratification in a fine scale with principal components and sequencing data. Genet Epidemiol 2013; 37:787-801. [PMID: 24123217 DOI: 10.1002/gepi.21764] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 08/26/2013] [Accepted: 09/10/2013] [Indexed: 01/09/2023]
Abstract
Population stratification is of primary interest in genetic studies to infer human evolution history and to avoid spurious findings in association testing. Although it is well studied with high-density single nucleotide polymorphisms (SNPs) in genome-wide association studies (GWASs), next-generation sequencing brings both new opportunities and challenges to uncovering population structures in finer scales. Several recent studies have noticed different confounding effects from variants of different minor allele frequencies (MAFs). In this paper, using a low-coverage sequencing dataset from the 1000 Genomes Project, we compared a popular method, principal component analysis (PCA), with a recently proposed spectral clustering technique, called spectral dimensional reduction (SDR), in detecting and adjusting for population stratification at the level of ethnic subgroups. We investigated the varying performance of adjusting for population stratification with different types and sets of variants when testing on different types of variants. One main conclusion is that principal components based on all variants or common variants were generally most effective in controlling inflations caused by population stratification; in particular, contrary to many speculations on the effectiveness of rare variants, we did not find much added value with the use of only rare variants. In addition, SDR was confirmed to be more robust than PCA, especially when applied to rare variants.
Collapse
Affiliation(s)
- Yiwei Zhang
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | | | | |
Collapse
|
39
|
Wright GEB, Koornhof PGJ, Adeyemo AA, Tiffin N. Ethical and legal implications of whole genome and whole exome sequencing in African populations. BMC Med Ethics 2013; 14:21. [PMID: 23714101 PMCID: PMC3668248 DOI: 10.1186/1472-6939-14-21] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 05/20/2013] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Rapid advances in high throughput genomic technologies and next generation sequencing are making medical genomic research more readily accessible and affordable, including the sequencing of patient and control whole genomes and exomes in order to elucidate genetic factors underlying disease. Over the next five years, the Human Heredity and Health in Africa (H3Africa) Initiative, funded by the Wellcome Trust (United Kingdom) and the National Institutes of Health (United States of America), will contribute greatly towards sequencing of numerous African samples for biomedical research. DISCUSSION Funding agencies and journals often require submission of genomic data from research participants to databases that allow open or controlled data access for all investigators. Access to such genotype-phenotype and pedigree data, however, needs careful control in order to prevent identification of individuals or families. This is particularly the case in Africa, where many researchers and their patients are inexperienced in the ethical issues accompanying whole genome and exome research; and where an historical unidirectional flow of samples and data out of Africa has created a sense of exploitation and distrust. In the current study, we analysed the implications of the anticipated surge of next generation sequencing data in Africa and the subsequent data sharing concepts on the protection of privacy of research subjects. We performed a retrospective analysis of the informed consent process for the continent and the rest-of-the-world and examined relevant legislation, both current and proposed. We investigated the following issues: (i) informed consent, including guidelines for performing culturally-sensitive next generation sequencing research in Africa and availability of suitable informed consent documents; (ii) data security and subject privacy whilst practicing data sharing; (iii) conveying the implications of such concepts to research participants in resource limited settings. SUMMARY We conclude that, in order to meet the unique requirements of performing next generation sequencing-related research in African populations, novel approaches to the informed consent process are required. This will help to avoid infringement of privacy of individual subjects as well as to ensure that informed consent adheres to acceptable data protection levels with regard to use and transfer of such information.
Collapse
Affiliation(s)
- Galen EB Wright
- South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| | - Pieter GJ Koornhof
- Department of Mercantile and Labour Law, University of the Western Cape, Bellville, South Africa
| | - Adebowale A Adeyemo
- Center for Research on Genomics and Global Health, National Institutes of Health/National Human Genome Research Institute, Bethesda, MD, USA
| | - Nicki Tiffin
- South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| |
Collapse
|
40
|
Alessandrini M, Asfaha S, Dodgen TM, Warnich L, Pepper MS. Cytochrome P450 pharmacogenetics in African populations. Drug Metab Rev 2013; 45:253-75. [PMID: 23590174 DOI: 10.3109/03602532.2013.783062] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The Cytochrome P450 (CYP450) family of enzymes is involved in the oxidative metabolism of many therapeutic drugs, carcinogens and various endogenous substrates. These enzymes are highly polymorphic at an inter-individual and inter-ethnic level. Polymorphisms or genetic variations account for up to 30% of inter-individual differences seen in a variety of drug responses. The frequencies of the different metabolizer categories (slow, intermediate, extensive and ultra-rapid), the distribution of genetic variants, genotype-phenotype correlations and the clinical importance of the CYP450 enzymes have been extensively documented in Caucasian and Oriental populations. Limited data exists for African populations, despite the fact that this knowledge is critically important for these populations who experience a heavy burden of communicable and non-communicable diseases. In addition, the costs incurred through adverse drug reactions and non-responsiveness to therapy could be reduced through the wide-scale application of pharmacogenetics. This review provides an overview and investigation of CYP450 genotypic and phenotypic reports published from 1980 to present in African populations. Our findings confirm the high degree of variability that is expected when comparing individuals of African origin to other ethnic groups and also highlight the distribution of clinically relevant CYP450 alleles amongst the various African populations. The notable discordance in genotypic and phenotypic data amongst African populations exemplifies the need for in-depth and well-orchestrated molecular and pharmacological investigations of these populations in the future, for which whole genome sequencing and association studies will be critical.
Collapse
Affiliation(s)
- Marco Alessandrini
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | | | | | | | | |
Collapse
|
41
|
Fakunle ES. iPSCs for personalized medicine: what will it take for Africa? Trends Mol Med 2012; 18:695-9. [PMID: 23137920 DOI: 10.1016/j.molmed.2012.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 10/11/2012] [Accepted: 10/11/2012] [Indexed: 11/15/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are cutting edge biotechnology that may revolutionize medicine, and creating iPSCs from ethnically diverse individuals would generate valuable therapeutic and drug development tools. However, challenges must be overcome in creating the infrastructure and scientific capacity needed to pursue innovative, leapfrogging strategies to make iPSCs available in Africa.
Collapse
Affiliation(s)
- Eyitayo S Fakunle
- Department of Chemical Physiology, Center for Regenerative Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
42
|
Khullar S, Tvedt D, Chapman K, Herlofson B. Sixty cases of extreme osteonecrosis and osteomyelitis of the mandible and maxilla in a West African population. Int J Oral Maxillofac Surg 2012; 41:978-85. [DOI: 10.1016/j.ijom.2012.04.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 02/16/2012] [Accepted: 04/26/2012] [Indexed: 10/28/2022]
|
43
|
Netea MG, Wijmenga C, O'Neill LAJ. Genetic variation in Toll-like receptors and disease susceptibility. Nat Immunol 2012; 13:535-42. [PMID: 22610250 DOI: 10.1038/ni.2284] [Citation(s) in RCA: 258] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) are key initiators of the innate immune response and promote adaptive immunity. Much has been learned about the role of TLRs in human immunity from studies linking TLR genetic variation with disease. First, monogenic disorders associated with complete deficiency in certain TLR pathways, such as MyD88-IRAK4 or TLR3-Unc93b-TRIF-TRAF3, have demonstrated the specific roles of these pathways in host defense against pyogenic bacteria and herpesviruses, respectively. Second, common polymorphisms in genes encoding several TLRs and associated genes have been associated with both infectious and autoimmune diseases. The study of genetic variation in TLRs in various populations combined with information on infection has demonstrated complex interaction between genetic variation in TLRs and environmental factors. This interaction explains the differences in the effect of TLR polymorphisms on susceptibility to infection and autoimmune disease in various populations.
Collapse
Affiliation(s)
- Mihai G Netea
- Department of Internal Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | | | | |
Collapse
|
44
|
Kempińska-Podhorodecka A, Knap O, Drozd A, Kaczmarczyk M, Parafiniuk M, Parczewski M, Ciechanowicz A. Analysis for genotyping Duffy blood group in inhabitants of Sudan, the fourth cataract of the Nile. Malar J 2012; 11:115. [PMID: 22510366 PMCID: PMC3375187 DOI: 10.1186/1475-2875-11-115] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 04/17/2012] [Indexed: 01/03/2023] Open
Abstract
Background Genetic polymophisms of the Duffy antigen receptor for the chemokines (DARC) gene successfully protected against blood stage infection by Plasmodium vivax infection. The Fy (a-, b-) phenotype is predominant among African populations, particularly those originating from West Africa, and it is rare among non-African populations. The aim of this study was to analyse the frequency of four Duffy blood groups based on SNPs (T-33C, G125A, G298A and C5411T) in two local tribes of Sudanese Arabs, the Shagia and Manasir, which are both from the region of the Fourth Nile cataract in Sudan. Methods An analysis of polymorphisms was performed on 217 individuals (126 representatives of the Shagia tribe and 91 of the Manasir). Real-time PCR and TaqMan Genotyping Assays were used to study the prevalence of alleles and genotypes. Results The analysis of allelic and genotype frequency in the T-33C polymorphisms demonstrated a significant dominance of the C allele and CC genotype (OR = 0.53 [0.32-0.88]; p = 0.02) in both tribes. The G125A polymorphism is associated with phenotype Fy(a-, b-) and was identified in 83% of Shagia and 77% of Manasir. With regard to G298A polymorphisms, the genotype frequencies were different between the tribes (p = 0,002) and no single AA homozygote was found. Based on four SNPs examined, 20 combinations of genotypes for the Shagia and Manasir tribes were determined. The genotype CC/AA/GG/CT occurred most often in Shagia tribe (45.9%) but was rare in the Manasir tribe (6.6%) (p < 0.001 Shagia versus Manasir). The FY*AES allele was identified in both analysed tribes. The presence of individuals with the FY*A/FY*A genotype was demonstrated only in the Shagia tribe. Conclusion This is probably the first report showing genotypically Duffy-negative people who carry both FY*BES and FY*AES. The identification of the FY*AES allele in both tribes may be due to admixture of the non-African genetic background. Taken as a whole, allele and genotype frequencies between the Shagia and the Manasir were statistically different. However, the presence of individuals with the FY*A/FY*A genotype was demonstrated only in the Shagia tribe.
Collapse
|
45
|
Gasmelseed N, Elsir AA, Deblasio P, Biunno I. Sub-Saharan centralized biorepository for genetic and genomic research. THE SCIENCE OF THE TOTAL ENVIRONMENT 2012; 423:210-213. [PMID: 21303714 DOI: 10.1016/j.scitotenv.2010.07.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 07/19/2010] [Accepted: 07/19/2010] [Indexed: 05/30/2023]
Abstract
Quality-assessed biomedical samples are essential for academia- and industry driven research on human diseases. The etiologies and the molecular genetic factors relevant in African diseases, including both infections and complex degenerative diseases as well as cancer, need to be studied using well annotated and well-preserved biosamples acquired from native African ethnic groups and compare the results with non-African populations and/or with Afro-Americans. However, a number of difficulties negatively impact on the possibility to obtain clinically annotated biological samples in most Sub-Saharan African countries. This is mainly due to major organizational problems, lack of clinical centres that can dedicate resources to research, as well as lack of facilities in which biomaterials can be properly processed and safely stored. Harmonization of biosample acquisition, storage phenotyping schemes and biocomputer infrastructures are the principal objectives of biological resource centers (BRCs). BRCs comprise biobanks of different formats (collection of blood, DNA, tissues, etc., annotated with medical, environmental, life-style and follow up data) a fundamental tool for molecular epidemiological studies aiming to increase excellence and efficacy of biomedical results, drug development and public health. BRCs provide large and highly controlled biomolecular resources necessary to meet the "omics" scientific platforms. Sudan may be a candidate nation to host such infrastructure, in view of its strategic geographical position and the already existing simple biobanking experiences connected with research groups in Central Sudan. Here, we describe the potential role of biobanks in African genetic studies aiming to dissect the eziopathogenesis of complex diseases in relation to environmental and life-style factors.
Collapse
Affiliation(s)
- Nagla Gasmelseed
- Department of Molecular Biology, National Cancer Institute University of Gezira, Wadmedani, Sudan
| | | | | | | |
Collapse
|
46
|
Elwali NE, Mariani Costantini R, Di Gioacchino M. Environment and immune-mediated diseases in Sub-Saharan Africa. THE SCIENCE OF THE TOTAL ENVIRONMENT 2012; 423:190-192. [PMID: 20961598 DOI: 10.1016/j.scitotenv.2010.08.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 08/26/2010] [Indexed: 05/30/2023]
|
47
|
Drögemöller BI, Wright GEB, Niehaus DJH, Emsley RA, Warnich L. Whole-genome resequencing in pharmacogenomics: moving away from past disparities to globally representative applications. Pharmacogenomics 2012; 12:1717-28. [PMID: 22118054 DOI: 10.2217/pgs.11.119] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Africa suffers from a high burden of disease; nonetheless, it has been one of the most under-represented continents with regard to genomic research. It can be argued that this disproportionate research is related to the fact that the genome architecture of African individuals is poorly suited to SNP-based genome-wide association studies, given existing genotyping platforms. However, this argument is no longer plausible with the arrival of next-generation sequencing technologies, which allow for the analysis of entire genomes. Using pharmacogenes to critically examine the merit of next-generation sequencing technologies in pharmacogenomics, we found a substantial amount of novel/uncharacterized variation, which was predicted to alter protein function. This variation was predominantly observed in African individuals, emphasizing the benefit of next-generation sequencing technologies specifically for these individuals. We also observed an improvement in the reliability of sequencing technologies in a relatively short time. Therefore, as sequencing technologies develop and decrease in cost, the ability to reliably detect variation will improve and these technologies will begin to replace other less comprehensive genotyping assays.
Collapse
Affiliation(s)
- Britt I Drögemöller
- Department of Genetics, Stellenbosch University, Private Bag XI, Matieland 7602, South Africa
| | | | | | | | | |
Collapse
|
48
|
Velez Edwards DR, Tacconelli A, Wejse C, Hill PC, Morris GAJ, Edwards TL, Gilbert JR, Myers JL, Park YS, Stryjewski ME, Abbate E, Estevan R, Rabna P, Novelli G, Hamilton CD, Adegbola R, Østergaard L, Williams SM, Scott WK, Sirugo G. MCP1 SNPs and pulmonary tuberculosis in cohorts from West Africa, the USA and Argentina: lack of association or epistasis with IL12B polymorphisms. PLoS One 2012; 7:e32275. [PMID: 22384203 PMCID: PMC3288089 DOI: 10.1371/journal.pone.0032275] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 01/25/2012] [Indexed: 11/20/2022] Open
Abstract
The monocyte chemotactic protein-1 (MCP-1) is a chemokine that plays an important role in the recruitment of monocytes to M. tuberculosis infection sites, and previous studies have reported that genetic variants in MCP1 are associated with differential susceptibility to pulmonary tuberculosis (PTB). We examined eight MCP1 single nucleotide polymorphisms (SNPs) in a multi-ethnic, case-control design that included: 321 cases and 346 controls from Guinea-Bissau, 258 cases and 271 controls from The Gambia, 295 cases and 179 controls from the U.S. (African-Americans), and an additional set of 237 cases and 144 controls of European ancestry from the U.S. and Argentina. Two locus interactions were also examined for polymorphisms in MCP1 and interleukin 12B (IL12B), another gene implicated in PTB risk. Examination of previously associated MCP1 SNPs rs1024611 (-2581A/G), rs2857656 (-362G/C) and rs4586 (+900C/T) did not show evidence for association. One interaction between rs2857656 and IL12B SNP rs2288831 was observed among Africans but the effect was in the opposite direction in Guineans (OR = 1.90, p = 0.001) and Gambians (OR = 0.64, p = 0.024). Our data indicate that the effect of genetic variation within MCP1 is not clear cut and additional studies will be needed to elucidate its role in TB susceptibility.
Collapse
Affiliation(s)
- Digna R. Velez Edwards
- Dr. John T. Macdonald Foundation Department of Human Genetics and Miami Institute of Human Genomics, University of Miami, Miami, Florida, United States of America
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Epidemiology Center, Institute of Medicine and Public Health, and Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Alessandra Tacconelli
- Centro di Genetica, Centro di Ricerca Scientifica, Ospedale San Pietro FBF, Rome, Italy
| | - Christian Wejse
- Bandim Health Project, Danish Epidemiology Science Centre and Statens Serum Institute, Bissau, Guinea-Bissau
- Department of Infectious Diseases, Aarhus University Hospital, Skejby, Denmark
- Center for Global Health, School of Public Health, Aarhus University, Skejby, Denmark
| | - Philip C. Hill
- MRC Laboratories, Fajara, The Gambia (West Africa)
- Centre for International Health, University of Otago School of Medicine, Dunedin, New Zealand
| | | | - Todd L. Edwards
- Dr. John T. Macdonald Foundation Department of Human Genetics and Miami Institute of Human Genomics, University of Miami, Miami, Florida, United States of America
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - John R. Gilbert
- Dr. John T. Macdonald Foundation Department of Human Genetics and Miami Institute of Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Jamie L. Myers
- Dr. John T. Macdonald Foundation Department of Human Genetics and Miami Institute of Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Yo Son Park
- Dr. John T. Macdonald Foundation Department of Human Genetics and Miami Institute of Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Martin E. Stryjewski
- Division of Infectious Diseases, Department of Medicine, Centro de Educación Médica e Investigaciones Clínicas “Norberto Quirno” (CEMIC), Buenos Aires, Argentina
| | - Eduardo Abbate
- Department of Medicine, Hospital F. J. Muñiz, Buenos Aires, Argentina
| | - Rosa Estevan
- Department of Medicine, Hospital F. J. Muñiz, Buenos Aires, Argentina
| | - Paulo Rabna
- Bandim Health Project, Danish Epidemiology Science Centre and Statens Serum Institute, Bissau, Guinea-Bissau
| | - Giuseppe Novelli
- Centro di Genetica, Centro di Ricerca Scientifica, Ospedale San Pietro FBF, Rome, Italy
- Dipartimento di Biopatologia e Diagnostica per Immagini, Università di Tor Vergata, Rome, Italy
| | - Carol D. Hamilton
- Family Health International 360, Research Triangle Park, North Carolina, United States of America
- Duke University Medical Center, Durham, North Carolina, United States of America
| | - Richard Adegbola
- MRC Laboratories, Fajara, The Gambia (West Africa)
- Bill & Melinda Gates Foundation, Seattle, Washington, United States of America
| | - Lars Østergaard
- Department of Infectious Diseases, Aarhus University Hospital, Skejby, Denmark
| | - Scott M. Williams
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - William K. Scott
- Dr. John T. Macdonald Foundation Department of Human Genetics and Miami Institute of Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Giorgio Sirugo
- Centro di Genetica, Centro di Ricerca Scientifica, Ospedale San Pietro FBF, Rome, Italy
- MRC Laboratories, Fajara, The Gambia (West Africa)
| |
Collapse
|
49
|
Deletion of the APOBEC3B gene strongly impacts susceptibility to falciparum malaria. INFECTION GENETICS AND EVOLUTION 2012; 12:142-8. [DOI: 10.1016/j.meegid.2011.11.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 09/19/2011] [Accepted: 11/03/2011] [Indexed: 01/28/2023]
|
50
|
Single nucleotide polymorphisms in the Toll-like receptor 3 and CD44 genes are associated with persistence of vaccine-induced immunity to the serogroup C meningococcal conjugate vaccine. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 19:295-303. [PMID: 22205660 DOI: 10.1128/cvi.05379-11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The rate of decay of antibody concentration following serogroup C meningococcal (MenC) polysaccharide-protein conjugate vaccination varies between individuals. This depends partly on vaccination age but may be influenced by human genetics. We studied 721 single nucleotide polymorphisms (SNPs) across 131 candidate genes in a first cohort of 905 Caucasians (11 to 21 years old; mean time after vaccination, 4.9 years) and 30 SNPs across 17 genes in a replication study using 155 children, aged 6 to 12 years (mean time after vaccination, 6.7 years), and 196 infants (1 year old; mean time after vaccination, 8 months). Individuals were classified as responders or nonresponders for total MenC IgG concentration and MenC serum bactericidal antibody (SBA) measurements. Associated genes were examined further for quantitative outcome measures. Fifty-nine SNPs in 37 genes were associated with IgG persistence (adjusted for age at measurement), and 56 SNPs in 36 genes were associated with SBA persistence (adjusted for age at measurement and vaccine used). Three SNPs each within the Toll-like receptor 3 (TLR3) (rs3775291, rs3775292, and rs5743312) and CD44 (rs11033013, rs353644, and rs996076) genes were associated with IgG (adjusted for age at measurement) or SBA (adjusted for age at measurement and vaccine used) persistence in the initial genetic study (P, 0.02 to 0.04). Single SNPs within the TLR3 (rs7657186) (P = 0.004 [unadjusted]) and CD44 (rs12419062) (P = 0.01 [unadjusted]) genes were associated with IgG persistence in the replication study. These results suggest that genetic polymorphisms in the TLR3 and CD44 genes are associated with the persistence of the immune response to MenC vaccines 1 to 6 years after vaccination.
Collapse
|